edwardsiella ictaluri is capable of persisting in channel

155
Louisiana State University Louisiana State University LSU Digital Commons LSU Digital Commons LSU Doctoral Dissertations Graduate School 7-8-2020 Edwardsiella Ictaluri Is Capable of Persisting in Channel Catfish Edwardsiella Ictaluri Is Capable of Persisting in Channel Catfish by Evading Host T Cell and Cell Death Responses by Evading Host T Cell and Cell Death Responses Elizabeth Watts Griggs Louisiana State University and Agricultural and Mechanical College Follow this and additional works at: https://digitalcommons.lsu.edu/gradschool_dissertations Part of the Veterinary Pathology and Pathobiology Commons Recommended Citation Recommended Citation Griggs, Elizabeth Watts, "Edwardsiella Ictaluri Is Capable of Persisting in Channel Catfish by Evading Host T Cell and Cell Death Responses" (2020). LSU Doctoral Dissertations. 5333. https://digitalcommons.lsu.edu/gradschool_dissertations/5333 This Dissertation is brought to you for free and open access by the Graduate School at LSU Digital Commons. It has been accepted for inclusion in LSU Doctoral Dissertations by an authorized graduate school editor of LSU Digital Commons. For more information, please contact[email protected].

Upload: others

Post on 14-Jun-2022

7 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Edwardsiella Ictaluri Is Capable of Persisting in Channel

Louisiana State University Louisiana State University

LSU Digital Commons LSU Digital Commons

LSU Doctoral Dissertations Graduate School

7-8-2020

Edwardsiella Ictaluri Is Capable of Persisting in Channel Catfish Edwardsiella Ictaluri Is Capable of Persisting in Channel Catfish

by Evading Host T Cell and Cell Death Responses by Evading Host T Cell and Cell Death Responses

Elizabeth Watts Griggs Louisiana State University and Agricultural and Mechanical College

Follow this and additional works at: https://digitalcommons.lsu.edu/gradschool_dissertations

Part of the Veterinary Pathology and Pathobiology Commons

Recommended Citation Recommended Citation Griggs, Elizabeth Watts, "Edwardsiella Ictaluri Is Capable of Persisting in Channel Catfish by Evading Host T Cell and Cell Death Responses" (2020). LSU Doctoral Dissertations. 5333. https://digitalcommons.lsu.edu/gradschool_dissertations/5333

This Dissertation is brought to you for free and open access by the Graduate School at LSU Digital Commons. It has been accepted for inclusion in LSU Doctoral Dissertations by an authorized graduate school editor of LSU Digital Commons. For more information, please [email protected].

Page 2: Edwardsiella Ictaluri Is Capable of Persisting in Channel

EDWARDSIELLA ICTALURI IS CAPABLE OF PERSISTING IN

CHANNEL CATFISH BY EVADING HOST T CELL AND CELL

DEATH RESPONSES

A Dissertation

Submitted to the Graduate Faculty of the

Louisiana State University and

Agricultural and Mechanical College

in partial fulfillment of the

requirements for the degree of

Doctor of Philosophy

in

Department of Pathobiological Sciences

by

Elizabeth Griggs

B.S., Louisiana Tech University, 2011

M.S., Louisiana Tech University, 2013

August 2020

Page 3: Edwardsiella Ictaluri Is Capable of Persisting in Channel

ii

ACKNOWLEDGMENTS

During my journey through this program I have received assistance and support from a

number of persons; members of the faculty, other students both past and present, friends, and

members of my family; and I would like to pause to recognize them for their contributions of

help and guidance along the way.

I would like to acknowledge Dr. Ronald Thune for his guidance and support throughout

this entire process, and who was always there to mentor and advise me during times of difficulty

as well as the times when everything went smoothly.

I would like to acknowledge Dr. Rebecca Giorno-McConnell who lit the spark of

microbiology in me and encouraged me to pursue it as a career; and who became my advisor for

my master’s program. Had I never encountered her, I would never have pursued this degree.

I’d also like to thank current and past members of the Thune lab, Doctors Christine

Darnall, Chanida Fongsaran, Lydia Dubytska, and Carolyn Chang, as well as other members of

the Department of Pathobiological Sciences, in particular Thaya, Marilyn, Trina and Jennifer.

I also seriously appreciate the care and patience of my parents and extended family who

have been constant sources of comfort throughout my life, and through this process.

I would be remiss if I did not also acknowledge my God and Creator, who has always

been with me and has led me to where I am today, and who allows my path to unfold according

to his will.

Page 4: Edwardsiella Ictaluri Is Capable of Persisting in Channel

iii

TABLE OF CONTENTS

ACKNOWLEDGMENTS .............................................................................................................. II

LIST OF TABLES .......................................................................................................................... V

LIST OF FIGURES ...................................................................................................................... VI

ABSTRACT ................................................................................................................................ VIII

CHAPTER 1. INTRODUCTION AND LITERATURE REVIEW ................................................1 Catfish Aquaculture ......................................................................................................................1 Edwardsiella Taxonomy ..............................................................................................................2 Edwardsiella ictaluri: Mechanisms of Pathogenesis ...................................................................9

Edwardsiella ictaluri Secretion Systems ...................................................................................12 Type III secretion systems and Edwardsiella ictaluri ................................................................13

Objectives and Hypotheses ........................................................................................................18 Literature Cited ..........................................................................................................................19

CHAPTER 2. ESEK HOST INTERACTION AND PROTECTIVE ABILITY AGAINST

SUBSEQUENT WILD-TYPE EXPOSURE .................................................................................28 Introduction ................................................................................................................................28

Materials and Methods ...............................................................................................................31 Results ........................................................................................................................................37

Discussion ..................................................................................................................................45 Literature Cited ..........................................................................................................................47

CHAPTER 3. NAÏVE HEAD KIDNEY DERIVED MACROPHAGES FAVOR M1

POLARIZATION IN RESPONSE TO EDWARDSIELLA ICTALURI INFECTION ...................51 Introduction ................................................................................................................................51

Methods ......................................................................................................................................53 Results ........................................................................................................................................55

Discussion ..................................................................................................................................60 Literature Cited ..........................................................................................................................73

CHAPTER 4. EDWARDSIALLA ICTALURI EVADES T CELL RESPONSES IN THE

CHANNEL CATFISH ...................................................................................................................78 Introduction ................................................................................................................................78 Methods ......................................................................................................................................80

Results ........................................................................................................................................85

Discussion ..................................................................................................................................91 Literature Cited ..........................................................................................................................98

CHAPTER 5. GENERAL CONCLUSIONS ...............................................................................105 Literature Cited ........................................................................................................................110

APPENDIX I: ABBREVIATIONS COMMONLY USED IN THIS DISSERTATION ............112

Page 5: Edwardsiella Ictaluri Is Capable of Persisting in Channel

iv

APPENDIX II: CHAPTER 1 SUPPLEMENTAL MATERIAL .................................................114

APPENDIX II: CHAPTER 4 SUPPLEMENTAL MATERIAL .................................................120

APPENDIX III: FAILED AND ONGOING EXPERIMENTS...................................................123

LIST OF REFERENCES .............................................................................................................126

VITA ............................................................................................................................................146

Page 6: Edwardsiella Ictaluri Is Capable of Persisting in Channel

v

LIST OF TABLES

Table 1.1 Biochemical testing results of Edwardsiella species. .....................................................5

Table 2.1: Strains and plasmids utilized in the scope of this research project. ..............................32

Table 2.2: Primer sets. ...................................................................................................................33

Table 2.3: Safety of ΔeseK vaccinated fish....................................................................................42

Table 3.1: Top 50 reads via GSA...................................................................................................57

Table 3.2 Top upregulated pathways Predicted by IPA showing the top predicted genes per

pathway. .........................................................................................................................................61

Table 3.3 Top Downregulated Pathways predicted by IPA showing the top predicted genes per

pathway. .........................................................................................................................................67

Page 7: Edwardsiella Ictaluri Is Capable of Persisting in Channel

vi

LIST OF FIGURES

Figure 1.1: T3SS of E. ictaluri Modified from Pallen et al ...........................................................14

Figure 2.1: Phyre 2 Model of EseK ...............................................................................................30

Figure 2.2: EseK::3XFLAG Confirmation ....................................................................................38

Figure 2.3: SYPRO Ruby Red Stain of Co-IP ...............................................................................39

Figure 2.4: Positional hits from mass spectrometry tryptic digest of proteins excised from bands

after Co-IP ......................................................................................................................................40

Figure 2.5: Preliminary Vaccination and Challenge ......................................................................41

Figure 2.6: Early Exposure to WT .................................................................................................44

Figure 2.7: Late WT challenge. .....................................................................................................45

Figure 3.1: Top 40 pathways according to overall -log(p-value) (y- axis), differentially regulated

in WT compared to uninfected HKDMs ........................................................................................59

Figure 3.2: Combined NF-κB virus and P13K/AKT signaling pathway .......................................66

Figure 3.3: Death Receptor signaling pathway ..............................................................................69

Figure 4.1: Representation of gating strategy with Day 0 samples ...............................................84

Figure 4.2: CD74 and MIF expression in HK per treatment graphed by the log2 transformation of

normalized RNA counts per day ....................................................................................................85

Figure 4.3: Expression of T cell Markers in HK per treatment graphed by the log2 transformation

of normalized RNA counts per day ...............................................................................................87

Figure 4.4: Expression of other cytokine indicators in HK per treatment graphed by the log2

transformation of normalized RNA counts per day .......................................................................88

Figure 4.5: CD74 and MIF expression in PBL per treatment graphed by the log2 transformation

of normalized RNA counts per day.. .............................................................................................89

Figure 4.6: Expression of T cell markers in PBL per treatment graphed by the log2

transformation of normalized RNA counts per day .......................................................................90

Figure 4.7: Expression of other cytokine indicators in PBL per treatment graphed by the log2

transformation of normalized RNA counts per day .......................................................................92

Figure 4.8: Percentage of CD3e+ lymphocytes grouped by treatment with SEM.........................93

Page 8: Edwardsiella Ictaluri Is Capable of Persisting in Channel

vii

Figure 5.1 Macrophage and T cell activation ..............................................................................109

Page 9: Edwardsiella Ictaluri Is Capable of Persisting in Channel

viii

ABSTRACT

Edwardsiella ictaluri is a gram-negative bacterium of the family Enterobacteriaceae that

infects and causes enteric septicemia (ESC) of channel catfish (Ictalurus punctatus), a fatal

disease costing the catfish industry millions of dollars in losses each year. Edwardsiella. ictaluri

is capable of replicating in catfish head-kidney-derived-macrophages (HKDM), and like many

other gram-negative bacteria, E. ictaluri encodes a Type III Secretion System (T3SS) that is

required for virulence and intracellular replication. In the case of E. ictaluri, the T3SS

translocates effectors from the Edwardsiella containing vacuole (ECV) through the bacterial cell

wall and the vacuolar membrane directly to the host cytoplasm. Of the nine known effectors, this

work analyzed one effector, EseK to determine the potential for cell-mediated immune (CMI)

responses in naïve fish and the fate of non-activated head-kidney-derived macrophages

(HKDMs) in response to infection. We determined that the binding partner of EseK is CD74, and

an EseK knockout strain provides catfish fingerlings with protection against subsequent WT

exposure. RNA sequencing using infected and uninfected HKDMs uncovered strong indications

for the M1 phenotype in response to infection. This data also indicates that E. ictaluri evades

typical programed-cell death measures, and suppresses the CD40 pathway, which is critical for T

cell dependent activation. Finally, in vivo immune responses detected several regulation

differences, notably downregulation of CD40L in infected catfish, further implicating the loss of

this pathway due to infection. Within the scope of this work, we did not discover any specific

differences in immune regulation that could be attributed to EseK, but this research supports the

theory that E. ictaluri modifies the macrophage environment in order to persist and replicate in

the host, and E. ictaluri evades CMI responses further aiding its survival in the host.

Page 10: Edwardsiella Ictaluri Is Capable of Persisting in Channel

1

CHAPTER 1. INTRODUCTION AND LITERATURE REVIEW

Catfish Aquaculture

Worldwide demand for sustainable seafood products is increasing, and to meet that

demand, reports indicate that global aquaculture production will need to increase by 36.7% by

2030, and 11% in the United States (1). In 2017, the value of United States aquaculture

production was approximately $1.2 billion USD, and the leading contributor to aquaculture

production is the channel catfish, Ictalurus punctatus (2). Catfish production typically accounts

for 1/3 to almost 1/2 of the total United States aquaculture production, but is currently on the

decline (2). For example, in 2008, catfish aquaculture was worth $390 million USD, 39.6% of

the total U.S. aquaculture production, but in 2017 catfish production was valued at

approximately $355 million USD, accounting for 29.2% of the total United States aquaculture

(2). The focus of this dissertation is on one culprit behind this decline, the gram-negative

bacterial pathogen Edwardsiella ictaluri. But before we focus on E. ictaluri, an understanding of

basic catfish farming practices is necessary.

Catfish farming began in the United States in the 1950s, and rapidly expanded in the

1960s, as the ability to properly rear and care for catfish was developed (3). Catfish farms can be

found across the U.S. but are most predominant in the southeastern states of Louisiana,

Arkansas, Alabama, and Mississippi (4). Catfish farms typically use channel catfish because they

provide one of the best dress out and feed conversion ratios , but also use other species such as

blue catfish Ictalurus furcatus (3). Farmers typically move fish three times during the catfish

rearing process. First, the farmers move eggs from spawning cans in the broodfish ponds to the

hatchery, then about 7-10 days post-hatch, move fry to fingerling ponds, and finally, move

Page 11: Edwardsiella Ictaluri Is Capable of Persisting in Channel

2

fingerlings to grow-out ponds where they will remain 18 months to 2 years until the fish reach

approximately 1.5 pounds each (3).

The catfish industry faces many challenges to production, including competition with

imported catfish species, such as basa (Pangasius bocourti) and swai/tra (Pangasius

hypopthalmus), and other imported fish species, increases in feed prices, and diseases, including

columnaris, channel catfish virus (CCV), and enteric septicemia of catfish (ESC) (4, 5). The

latter of these ESC, caused by the bacterium Edwardsiella ictaluri, is the focus of this work.

Several Edwardsiella species can infect channel catfish, but E. ictaluri, is still the most

prevalent (6). Enteric septicemia of catfish was first described from diseased catfish reared in

ponds in 1979 and is a continuous source of loss to catfish production (5, 7, 8). As of 2010,

catfish operations reported ESC outbreaks on 19.3% of fry/fingerling operations, 40.4% of

fingerling operations, and 36.6% of operations producing food size fish (5). It is important to

note that factors affecting these statistics include: the use of more resistant hybrid fish,

underreporting/not diagnosing infected fish, taking infected fish off feed, and in some rare cases,

on about 3% of reporting operations, vaccination (5). Given this information and the need to

sustain or increase catfish production in the coming years, it would be wise to increase our

understanding of ESC, a disease that still wreaks havoc in the industry. A better understanding

of E. ictaluri should lead to improved prevention and control methods to help this valuable

industry.

Edwardsiella Taxonomy

Edwardsiella tarda, first identified in 1965, was the first species of the genus

Edwardsiella described and initially was the lone species in the genus (9). As early as 1959,

several groups studying unknown cultures known as ‘bacterium 1483-59’ found that these

Page 12: Edwardsiella Ictaluri Is Capable of Persisting in Channel

3

cultures did not match anything known in the literature (9). Through diagnostics, researchers

determined that this organism belonged to the family Enterobacteriaceae, but was different from

the previously classified genus and species of this group because the IMViC, a four-part test

specific for identifying organisms of the coliform group, and additional tests showed 20 points of

difference from Escherichia coli, including production of hydrogen sulfide. This data supported

the description of a new genus, Edwardsiella, and the first species, Edwardsiella tarda (9).

Phenotypic traits of E. tarda can be found in Table 1.1, but based on the original analysis, major

traits of E. tarda are that it is gram-negative, rod-shaped, β-hemolytic, ferments glucose, and

reduces nitrate to nitrite (9).

Through the early 2000’s E. tarda was isolated from fresh and brackish water

environments, as well as from healthy and diseased animals in and connected to these

environments. Affected animals include fish, reptiles, and humans, and was described as the

cause of emphysematous putrefactive disease (EPDC) in catfish (10), red disease in eels (11),

and gastroenteritis in humans (12-14). However, as of 2009, EPDC, now often called

Edwardsiellosis (15), has been described in a variety of fish, including the Japanese flounder, red

sea bream, Japanese eel, ayu, and Nile tilapia (cited by)(16).

For a little over a decade, E. tarda was the only species in the genus until the description

of Edwardsiella hoshinae and Edwardsiella ictaluri in 1980 and 1981, respectively (17, 18).

Because it was able to produce acid from mannitol, sucrose, trehalose, and salicin, and was able

to utilize malonate (Table 1.1) E. hoshinae was determined to be a different species from E.

tarda (the typical strain) (17). Birds, reptiles (17), and in some cases, humans (13) can carry E.

hoshinae. Unfortunately, there are only a handful of E. hoshinae papers in the literature because

it is not associated with major pathogenesis in any of the described hosts.

Page 13: Edwardsiella Ictaluri Is Capable of Persisting in Channel

4

Researchers, in a 1981 paper, identified a new species of Edwardsiella, E. ictaluri that infects

channel catfish (Ictalurus punctatus), although evidence suggests its presence as early as 1969

in the southeastern United States (7, 18). The defining differences of E. ictaluri from E. tarda

are: it is α-hemolytic, does not grow at higher temperatures of 30 and 40C, is less resistant to

NaCl, is indole negative, and cannot produce H2S on TSI slants (Table 1.1) (18). Since the

discovery of E. ictaluri, the list of susceptible fish expanded to include tilapia, zebrafish, barbs,

green knife fish and other catfish species; but channel catfish remains the most relevant

susceptible species to E. ictaluri infection. E. ictaluri does not infect non-fish species (7, 19).

The geographical range of E. ictaluri includes the United States (7), Vietnam (20), Thailand (21),

Indonesia (22), Turkey (23), and Japan (16, 24) (tilapia isolates of Soto, Western hemisphere

Caribbean?). An in-depth review of this pathogen will follow this section, but briefly, E. ictaluri

invades the host via several potential entry points: orally and through the gills, nares, and skin

abrasions (7). The transmission is primarily fecal/oral and after ingestion it subsequently

breaches the epithelial barrier in the intestine, where it gains entry to the bloodstream and

disseminates throughout the rest of the body (25-27).

For over 30 years, the Edwardsiella genus only contained these three species, although

tests suggest considerable variability in E. tarda strains (28, 29). The evolution of procedures

used to characterize Edwardsiella species and strains and the naming systems commonly used in

the literature to discuss E. tarda has led to the current nomenclature. Previous methods

commonly used, such as SNPs in the gyrB gene, a combination of differences in antibiotic

susceptibility, generally consistent differences in the two plasmids carried by E. ictaluri, and the

presence or absence of particular genes, such as an absence of esrC in most zebrafish isolates,

and absence of eseI, escD, and virD4 from tilapia and zebrafish allowed for better

Page 14: Edwardsiella Ictaluri Is Capable of Persisting in Channel

5

Table 1.1 Biochemical testing results of Edwardsiella species. Table modified primarily from

Ucko et al (44). Legend as retrieved from Ucko et al and Abayneh et al. : Fg, ferments glucose;

+d, delayed reaction; nd, not determined; [-], negative 75 to 89% of the time.

E. tarda

E.

hoshinaea

E.

ictaluri

E.

piscicida

E.

anguillarum

E.

piscicida-

like

Type Strain

Bacterium

1483-59 CIP 78.56

GA 77-

52 ET883 ET080813T

EA18101

1

Naming

Source

Ewing

1965

Grimont

1980

Hawke

1981

Abayneh

2013 Shao 2015

Ucko

2016

Traits

Gram stain - - - - - -

sMorphology Rod Rod Rod Rod Rod Rod

Red blood cell

hemolysis Beta nd Alpha Beta nd Gamma

Motility + nd

at 25°C + + + -

at 37°C + - + -

OF glucose Fg nd Fg Fg nd Fg

Growth at

20oC + nd + + nd +

25oC + nd + + nd +

30oC + nd - + + +

40oC + nd - + + (37-42oC) +

Growth in

NaCl

1% + nd + + + +

2% + nd - + + +

3% + nd - + + +

API20E

Arginine - - - - - -

Lysine + + + + + +

Ornithine + + + + + -

Citrate

(Simmons') - - - - + +

H2S on TSI + + - + + +

Urease - nd - - - -

Phenylalanine - nd - - nd nd

Indole + [-] - + + -

Gelatin - nd - - - -

Glucose + nd + + + +

Mannitol - + - - + +d

Inositol - nd - - - -

(table cont’d)

Page 15: Edwardsiella Ictaluri Is Capable of Persisting in Channel

6

Table 1.1 continued

E. tarda

E.

hoshinaea

E.

ictaluri

E.

piscicida

E.

anguillarum

E.

piscicida-

like

Sorbitol - - - - - -

Rhamnose - - - - - -

Melebiose - - - - - -

Arabinose - [-] - - + -

cytochrome

oxidase

(CyOx) - - - - - -

Metabolism

and others

Lactose - - - - - -

Sucrose - + - - - -

Maltose + nd + + + +d

Xylose - nd - - nd -

Trehalose - + - - - -

Cellobiose - nd - - nd -

Erythritol - nd - - nd nd

Dulcitol - nd - - nd -

Salicin - [+] - - nd -

Adonitol - nd - - nd -

Malonate - + - - nd -

Galactose + nd + + + +

Mannose + nd + + - +

Nitrate

reduction to

nitrite + nd + + nd +

o-

Nitrophenyl-

b-D -

galactosidase

(ONPG) - nd - - nd -

Methyl red

test + + + + + nd

Voges-

Proskauer

(VP) - nd - - + - a Abayneh 2013 was used in addition to Grimont 1980 to provide more biochemical testing

results for E. hoshinae.

Page 16: Edwardsiella Ictaluri Is Capable of Persisting in Channel

7

differentiation of E. ictaluri strains from different fish species (30). Unfortunately, many of these

tests are also limited for in depth categorization of Edwardsiella species and strains (30). For

instance, one group used multiplexed PCR to identify major fish pathogens including

Flavobacterium columnare, E. ictaluri, and Aeromonas hydrophila (31). Unfortunately, E.

ictaluri 16S rDNA, the target, evolved slowly (30), making it implausible to use to detect

divergence between the Edwardsiella species E. tarda and E. ictaluri (32).

Advances in diagnostics have led to a better differentiation of E. tarda. Some researchers,

have detailed the history and utilized various identification methods in the ongoing quest to

better characterize Edwardsiella strains, with particular attention given to E. tarda (32, 33). This

effort led to the use of an important method for differentiation, Multi-Locus Sequence Analysis

(MLSA) (32-34). Ultimately, using MLSA, targeting several genes including gyrB, mdh, adK,

and metG, led to the conclusion that many E. tarda isolates from fish were misclassified (32).

Three commonly used strategies exist for differentiating strains of E. tarda. First, at the

inception of the genus naming, serovar/serotyping to differentiate strains found 21 serotypes

based on a preliminary investigation of 37, strains including those from O and H antigen groups

(9). Also, using this system on 445 isolates from eels, water, and sediments, divided E. tarda

strains into four different serotypes, A, B, C, and D using LPS O-antigen (35). Another group

characterized 61 O groups and 45 H antigens, and suggested this strategy for international

application (36).

Another grouping system relied on phenotypic characteristics that led to the identification

of typical versus atypical E. tarda strains. Typical E. tarda strains are motile, usually come from

freshwater sources, and in experiments testing virulence against three species, yellowtail,

Japanese flounder, and red sea bream, is virulent in the first 2 species, but not red sea bream (37,

Page 17: Edwardsiella Ictaluri Is Capable of Persisting in Channel

8

38). Atypical E. tarda strains are nonmotile, usually come from marine isolates, and in a study

testing virulence against three species, yellowtail, Japanese flounder, and red sea bream, were

virulent in all three (37, 38). There are also some genetic differences between these two groups.

MLSA has revealed a genetic diversity among E. tarda isolates from different host and

geographical regions, and pointed out a split between E. tarda groups with the fish isolates

grouping more closely with E. ictaluri as a separate species leading to a suggestion that E. tarda

is actually comprised of multiple species (32). Further expansion on this theory using MLSA in

combination with repetitive element sequence-based PCR (rep-PCR) identified two distinct E.

tarda groups called DNA group I, and DNA group II (33). This grouping is supported by similar

findings from other investigators (28, 39-42). It was proposed that DNA group I evolved first,

and that DNA group II and E. ictaluri evolved later, and gave further support to a new species

previously hidden within the E. tarda species (33). Data presented by Griffin et al. also

suggested that the typical and atypical strains previously reported by Sakai et al. both fall into

DNA group II , which is more closely related to E. ictaluri (16, 33). This ultimately led to the

rapid identification of two new Edwardsiella species, E. piscicida, type strain ET883, and E.

anguillarum, type strain ET080813T (32, 33).

Phenotypically E. piscicida is highly similar to E. tarda (Table 1.1). Researchers

reclassified E. tarda strains using species-specific gyrB primer sets for E. piscicida, leading to

the observation that E. piscicida has increased prevalence as a catfish pathogen compared to E.

tarda (6).

Additionally, recently identified E. anguillarum, (identified from eel isolates) is distinctly

different from E. tarda and E. piscicida (43). Biochemically, E. anguillarum differentiates from

E. tarda and other Edwardsiella species because it is citrate and arabinose positive (Table 1.1).

Page 18: Edwardsiella Ictaluri Is Capable of Persisting in Channel

9

E. anguillarum also has 3 Type VI Secretion Systems (T6SS) and 2 Type III Secretion Systems

(T3SS); with the T6SS1 and T3SS2 highly conserved among E. anguillarum, piscicida, and

ictaluri. The T6SS2, T6SS3 and T3SS1 however, are unique to E. anguillarum (43). The T3SS1

is also similar to the Locus of Enterocyte Effacement (LEE) of E. coli, and T6SS1 and T6SS2

are similar to Pantoea ananatis and may be involved in lateral gene transfer (43).

There is also one additional, yet to be named species that is E. piscicida-like, type strain

EA181011 that is also highly similar to E. piscicida but is indole negative and gamma hemolytic

(44). With more of the narrative coming together, a recent study identified genes that can be

specific to differentiating between the five named strains, in particular, dnaJ, aiding the

reclassification of several E. tarda isolates as E. piscicida and E. anguillarum (45).

Edwardsiella ictaluri: Mechanisms of Pathogenesis

E. ictaluri is pathogenic to catfish and several other fish species. It typically presents as

an acute gastrointestinal enteritis which progresses into septicemia often associated with high

mortality. It can also appear, often in fingerling fish, as a chronic “hole-in-the-head” lesion

which is a result of meningoencephalitis (46-48). Catfish populations are at the greatest risk for

ESC in the spring and fall months when the water temperature is moderate and in the range of 22

to 28°C (49). This is the so called “ESC window”. This bacterium can survive several days in

water, and has been shown to be capable of surviving up to 95 days in sterile pond mud (48, 50).

Fish can be visually identified as infected with ESC by observing behavioral changes, including

swimming in circles or with the head up and tail down, or by not eating (7). Gross clinical signs

include: skin ulcerations, petechial hemorrhages in the skin, muscle, intestine, and fat tissues; a

swollen abdomen indicative of fluid accumulation in the body cavity; and/or mottled areas in the

liver indicative of necrotic foci (7).

Page 19: Edwardsiella Ictaluri Is Capable of Persisting in Channel

10

As stated previously, the general route of entry of E. ictaluri is oral, but may be also via

the gills, and nares (7). Once E. ictaluri is ingested it migrates to the intestine and invades

through the epithelial lining and disseminates through the bloodstream (25, 26). Other

experiments have shown that water borne exposure can lead to invasion of the olfactory sac

followed by damage to the epithelium and inflammation of the olfactory sacs, and ultimately

entry to the brain from the olfactory bulb and dissemination resulting in meningoencephalitis

(25, 47, 48, 51). Experiments have suggested that uptake and dissemination can occur quickly.

One experiment infecting fish by gastric intubation was able to detect E. ictaluri in kidney

samples 15 minutes post infection and in liver samples 30 minutes post infection (52). This

group also showed that after infection, E. ictaluri can be detected along the intestinal brush

border at 30 minutes post infection, with phagocytic cells present near the basement membrane

(53). Twenty-four hours after fish are exposed to E. ictaluri, infiltration of mononuclear cells can

be observed in the intestine (47, 51). Other tissues affected by E. ictaluri include the skin,

muscle, spleen, intestine, and brain (54). Fish that survive an outbreak have the potential to carry

E. ictaluri up to 200 days (7).

E. ictaluri is internalizes in several cells, including the channel catfish ovary cell line

(CCO), neutrophils, and macrophages (55, 56). Head kidney derived macrophages (HKDM) are

used in many experimental assays to obtain a better understanding of pathogenesis in this cell

type. E. ictaluri can infiltrate, survive, and replicate in catfish HKDM (57). While Booth et al.

speculated that E. ictaluri likely has some ligands or receptors involved in uptake into

macrophages, they showed that opsonization of live E. ictaluri with normal catfish serum greatly

increases E. ictaluri’s uptake into catfish macrophages (57). Additionally, they found that E.

ictaluri replicates in spacious vacuoles within the macrophage similar to S. typhimurium in

Page 20: Edwardsiella Ictaluri Is Capable of Persisting in Channel

11

Salmonella containing vacuoles SCV’s, leading to the naming of Edwardsiella containing

vacuoles (ECV) (57).

The ECV initially acidifies the vacuolar environment to a pH of 4, 30 to 210 minutes post

infection, a process that is dependent on the vacuolar H+ -ATPases of the HKDM (58). While E.

ictaluri survives well at low pH, it cannot replicate at pH lower than 6, but with the addition of

exogenous urea in the media, E. ictaluri increases the pH to a neutral condition favorable for

replication (59).

E. ictaluri is considered urease negative, biochemically, but it was found to have a

pathogenicity island containing nine urease associated genes, including an acid activated urease

enzyme similar to the Y. enterocolitica class of ureases (59). The typical function of a bacterial

urease is to hydrolyze urea to ammonia and carbamate. The carbamate then breaks down to

carbonic acid and more ammonia in solution, which ultimately gives rise to ammonium and an

increase in pH as the reaction comes to equilibrium (60). Macrophage-encoded arginase is the

source of urea, which modulates the supply of arginine and regulates iNOS and NO production

(61, 62). Arginase degradation of arginine in favor of production of urea may promote E.

ictaluri’s resistance to killing in HKDM by reducing substrates available for NO production, and

simultaneously provide urea for ammonia production by urease, which aids E. ictaluri’s ability

to modify pH regulation (59). This allows E. ictaluri to modulate the low pH environment in the

HKDM vacuoles and possibly the GI tract (63).

E. ictaluri has several other genes involved with virulence, first identified using Signature

Tagged Mutagenesis (63), but we will now focus on those involved with secretion systems.

Page 21: Edwardsiella Ictaluri Is Capable of Persisting in Channel

12

Edwardsiella ictaluri Secretion Systems

A recent paper, in addition to the NCBI database, shows that E. ictaluri has 5 known

types of secretion systems: Type I, Type II, Type III, Type IV and Type VI (T1SS, T2SS, T3SS,

T4SS and T6SS), all of which are typically involved in virulence or aiding the bacterium in some

manner (64, 65). In E. ictaluri, several papers describe the role of virulence in the T3SS (58, 63,

66), important to this work, and also the T6SS (66, 67).

Our focus will be on the T3SS, but first a brief discussion of the T6SS. The T6SS

transfers substrates in a contact dependent manner. T6SS’s consist of two main proteins,

hemolysin coregulated protein (Hcp) and valine-glycine repeat protein (VgrG) (68). Together

they form part or all of the T6SS apparatus, and they resemble bacteriophage tail and spike

proteins (68). The Hcp hexamer is about 4 nm (internal diameter) and can accommodate a

globular protein about 50 kDa, but Vgr is too narrow to allow it to pass so, it is proposed that

VgrG breaks off after target cell puncture (68). T6SS’s are known to target other bacteria and/or

eukaryotic cells, and some are involved in functions such as conjugation, gene regulation, and

cellular adhesion (68). T6SS’s may not always be relevant to pathogenicity because the majority

of bacteria that have T6SS are not pathogens and are found in bacteria inhabiting marine

environments, soil, and are often symbionts or commensals (68).

Pathogenic implications for the T6SS in Edwardsiella spp., exist (69). A study disrupting

T6SS effector evpP in E. tarda showed an attenuated effect in blue gourami, and additional

evidence showed that the T6SS is repressed by Fur, a protein that modulates gene expression in

bacteria (68). The T6SS of E. ictaluri was identified as homologous to the one found in E. tarda

and is necessary for virulence. Additionally, another study suggested that the T6SS is in a single

Page 22: Edwardsiella Ictaluri Is Capable of Persisting in Channel

13

transcript and is regulated by EsrC, a T3SS protein (66). Although the T6SS is involved in

virulence and is the subject of much current research, our work focuses on the T3SS.

Type III secretion systems and Edwardsiella ictaluri

T3SS’s are found in a wide array of gram-negative bacteria, including Yersinia,

Salmonella, Escherichia, Pseudomonas, and Edwardsiella, and act to target specific functions in

the host cell that allow persistence and replication of the bacterium within the cell (70). T3SS’s

are comprised of a group of proteins that come together to form a needle like apparatus known as

the injectosome, which is used to inject effector proteins into the host cell either from outside of

the cell or from a vacuole in the cell (Figure 1.1) (71). Early research on T3SS’s in Salmonella

showed that it has two types, SPI-1, which is used for bacterial uptake into the cell and invasion

in the gut, and SPI-2, which is required for intracellular replication (51). Researchers were able

to show that some of the effectors were specific to the SPI-2 T3SS, and concluded that SPI-2 is

required for proliferation in macrophages in vivo, but could also have other functions (72).

Salmonella has a variety of effectors; some have a translocation sequence in their amino termini,

some contain leucine-rich repeats (LRR), and others are translocon proteins involved in the

assembly of the needle-like apparatus of the T3SS (73-75). E. tarda also codes a T3SS similar to

the SPI-2 T3SS found in Salmonella, and was found to have proteins homologous to

Salmonella’s apparatus forming proteins, chaperone proteins, and effector proteins (76, 77).

E. ictaluri encodes a T3SS structurally homologous to SPI-2 and E. tarda T3SS’s, where

E. ictaluri and Salmonella proteins share 42% to 77% identity at the protein level and genes are

arranged in similar operons (63, 66). The E. ictaluri genome encodes T3SS effector proteins both

in and outside of the T3SS pathogenicity island, and like the Salmonella SPI-2 T3SS, transports

Page 23: Edwardsiella Ictaluri Is Capable of Persisting in Channel

14

effectors from the ECV to the host cytoplasm (66). As is true for Salmonella, the E. ictaluri

T3SS is necessary for replication and virulence, but regardless of similarity to other systems,

Figure 1.1: T3SS of E. ictaluri Modified from Pallen et al. (71). T3SS’s are comprised of a

group of proteins that come together to form a needle-like apparatus, given the ‘Esa’ designation,

known as the injectosome, which spans the inner and outer membrane of the bacterial cell wall

as well as forming a pore through the host vacuole membrane. The injectosome is used to inject

effector proteins, with the ‘Ese’ designation, from a vacuole in the cell. Some of these effector

proteins are shuttled to the injectosome by chaperone proteins with ‘Esc’ designations, while

others may contain translocation sequences.

Page 24: Edwardsiella Ictaluri Is Capable of Persisting in Channel

15

effectors can carry out an array of different functions that do not necessarily translate between

species, so determining the function of E. ictaluri’s secretion system and it’s effectors is

important (63).

A study determined that the E. ictaluri T3SS was necessary for virulence in the host

because three genes, EsaU, and the two plasmids encoding effectors of the T3SS, pEI1 and pEI2,

were all attenuated when mutated (63). Baumgartner et al. found that the drop in pH in the

vacuole, driven by ATPase response to infection by the macrophage, triggers transcription of the

T3SS and activates the urease enzyme, which then increase the pH, ultimately allowing E.

ictaluri replication (58). The E. ictaluri T3SS is regulated by a two-component system EsrA, a

sensor kinase, and EsrB. This response regulator is homologous to two proteins responsible for

T3SS regulation in E. tarda and Salmonella (66). In low pH, EsrB is phosphorylated and

upregulates expression of the translocon genes and the transcriptional regulatory gene, esrC (66).

EsrC is then involved in regulating the T3SS, ultimately leading to the expression of the

remaining apparatus genes and the formation of the injectosome complex (66).

E. ictaluri T3SS effectors are also similar to Salmonella, where some are shuttled to the

injectosome by chaperone proteins, others contain translocation sequences, and some contain

LRR sequences (66, 70, 78, 79). Work to characterize the function of these effectors is ongoing

and will be of benefit for downstream applications to protect fish from ESC. A mutation in esaU,

which is part of the T3SS apparatus, prevents effector translocation and is completely attenuated

in vivo but does not protect fish after subsequent exposure to wild-type (WT) (63). This would

suggest that, in addition to being critical for virulence, one, if not several, of these effectors plays

an important role in stimulating a protective immune response in the catfish.

Page 25: Edwardsiella Ictaluri Is Capable of Persisting in Channel

16

Currently, E. ictaluri is known to have nine T3SS effector proteins (Table 1.2) (63, 80).

Two effectors, EseH and EseI, were found in Open-reading frames on the plasmids, pEI1 and

pEI2, respectively (63). EseH was found to be a LRR, and EseI was determined to be highly

homologous to Shigella protein OspB (63). The other 7 are found in the chromosome (80).

Table 1.2: T3SS effectors of E. ictaluri. Modified from Dubytska et al. (80).

Effector GenBank accession

no.

Size (aa) Known or Putative

activity/description (reference)

EseG ABC60070 301 Vacuolar localization (63)

EseI AGF34188 151 Shigella OspB family of T3SS

effectors (81)

EseN ACR69124 214 Major Vault Protein C;

ERK1/2C (82)

EseO ACR69900 667 Shigella enterotoxin 2 and

ankyrin repeat protein

EseH AAF85955 619 aLRR, 5 repeats, E3 ubiquitin

ligase (81)

EseJ

ACR69526

599 LRR, 12 repeats, E3 ubiquitin

ligase

EseK Genomeb 930 LRR, 18 repeats, E3 ubiquitin

ligase; interacts with CD74

EseL

ACR68523 705 LRR, 6 repeats, E3 ubiquitin

ligase

EseM ACR68525 793 LRR, 11 repeats, E3 ubiquitin

ligase aLRR, leucine-rich repeat protein. bEseK was identified during E. ictaluri genome sequencing but was not found in the final

genome assembly. Analysis of genomic DNA using specific PCR primers confirmed its

presence in the genome.

Four of the effectors EseG, EseI, EseN, and EseO are unlike the other five, in that they

are not LRR’s. Currently EseN is the only effector with a described function. Recent work found

that EseN interacts with Major Vault Protein (MVP) as well as ERK1/2 (82). EseG, EseI, and

EseO currently have unknown functions, but we can speculate what their roles are based on their

appearance and known role in other species. EseG shares a high degree of homology with its

namesake in E. tarda, which functions to destabilize microtubules (83). EseI shares similarity

Page 26: Edwardsiella Ictaluri Is Capable of Persisting in Channel

17

with Shigella flexneri OspB, which reduces inflammatory cytokine production in host cells (51),

and EseO is an ankyrin repeat protein that shares homology with ShET2 in Shigella and E. coli,

which is responsible for upregulating IL-8 in epithelial cells in Shigella infections (80).

BLAST analysis of the LRR effectors showed that all five have a translocation domain

in the amino terminus, an E3 ubiquitin ligase domain in their carboxy terminus, and 5 to 18

leucine-rich repeats in the core of the protein (80). Each of these effectors were shown to share

homology with similar proteins in other species. For example, EseK shares 58% identity with

SspH2 from S. enterica, and 42% identity with SlrP of S. typhimurium. Leucine rich repeats

drive the overall size and shape of the solenoid structure of each effector and determine the

nature of the target protein for each effector. As a result, the effectors bind entirely different

proteins and carry out vastly different functions despite a high degree of amino acid homology

(84). Additionally the translocation domain was shown to be functional, meaning each LRR

effector can translocate out of the ECV without the need of a chaperone (80). The ubiquitin

ligase domain likely plays a role in determining the fate of the host proteins bound by the

effector (80).

Additionally, research showed that all effector deletions tested had an attenuated effect

on replication in CCO’s, and three effectors, EseJ, EseK, and EseN also had attenuated

replication, albeit not as severely, in HKDM compared to WT (80). EseK was secreted by the

T3SS and was translocated in HKDM, but not in channel catfish ovary cells (CCOs) (80). This

could suggest that EseK plays a specific role in antigen-presenting cells (APCs) or phagocytes.

Also, unpublished work from other members of the Thune lab using a yeast two-hybrid assay on

all effectors found that EseK likely interacts with CD74 of the host cell.

Page 27: Edwardsiella Ictaluri Is Capable of Persisting in Channel

18

Limited research on the LRR’s means their effects on pathogenesis are largely unknown.

Because of the potential ramification of EseK’s interaction with CD74, and the limited

information suggesting that EseK plays an important role in virulence of E. ictaluri, one focus of

this work is to determine the effect of EseK on cell mediated immunity.

Objectives and Hypotheses

The hypothesis driving this work is that E. ictaluri is capable of persisting in and

replicating in the channel catfish because it can evade cell-mediated immune (CMI) responses of

the host. Secondly, we hypothesis that EseK aids WT E. ictaluri in the evasion of CMI

responses. Therefore, the objective of this research is to better characterize an E. ictaluri

infection, focusing on the potential for cellular responses and, additionally, to characterize the

T3SS effector, EseK, relative to WT. In order to do this, work focused on uncovering more

information related to E. ictaluri’s role in cellular responses both in the HKDM and in vivo, and

to characterize the role of EseK in this response.

The second chapter of this dissertation characterizes EseK, both confirming its host

protein binding partner and the potential use of an EseK mutant to establish protection of catfish

against WT exposure. Previous work in the lab showed that EseK was translocated in a pH-

dependent manner (80) and suggested, using a yeast-2-hybrid assay, that the binding partner of

EseK is CD74 (unpublished). A co-immunoprecipitation assay was carried out to verify EseK

and CD74 interaction. Because CD74 is highly involved in adaptive immune responses, an eseK

mutant strain was compared to WT to determine relative virulence, and vaccination assays were

carried out with ΔeseK to determine if a lack of EseK could provide protection against WT

exposure, possibly indicating a role for this protein in reducing cell-mediated immunity (CMI).

Page 28: Edwardsiella Ictaluri Is Capable of Persisting in Channel

19

The third chapter of this dissertation aims to determine the modulation of HKDM in the

presence of WT, and ΔeseK strains with special attention given to pathways involved in

immunity and cell death. The fourth chapter of this dissertation aims to characterize the immune

modulation of fish infected with WT relative to a control, and to what end EseK may be involved

in that response. The experiments in this chapter focused primarily on genes related to T cells

and cellular responses in HK and peripheral blood leukocyte (PBL) samples and supporting data

of leukocyte cell dispersion was collected using flow cytometry.

Finally, the fifth chapter provides overall conclusions for this work, tying the previous

chapters together. Briefly, we found that E. ictaluri is downregulating the CD40-CD40L

pathway, ultimately allowing it to evade most T cell responses, and additionally, pushes HKDM

towards autophagy instead of apoptosis, allowing it to persist in the cell. We also found that

EseK interacts with CD74 in channel catfish HKDM, which is a highly pleiotropic protein

involved in many immune related responses. The ΔeseK strain does provide some protection

against WT exposure, giving credence to important functions for the EseK-CD74 interaction, but

under the tested parameters, EseK, was not implicated in evasive immune responses at the

transcriptional level.

Literature Cited

1. FAO. 2018. The State of World Fisheries and Aquaculture 2018: Meeting the Sustainable

Development Goals. Rome.

2. FAO. 2011-2012. Fisheries and aquaculture software. FishStatJ, Rome.

3. Tucker CC, Robinson EH. 1990. Channel catfish farming handbook. Springer Science &

Business Media.

4. Paul G. 2012. Aquaculture in the United States of America: present status and future

opportunities. Bull Fish Res Agen No 35:7-13.

Page 29: Edwardsiella Ictaluri Is Capable of Persisting in Channel

20

5. USDA. 2010. USDA Catfish 2010: Part III: Changes in Catfish Health and Production

Practices in the United States, 2002-2009.

6. Griffin MJ, Ware C, Quiniou SM, Steadman JM, Gaunt PS, Khoo LH, Soto E. 2014.

Edwardsiella piscicida identified in the Southeastern USA by gyrB sequence, species-

specific and repetitive sequence-mediated PCR. Dis Aquat Org 108:23-35.

7. J. P. Hawke RMD, R. L. Thune, A, C. Camus. 1998. ESC- Enteric Septecemia of Catfish.

8. Hawke JP. 1979. A Bacterium Associated with Disease of Pond Cultured Channel

Catfish, Ictalurus punctatus. Can J Fish Aquat Sci 36:1508-1512.

9. H. Ewing W, C. Mcwhorter A, R. Escobar M, H. Lubin A. 1965. Edwardsiella, a new

genus of Enterobacteriaceae based on a new species, E. tarda. Int J Syst Evol Microbiol

15.

10. Meyer FP, Bullock GL. 1973. Edwardsiella tarda, a new pathogen of channel catfish

(Ictalurus punctatus). Appl Microbiol 25:155-6.

11. Wakabayashi H. 1973. Edwardsiella tarda (Paracolobacterium anguillimortiferum)

associated with pond-cultured eel diseases. Bul Japan Soc Sci Fish 39:931-939.

12. Bockemuhl J, Pan-Urai R, Burkhardt F. 1971. Edwardsiella tarda associated with human

disease. Pathol Microbiol 37:393-401.

13. Janda JM, Abbott SL, Kroske-Bystrom S, Cheung WK, Powers C, Kokka RP, Tamura K.

1991. Pathogenic properties of Edwardsiella species. J Clin Microbiol 29:1997-2001.

14. Jordan GW, Hadley WK. 1969. Human infection with Edwardsiella tarda. Ann Intern

70:283-8.

15. Mohanty BR, Sahoo PK. 2007. Edwardsiellosis in fish: a brief review. Biosci 32:1331-

44.

16. Sakai T, Yuasa K, Sano M, Iida T. 2009. Identification of Edwardsiella ictaluri and E.

tarda by species-specific polymerase chain reaction targeted to the upstream region of the

fimbrial gene. J Aquat Anim Health 21:124-32.

Page 30: Edwardsiella Ictaluri Is Capable of Persisting in Channel

21

17. Grimont P, Grimont F, Richard C, Sakazaki R. 1980. Edwardsiella hoshinae, a new

species of Enterobacteriaceae, vol 4.

18. Hawke J, McWhorter A, Steigerwalt AG, Brenner D. 1981. Edwardsiella ictaluri sp.

nov., the causative agent of Enteric Septicemia of Catfish. Int J Syst Bacteriol 31:396-

400.

19. Kent M, Lyons J. 1982. Edwardsiella ictaluri in the green knifefish, Eigemannia

virescens. Fish Health News.

20. Ferguson HW, Turnbull JF, Shinn A, Thompson K, Dung TT, Crumlish M. 2001.

Bacillary necrosis in farmed Pangasius hypophthalmus (Sauvage) from the Mekong

Delta, Vietnam. J Fish Dis 24:509-513.

21. Kasornchandra J, Rogers WA, Plumb JA. 1987. Edwardsiella-Ictaluri from Walking

Catfish, Clarias-Batrachus L, in Thailand. J Fish Dis 10:137-138.

22. Yuasa K, Kholidin EB, Panigoro N, Hatai K. 2003. First isolation of Edwardsiella

ictaluri from cultured striped catfish Pangasius hypophthalmus in Indonesia. Fish Pathol

38:181-183.

23. Keskin O, Secer SU, Izgur M, Turkyilmaz S, Mkakosya RS. 2004. Edwardsiella ictaluri

infection in rainbow trout (Oncorhynchus mykiss). Turk J Vet Anim Sci 28:649-653.

24. Nagai T, Iwamoto E, Sakai T, Arima T, Tensha K, Iida Y, Iida T, Nakai T. 2008.

Characterization of Edwardsiella ictaluri Isolated from Wild Ayu Plecoglossus altivelis

in Japan. Fish Pathol 43:158-163.

25. Miyazaki T, Plumb JA. 1985. Histopathology of Edwardsiella-Ictaluri in channel catfish,

Ictalurus-Punctatus (Rafinesque). J Fish Dis 8:389-392.

26. Zhou T, Yuan Z, Tan S, Jin Y, Yang Y, Shi H, Wang W, Niu D, Gao L, Jiang W, Gao D,

Liu Z. 2018. A review of molecular responses of catfish to bacterial diseases and abiotic

stresses. Front Physiol 9:1113.

27. Ramona TS, Thomas JB. 2002. Edwardsiella ictaluri invasion of IEC-6, Henle 407,

fathead minnow and channel catfish enteric epithelial cells. Dis Aquat Org 51:161-167.

Page 31: Edwardsiella Ictaluri Is Capable of Persisting in Channel

22

28. Acharya M, Maiti NK, Mohanty S, Mishra P, Samanta M. 2007. Genotyping of

Edwardsiella tarda isolated from freshwater fish culture system. Comp Immunol

Microbiol Infect Dis 30:33-40.

29. Wei L, Musa N. 2008. Phenotyping, genotyping and whole cell protein profiling of

Edwardsiella tarda isolated from cultured and natural habitat freshwater fish. Am

Eurasian J Agric Environ Sci 3.

30. Griffin MJ, Reichley SR, Greenway TE, Quiniou SM, Ware C, Gao DX, Gaunt PS,

Yanong RP, Pouder DB, Hawke JP, Soto E. 2016. Comparison of Edwardsiella ictaluri

isolates from different hosts and geographic origins. J Fish Dis 39:947-69.

31. Victor SP, Craig AS, Vicky LvS, Kevin D, Phillip HK. 2007. Multiplex-PCR for

simultaneous detection of 3 bacterial fish pathogens, Flavobacterium columnare,

Edwardsiella ictaluri, and Aeromonas hydrophila. Dis Aquat Org 74:199-208.

32. Abayneh T, Colquhoun DJ, Sorum H. 2012. Multi-locus Sequence Analysis (MLSA) of

Edwardsiella tarda isolates from fish. Vet Microbiol 158:367-75.

33. Griffin MJ, Quiniou SM, Cody T, Tabuchi M, Ware C, Cipriano RC, Mauel MJ, Soto E.

2013. Comparative analysis of Edwardsiella isolates from fish in the eastern United

States identifies two distinct genetic taxa amongst organisms phenotypically classified as

E. tarda. Vet Microbiol 165:358-72.

34. Cooper JE, Feil EJ. 2004. Multilocus sequence typing-what is resolved? Trends

Microbiol 12:373-7.

35. Park S-I, Wakabayashi H, Watanabe Y. 1983. Serotype and virulence of Edwardsiella

tarda isolated from eel and their environment. Fish Pathol 18:85-89.

36. Tamura K, Sakazaki R, McWhorter AC, Kosako Y. 1988. Edwardsiella tarda serotyping

scheme for international use. J Clin Microbiol 26:2343-6.

37. Yasunaga N. 1982. Characteristics of the fish pathogen Edwardsiella isolated from

several species of cultured marine fish. Bull Nagasaki Prefect Inst Fish 8:67-73.

38. Matsuyama T, Kamaishi T, Ooseko N, Kurohara K, Iida T. 2005. Pathogenicity of motile

and non-motile Edwardsiella tarda to some marine fish. Fish Pathol 40:133-135.

Page 32: Edwardsiella Ictaluri Is Capable of Persisting in Channel

23

39. Yang M, Lv Y, Xiao J, Wu H, Zheng H, Liu Q, Zhang Y, Wang Q. 2012. Edwardsiella

comparative phylogenomics reveal the new intra/inter-species taxonomic relationships,

virulence evolution and niche adaptation mechanisms. PLoS One 7:e36987.

40. Maiti NK, Mandal A, Mohanty S, Mandal RN. 2009. Phenotypic and genetic

characterization of Edwardsiella tarda isolated from pond sediments. Comp Immunol

Microbiol Infect Dis 32:1-8.

41. Panangala VS, Shoemaker CA, McNulty ST, Arias CR, Klesius PH. 2006. Intra‐and

interspecific phenotypic characteristics of fish‐pathogenic Edwardsiella ictaluri and E.

tarda. Aquacult Res 37:49-60.

42. Castro N, Toranzo AE, Bastardo A, Barja JL, Magarinos B. 2011. Intraspecific genetic

variability of Edwardsiella tarda strains from cultured turbot. Dis Aquat Org 95:253-8.

43. Shao S, Lai Q, Liu Q, Wu H, Xiao J, Shao Z, Wang Q, Zhang Y. 2015. Phylogenomics

characterization of a highly virulent Edwardsiella strain ET080813(T) encoding two

distinct T3SS and three T6SS gene clusters: propose a novel species as Edwardsiella

anguillarum sp. nov. Syst Appl Microbiol 38:36-47.

44. Ucko M, Colorni A, Dubytska L, Thune RL. 2016. Edwardsiella piscicida-like pathogen

in cultured grouper. Dis Aquat Org 121:141-148.

45. Bujan N, Mohammed H, Balboa S, Romalde JL, Toranzo AE, Arias CR, Magarinos B.

2018. Genetic studies to re-affiliate Edwardsiella tarda fish isolates to Edwardsiella

piscicida and Edwardsiella anguillarum species. Syst Appl Microbiol 41:30-37.

46. Newton J, Wolfe L, Grizzle J, Plumb J. 1989. Pathology of experimental enteric

septicaemia in channel catfish, Ictalurus punctatus (rafinesque), following immersion‐

exposure to Edwardsiella ictaluri. J Fish Dis 12:335-347.

47. Shotts E, Blazer V, Waltman W. 1986. Pathogenesis of experimental Edwardsiella

ictaluri infections in channel catfish (Ictalurus punctatus). Can J Fish Aquat Sci 43:36-

42.

48. Thune R, A. Stanley L, Cooper R. 1993. Pathogenesis of Gram-negative bacterial

infection in warm water fish. Annu Rev Fish Dis 3:37-68.

Page 33: Edwardsiella Ictaluri Is Capable of Persisting in Channel

24

49. Francis-Floyd R, Beleau MH, Waterstrat PR, Bowser PR. 1987. Effect of water

temperature on the clinical outcome of infection with Edwardsiella ictaluri in channel

catfish. J Am Vet Med Assoc 191:1413-6.

50. Plumb JA, Quinlan EE. 1986. Survival of Edwardsiella ictaluri in pond water and bottom

mud. Prog Fish-Cult 48:212-214.

51. Rogge M. 2009. Expression of the Edwardsiella ictaluri type III secretion system and its

relationship to the native plasmids. Doctor of Philosophy. Louisiana State University and

Agricultural and Mechanical College

52. Baldwin TJ. 1992. Edwardsiella Ictaluri: pathogenic mechanisms and antigens

recognized by infected channel catfish. Doctor of Philosophy Louisiana State University

53. Baldwin TJ, Newton JC. 1993. Pathogenesis of Enteric Septicemia of Channel Catfish,

caused by Edwardsiella ictaluri: bacteriologic and light and electron microscopic

findings. J Aquat Anim Health 5:189-198.

54. Mqolomba TN, Plumb JA. 1992. Longevity of Edwardsiella-Ictaluri in the organs of

experimentally infected channel catfish, Ictalurus-Punctatus. Aquaculture 101:1-6.

55. Ainsworth AJ, Chen DX. 1990. Differences in the phagocytosis of four bacteria by

channel catfish neutrophils. Dev Comp Immunol 14:201-9.

56. Waterstrat PR, Ainsworth AJ, Capley G. 1991. In vitro responses of channel catfish,

Ictalurus punctatus, neutrophils to Edwardsiella ictaluri. Dev Comp Immunol 15:53-63.

57. Booth NJ, Elkamel A, Thune RL. 2006. Intracellular replication of Edwardsiella ictaluri

in channel catfish macrophages. J Aquat Anim Health 18:101-108.

58. Baumgartner WA, Dubytska L, Rogge ML, Mottram PJ, Thune RL. 2014. Modulation of

vacuolar pH is required for replication of Edwardsiella ictaluri in channel catfish

macrophages. Infect Immun 82:2329-36.

59. Booth NJ, Beekman JB, Thune RL. 2009. Edwardsiella ictaluri encodes an acid-

activated urease that is required for intracellular replication in channel catfish (Ictalurus

punctatus) macrophages. Appl Environ Microbiol 75:6712-20.

60. Burne RA, Chen Y-YM. 2000. Bacterial ureases in infectious diseases. Microb Infect

2:533-542.

Page 34: Edwardsiella Ictaluri Is Capable of Persisting in Channel

25

61. Chang C-I, Liao JC, Kuo L. 1998. Arginase modulates nitric oxide production in

activated macrophages. Am J Physiol Heart Circ Physiol 274:H342-H348.

62. Mori M. 2007. Regulation of nitric oxide synthesis and apoptosis by arginase and

arginine recycling. J Nutr 137:1616S-1620S.

63. Thune RL, Fernandez DH, Benoit JL, Kelly-Smith M, Rogge ML, Booth NJ, Landry CA,

Bologna RA. 2007. Signature-tagged mutagenesis of Edwardsiella ictaluri identifies

virulence-related genes, including a salmonella pathogenicity island 2 class of type III

secretion systems. Appl Environ Microbiol 73:7934-46.

64. National Library of Medicine (US) NCfBI. 1988. https://www.ncbi.nlm.nih.gov/.

Accessed

65. Tekedar HC, Blom J, Kalindamar S, Nho S, Karsi A, Lawrence ML. 2020. Comparative

genomics of the fish pathogens Edwardsiella ictaluri 93-146 and Edwardsiella piscicida

C07-087. Microb Genom 6.

66. Rogge ML, Thune RL. 2011. Regulation of the Edwardsiella ictaluri type III secretion

system by pH and phosphate concentration through EsrA, EsrB, and EsrC. Appl Environ

Microbiol 77:4293-302.

67. Kordon AO, Kalindamar S, Majors K, Abdelhamed H, Tan W, Karsi A, Pinchuk LM.

2019. Effects of live attenuated vaccine and wild type strains of Edwardsiella ictaluri on

phagocytosis, bacterial killing, and survival of catfish B cells. Front Immunol 10:2383.

68. Silverman JM, Brunet YR, Cascales E, Mougous JD. 2012. Structure and Regulation of

the Type VI Secretion System. Annual review of microbiology 66:453-472.

69. Zhang A, Chen D, Wei H, Du L, Zhao T, Wang X, Zhou H. 2012. Functional

characterization of TNF-α in grass carp head kidney leukocytes: induction and

involvement in the regulation of NF-κB signaling. Fish Shellfish Immunol 33:1123-1132.

70. Vishwakarma V, Pati NB, Chandel HS, Sahoo SS, Saha B, Suar M. 2012. Evaluation of

Salmonella enterica serovar Typhimurium TTSS-2 deficient fur mutant as safe live-

attenuated vaccine candidate for immunocompromised mice. PLoS One 7:e52043.

Page 35: Edwardsiella Ictaluri Is Capable of Persisting in Channel

26

71. Pallen MJ, Beatson SA, Bailey CM. 2005. Bioinformatics, genomics and evolution of

non-flagellar type-III secretion systems: a Darwinian perspective. FEMS Microbiol Rev

29:201-29.

72. Hensel M, Shea JE, Waterman SR, Mundy R, Nikolaus T, Banks G, Vazquez-Torres A,

Gleeson C, Fang FC, Holden DW. 1998. Genes encoding putative effector proteins of the

type III secretion system of Salmonella pathogenicity island 2 are required for bacterial

virulence and proliferation in macrophages. Mol Microbiol 30:163-74.

73. Brumell JH, Marcus SL, Finlay BB. 2000. N‐terminal conservation of putative type III

secreted effectors of Salmonella typhimurium. Mol Microbiol 36:773-774.

74. Kobe B, Kajava AV. 2001. The leucine-rich repeat as a protein recognition motif. Curr

Opin Struct Biol 11:725-732.

75. Miao EA, Miller SI. 2000. A conserved amino acid sequence directing intracellular type

III secretion by Salmonella typhimurium. Proceedings of the National Academy of

Sciences of the United States of America 97:7539-7544.

76. Srinivasa Rao P, Yamada Y, Tan YP, Leung KY. 2004. Use of proteomics to identify

novel virulence determinants that are required for Edwardsiella tarda pathogenesis. Mol

Microbiol 53:573-586.

77. Zheng J, Li N, Tan YP, Sivaraman J, Mok Y-K, Mo ZL, Leung KY. 2007. EscC is a

chaperone for the Edwardsiella tarda type III secretion system putative translocon

components EseB and EseD. Microbiology 153:1953-1962.

78. Bhavsar AP, Brown NF, Stoepel J, Wiermer M, Martin DD, Hsu KJ, Imami K, Ross CJ,

Hayden MR, Foster LJ, Li X, Hieter P, Finlay BB. 2013. The Salmonella type III effector

SspH2 specifically exploits the NLR co-chaperone activity of SGT1 to subvert immunity.

PLoS Path 9:e1003518.

79. McShan AC, De Guzman RN. 2015. The bacterial type III secretion system as a target for

developing new antibiotics. Chem Biol Drug Des 85:30-42.

80. Dubytska LP, Rogge ML, Thune RL. 2016. Identification and characterization of putative

translocated effector proteins of the Edwardsiella ictaluri type III secretion system.

mSphere 1:e00039-16.

Page 36: Edwardsiella Ictaluri Is Capable of Persisting in Channel

27

81. Fernandez DH, Pittman-Cooley L, Thune RL. 2001. Sequencing and analysis of the

Edwardsiella ictaluri plasmids. Plasmid 45:52-6.

82. Dubytska LP, Thune RL. 2018. Edwardsiella ictaluri type III secretion system (T3SS)

effector EseN is a phosphothreonine lyase that inactivates ERK1/2. Dis Aquat Org

130:117-129.

83. Xie HX, Yu HB, Zheng J, Nie P, Foster LJ, Mok Y-K, Finlay BB, Leung KY. 2010.

EseG, an effector of the type III secretion system of Edwardsiella tarda, triggers

microtubule destabilization. Infect Immun 78:5011-5021.

84. Dubytska LP, Rogge ML, Thune RL. 2016. Identification and Characterization of

Putative Translocated Effector Proteins of the Edwardsiella ictaluri Type III Secretion

System. mSphere 1.

Page 37: Edwardsiella Ictaluri Is Capable of Persisting in Channel

28

CHAPTER 2. ESEK HOST INTERACTION AND PROTECTIVE ABILITY

AGAINST SUBSEQUENT WILD-TYPE EXPOSURE

Introduction

Edwardsiella ictaluri is a gram-negative bacterium of the phylum proteobacteria and

family Enterobacteriaceae, which includes other pathogens such as Salmonella, Escherichia, and

Yersinia spp. The bacterium Edwardsiella ictaluri causes the disease Enteric Septicemia of

Catfish (ESC), which causes 20-30% loss to catfish production amounting to millions of dollars

in loss annually (1). This is pertinent because the United States catfish industry has consistently

accounted for one-third to almost one-half of U.S. generated aquaculture revenue annually (2).

Reports indicate that production of all U.S. aquaculture, including catfish production, will need

to increase in the coming years to meet market demand, and improving the efficiency of

production will be essential to this process (3, 4). One avenue that will aid in catfish production

will be the reduction of loss to disease. Several common strategies currently in use to reduce

disease losses are antibiotic treatment, food restriction, and hybrid catfish production (1, 5).

Another avenue that will aid the reduction in loss of catfish will be a better understanding of the

bacterium, specifically concerning modifications of the host function and physiology.

Knowledge of the effects of protein modifications by the bacterium on host metabolism will aid

in designing smarter vaccine candidates in the future, whether that be a single attenuated mutant

or coupled mutations. One strategy is using a live attenuated vaccine to promote natural

immunity to E. ictaluri. Aquavac-ESC, a previously developed live vaccine, showed limited

efficacy, but significant research is ongoing to develop an efficacious product (6-8).

One potential knock-out target for such a vaccine is the Type III Secretion System

(T3SS), which is required for virulence and intracellular replication (9). Gram-negative bacteria,

Page 38: Edwardsiella Ictaluri Is Capable of Persisting in Channel

29

including Salmonella, Escherichia, Yersinia, and Edwardsiella have T3SS’s that can target

specific functions in the host cell to allow persistence and replication of the bacterium within the

cell (9, 10). T3SS’s are composed of a group of proteins that come together to form a needle-like

apparatus known as the injectosome, which is used to inject effector proteins into the host cell

cytosol, either from outside of the cell or from a vacuole within the cell (11). The latter is the

case of E. ictaluri, which utilizes the T3SS to translocate effectors from the Edwardsiella-

containing vacuole (ECV) to the host cytoplasm (12).

Some of these effector proteins are shuttled to the injectosome by chaperone proteins,

which can be specific to particular effectors, while others may contain a translocation sequence

(10, 13-15). The T3SS of E. ictaluri has nine known effectors, five of which contain leucine-rich

repeats (LRR’s), causing them to share a high degree of homology both with each other and with

effectors of other species such as Salmonella (12). Although high degrees of homology exist

among effectors from different organisms, each effector is still unique to a particular pathogen

and they often carry out different functions in the host despite their homology (12).

For instance, the effector protein EseK was identified on the chromosome of E. ictaluri

and was determined to contain 18 leucine-rich repeats (LRR), a ubiquitin ligase domain in the

carboxyl terminus, and a functional translocation sequence at the amino terminus (12). Phyre 2

modeling showed 77% of the residues at greater than 90% confidence and indicated that EseK

shares a 58% identity with SspH2 from S. enterica, and a 42% identity to the S. typhimurium

effector SlrP (Figure 2.1) (16). While EseK shares homology with SspH2 and SlrP, the function

of EseK can differ from SspH2 and SlrP because both the bacterium as well as the number of

LRRs affects the target protein for EseK by changing the overall binding shape of the protein

Page 39: Edwardsiella Ictaluri Is Capable of Persisting in Channel

30

(12, 17). A translocation assay showed that EseK was translocated under neutral pH to the host

cell cytoplasm in HKDM (18).

Figure 2.1: Phyre 2 Model of EseK. Protein residues are colored in rainbow format with red on

the N terminus and blue on the C terminus Phyre 2 modeling was able to model 77% of the

Page 40: Edwardsiella Ictaluri Is Capable of Persisting in Channel

31

residues at greater than 90%confidence and suggests that EseK shares a 58% identity with SspH2

from S. enterica, and a 42% identity to the S. typhimurium effector SlrP.

Additionally, previous yeast two-hybrid experiments in the lab suggest a protein-protein

interaction with CD74, which could mean that EseK plays a specific role in antigen presentation

in dendritic cells and macrophages (data unpublished). The overarching hypothesis of this work

is that WT E. ictaluri can evade cell-mediated immune responses, and this preliminary evidence

led to the specific hypothesis that EseK could aid E. ictaluri in evasion of the host immune

response. In this chapter we began to address the potential for EseK to be involved in virulence

by confirming its host binding partner and potential protection of catfish against WT exposure.

Initially an EseK::3XFLAG fusion was constructed that could be specifically detected with

3XFLAG antibody in the presence of the four other LRR effectors, and used to confirm that

CD74 is the specific binding partner of EseK. Given the role of CD74 in host cell immunity, we

carried out several vaccination experiments, believing an eseK mutant strain (ΔeseK) could be

protective against ESC. We observed that ΔeseK has an attenuated effect on pathogenesis at

target doses while providing protection from subsequent exposure to WT.

Materials and Methods

Bacterial Growth. Strains and plasmids used in this work are listed in Table 2.1.

Edwardsiella ictaluri strains were grown overnight (O/N) at 28C with aeration from frozen

cultures stored at -80C in 5mL aliquots of BHI-porcine with 15% glycerol (Sigma-Aldrich, St.

Louis, MO). Escherichia coli strains were grown in Luria-Bertani (LB) broth at 37C with

aeration and antibiotics added where appropriate. Antibiotics used were 200 ug/mL Antibiotics

used were 200 µg/mL Ampicillin (Amp) and 20 µg/mL Colistin (Col). These starter cultures

were then used for downstream assays.

Page 41: Edwardsiella Ictaluri Is Capable of Persisting in Channel

32

Table 2.1: Strains and plasmids utilized in the scope of this research project.

Strain or

Plasmid

Description Source

Edwardsiella

ictaluri 93-146

Wild-type E. ictaluri isolated in 1993

from moribund channel catfish in a

natural outbreak of ESC on a

commercial farm

LSU aquatic animal diagnostic

laboratory

eseK::3XFLAG eseK::3XFLAG in ΔeseK E. ictaluri This study

ΔeseK E. ictaluri 93-146 Δ(eseK1-2202) This Study

65ST 93-146 esaU::Tn5Km2 (9)

pRE107 R6K ori mob sacB1; Apr; suicide vector

used for allelic exchange

(19)

CC118 λpir Δ (ara-leu) araD Δ lacX74 galE galK

phoA20 thi-1 rpsE rpoB argE(Am)

recA1 λpir lysogen

(20)

S17-1 λpir Tp Sm thi pro recA HsdR-HsdM+; RP4-

2-Tc::Mu::Km Tn7 λpir

(21)

pBBR1-MCS4 Broad host range expression vector; Apr (22)

Strain Generation. An eseK mutant strain (ΔeseK) was generated from wild-type E.

ictaluri 93-146 (WT) (Appendix 2). Overlap-deletion PCR using the primer sets P190, P191,

P192, and P197 allowed for the amplification of a 5’ portion of eseK and a 3’ portion of eseK

(Table 2.2 and Appendix 2, Supplemental Figures 2.1, 2.2,and 2.3). These two fragments

contained overlapping sequences of the other fragment as linkers that were mixed and amplified

using primers P190 and P192 to delete 2200 bp of eseK beginning at the ATG start site at the N-

terminus. This truncated eseK product was then ligated to pRE107, a suicide vector, and

transformed into E. coli CC118 λpir for sequence verification before transformation into E. coli

S17-1 λpir. E. coli S17-1 λpir containing the construct was then conjugated to WT on BA plates

O/N at 28C. Selection occurred by a double crossover generating the ΔeseK strain.

Page 42: Edwardsiella Ictaluri Is Capable of Persisting in Channel

33

Briefly, the plasmid and WT were selected on BHIColAmp at 28C. Colonies were then

patched to BHICol at 28C. Finally, patched colonies were struck onto BHIcolsucrose at 28C to

allow for selection of a second crossover of ΔeseK into the WT chromosome and deletion of the

pRE107 plasmid. Verification of the construct was done by PCR and DNA sequencing. The

eseK::3XFLAG fusion strain was generated using a forward primer upstream of the native

promoter (Table 2.2) and a downstream promoter, which was designed to include a 3X FLAG

sequence in frame with the eseK sequence and followed by a stop codon. This amplicon was

added to the pBBR1-MCS4 plasmid and inserted into the ΔeseK E. ictaluri strain to create a

complemented strain (Appendix 2, Supplemental Figure 2.4).

Table 2.2: Primer sets.

Primer name/

description

Sequence Source

P119. eseK

Forward SacI RE

gccgatgagctcGTGATCTACACAACGAATGCTATCGAGT Thune

Lab

OL1.

eseK::3XFLAG

Reverse XbaI RE

gaagattctagaCTActtgtcatcgtcatccttgtagtcgatgtcatgatctttataatca

ccgtcatggtctttgtagtcTACAGGGGACGCATCTAGCAA

This

Study

P190. muteseK

Forward 5’ SacI RE

GGACTATCTGAGCTCTCTGGCTCAATGTGCTGACAG

AGCTGAAG

This

Study

P191. muteseK

Forward 3’

CCAATGAATCAATAGGAAAATTGTACTAACGATATC

CCGGGGCTCGTGTCGGTAG

This

Study

P192. muteseK

Reverse 3’ XbaI RE

CACGATGCCTCTAGATCAGTTTATGCCAGGGAATGC

TATACAGGGGACGCATC

This

Study

P197. GC-muteseK

Reverse 5’

CTACCGACACGAGCCCCGGGATATCGTTAGTACAAT

TTTCCTATTGATTCATTGG

This

Study

Secretion Assays. E. ictaluri cultures were grown according to Dubytska et al. under low

phosphate conditions in a defined minimal media (MMP) specific for E. ictaluri growth (18, 23).

Page 43: Edwardsiella Ictaluri Is Capable of Persisting in Channel

34

Briefly, E. ictaluri starter cultures were pelleted at 3,000 rpm (accuSpin™ 3R, Fisher Scientific)

washed once in MMP, and resuspended in MMP at 1 part starter culture to 5 parts MMP at 28C

with aeration for 4 hours. The pH of this culture was then neutralized to pH 7 to promote effector

secretion (18). After 90 minutes, cultures were pelleted, and the supernatant containing the

secreted proteins were collected. This was then used for immunoblot and Co-IP assays.

HKDM Collection. HKDMs were collected and used for downstream assays according

to Booth et al (24). Briefly, fish were euthanized in 1g/L Tricaine-S (Pentair). Head kidneys were

removed, homogenized through a sterile double stainless-steel mesh tissue sieve (280 and 140

µm) (Sigma Chemical co.), and suspended at approximately 1 X 107 cells/mL in channel catfish

macrophage medium (CCMM) (25). Five ml per well was added to 6 well plates (Sigma-

Aldrich). Cells were allowed to adhere overnight at 28C in a 5% CO2 atmosphere, washed three

times in RPMI, resuspended in CCMM and counted.

Co-IP. Co-IP experiments, performed on three separate occasions, were carried out using

the Dynabeads® Co-Immunoprecipitation Kit (Thermo Fisher Scientific) in conjugation with pre-

coated anti-FLAG® M2 magnetic beads (Sigma-Aldrich). Briefly, HKDM were lysed using lysis

buffer B, provided by the kit. Lysates were then allowed to incubate with fresh anti-FLAG

coated magnetic beads, to allow for pre-adsorption of non-specific protein binding. Pre-adsorbed

lysates were then allowed to incubate with fresh anti-FLAG coated magnetic beads, and

supernatant produced from eseK::3XFLAG that was concentrated approximately 1:100 using an

Amicon® Ultra-15 10K centrifugal filter device (EMD Millipore Corp). After incubation, the

beads were washed, and the co-immunoprecipitated proteins were eluted from the beads with a

proprietary kit-eluate containing lithium dodecyl sulfite (LDS). Eluate was run on LDS-PAGE,

and proteins of interest were verified by mass spectrometry.

Page 44: Edwardsiella Ictaluri Is Capable of Persisting in Channel

35

Polyacrylamide Gel Electrophoresis. Analysis with sodium dodecyl sulfate (SDS) or

LDS-PAGE was performed using 4-20% gradient polyacrylamide gels (Bio-Rad) following the

manufacture’s protocol. Following separation, proteins were either stained or transferred to a

PVDF membrane. Direct staining was carried out using Blue BANDit™ (VWR Life Science),

SYPRO® Ruby Red (Invitrogen), or Silver Reagent Concentrate (Bio-Rad). Gel transfer for

Western blotting was carried out using the iBlot™ (Invitrogen) according to manufacturer

protocols.

Mass spectrometry was completed in the LSU Mass Spectrometry Core Facility in the

Department of Chemistry. Briefly, protein samples cut from a polyacrylamide gel underwent

tryptic digest. Tryptic peptide peaks were identified by MALDI-TOF MS analysis. Observed

peaks were compared with theoretical mass values from in-silico digestion using the Peptide

Mass program (http://web.expasy.org/peptide_mass/).

Western Blot Analysis. Briefly, membranes were allowed to block in 5% BSA or 5%

milk for one hour at room temperature (RT) or overnight (O/N) at 4C. The anti-FLAG primary

antibody, THETM DYKDDDDK Tag, Mouse mAb (Genscript Biotech) was added at 1 ug/mL for

one hour at RT. Following this, mouse secondary antibody, IRDye 680RD donkey anti-Mouse

IgG (H+L) (LI-COR Biosciences) was used for detection on an Odyssey® CLx imager (LI-COR

Biosciences).

SimpleWES Electrophoresis and Detection. The WES system (Protein Simple) was

used for putative target protein detection. Briefly, 5 uL of equal concentrations of each sample

were loaded into wells and separated by capillary force with the 12-230 kDa Separation Module

(Protein Simple). Detection occurred using Anti-FLAG primary with the Anti-Mouse Detection

Module (Protein Simple). Detection followed standard WES cycling parameters.

Page 45: Edwardsiella Ictaluri Is Capable of Persisting in Channel

36

Fish Care. Channel catfish egg masses were received from a Morehead, Mississippi

catfish farm with no history of ESC outbreaks. The eggs were then disinfected with 100 ppm free

iodine and hatched in a closed recirculating system in the specific-pathogen-free (SPF)

laboratory at the LSU School of Veterinary Medicine. Fish were reared in systems containing

four 350 L fiberglass tanks connected to a 45 L recirculating-bead filter (Aquaculture Systems

Technologies). Water quality was tested at least three times a week, and the temperature was

constantly monitored to ensure optimal conditions for fish wellness. Specific pathogen-free

channel catfish fingerlings ranging from 12.5, 20, and 32.5 g in separate experiments were

moved at 25 fish each per 20 L flow-through tank. Catfish were fed 2% to 3% body weight every

other day and allowed to acclimate 28 days before vaccination (26). Pre-vaccination, catfish

were re-sorted to assure 25 fish per tank. All experiments were carried out following protocols

approved by the Institutional Animal Care and Use Committee and the Inter-Institutional

Biological and Recombinant DNA Safety Committee of Louisiana State University.

Immersion Assays. Starter cultures of the WT strain E. ictaluri 93-146 and the ΔeseK

strain were used to inoculate up to 400 mL BHI per 2 L flask at 28C and cultures were allowed

to incubate until each bacterial treatment reached an OD600, reflecting a concentration of 1 X 1010

colony-forming unit’s (CFU) per mL. The culture was then diluted as required per dose-

treatment for the inoculation of the catfish. The bacterial concentration was subsequently verified

by drop plating ten-fold serial dilutions in triplicate on trypticase soy agar supplemented with 5%

defibrinated sheep blood (BA) (27).

Vaccination. During vaccination, each tank was lowered to 4 L or 8 L of water using a

standardized adapter. Cultures of ΔeseK grown in BHI-porcine was then added to 4 tanks each

for the preliminary experiment, and six tanks each for Trials 1 and 2 assigned randomly. This

Page 46: Edwardsiella Ictaluri Is Capable of Persisting in Channel

37

allowed for the following doses: preliminary experiment, ΔeseK 1.8 X 108 cfu/mL, and WT 1.7

X 108; for Trial 1, E. ictaluri ΔeseK 1.1 X 107 cfu/mL; for Trial 2, ΔeseK 5.1 X 106 cfu/mL. An

uninfected BHI-Sham served as a negative control. After one hour the tanks were allowed to

return to 20 L. Catfish were monitored daily for mortalities, which typically occurred within 14

days (26). E. ictaluri presence was confirmed in mortalities by inoculating liver samples onto BA

plates.

Challenge. At 4 and 8 weeks following vaccination, 3 tanks per vaccination treatment

were challenged using WT at the following doses: preliminary 3 X108 cfu/mL, Trial 1 challenge

4 week: 1.5 X108 cfu/mL and challenge 8 week: 1.6 X108 cfu/mL, Trial 2 challenge 4 week:

eseK: 1 X108 cfu/mL, challenge 8 week: 1.2 X108 cfu/mL following the same bacterial

immersion protocol described for the vaccination. Catfish were monitored for 14 days, and E.

ictaluri presence was confirmed in mortalities by streaking liver samples onto BA plates.

Statistics. Significance among treatments was determined by taking the average cumulative

percent survival of each treatment and carrying out a one-way ANOVA with Tukey’s posthoc

test and confidence interval of 95% using PRISM, Graphpad version 5.00.288.

Results

EseK::3XFLAG Functionality. The EseK::3XFLAG strain was determined to have

three ]plasmids, the two native plasmids pEI1 and pEI2, which are 4807 bp and 5643 bp

respectively, and a third plasmid approximately 8261 bp in size which correlates to the size

expected for the pBBR1-MCS4 plasmid carrying the EseK::3XFLAG fusion (Figure 2.2A) (28).

Catfish infected with eseK::3XFLAG at 1 X108 cfu/mL and WT at 1 X108 cfu/mL showed no

significant difference in mortality, with 26.6 and 21.3 percent survival observed, respectively

(Figure 2.2B). Functional secretion of EseK::3XFLAG, 105 kDa, was determined by

Page 47: Edwardsiella Ictaluri Is Capable of Persisting in Channel

38

immunoblot using the eseK::3XFLAG strain and WT strain grown at conditions favorable for

EseK translocation, detected using the anti-FLAG monoclonal antibody, and also using the WES

detection system (Figure 2.2C,D). No detection occurred in the WT strain, while detection was

visible in the EseK::3XFLAG strain. This band was verified as EseK with a FLAG fusion by

mass spectrometry (data not shown).

Figure 2.2: EseK::3XFLAG Confirmation. A) MW marker (1) followed by plasmid preps of

supercoiled DNA from ΔeseK (2), WT (3), and eseK::3XFLAG (4). eseK::3XFLAG regulates 3

plasmids, the native pEI1, 4807 bp, and pEI2, 5643 bp, in addition to pBBR1-

MCS4+EseK::3XFLAG, 8261 bp. B) Survival of catfish infected with WT or EseK::3XFLAG

strains each at 1 X108 cfu/mL. No significant difference in survival is detected. C) Western blot

detection of MW marker (1), eseK::3XFLAG using anti-FLAG in the supernatant of

EseK::3XFLAG at pH 5 (2), EseK::3XFLAG at pH 7 (3), WT at pH 5 (4), WT at pH 7 (5);

compared to standard protein marker (1). D) WES detection of EseK::3XFLAG in

Page 48: Edwardsiella Ictaluri Is Capable of Persisting in Channel

39

eseK::3XFLAG pH7 (102 kDa) and WT pH7 supernatants with an ERK1/2 (47 kDa) control

used for WES assay performance control.

EseK and CD74 Interaction. Co-IP using FLAG antibody-coated magnetic beads

confirmed CD74 and EseK interaction. Staining detected two distinct bands in a polyacrylamide-

gel of the co-immunoprecipitated sample (Figure 2.3). Mass spectrometry analysis confirmed

the two proteins as EseK and CD74 (Figure 2.4).

Figure 2.3: SYPRO Ruby Red Stain of Co-IP: of protein ladder (1), concentrated

EseK::3XFLAG supernatant (2), and co-immunoprecipitated proteins (3). EseK::3XFLAG is

predicted to be 105 kDa, and CD74 is predicted to be 26 kDa.

Page 49: Edwardsiella Ictaluri Is Capable of Persisting in Channel

40

Figure 2.4: Positional hits from mass spectrometry tryptic digest of proteins excised from bands

after Co-IP. The top amino acid sequence is EseK showing the positional hits indicated by Mass

spectrometry, including the 3XFLAG region. The bottom amino acid sequence is channel catfish

CD74, showing the positional hits indicated by Mass spectrometry.

Preliminary Vaccination. A preliminary vaccination experiment determined virulence

of ΔeseK at varying doses. While ΔeseK and WT at the highest dose showed no significant

difference, with a mean of 28 and 8% respectively, ΔeseK had fewer mortalities, a correlation

also seen in prior preliminary lab experiments (data not shown). The remaining doses proved to

be significantly attenuated, where survival improved, showing 95% survival in ΔeseK 1.8 X107

Page 50: Edwardsiella Ictaluri Is Capable of Persisting in Channel

41

cfu/mL, 99% survival in ΔeseK 1.8 X104 cfu/mL, and 100% survival in all other doses (Figure

2.5A).

Figure 2.5: Preliminary Vaccination and Challenge A) Fish were infected with E. ictaluri wild

type 1.8 X108 cfu/mL, ΔeseK 1.8 X108 cfu/mL, ΔeseK 1.8X107 cfu/mL, ΔeseK 1.8 X106 cfu/mL,

ΔeseK 1.8 X105 cfu/mL, ΔeseK 1.8 X104 cfu/mL, Sham-BHI, or Control. Deaths were monitored

for 14 days post vaccination. B) All channel Catfish were challenged with wild type 2.9 X108

cfu/mL. Deaths were monitored for 14 days PV.

Preliminary Challenge. Seven weeks post-vaccination (PV), catfish were exposed to

WT at a dose of 2.9 X108 cfu/mL. Fish vaccinated with doses as low as 1.8 X105 cfu/mL had

84.5% survival, similar to 71.7% observed in 65ST. Doses as low as ΔeseK 1.8 X105 cfu/mL

Page 51: Edwardsiella Ictaluri Is Capable of Persisting in Channel

42

provided protection, as 83.5% of catfish survived, and 99 to 100% survived in the ΔeseK 1.8

X106 cfu/mL and ΔeseK 1.8 X107 cfu/mL treatments (Figure 2.5B). Interestingly, 54.3% of

catfish survived the ΔeseK 1.8 X104 cfu/mL dose, so remaining experiments incorporated this

dosage range.

Safety of ΔeseK. Two separate experiments were then performed on catfish fingerlings

using the optimal dosage for protection from the preliminary data of 1.1 X107 cfu/mL to 5.1

X104 cfu/mL.

In both experiments, 6 tanks each containing 25 fish were vaccinated with ΔeseK at doses

from 1.1 X105 cfu/mL to 1.1 X107 cfu/mL for the first experiment (Table 2.3), and 5.1 X104

cfu/mL to 5.1 X106 cfu/mL for the second experiment (Table 2.3). This range appeared safe,

where mortalities were only seen in the highest doses of 5.1 X106 cfu/mL to 1.1 X107 cfu/mL

between the two experiments, 4.7%, and 0.7% respectively.

Table 2.3: Safety of ΔeseK vaccinated fish. Percent survival observed per treatment per tank

from Vaccinations during Trial 1 and Trial 2 followed by average (Avg) percent survival

observed per treatment.

Trial 1 Vaccination Survival Per Tank Avg

ΔeseK 1.1 X107

cfu/mL 100 100 100 96 100 100 99.3

ΔeseK 1.1 X106

cfu/mL 100 100 100 100 100 100 100.0

ΔeseK 1.1 X105

cfu/mL 100 100 100 100 100 100 100.0

Sham-BHI 100 100 100 100 100 100 100.0

Trial 2 Vaccination Survival Per Tank Avg

ΔeseK 5.1 X106

cfu/mL 100 92 80 100 100 100 95.3

ΔeseK 5.1 X105

cfu/mL 100 100 100 100 100 100 100.0

ΔeseK 5.1 X104

cfu/mL 100 100 100 100 100 100 100.0

Sham-BHI 100 100 100 100 100 100 100.0

Page 52: Edwardsiella Ictaluri Is Capable of Persisting in Channel

43

Early WT Challenge. Challenges were carried out on three of the six tanks per dose 4

weeks PV to determine if catfish can mount a relatively quick response to subsequent WT

challenge. In the first experiment, fish received WT at 1.5 X108 cfu/mL, and in the second trial,

fish received WT at 1 X108 cfu/mL. In the first experiment (figure 2.6A), the ΔeseK 1.1 X107

cfu/mL vaccine dose had 100% survival, the ΔeseK 1.1 X106 cfu/mL dose had 92% survival, and

the ΔeseK 1.1 X105 cfu/mL dose had 74.3% survival. The second trial (figure 2.6B) followed a

similar trend where 100% survival was observed in ΔeseK 5.1 X106 cfu/mL, 97.3% survival was

observed in ΔeseK 5.1 X105 cfu/mL, and 78.7% survival was observed in ΔeseK 5.1 X104

cfu/mL. This compared to the Sham-Challenged, which only had 17.3% and 4% survival for the

first and second trials. This suggests that doses of 1.1 X107 cfu/mL to 5 X105 cfu/mL produce

high levels of protection against subsequent exposure to WT, while survival begins to decline

between the 5.1 X105 cfu/mL and 1.1 X105 cfu/mL dose to 74.6%, where the lower doses of 1.1

X105 cfu/mL to 5.1 X104 cfu/mL begin to decline in protection.

Late WT Challenge. Assessment of late survival occurred at 8 weeks post-vaccination,

where the final 3 of 6 tanks of catfish received the WT dose at 1.6 X108 cfu/mL for Trial 1

(Figure 2.7A) and 1.2 X108 cfu/mL for Trial 2 (Figure 2.7B). In the first experiment it was

determined that 97.3% of the ΔeseK 1.8 X107 cfu/mL fish survived, 100% of the ΔeseK 1.1 X106

cfu/mL treatment survived, and 94.6% of the ΔeseK 1.1 X105 cfu/mL fish survived (figure 2.7B).

During the second Trial 100% of the ΔeseK 5.1 X106 cfu/mL fish survived, but reduced

survivability of 74.7% and 28% resulted in the ΔeseK 5.1 X105 cfu/mL, and the ΔeseK 5.1 X104

cfu/mL respectively (Figure 2.7B). Together the trend suggests that the highest doses of ΔeseK

1.1 X107 cfu/mL to 1.1 X105 cfu/mL are protective, but this decreases at the 1.1 X105 doses

through the 5.1 X104 doses.

Page 53: Edwardsiella Ictaluri Is Capable of Persisting in Channel

44

Figure 2.6: Early Exposure to WT. A) Trial 1. Mean cumulative percent survival with standard

error of the mean of 3 tanks per dose of ΔeseK 1.1 X107 cfu/mL, ΔeseK 1.1 X106 cfu/mL, ΔeseK

1.1 X105 cfu/mL challenged with WT 1.5 X108 cfu/mL. B) Trial 2. Mean cumulative percent

survival with standard error of the mean of 3 tanks per dose of ΔeseK 5.1 X106 cfu/mL, ΔeseK

5.1 X105 cfu/mL, ΔeseK 5.1 X104 cfu/mL challenged with WT 1 X108 cfu/mL. Significant

differences in groups represented by letters.

Page 54: Edwardsiella Ictaluri Is Capable of Persisting in Channel

45

Figure 2.7: Late WT challenge. A) Trial 1. Mean cumulative percent survival with standard error

of the mean of 3 tanks per dose of ΔeseK 1.1 X107 cfu/mL, ΔeseK 1.1 X106 cfu/mL, ΔeseK 1.1

X105 cfu/mL challenged with WT 1.6 X108 cfu/mL. B) Trial 2. Mean cumulative percent

survival with standard error of the mean of 3 tanks per dose of ΔeseK 5.1 X106 cfu/mL, ΔeseK

5.1 X105 cfu/mL, ΔeseK 5.1 X104 cfu/mL challenged with WT 1.2 X108 cfu/mL. Significant

differences in groups represented by letters.

Discussion

The high degree of homology among the E. ictaluri T3SS LRR effectors makes them

impossible to detect from one another with specific antibody. The 3XFLAG tag was chosen

Page 55: Edwardsiella Ictaluri Is Capable of Persisting in Channel

46

because tagging can alter protein function and expression, and the 3XFLAG tag strategy proved

effective on T3SS effectors SopB and SspH1 in Salmonella (13, 29). Using an eseK::3XFLAG

fusion created under the control of EseK’s native promoter on pBBR1-MCS4 and inserted into

the ΔeseK strain, effectively resulted in a complemented strain. The plasmids of eseK::3XFLAG

exhibited regulation at the same relative abundance of the two native plasmids in the WT and

ΔeseK strain. The virulence of eseK::3XFLAG was the same as the WT, and the protein was

able to be secreted and detected specifically in the supernatant. This allowed completion of a Co-

IP assay while minimizing specificity issues associated with false negatives (30). A previous

experiment in the lab utilizing a yeast-2-hybrid assay gave the first indication that EseK interacts

with CD74. The Co-IP assay using bacterial supernatant and a HKDM lysate confirmed the

interaction between EseK and CD74. Carrying out this experiment with HKDM leaves other

potential target proteins unidentified, but reduced the potential for non-specific protein binding

from an increased protein pool (30). Mass spectrometry then identified the proteins as CD74 and

EseK. This is of interest because CD74 carries out a wide array of roles in the host immune

response (31), including chaperoning MHC class II during its assembly and migration from the

ER to the cell surface (32).

CD74 also acts as a ligand-receptor for macrophage migration inhibitory factor (MIF)

regulation and exhibits a role in cross-presentation to MHC class I (31). It is possible that the

interaction of EseK with CD74 means that it is involved with suppression of cell-mediated

immune responses, and that its deletion may improve protection. Based on this hypothesis, the

virulence of ΔeseK was determined relative to the WT, T3SS null mutant (65ST) and Sham-BHI.

Early exposure to ΔeseK at 4 weeks PV found that protection is very high for all doses. A later

exposure at 8 weeks PV saw a decline in protection from WT. The reduced efficacy at eight

Page 56: Edwardsiella Ictaluri Is Capable of Persisting in Channel

47

weeks could result because the dosage given 8 weeks prior to challenge was too low to mount an

adequate antibody response after 8 weeks. A 1993 study found that fish re-exposed to WT after

a natural ESC outbreak showed a correlation of antibody titer to susceptibility to disease after re-

infection, and that some fish produce low or no antibody titer after initial exposure (33). It was

also speculated that re-stimulation with antigen could improve antibody titers. Literature

suggests that antibody accumulation is less important than cell-mediated responses in preventing

ESC because E. ictaluri is an intracellular pathogen that survives and replicates within host cells,

where it is protected from antibody (34, 35). Moreover, previous reports of fish surviving

outbreaks of ESC following immersion challenge were determined not to be antibody-related

(36), giving further credence to a major role for CMI (35). This could suggest that ΔeseK is

producing CMI responses at higher doses, but not robustly enough at lower doses to be

protective at 8 weeks post- vaccination. These data support the overarching hypothesis that WT

is capable of preventing CMI to persist in the host, because this is likely the arm of immunity

most capable of targeting an intracellular pathogen. Additionally, this immunity is of interest in

the case of EseK because an interaction with CD74 could have implications on limiting antigen

presentation and preventing a strong T cell response.

Our current findings determined that EseK interacts with catfish CD74 in HKDM, and that

protection could be observed in response to exposure to the ΔeseK strain. Future work will aid

our understanding of EseK’s role in the cell-mediated immune response.

Literature Cited

1. USDA. 2010. USDA Catfish 2010: Part III: Changes in Catfish Health and Production

Practices in the United States, 2002-2009.

2. FAO. 2011-2012. Fisheries and aquaculture software. FishStatJ, Rome.

Page 57: Edwardsiella Ictaluri Is Capable of Persisting in Channel

48

3. FAO. 2018. The State of World Fisheries and Aquaculture 2018: Meeting the Sustainable

Development Goals. Rome.

4. Paul G. 2012. Aquaculture in the United States of America: present status and future

opportunities. Bull Fish Res Agen No 35:7-13.

5. J. P. Hawke RMD, R. L. Thune, A, C. Camus. 1998. ESC- Enteric Septecemia of Catfish.

6. Yang Q, Pan YL, Wang KY, Wang J, He Y, Wang EL, Liu T, Yi G, Chen DF, Huang

XL. 2016. OmpN, outer membrane proteins of Edwardsiella ictaluri are potential vaccine

candidates for channel catfish (Ictalurus punctatus). Mol Immunol 78:1-8.

7. Wise DJ, Greenway TE, Byars TS, Griffin MJ, Khoo LH. 2015. Oral vaccination of

channel catfish against Enteric Septicemia of Catfish using a live attenuated Edwardsiella

ictaluri isolate. J Aquat Anim Health 27:135-43.

8. Nho SW, Abdelhamed H, Karsi A, Lawrence ML. 2017. Improving safety of a live

attenuated Edwardsiella ictaluri vaccine against enteric septicemia of catfish and

evaluation of efficacy. Vet Microbiol 210:83-90.

9. Thune RL, Fernandez DH, Benoit JL, Kelly-Smith M, Rogge ML, Booth NJ, Landry CA,

Bologna RA. 2007. Signature-Tagged Mutagenesis of Edwardsiella ictaluri Identifies

Virulence-Related Genes, Including a Salmonella Pathogenicity Island 2 Class of Type

III Secretion Systems. Applied and Environmental Microbiology 73:7934-7946.

10. Vishwakarma V, Pati NB, Chandel HS, Sahoo SS, Saha B, Suar M. 2012. Evaluation of

Salmonella enterica serovar Typhimurium TTSS-2 deficient fur mutant as safe live-

attenuated vaccine candidate for immunocompromised mice. PLoS One 7:e52043.

11. Wilson BA, Salyers AA, Whitt DD, Winkler ME. 2011. Bacterial pathogenesis: a

molecular approach. American Society for Microbiology (ASM).

12. Dubytska LP, Rogge ML, Thune RL. 2016. Identification and Characterization of

Putative Translocated Effector Proteins of the Edwardsiella ictaluri Type III Secretion

System. mSphere 1.

13. Bhavsar AP, Brown NF, Stoepel J, Wiermer M, Martin DD, Hsu KJ, Imami K, Ross CJ,

Hayden MR, Foster LJ, Li X, Hieter P, Finlay BB. 2013. The Salmonella type III effector

SspH2 specifically exploits the NLR co-chaperone activity of SGT1 to subvert immunity.

PLoS Path 9:e1003518.

Page 58: Edwardsiella Ictaluri Is Capable of Persisting in Channel

49

14. McShan AC, De Guzman RN. 2015. The bacterial type III secretion system as a target for

developing new antibiotics. Chem Biol Drug Des 85:30-42.

15. Rogge ML, Thune RL. 2011. Regulation of the Edwardsiella ictaluri type III secretion

system by pH and phosphate concentration through EsrA, EsrB, and EsrC. Appl Environ

Microbiol 77:4293-302.

16. Kelley LA, Mezulis S, Yates CM, Wass MN, Sternberg MJE. 2015. The Phyre2 web

portal for protein modeling, prediction and analysis. Nat Protoc 10:845-858.

17. Kobe B, Kajava AV. 2001. The leucine-rich repeat as a protein recognition motif.

Current Opinion in Structural Biology 11:725-732.

18. Dubytska LP, Rogge ML, Thune RL. 2016. Identification and characterization of putative

translocated effector proteins of the Edwardsiella ictaluri type III secretion system.

mSphere 1:e00039-16.

19. Edwards RA, Keller LH, Schifferli DM. 1998. Improved allelic exchange vectors and

their use to analyze 987P fimbria gene expression. Gene 207:149-157.

20. Herrero M, de Lorenzo V, Timmis KN. 1990. Transposon vectors containing non-

antibiotic resistance selection markers for cloning and stable chromosomal insertion of

foreign genes in gram-negative bacteria. J Bacteriol 172:6557-6567.

21. de Lorenzo V, Herrero M, Jakubzik U, Timmis KN. 1990. Mini-Tn5 transposon

derivatives for insertion mutagenesis, promoter probing, and chromosomal insertion of

cloned DNA in gram-negative eubacteria. J Bacteriol 172:6568-72.

22. Kovach ME, Elzer PH, Hill DS, Robertson GT, Farris MA, Roop RM, 2nd, Peterson KM.

1995. Four new derivatives of the broad-host-range cloning vector pBBR1MCS, carrying

different antibiotic-resistance cassettes. Gene 166:175-6.

23. Collins LA, Thune RL. 1996. Development of a defined minimal medium for the growth

of Edwardsiella ictaluri. Appl Environ Microbiol 62:848-52.

24. Booth NJ, Elkamel A, Thune RL. 2006. Intracellular replication of Edwardsiella ictaluri

in channel catfish macrophages. J Aquat Anim Health 18:101-108.

Page 59: Edwardsiella Ictaluri Is Capable of Persisting in Channel

50

25. Wang Y, Wang Q, Baoprasertkul P, Peatman E, Liu Z. 2006. Genomic organization, gene

duplication, and expression analysis of interleukin-1beta in channel catfish (Ictalurus

punctatus). Mol Immunol 43:1653-64.

26. Rogge M. 2009. Expression of the Edwardsiella ictaluri type III secretion system and its

relationship to the native plasmids. Doctor of Philosophy. Louisiana State University and

Agricultural and Mechanical College

27. Booth NJ, Beekman JB, Thune RL. 2009. Edwardsiella ictaluri encodes an acid-

activated urease that is required for intracellular replication in channel catfish (Ictalurus

punctatus) macrophages. Appl Environ Microbiol 75:6712-20.

28. Fernandez DH, Pittman-Cooley L, Thune RL. 2001. Sequencing and analysis of the

Edwardsiella ictaluri plasmids. Plasmid 45:52-6.

29. Finn CE, Chong A, Cooper KG, Starr T, Steele-Mortimer O. 2017. A second wave of

Salmonella T3SS1 activity prolongs the lifespan of infected epithelial cells. PLoS Path

13:e1006354.

30. Moresco JJ, Carvalho PC, Yates JR, 3rd. 2010. Identifying components of protein

complexes in C. elegans using co-immunoprecipitation and mass spectrometry. J

Proteomics 73:2198-204.

31. Schroder B. 2016. The multifaceted roles of the invariant chain CD74-more than just a

chaperone. Biochim Biophys Acta 1863:1269-81.

32. Abul K. Abbas AHHL, Shic Pillai. 2012. Basic Immunology: Functions and Disorders of

the Immune System, 4 ed.

33. Vinitnantharat S, Plumb JA. 1993. Protection of channel catfish Ictalurus punctatus

following natural exposure to Edwardsiella ictaluri and effects of feeding antigen on

antibody titer. Dis Aquat Org 15:31-34.

34. Kaufmann SH. 1993. Immunity to intracellular bacteria. Annu Rev Immunol 11:129-163.

35. Thune RL, Fernandez DH, Battista JR. 1999. An aroA mutant of Edwardsiella ictaluri is

safe and efficacious as a live, attenuated vaccine. J Aquat Anim Health 11:358-372.

36. Klesius PH, Sealey WM. 1995. Characteristics of serum antibody in Enteric Septicemia

of Catfish. J Aquat Anim Health 7:205-210.

Page 60: Edwardsiella Ictaluri Is Capable of Persisting in Channel

51

CHAPTER 3. NAÏVE HEAD KIDNEY DERIVED MACROPHAGES

FAVOR M1 POLARIZATION IN RESPONSE TO EDWARDSIELLA

ICTALURI INFECTION

Introduction

Macrophages are important facilitators of Edwardsiella ictaluri infection, where E.

ictaluri can invade, survive, and replicate (1). Opsonization of live E. ictaluri with normal catfish

serum greatly increases E. ictaluri’s uptake into catfish macrophages (1). Once internalized, E.

ictaluri can replicate in spacious vacuoles within the macrophage called Edwardsiella-

containing-vacuoles (ECV) (1). Researchers use macrophages often in experiments indicating

that E. ictaluri survives in macrophages from several tissues, including intestinal macrophages

(2), peritoneal macrophages (3), and head-kidney-derived macrophages (HKDM) (1).

Macrophages are important facilitators of host immune functions, capable of activating and

polarizing in order to aid host defenses (4). Classically, macrophages polarize to the M1 pro-

inflammatory response which promotes killing of invading microbes, or the M2 anti-

inflammatory response which promotes tissue repair (4).

In particular, HKDM are of interest because of their necessity and importance to the

natural disease process, ease of cultivation in laboratory settings, and our understanding of the

dynamics of the host-pathogen interaction (1, 5-7). HKDMs also allow ex vivo modeling of

disease to aid in a better prediction of disease outcome before conducting in vivo assays on

catfish, where host death is usually the end-result.

E. ictaluri has multiple virulence factors that allow it to evade innate immune defense

mechanisms and facilitate replication within macrophages (1, 8-10). Some of the E. ictaluri

virulence factors involved in HKDMs include a urease pathogenicity island (PAI) that encodes

Page 61: Edwardsiella Ictaluri Is Capable of Persisting in Channel

52

an acid activated urease that allows it to modulate pH (11), a Type III secretion system, (12, 13),

and a Type VI secretion system (12). The Thune lab primarily focuses on the role of the E.

ictaluri T3SS in pathogenesis, and, specifically in this study, the involvement of T3SS effector

EseK.

Recent studies in catfish indicate that HKDM may help model immune system

modulation. Russo et al. showed that macrophages exposed to convalescent serum produced

tenfold more reactive oxygen and nitrogen species and killed E. ictaluri better than macrophages

from non-vaccinated fish (14). In another study, Russo et al. found an importance for amino

acids such as glutamine and arginine in increasing the phagocytic capability of macrophages and

subsequent killing of E. ictaluri (7, 14). These findings, together with the common knowledge of

macrophage clearance of intracellular pathogens, suggest a necessity for an M1/ cell-mediated

immune response to clear E. ictaluri in catfish macrophages (14). The literature mentioned

above, however, also strongly indicates that E. ictaluri is very capable of persisting in the naïve

host, particularly in HKDM. Consequently, this experiment aims to address the native response

of HKDMs to infection with E. ictaluri. Based on the literature, we hypothesized that WT

persists and replicates in channel catfish HKDM because it prevents or evades polarizing effects

implemented by HKDMs. Preliminary results and results presented in the previous chapter

suggest that EseK aids WT E. ictaluri in this response. The experimental design for this work

involved using RNA sequence analysis (RNA seq) on WT, ΔeseK, and uninfected HKDM

samples. RNA seq allows for a more in-depth analysis of target samples and a more complete

picture of host-pathogen dynamics (15-21). The predicted regulation of several pathways

indicates a role for M1 polarization. Additionally, we uncovered potential mechanisms for

preventing apoptosis and an effect on initiation of the adaptive immune responses.

Page 62: Edwardsiella Ictaluri Is Capable of Persisting in Channel

53

Methods

Bacterial Growth. Strains used in this study were Edwardsiella ictaluri LADL93-146

isolated in 1993from moribund channel catfish in a natural outbreak of ESC on a commercial

farm, and ΔeseK in E. ictaluri with a deletion of the first 2200 bp, created in the Thune lab. E.

ictaluri strains were grown from frozen stock cultures stored at -80C in 5mL BHI-porcine

(Sigma-Aldrich) overnight (O/N) at 28C with aeration.

Fish Care. Channel catfish egg masses were obtained from a Morehead, Mississippi

catfish farm with no history of ESC outbreaks. The eggs were disinfected with 100 ppm free

iodine and hatched in a closed recirculating system in the specific pathogen-free (SPF)

laboratory at the LSU School of Veterinary Medicine. Fish were reared in systems containing

four, 350 L fiberglass tanks, connected to a 45 L bead filter (Aquaculture Systems

Technologies). Water quality was tested at least three times a week, and the temperature was

constantly monitored to ensure optimal conditions for fish wellness. All experiments were

carried out following protocols approved by from the Institutional Animal Care and Use

Committee and the Inter-Institutional Biological and Recombinant DNA Safety Committee of

Louisiana State University.

HKDM Collection. HKDMs were collected according to the method of Booth et al. 2006

(1). Briefly, fish were euthanized in 1 g/L Tricaine-S (Pentair). Head kidneys were removed,

homogenized through a sterile double stainless-steel mesh tissue sieve (280 and 140 µm) (Sigma

Chemical Co.), and suspended at approximately 1 X107 cells/mL in channel catfish macrophage

medium (CCMM) (22). Five mL per well was added to each well of a 6-well plate (Sigma-

Page 63: Edwardsiella Ictaluri Is Capable of Persisting in Channel

54

Aldrich), and cells were allowed to adhere overnight at 28C in a 5% CO2 atmosphere. Following

this, cells were washed three times in RPMI, re-suspended in 5 mL CCMM and counted.

HKDM Assays. Both WT and ΔeseK were opsonized with normal channel catfish serum

for 30 minutes before being added to 6 well plates containing HKDMs at a multiplicity of

infection (MOI) of 10 bacteria/cell. Plates were then centrifuged at 400x g for 5 minutes to

promote bacterial contact with the cell sheet, then incubated for 30 minutes before a 100 µg/mL

killing dose of gentamicin was added for 1 hour. Following this, HKDMs were washed one time

in RPMI (Sigma-Aldrich) and resuspended in CCMM containing a 1 µg/mL static dose of

gentamicin. Cells were allowed to persist for 5 hours, then washed once with RPMI before

proceeding to RNA extractions. Sampling was performed on three separate occasions according

to the collection and assay protocols.

RNA Extractions. Total RNA extractions were carried out on HKDM samples using

RNAzol® RT Isolation Reagent (Molecular Research Center) in conjunction with the RNA

Clean and Concentrator-5 kit (Zymo Research) following manufacturer protocols. Samples were

resuspended in DEPC-treated H20 (Ambion™). The RNA quality was determined by fragment

analysis using the Agilent Fragment Analyzer (Advanced Technologies, Inc) with the lowest

RNA quality number (RQN) value of 7.4. After fragment analysis, mRNA extractions and runs

were carried out by LSU School of Veterinary Medicine Genelab Core. mRNA was extracted

using the RiboMinus Eukaryote Kit v2 (Ambion™) and was sequenced on an Ion Proton Next-

Generation Sequencer.

Data Analysis. Data was compiled and analyzed by the LSU Center for Computational

Technology using Partek Flow based on researcher input. Unaligned reads were converted to

FASTQ format, trimmed using a quality score and read length (Phred>20; >25bp), then aligned

Page 64: Edwardsiella Ictaluri Is Capable of Persisting in Channel

55

to the Ictalurus punctatus reference genome using STAR (Version 2.6.1d)(23). The reads were

then quantified to the Ictalurus punctatus genome, calculating the fragments per kilobase of

transcript per million aligned reads (FPKM) using the Partek Flow quantification algorithm,

based on the hierarchical model described by Leng et. al (24). Differential analysis between each

macrophage treatment (WT versus uninfected, ΔeseK versus uninfected, and WT versus ΔeseK)

was carried out using Gene Specific Analysis (GSA), and transcripts were selected that had 200

or more total counts with a p-value of 0.2 or less. Pathway analysis was carried out using

Ingenuity Pathway Analysis (IPA), with a q-value cut-off of 0.1, to identify molecular pathways

containing the differentially expressed genes. In IPA, expression analysis allowed the calculation

of the fold change in expression between treatments which were then used to calculate z-scores.

Results

GSA Analysis. Comparisons of the three treatments of samples using GSA analysis

uncovered approximately 45,000 genes, with 2126 genes having p-values of 0.2 or less (Table

3.1). IPA was then carried out to predict the most relevant pathways effected by infection with

WT and ΔeseK compared to uninfected HKDMs. While there were specific genes differentially

regulated between the WT and ΔeseK, none of these were significantly differentially regulated

when analyzed with a q value of 0.1 in the IPA.

Pathway Analysis. To further analyze the data, pathways of interest were examined by

z-score value. Because relatively few differences were observed in the WT versus ΔeseK

treatments, the remaining results focus on pathways significantly changed in the WT compared

to the uninfected HKDMs. Similar results would be expected for ΔeseK compared to uninfected

HKDMs. Approximately 180 pathways were analyzed (Figure 3.1). Several pathways showed a

Page 65: Edwardsiella Ictaluri Is Capable of Persisting in Channel

56

significant difference in regulation. For the sake of this work, the top 40 pathways were

investigated more closely because they had strong combined p-values and z-scores.

Page 66: Edwardsiella Ictaluri Is Capable of Persisting in Channel

57

Table 3.1: Top 50 reads via GSA. P-values and fold-changes given for WT versus uninfected, ΔeseK versus uninfected (Un), and WT

versus ΔeseK.

GENE

SYMBOL Transcript ID

P-value

(WT vs.

Un.)

Fold change

(WT vs. Un.)

P-value

(ΔeseK

vs. Un.)

Fold change

(ΔeseK vs. Un.)

P-value

(WT vs.

ΔeseK)

Fold change (WT

vs. ΔeseK)

JUNB XM_017458253.1 1.83E-07 35.6 1.12E-07 41.9 4.71E-01 -1.2

ENPEP XM_017465049.1 9.93E-07 687.4 8.71E-07 693.0 8.72E-01 -1.0

IER3 XM_017451685.1 1.05E-06 17.5 5.40E-07 21.4 4.12E-01 -1.2

KDM6B XM_017472048.1 1.09E-06 17.9 1.08E-06 17.6 9.96E-01 1.0

LOC108269347 XM_017475170.1 1.65E-06 460.9 1.92E-06 403.7 8.59E-01 1.1

LOC108257234 XM_017454847.1 2.37E-06 4.0 1.54E-06 4.3 6.21E-01 -1.1

LOC108267131 XM_017471106.1 2.46E-06 32.0 1.68E-06 39.5 6.67E-01 -1.2

RNF19B XM_017454608.1 2.60E-06 6.1 1.05E-06 7.1 3.05E-01 -1.2

LOC108269240 XM_017474985.1 2.92E-06 9.7 8.91E-07 12.6 1.87E-01 -1.3

LOC108260811 XM_017461423.1 2.95E-06 21646.6 2.52E-06 23802.9 8.63E-01 -1.1

COBLL1 XM_017470098.1 3.11E-06 330.2 2.85E-06 330.9 9.23E-01 -1.0

NR4A3 XM_017453779.1 3.83E-06 5.1 6.28E-07 7.3 5.60E-02 -1.4

LOC108260305 XM_017460480.1 4.48E-06 8.6 3.76E-06 9.0 8.53E-01 -1.0

TTC13 XM_017449996.1 5.51E-06 124.9 3.44E-06 142.8 6.20E-01 -1.1

NFKBIA XM_017475822.1 5.53E-06 23.6 2.84E-06 28.8 4.81E-01 -1.2

TTC13 XM_017450006.1 5.61E-06 123.2 3.50E-06 140.8 6.20E-01 -1.1

TNFAIP3 XM_017464718.1 6.95E-06 9.1 3.82E-06 10.1 5.37E-01 -1.1

CD82 XM_017478912.1 7.02E-06 8.2 5.00E-06 8.8 7.28E-01 -1.1

ABTB2 XM_017485648.1 7.21E-06 6.2 1.51E-06 9.3 1.12E-01 -1.5

LOC108278857 XM_017492510.1 7.63E-06 30.3 3.42E-06 41.9 4.12E-01 -1.4

AR XM_017459546.1 7.86E-06 37.4 1.28E-05 28.5 6.35E-01 1.3

ITGA2 XM_017488883.1 8.68E-06 16.7 5.28E-06 21.1 6.16E-01 -1.3

LOC108257234 XM_017454838.1 8.70E-06 3.6 3.05E-06 4.2 2.88E-01 -1.2

LOC108269063 XM_017474584.1 9.11E-06 1254.4 6.99E-06 1523.8 7.92E-01 -1.2

RAD17 XM_017451562.1 1.27E-05 246.8 7.42E-06 306.2 6.03E-01 -1.2

DLL1 XM_017463168.1 1.36E-05 130.2 1.28E-05 128.2 9.55E-01 1.0

(table cont’d)

Page 67: Edwardsiella Ictaluri Is Capable of Persisting in Channel

58

Table 3.1 continued

GENE

SYMBOL Transcript ID

P-value

(WT vs.

Un.)

Fold change

(WT vs.

Un.)

P-value

(ΔeseK vs.

Un.)

Fold change

(ΔeseK vs.

Un.)

P-value

(WT vs.

ΔeseK)

Fold change (WT vs.

ΔeseK)

LOC108277114 XM_017489499.1 1.42E-05 146.5 1.82E-05 118.8 8.16E-01 1.2

ACSS2 XM_017486747.1 1.45E-05 7.3 9.35E-06 8.0 6.73E-01 -1.1

LOC108266341 XM_017469480.1 1.45E-05 3.6 6.54E-06 3.9 4.39E-01 -1.1

LOC108270133 XM_017476502.1 1.51E-05 270.7 1.82E-05 227.2 8.62E-01 1.2

NFKBIB XM_017459598.1 1.62E-05 3.6 5.97E-06 4.2 3.39E-01 -1.2

LOC108263482 XM_017464303.1 1.71E-05 260.5 1.71E-05 250.4 9.99E-01 1.0

BCOR XM_017459001.1 1.73E-05 4.4 4.82E-06 5.7 2.26E-01 -1.3

KLF12 XM_017470462.1 1.77E-05 18.1 7.02E-06 24.1 3.81E-01 -1.3

NDUFV3 XM_017470327.1 1.80E-05 67.8 1.69E-05 68.8 9.51E-01 -1.0

NDUFV3 XM_017470326.1 2.08E-05 42.4 1.86E-05 43.1 9.18E-01 -1.0

CBS XM_017458119.1 2.36E-05 16.5 1.04E-05 20.3 4.48E-01 -1.2

CFLAR XM_017469517.1 2.44E-05 14.6 1.10E-05 17.5 4.61E-01 -1.2

LOC108265972 XM_017468874.1 2.47E-05 18.6 1.35E-05 21.7 5.76E-01 -1.2

DOPEY2 XM_017469589.1 2.61E-05 3.5 8.17E-06 4.3 2.86E-01 -1.2

LOC108255781 XM_017451987.1 3.22E-05 7.8 7.88E-06 11.8 2.06E-01 -1.5

ATG4D XM_017457236.1 3.28E-05 3.2 1.68E-05 3.4 5.47E-01 -1.1

CD82 XM_017478916.1 3.28E-05 8.6 1.93E-05 9.9 6.33E-01 -1.2

PTGIS XM_017480375.1 3.59E-05 13.3 3.29E-05 13.5 9.40E-01 -1.0

XKR7 XM_017486463.1 3.72E-05 36.3 3.91E-05 35.9 9.65E-01 1.0

LOC108269063 XM_017474585.1 3.74E-05 97.4 2.43E-05 116.7 7.03E-01 -1.2

LOC108257234 XM_017454818.1 3.99E-05 3.2 1.97E-05 3.5 5.35E-01 -1.1

LOC108265925 XM_017468777.1 4.03E-05 4.3 1.65E-05 5.0 4.31E-01 -1.2

RAI14 XM_017492770.1 4.15E-05 3.3 1.21E-05 4.0 2.78E-01 -1.2

MAP3K1 XM_017489827.1 4.37E-05 3.6 1.00E-05 4.4 1.98E-01 -1.2

Page 68: Edwardsiella Ictaluri Is Capable of Persisting in Channel

59

Figure 3.1: Top 40 pathways according to overall -log(p-value) (y- axis), differentially regulated in WT compared to uninfected

HKDMs. Intensity of color indicate positive (orange), negative (blue), or no change (white) in z-score.

Page 69: Edwardsiella Ictaluri Is Capable of Persisting in Channel

60

Notable pathways within the top 40 which are predicted to be upregulated in the WT compared

to uninfected HKDMs were both TNFR1 and TNFR2 signaling, Acute Phase Response

signaling, IL-6 signaling, P13K/AKT signaling (Figure 3.2), Production of Nitric Oxide and

Reactive Oxygen Species in Macrophages (which was not indicated in pathway analysis), IL-15

signaling, INOS signaling, NF-κB Activation by Viruses (Figure 3.2, Table 3.2), IL-1 signaling,

and Tec Kinase signaling.

Notable Pathways within the top 40 which are predicated to be downregulated in the WT

compared to uninfected HKDMs were Death Receptor signaling (Figure 3.3), CD40 signaling,

and PPAR signaling (Figure 3.1, Table 3.3).

Discussion

The RNA-seq data generated in this study has yielded an abundance of valuable information,

including several genes that exhibited high degrees of differential expression that were not

within the previously prescribed pathways, and will require a great deal of further investigation.

Because no pathways were predicted as differentially regulated between the WT and ΔeseK

infected HKDMs, we concluded that EseK has no bearing on transcription under these

parameters. Therefore, analysis of this work focused on WT compared to uninfected HKDMs.

The differential regulation of several important pathways and their related transcripts in response

to E. ictaluri infection have implications in M1 polarization. Upregulation of the iNOS pathway

is a classical indicator of M1 activation in macrophages (25, 26). Likewise, the upregulation of

both the acute phase response pathway (27), and NF-kB viral pathway have indications in M1

polarization. Further support for M1 polarization comes from the predicted downregulation of

the proliferator receptor (PPAR) signaling pathway (4). Unlike the other pathways, the PPARγ

Page 70: Edwardsiella Ictaluri Is Capable of Persisting in Channel

61

Table 3.2 Top upregulated pathways Predicted by IPA showing the top predicted genes per pathway, and its corresponding expression

value in WT compared to uninfected HKDMs, the false discovery rate (q- value), and the fold change between the WT and uninfected

HKDMs. IPA also indicated the expected regulation, location, and type of molecule per gene.

Symbol Gene Name p-value q-value Fold Change Expected Location Type(s)

TNFR1 and TNFR2 Signaling Pathways

BIRC2

baculoviral IAP repeat

containing 2 6.48E-04 7.22E-02 4.3

Down Cytoplasm enzyme

MAP3K1

mitogen-activated protein

kinase kinase kinase 1 4.37E-05 2.12E-02 3.6

Up Cytoplasm kinase

NFKB2

nuclear factor kappa B

subunit 2 5.38E-05 2.33E-02 4.8

Up Nucleus

transcription

regulator

NFKBIA NFKB inhibitor alpha 5.53E-06 9.52E-03 23.6

Up Cytoplasm

transcription

regulator

NFKBIB NFKB inhibitor beta 1.62E-05 1.31E-02 3.6

Up Nucleus

transcription

regulator

NFKBIE NFKB inhibitor epsilon 2.61E-04 4.67E-02 3.5

Up Nucleus

transcription

regulator

TANK

TRAF family member

associated NFKB activator 1.03E-04 3.02E-02 4.2

Down Cytoplasm other

TNFAIP3 TNF alpha induced protein 3 6.95E-06 1.04E-02 9.1 Nucleus enzyme

Acute Phase Response Signaling

IL1B interleukin 1 beta 5.31E-05 2.32E-02 35.4

Up

Extracellul

ar Space cytokine

JAK2 Janus kinase 2 2.24E-04 4.31E-02 4.5 Up Cytoplasm kinase

MAP3K1

mitogen-activated protein

kinase kinase kinase 1 4.37E-05 2.12E-02 3.6

Up Cytoplasm kinase

MAP3K5

mitogen-activated protein

kinase kinase kinase 5 1.20E-04 3.17E-02 4.3

Up Cytoplasm kinase

NFKB2

nuclear factor kappa B

subunit 2 5.38E-05 2.33E-02 4.8

Up Nucleus

transcription

regulator

(table cont’d)

Page 71: Edwardsiella Ictaluri Is Capable of Persisting in Channel

62

Table 3.2 continued

Symbol Gene Name p-value q-value Fold Change Expected Location Type(s)

NFKBIA NFKB inhibitor alpha 5.53E-06 9.52E-03 23.6

Up Cytoplasm

transcription

regulator

NFKBIB NFKB inhibitor beta 1.62E-05 1.31E-02 3.6

Down Nucleus

transcription

regulator

NFKBIE NFKB inhibitor epsilon 2.61E-04 4.67E-02 3.5

Down Nucleus

transcription

regulator

SHC1 SHC adaptor protein 1 9.37E-04 8.91E-02 2.5 Up Cytoplasm other

SOCS3

suppressor of cytokine

signaling 3 5.01E-05 2.24E-02 5.4

Down Cytoplasm phosphatase

STAT3

signal transducer and

activator of transcription 3 9.23E-04 8.84E-02 2.2

Up Nucleus

transcription

regulator

IL-6 Signaling

IL1B interleukin 1 beta 5.31E-05 2.32E-02 35.4

Up

Extracellul

ar Space cytokine

JAK2 Janus kinase 2 2.24E-04 4.31E-02 4.5 Up Cytoplasm kinase

NFKB2

nuclear factor kappa B

subunit 2 5.38E-05 2.33E-02 4.8

Up Nucleus

transcription

regulator

NFKBIA NFKB inhibitor alpha 5.53E-06 9.52E-03 23.6

Up Cytoplasm

transcription

regulator

NFKBIB NFKB inhibitor beta 1.62E-05 1.31E-02 3.6

Up Nucleus

transcription

regulator

NFKBIE NFKB inhibitor epsilon 2.61E-04 4.67E-02 3.5

Up Nucleus

transcription

regulator

SHC1 SHC adaptor protein 1 9.37E-04 8.91E-02 2.5 Up Cytoplasm other

SOCS3

suppressor of cytokine

signaling 3 5.01E-05 2.24E-02 5.4

Down Cytoplasm phosphatase

STAT3

signal transducer and

activator of transcription 3 9.23E-04 8.84E-02 2.2

Up Nucleus

transcription

regulator (table cont’d)

Page 72: Edwardsiella Ictaluri Is Capable of Persisting in Channel

63

Table 3.2 continued

Symbol Gene Name p-value q-value Fold Change Expected Location Type(s)

NF-kB Virus Pathway

ITGA2 integrin subunit alpha 2 8.68E-06 1.06E-02 16.7

Up

Plasma

Membrane

transmembrane

receptor

MAP3K1

mitogen-activated protein

kinase kinase kinase 1 4.37E-05 2.12E-02 3.6

Up Cytoplasm kinase

NFKB2

nuclear factor kappa B

subunit 2 5.38E-05 2.33E-02 4.8

Up Nucleus

transcription

regulator

NFKBIA NFKB inhibitor alpha 5.53E-06 9.52E-03 23.6

Up Cytoplasm

transcription

regulator

NFKBIB NFKB inhibitor beta 1.62E-05 1.31E-02 3.6

Up Nucleus

transcription

regulator

NFKBIE NFKB inhibitor epsilon 2.61E-04 4.67E-02 3.5

Up Nucleus

transcription

regulator

P13K/AKT Pathway

NFKB2

nuclear factor kappa B

subunit 2 5.38E-05 2.33E-02 4.8

Up Nucleus

transcription

regulator

NFKBIA NFKB inhibitor alpha 5.53E-06 9.52E-03 23.6

Up Cytoplasm

transcription

regulator

NFKBIB NFKB inhibitor beta 1.62E-05 1.31E-02 3.6

Up Nucleus

transcription

regulator

NFKBIE NFKB inhibitor epsilon 2.61E-04 4.67E-02 3.5

Up Nucleus

transcription

regulator

PIK3AP1

phosphoinositide-3-kinase

adaptor protein 1 6.30E-04 7.13E-02 3.2

Up Cytoplasm other

PLEKHA1

pleckstrin homology domain

containing A1 2.75E-04 4.73E-02 3.4

Cytoplasm other

SYK

spleen associated tyrosine

kinase 9.24E-04 8.84E-02 2.5

Up Cytoplasm kinase (table cont’d)

Page 73: Edwardsiella Ictaluri Is Capable of Persisting in Channel

64

Table 3.2 continued

Symbol Gene Name p-value q-value Fold Change Expected Location Type(s)

IL-15 Pathway

IL2RG

interleukin 2 receptor

subunit gamma 3.22E-04 5.16E-02 3.1

Up

Plasma

Membrane

transmembrane

receptor

JAK2 Janus kinase 2 2.24E-04 4.31E-02 4.5 Up Cytoplasm kinase

NFKB2

nuclear factor kappa B

subunit 2 5.38E-05 2.33E-02 4.8

Up Nucleus

transcription

regulator

SHC1 SHC adaptor protein 1 9.37E-04 8.91E-02 2.5 Up Cytoplasm other

STAT3

signal transducer and

activator of transcription 3 9.23E-04 8.84E-02 2.2

Up Nucleus

transcription

regulator

SYK

spleen associated tyrosine

kinase 9.24E-04 8.84E-02 2.5

Up Cytoplasm kinase

iNOS Signaling Pathway

JAK2 Janus kinase 2 2.24E-04 4.31E-02 4.5 Up Cytoplasm kinase

NFKB2

nuclear factor kappa B

subunit 2 5.38E-05 2.33E-02 4.8

Up Nucleus

transcription

regulator

NFKBIA NFKB inhibitor alpha 5.53E-06 9.52E-03 23.6

Up Cytoplasm

transcription

regulator

NFKBIB NFKB inhibitor beta 1.62E-05 1.31E-02 3.6

Up Nucleus

transcription

regulator

NFKBIE NFKB inhibitor epsilon 2.61E-04 4.67E-02 3.5

Up Nucleus

transcription

regulator

IL-1 Signaling Pathway

GNAI2 G protein subunit alpha i2 7.49E-04 7.83E-02 2.3

Plasma

Membrane enzyme

MAP3K1

mitogen-activated protein

kinase kinase kinase 1 4.37E-05 2.12E-02 3.6

Up Cytoplasm kinase

NFKB2

nuclear factor kappa B

subunit 2 5.38E-05 2.33E-02 4.8

Up Nucleus

transcription

regulator

(table cont’d)

Page 74: Edwardsiella Ictaluri Is Capable of Persisting in Channel

65

Table 3.2 continued

Symbol Gene Name p-value q-value Fold Change Expected Location Type(s)

NFKBIA NFKB inhibitor alpha 5.53E-06 9.52E-03 23.6

Up Cytoplasm

transcription

regulator

NFKBIB NFKB inhibitor beta 1.62E-05 1.31E-02 3.6

Up Nucleus

transcription

regulator

NFKBIE NFKB inhibitor epsilon 2.61E-04 4.67E-02 3.5

Up Nucleus

transcription

regulator

GNAI2 G protein subunit alpha i2 7.49E-04 7.83E-02 2.3

Plasma

Membrane enzyme

Tec Kinase Signaling

ITGA2 integrin subunit alpha 2 8.68E-06 1.06E-02 16.7

Up

Plasma

Membrane

transmembrane

receptor

JAK2 Janus kinase 2 2.24E-04 4.31E-02 4.5 Up Cytoplasm kinase

NFKB2

nuclear factor kappa B

subunit 2 5.38E-05 2.33E-02 4.8

Up Nucleus

transcription

regulator

RHOQ

ras homolog family member

Q 4.87E-04 6.34E-02 3.2

Up

Plasma

Membrane enzyme

RHOT2

ras homolog family member

T2 1.02E-03 9.22E-02 5.9

Up Cytoplasm enzyme

STAT3

signal transducer and

activator of transcription 3 9.23E-04 8.84E-02 2.2

Up Nucleus

transcription

regulator

Page 75: Edwardsiella Ictaluri Is Capable of Persisting in Channel

66

Figure 3.2: Combined NF-κB virus and P13K/AKT signaling pathway. Molecules predicted to

be upregulated in WT infected HKDM compared to uninfected HKDMs appear in Red/pink. IPA

predicts that integrin and cytokine activation leads to AKT, which activates the COX kinase,

activating the IKK kinase, which in turn activates the NF-κB, IκB complex, ultimately leading to

cell growth and survival.

Page 76: Edwardsiella Ictaluri Is Capable of Persisting in Channel

67

Table 3.3 Top Downregulated Pathways predicted by IPA showing the top predicted genes per pathway, and its corresponding

expression value in WT compared to uninfected HKDMs, the false discovery rate (q- value), and the fold change between the WT and

uninfected HKDMs. IPA also indicated the expected regulation, location, and type of molecule per gene.

Symbol Gene Name p-value q-value

Fold

Change Expected Location Type(s)

Death Cell Receptor Signaling Pathway

BIRC2 baculoviral IAP repeat containing 2

6.48E-

04 7.22E-02 4.3 Down Cytoplasm enzyme

CFLAR

CASP8 and FADD like apoptosis

regulator

2.44E-

05 1.57E-02 14.6 Down Cytoplasm other

MAP3K5

mitogen-activated protein kinase

kinase kinase 5

1.20E-

04 3.17E-02 4.3 Up Cytoplasm kinase

NFKB2 nuclear factor kappa B subunit 2

5.38E-

05 2.33E-02 4.7 Up Nucleus

transcription

regulator

NFKBIA NFKB inhibitor alpha

5.53E-

06 9.52E-03 23.5 Down Cytoplasm

transcription

regulator

NFKBIB NFKB inhibitor beta

1.62E-

05 1.31E-02 3.5 Down Nucleus

transcription

regulator

NFKBIE NFKB inhibitor epsilon

2.61E-

04 4.67E-02 3.5 Down Nucleus

transcription

regulator

TANK

TRAF family member associated

NFKB activator

1.03E-

04 3.02E-02 4.2 Cytoplasm other

CD40 Signaling Pathway

NFKB2 nuclear factor kappa B subunit 2

5.38E-

05 2.33E-02 4.7 Up Nucleus

transcription

regulator

NFKBIA NFKB inhibitor alpha

5.53E-

06 9.52E-03 23.5 Down Cytoplasm

transcription

regulator

NFKBIB NFKB inhibitor beta

1.62E-

05 1.31E-02 3.5 Down Nucleus

transcription

regulator

NFKBIE NFKB inhibitor epsilon

2.61E-

04 4.67E-02 3.5 Down Nucleus

transcription

regulator

(table cont’d)

Page 77: Edwardsiella Ictaluri Is Capable of Persisting in Channel

68

Table 3.3 continued

Symbol Gene Name p-value q-value

Fold

Change Expected Location Type(s)

STAT3

signal transducer and activator of

transcription 3

9.23E-

04 8.84E-02 2.2 Up Nucleus

transcription

regulator

TANK

TRAF family member associated

NFKB activator

1.03E-

04 3.02E-02 4.2 Down Cytoplasm other

TNFAIP3 TNF alpha induced protein 3

6.95E-

06 1.04E-02 9.0 Down Nucleus enzyme

PPAR Signaling Pathway

IL1B interleukin 1 beta

5.31E-

05 2.32E-02 35.3 Down

Extracellula

r Space cytokine

NFKB2 nuclear factor kappa B subunit 2

5.38E-

05 2.33E-02 4.7 Down Nucleus

transcription

regulator

NFKBIA NFKB inhibitor alpha

5.53E-

06 9.52E-03 23.5 Down Cytoplasm

transcription

regulator

NFKBIB NFKB inhibitor beta

1.62E-

05 1.31E-02 3.5 Down Nucleus

transcription

regulator

NFKBIE NFKB inhibitor epsilon

2.61E-

04 4.67E-02 3.5 Down Nucleus

transcription

regulator

SHC1 SHC adaptor protein 1

9.37E-

04 8.91E-02 2.4 Down Cytoplasm other

Page 78: Edwardsiella Ictaluri Is Capable of Persisting in Channel

69

Figure 3.3: Death Receptor Signaling Pathway. Molecules predicted to be upregulated in WT

infected HKDM compared to uninfected HKDM appear in Red/pink. IPA predicts that ultimately

NF-κB complex production leads to the production of the XIAP inhibitory complex preventing

cell death through apoptosis.

transcript is important for macrophage polarization to the M2 phenotype, and the absence of

PPARγ has been shown to cause M1 polarization, so the downregulation of this pathway would

suggest M1 polarization (4). Although some of these pathways can be indicated in both M1 and

M2 response, a recent study looking at differences in M1 and M2 activation as it relates to

classical and alternative activation in mouse bone marrow-derived macrophages supports our

Page 79: Edwardsiella Ictaluri Is Capable of Persisting in Channel

70

findings for the M1/ classical activation phenotype (27). Just as in their study, we saw activation

of the previously mentioned pathways in response to E. ictaluri as well as the TNFR1/2

pathways, IL-1 pathway, IL-6 pathway and PI3K/AKT pathway (27). Conversely, it is important

to point out that some of these pathways have additional indications towards the M2 phenotype

such as the upregulation of the PI3K/AKT pathway (28), and may point to an attempt to maintain

a neutral regulation of the macrophage as these pathways tend to lean more towards an M2

polarization response.

The transcripts belonging to the NF-κB family, NFKB2, and inhibitor family, NFKBIA,

NFKBIB, and NFKBIE, were highly expressed and indicated in most IPA analyses, favoring

both the likelihood of an M1 phenotype, and the potential suppression of apoptosis in favor of

cell survival. Activation of NF-κB family proteins, including NFKB2, is primarily initiated by

the upstream IκB kinase (IKK) complex. This complex, along with the IκB family of inhibitors,

such as NFKBIA, NFKBIB, and NFKBIE, which transcribe 3 of the 5 IκB proteins, allow the

modulation of inflammatory responses due to infection (29-31). A study using channel catfish

mucosal tissues identified 13 NF-κB related genes, and determined that all NF-κB genes existed

as a single copy with the exception of NFKBIA (29). This is of interest because NFKBIA

exhibited the greatest fold change out of the NF-κB family in response to E. ictaluri infection.

Two studies that collected data on catfish mucosal tissue in response to F. columnare

infection (19, 29), found that, in general, NF-κB was induced in all tissues, but different genes

within the family were significantly regulated suggesting variability in NF-κB activation in

various tissue (29). They also noticed this difference in resistant and susceptible catfish (29). The

NF-κB pathway can be initiated by several upstream modulators including LPS activation of

TLRs (32). It is likely that the LPS of E. ictaluri is involved with the downstream activation of

Page 80: Edwardsiella Ictaluri Is Capable of Persisting in Channel

71

the NF-κB pathway, however, the current analysis has not provided an obvious answer. While

TLRs are known to activate the NF-κB pathway, only small fold changes in some TLRs were

observed in the GSA data in the course of this experiment (29). This is consistent with other

studies that have indicated modest activation of TLRs in catfish tissues in response to both E.

ictaluri and F. columnare infections (17-19). Sun et al. suggested that by comparison, more

highly expressed lectins and non-traditional immune factors such as MFAP4 could be more

important for this recognition (19). Further data mining will be required to uncover which

TLR’s, or other PRR’s, may be involved with induced signaling.

Another possible answer this analysis uncovered is the fate of the HKDMs after E.

ictaluri infection. Classically, apoptosis is both a normally occurring event to maintain cell

populations, as well as a defense mechanism employed in response to damaged or diseased cells

(33, 34). However, the strong regulation of NF-κB and downregulation of two other important

pathways in the IPA analysis, the Tec kinase signaling pathway and the death receptor pathway,

indicate that HKDMs are prevented from undergoing apoptosis. In the context of macrophages,

Tec family kinases, whose pathway was upregulated in this study, have been shown to promote

macrophage survival (35). TNFR1 also contains a death domain and is involved in apoptosis,

although this process is typically suppressed by NF-κB induced expression of FLIP and receptor

pathway, and the production of FLIP in the TNFR1 pathway would favor production of NF-κB

(36). Additionally, pathway analysis of the PI3K/AKT and NF-κB pathway indicates that NF-κB

production ultimately leads to cell growth and survival.

Indications from several pathways suggest that HKDM favor autophagy, which is often

involved in routine maintenance and phagocytosis (37, 38). The upregulation of the IL-1

signaling pathway, of which IL-1β is highly expressed, and the iNOS pathway also indicate

Page 81: Edwardsiella Ictaluri Is Capable of Persisting in Channel

72

autophagy is more likely (39). NF-κB is also implicated in these pathways and can regulate

autophagy, and is typically involved with cell survival, proliferation, inflammatory responses,

and angiogenesis by activating several cytokines including IL-6 (39). Previous reports have

shown the ability of E. ictaluri to survive and replicate in ECV within HKDM (1), so while it

possible that the host eventually overcomes bacterial evasion, it seems more likely that E.

ictaluri is unhindered by the autophagic environment of the HKDM, like many other bacteria

(37).

In a related study looking at ECV surface markers, Dubytska and Thune (Submitted)

found that WT E. ictaluri suppresses apoptosis and allows limited recruitment of the autophagy

marker LC3 on WT containing ECVs along with limited phosphorylation of LC3-I to LC3-II.

This further suggests that E. ictaluri may be allowing the HKDM to move towards an autophagic

response but is capable of avoiding degradation by this pathway.

The current analysis also uncovered interesting possibilities regarding the fate of host

signaling in response to bacterial infection. The CD40 pathway in the macrophage is

downregulated in response to E. ictaluri infection. CD40 is a key molecule involved with

activating T cells, so this suggests a limited ability to fully mature and activate T-cells in the

presence of E. ictaluri (40, 41).

In conclusion, it appears that strong regulation of NK-κB family members drive an M1

phenotype in HKDM in response to E. ictaluri infection, and that this also drives the cell to favor

autophagy, possibly allowing the bacteria to survive and replicate longer. In addition,

downregulation of the CD40 pathway could have interesting ramifications on the fate of T cell

dependent immune responses. This observation will be aided by deeper analysis of the current

Page 82: Edwardsiella Ictaluri Is Capable of Persisting in Channel

73

data as well as in vivo experimentation to better elucidate differences in early adaptive cell-

mediated immunity of the host compared to the two infected treatments.

Literature Cited

1. Booth NJ, Elkamel A, Thune RL. 2006. Intracellular replication of Edwardsiella ictaluri

in channel catfish macrophages. J Aquat Anim Health 18:101-108.

2. Santander J, Martin T, Loh A, Pohlenz C, Gatlin III DM, Curtiss III R. 2013.

Mechanisms of intrinsic resistance to antimicrobial peptides of Edwardsiella ictaluri and

its influence on fish gut inflammation and virulence. Microbiology 159:1471.

3. Kordon AO, Abdelhamed H, Ahmed H, Park JY, Karsi A, Pinchuk LM. 2017.

Phagocytic and bactericidal properties of channel catfish peritoneal macrophages exposed

to Edwardsiella ictaluri live attenuated vaccine and wild-type strains. Front Microbiol

8:2638.

4. Yunna C, Mengru H, Lei W, Weidong C. 2020. Macrophage M1/M2 polarization. Eur J

Pharmacol 877:173090.

5. Baumgartner WA, Dubytska L, Rogge ML, Mottram PJ, Thune RL. 2014. Modulation of

vacuolar pH is required for replication of Edwardsiella ictaluri in channel catfish

macrophages. Infect Immun 82:2329-36.

6. Blackmon LE, Quiniou SMA, Wilson M, Bengten E. 2020. Catfish lymphocytes

expressing CC41-reactive leukocyte immune-type receptors (LITRs) proliferate in

response to Edwardsiella ictaluri infection in vitro. Dev Comp Immunol 106:103610.

7. Pohlenz C, Buentello A, Mwangi W, Gatlin DM, 3rd. 2012. Arginine and glutamine

supplementation to culture media improves the performance of various channel catfish

immune cells. Fish Shellfish Immunol 32:762-8.

8. Thune RL, Fernandez DH, Benoit JL, Kelly-Smith M, Rogge ML, Booth NJ, Landry CA,

Bologna RA. 2007. Signature-tagged mutagenesis of Edwardsiella ictaluri identifies

virulence-related genes, including a salmonella pathogenicity island 2 class of type III

secretion systems. Appl Environ Microbiol 73:7934-46.

9. Akgul A, Akgul A, Lawrence ML, Karsi A. 2018. Stress-related genes promote

Edwardsiella ictaluri pathogenesis. PLoS One 13:e0194669.

Page 83: Edwardsiella Ictaluri Is Capable of Persisting in Channel

74

10. Baldwin TJ, Newton JC. 1993. Pathogenesis of Enteric Septicemia of Channel Catfish,

caused by Edwardsiella ictaluri: bacteriologic and light and electron microscopic

findings. J Aquat Anim Health 5:189-198.

11. Booth NJ, Beekman JB, Thune RL. 2009. Edwardsiella ictaluri encodes an acid-

activated urease that is required for intracellular replication in channel catfish (Ictalurus

punctatus) macrophages. Appl Environ Microbiol 75:6712-20.

12. Rogge ML, Thune RL. 2011. Regulation of the Edwardsiella ictaluri type III secretion

system by pH and phosphate concentration through EsrA, EsrB, and EsrC. Appl Environ

Microbiol 77:4293-302.

13. Thune RL, Fernandez DH, Benoit JL, Kelly-Smith M, Rogge ML, Booth NJ, Landry CA,

Bologna RA. 2007. Signature-Tagged Mutagenesis of Edwardsiella ictaluri Identifies

Virulence-Related Genes, Including a Salmonella Pathogenicity Island 2 Class of Type

III Secretion Systems. Applied and Environmental Microbiology 73:7934-7946.

14. Russo R, Shoemaker CA, Panangala VS, Klesius PH. 2009. In vitro and in vivo

interaction of macrophages from vaccinated and non-vaccinated channel catfish

(Ictalurus punctatus) to Edwardsiella ictaluri. Fish Shellfish Immunol 26:543-52.

15. Tan S, Wang W, Zhou T, Yang Y, Gao D, Dunham R, Liu Z. 2019. Polyadenylation sites

and their characteristics in the genome of channel catfish (Ictalurus punctatus) as revealed

by using RNA-Seq data. Comp Biochem Physiol D: Genomics Proteomics 30:248-255.

16. Tan S, Wang W, Tian C, Niu D, Zhou T, Yang Y, Gao D, Liu Z. 2019. Post-

transcriptional regulation through alternative splicing after infection with Flavobacterium

columnare in channel catfish (Ictalurus punctatus). Fish Shellfish Immunol 91:188-193.

17. Peatman E, Terhune J, Baoprasertkul P, Xu P, Nandi S, Wang S, Somridhivej B,

Kucuktas H, Li P, Dunham R, Liu Z. 2008. Microarray analysis of gene expression in the

blue catfish liver reveals early activation of the MHC class I pathway after infection with

Edwardsiella ictaluri. Mol Immunol 45:553-566.

18. Li C, Zhang Y, Wang R, Lu J, Nandi S, Mohanty S, Terhune J, Liu Z, Peatman E. 2012.

RNA-seq analysis of mucosal immune responses reveals signatures of intestinal barrier

disruption and pathogen entry following Edwardsiella ictaluri infection in channel

catfish, Ictalurus punctatus. Fish Shellfish Immunol 32:816-827.

Page 84: Edwardsiella Ictaluri Is Capable of Persisting in Channel

75

19. Sun F, Peatman E, Li C, Liu S, Jiang Y, Zhou Z, Liu Z. 2012. Transcriptomic signatures

of attachment, NF-κB suppression and IFN stimulation in the catfish gill following

columnaris bacterial infection. Dev Comp Immunol 38:169-180.

20. Spencer DA, Quiniou SMA, Crider J, Musungu B, Bengten E, Wilson M. 2019. Insights

into the dynamics of memory, effector and apoptotic cytotoxic T lymphocytes in channel

catfish, Ictalurus punctatus. Dev Comp Immunol 92:116-128.

21. Xu Z, Chen J, Li X, Ge J, Pan J, Xu X. 2013. Identification and characterization of

microRNAs in channel catfish (Ictalurus punctatus) by using Solexa sequencing

technology. PloS One 8:e54174-e54174.

22. Yasunaga N. 1982. Characteristics of the fish pathogen Edwardsiella isolated from

several species of cultured marine fish. Bull Nagasaki Prefect Inst Fish 8:67-73.

23. Liu Z, Liu S, Yao J, Bao L, Zhang J, Li Y, Jiang C, Sun L, Wang R, Zhang Y, Zhou T,

Zeng Q, Fu Q, Gao S, Li N, Koren S, Jiang Y, Zimin A, Xu P, Phillippy AM, Geng X,

Song L, Sun F, Li C, Wang X, Chen A, Jin Y, Yuan Z, Yang Y, Tan S, Peatman E, Lu J,

Qin Z, Dunham R, Li Z, Sonstegard T, Feng J, Danzmann RG, Schroeder S, Scheffler B,

Duke MV, Ballard L, Kucuktas H, Kaltenboeck L, Liu H, Armbruster J, Xie Y, Kirby

ML, Tian Y, Flanagan ME, et al. 2016. The channel catfish genome sequence provides

insights into the evolution of scale formation in teleosts. nat commun 7:11757.

24. Leng N, Dawson JA, Thomson JA, Ruotti V, Rissman AI, Smits BM, Haag JD, Gould

MN, Stewart RM, Kendziorski C. 2013. EBSeq: an empirical Bayes hierarchical model

for inference in RNA-seq experiments. Bioinformatics 29:1035-1043.

25. Bogdan C, Röllinghoff M, Diefenbach A. 2000. The role of nitric oxide in innate

immunity. Immunol Rev 173:17-26.

26. Xue Q, Yan Y, Zhang R, Xiong H. 2018. Regulation of iNOS on immune cells and its

role in diseases. Int J Mol Sci 19:3805.

27. Orecchioni M, Ghosheh Y, Pramod AB, Ley K. 2019. Macrophage polarization: different

gene signatures in M1(LPS+) vs. classically and M2(LPS–) vs. alternatively activated

macrophages. Front Immunol 10.

28. Vergadi E, Ieronymaki E, Lyroni K, Vaporidi K, Tsatsanis C. 2017. Akt signaling

pathway in macrophage activation and M1/M2 polarization. J Immunol 198:1006-1014.

Page 85: Edwardsiella Ictaluri Is Capable of Persisting in Channel

76

29. Wang X, Liu S, Yang Y, Fu Q, Abebe A, Liu Z. 2017. Identification of NF-kappaB

related genes in channel catfish and their expression profiles in mucosal tissues after

columnaris bacterial infection. Dev Comp Immunol 70:27-38.

30. Baldwin Jr AS. 1996. The NF-κB and IκB proteins: new discoveries and insights. Annu

Rev Immunol 14:649-681.

31. Israël A. 2010. The IKK complex, a central regulator of NF-κB activation. Cold Spring

Harb Perspect Med 2:a000158.

32. Liu T, Zhang L, Joo D, Sun S-C. 2017. NF-κB signaling in inflammation. Signal

Transduct Target Ther 2:17023.

33. Ashida H, Mimuro H, Ogawa M, Kobayashi T, Sanada T, Kim M, Sasakawa C. 2011.

Cell death and infection: a double-edged sword for host and pathogen survival. J Cell

Biol 195:931-942.

34. Elmore S. 2007. Apoptosis: a review of programmed cell death. Toxicol Pathol 35:495-

516.

35. Melcher M, Unger B, Schmidt U, Rajantie IA, Alitalo K, Ellmeier W. 2008. Essential

roles for the Tec family kinases Tec and Btk in M-CSF receptor signaling pathways that

regulate macrophage survival. Journal of Immunology 180:8048-8056.

36. Wajant H, Siegmund D. 2019. TNFR1 and TNFR2 in the control of the life and death

balance of macrophages. Front Cell Dev Biol 7:91-91.

37. Kimmey JM, Stallings CL. 2016. Bacterial pathogens versus autophagy: Implications for

therapeutic interventions. Trends Mol Med 22:1060-1076.

38. Mariño G, Niso-Santano M, Baehrecke EH, Kroemer G. 2014. Self-consumption: the

interplay of autophagy and apoptosis. Nat Rev Mol Cell Biol 15:81-94.

39. Wu M-Y, Lu J-H. 2020. Autophagy and macrophage functions: inflammatory response

and phagocytosis. Cells 9:70.

40. Mackey MF, Barth Jr RJ, Noelle RJ. 1998. The role of CD40/CD154 interactions in the

priming, differentiation, and effector function of helper and cytotoxic T cells. J

Leukocyte Biol 63:418-428.

Page 86: Edwardsiella Ictaluri Is Capable of Persisting in Channel

77

41. Lagos LX, Iliev DB, Helland R, Rosemblatt M, Jorgensen JB. 2012. CD40L--a

costimulatory molecule involved in the maturation of antigen presenting cells in Atlantic

salmon (Salmo salar). Dev Comp Immunol 38:416-30.

Page 87: Edwardsiella Ictaluri Is Capable of Persisting in Channel

78

CHAPTER 4. EDWARDSIALLA ICTALURI EVADES T CELL RESPONSES

IN THE CHANNEL CATFISH

Introduction

Edwardsiella ictaluri is a gram-negative facultative intracellular bacterium and the

causative agent of enteric septicemia in the channel catfish (ESC). Reports indicate that ESC

infects several fish species, including tilapia, zebrafish, barbs, and other catfish species (1), but is

of economic significance in the catfish industry, where reported outbreaks on 19.3% of

fry/fingerling operations, 40.4% of fingerling operations, and 36.6% of operations producing

food size fish (2). In the catfish host, ESC mainly affects the intestine, but can also be isolated

from the head kidney, brain, and liver, and is characterized by internal and external physical and

physiologic changes, including ulcerations on the skin and swelling of the abdomen (1).

Previous research indicated that E. ictaluri persists in catfish due to an ability to evade

and manipulate the immune response, but little research has been conducted on the function of

CMI responses in naïve fish. In mammals, the immune system is extremely complex,

encompassing many different cell types and regulatory components. Antigen presentation to T

cells is an important cell-mediated linkage between the innate and adaptive arms of immunity

(3). One example of this is antigen presentation by antigen-presenting cells (APCs) using the

Major Histocompatibility Complex class II (MHC class II) carrying foreign peptides, which are

recognized by CD4 T cells (3). Antigen presentation is often considered a major initiator of the

adaptive immune response and is a function of APCs, such as dendritic cells, macrophages, and

B cells (4). Our understanding of fish immunity has increased over the years and revealed that

the teleost immune system is highly similar to mammals, with innate, adaptive, humoral, and

cell-mediated components (5). Teleost immune systems include many of the same white blood

Page 88: Edwardsiella Ictaluri Is Capable of Persisting in Channel

79

cell types as mammals, such as macrophages, neutrophils, dendritic cells, B cells, CD8 T cells,

and CD4 T cells (6-12). Teleosts also have a vast array of cytokines, and many carry out the

same or similar functions to their mammalian counterparts (13-15).

Recent work showed that the E. ictaluri T3SS effector EseK binds to the hosts CD74

protein, (see chapter 2) (16). This is of potential interest because CD74 plays several important

roles in immune related pathways, such as binding to MHC class II and preventing endogenous

protein binding while allowing MHC to assemble, acting as the binding ligand for macrophage

migration inhibitory factor (MIF) and aiding in cross-presentation to MHC class I (17-19).

Therefore, this experiment hoped to address the actual response of naïve catfish to E. ictaluri

infection using both the WT and the EseK null strain, ΔeseK.

The nCounter® Analysis system boasts a highly sensitive, accurate, reproducible

platform that introduces less bias from amplification steps required in RNA sequencing and

qRT-PCR counterpart technologies (20-22). Instead, probe pairs consisting of a Reporter probe

and a Capture probe containing barcode sequences unique to each target of interest are directly

hybridized to mRNA, which also allows multiple targets per sample. In a study by Bouzas et al.,

additional positive respiratory syncytial virus (RSV) samples were detected by this platform that

were not detected by RT-PCR methods, a result that the researchers believed to be genuine (20).

Because the nCounter® system hybridizes a probe directly to mRNA, it increases accuracy by

removing bias associated with cDNA conversion (22). A study by Chen et al. using formalin-

fixed paraffin-embedded (FFPE) samples found that technical replicates were highly

reproducible, with a correlation of 0.983. This platform uses low amounts of total RNA (21, 22)

and can accurately detect mRNA expression, even in highly degraded RNA samples (23). Lastly,

this system occupies a niche between conventional RNA-seq and qRT-PCR methods where up to

Page 89: Edwardsiella Ictaluri Is Capable of Persisting in Channel

80

800 probes can be targeted to each RNA sample, using a multitude of samples per experiment

(24).

In addition, flow cytometry allowed for identification of T cell populations. Flow

cytometry is a useful tool for determining individual cell populations from a heterogenous

population for downstream assays. While a limited number of antibodies specific to channel

catfish proteins exist, we were unable to procure one specific to all T cells at the time of this

experiment. Therefore, a commercially available CD3ε T cell marker was used that preliminary

experiments showed was effective for targeting catfish CD3ε.

The goal of these experiments is to better characterize the role of the WT in the host

immune response and to determine the potential involvement of EseK in the host immune

response. A combination of transcriptional analysis with nCounter® and flow cytometry were

used to determine that E. ictaluri suppresses certain T cell immune responses, which likely

allows it to persist in the host.

Methods

Bacterial Strains. Bacterial strains used in this study were wild-type (WT) Edwardsiella

ictaluri LADL 93-146, isolated in 1993 from moribund channel catfish in a natural outbreak of

ESC on a commercial farm, and ΔeseK, a mutated strain of E. ictaluri 93-146 containing a

deletion of the first 2200 bp of EseK. Strains were grown from -80C frozen back cultures in 5mL

BHI-porcine (Sigma-Aldrich) overnight (O/N) at 28C with aeration. These starter cultures were

used for downstream assays.

Fish Care. Channel catfish eggs obtained from a Morehead Mississippi catfish farm with

no history of ESC outbreaks were disinfected with 100 ppm free iodine and hatched in a closed

recirculating system in the specific pathogen-free (SPF) laboratory at the LSU School of

Page 90: Edwardsiella Ictaluri Is Capable of Persisting in Channel

81

Veterinary Medicine. Fish were reared in systems containing four, 350 L fiberglass tanks,

connected to a 45 L bead filter (Aquaculture Systems Technologies). Water quality was tested at

least three times a week, and the temperature was constantly monitored to ensure optimal

conditions for fish wellness. Fish weighing approximately 40g were moved to challenge tanks at

25 fish per 20 L flow-through tank. Catfish were fed 2% body weight every other day and

allowed to acclimate 28 days before inoculation (25). All experiments were carried out following

protocols approved by the Institutional Animal Care and Use Committee and the Inter-

Institutional Biological and Recombinant DNA Safety Committee of Louisiana State University.

Immersion Assays. Starter cultures of WT and the ΔeseK strain were used to inoculate

400 mL of porcine-BHI per 2 L flask at 28C, and cultures were allowed to incubate until each

culture reached an OD600 1.9, reflecting a concentration of 1 X1010 colony-forming unit’s (CFU)

per mL. The culture was then diluted as required per treatment dose for the inoculation of catfish.

The bacterial concentration was subsequently verified by drop plating using ten-fold serial

dilutions in triplicate on Trypticase Soy Agar supplemented with 5% defibrinated sheep blood

(BA) (26).

Fish Sampling. Fish were anesthetized in 100 mg/L Tricaine S (Pentair). Blood was

collected by caudal vein puncture into K3 EDTA tubes (Becton, Dickinson and Company) and

processed for peripheral blood leukocytes (PBLs). Fish were then euthanized in 1 g/L Tricaine S

for tissue collection. One fish per tank was collected per day.

PBL Collection. PBLs were collected by gradient centrifugation as modified from Miller

et al. (26). Briefly, blood was mixed equal parts with modified PBS (mPBS) consisting of 9

parts 1XPBS:1part H20, with 2% heat inactivated, filter-sterilized channel catfish serum. The

mixture was layered over a Lymphoprep (Stem Cell Technologies) density gradient with a

Page 91: Edwardsiella Ictaluri Is Capable of Persisting in Channel

82

specific density of 1.077 g/mL. Centrifugation was carried out at 800x g (accuSpin™ 3R, Fisher

Scientific) for 30 minutes at room temperature. The buffy layer was then removed and washed

twice in mPBS followed by centrifugation at 800x g for 8 minutes.

For nCounter experiments, flash lysis was used following the final wash step to remove residual

red blood cells. PBLs were centrifuged at 800x g for 1 min in distilled water (28). The

supernatant was removed, and cells were immediately placed in RNAzol for RNA processing.

Tissue Collection. Head Kidney (HK) tissue was aseptically removed from sampled

catfish. The HK was dissected into 3 portions and weighed for the nCounter® experiment. One

portion was used to determine CFU per gram of tissue, one portion was used for RNA extraction,

and one portion was flash-frozen using liquid nitrogen and stored at -80C. For flow cytometry,

HK was weighed and used for CFU counts.

CFU counts. Bacterial counts were quantified per each HK sample by homogenizing

weighed HK tissue in sterile saline using a bead beater (Bullet Blender 24, Next Advance Inc,) at

a speed setting of 8 for 4 minutes. Ten-fold serial dilutions were prepared of each homogenate,

plated in triplicate onto BA plates, and grown at 28C for 48 hours. Colony counts per gram of

tissue were recorded.

RNA Extractions. Total RNA extractions were carried out on HK and PBL samples

using RNAzol® RT Isolation Reagent (Molecular Research Center) in conjunction with the RNA

Clean and Concentrator-5 Kit (Zymo Research) following manufacturer protocols. Samples were

re-suspended in DEPC-treated H20 (Ambion™). The HK tissue had an additional initial step of

homogenization, which was carried out by adding HK to 1 mL of RNAzol RT and homogenizing

on a bead beater (Bullet Blender 24, Next Advance Inc,) at a speed setting of 5 for 2 minutes. All

RNA samples were quantified on a NanoDrop (Thermo Fisher Scientific), and the 260/280 value

Page 92: Edwardsiella Ictaluri Is Capable of Persisting in Channel

83

was taken to determine that samples had a value of approximately 1.9 or greater to establish the

purity of samples. Additionally, a random selection of RNA samples were tested for RNA

quality using the Agilent Fragment Analyzer (Advanced Technologies, Inc).

Probe Creation. Total RNA of each sample was used for hybridization of catfish mRNA

to probes (Appendix 2, Table 1) created as a custom codeset (29), designed by NanoString

Technologies. nCounter® analysis was carried out at the LSU Health Science Center, New

Orleans, LA. Briefly, 15 ng of each sample was mixed with each probe as well as eight positive

spike-in controls, and six negative spike-in controls.

Flow Cytometry. For flow cytometry, PBLs were counted on a Countess automated cell

counter (Invitrogen) and approximately 1 X 106 PBL cells were isolated for use. PBLs were

blocked with Fc Blocker (anti CD16/CD32), fixed with 4% formaldehyde, and permeabilized

with 0.5% saponin. Staining was performed using 2 µg human anti-CD3ε (Novus), with 1:200

anti-human conjugated FITC. Cells were analyzed on a FACSCalibur flow-cytometer (Becton-

Dickinson,) and gating was performed using forward and side scatter for separation of size and

internal granularity (Figure 4.1). A gate was then set over lymphocyte populations from which

the percentage of CD3+ cells were identified.

Analysis and Statistics. Raw nCounter data was analyzed using nSolver Analysis

Software 4.0.70 (NanoString Technologies). Positive spike-in controls were used to correct

technical variation, and negative spike-in controls were used to correct for background variation.

Codeset content normalization was carried out using three fish housekeeping genes (B2M,

EEF1A, and ACTB). Seven of the 192 samples were excluded. Three samples were excluded

from analysis because there were no colonies recovered from BA plates (D2-T7, D6-T7, and D7-

Page 93: Edwardsiella Ictaluri Is Capable of Persisting in Channel

84

T9). One sample, out of 192, failed to run correctly before analysis (D6-T12H). Additionally,

samples that

were flagged by the software were removed (D2-T12P, D5-T5P, and D5-T10P). After sample

exclusion, at least 3 samples per treatment group per day for analysis remained. Fish infected

with WT, ΔeseK, or BHI-Sham were sampled at time 0 and for 7 days following infection, and

gene function was analyzed in HK tissue and PBLs. Using PRISM, GraphPad version 5.00.288,

a two-way ANOVA was performed with Tukey’s posthoc test and confidence interval of 95% to

determine significance among treatments per day. Flow cytometry data were gated and analyzed

using FlowJo™ Software version 10.6.1 (Becton, Dickinson and Company). Using PRISM,

GraphPad version 5.00.288, a two-way ANOVA was performed with Tukey’s posthoc test and

confidence interval of 95% to determine significance among gate types and treatments per day.

Figure 4.1: Representation of gating strategy with Day 0 samples. Using FlowJo, gates were

drawn over lymphoid and myeloid/precursor populations using FSC versus SSC. Lymphoid

populations were then analyzed for CD3ε+ cells based on FITC fluorescence. Percentage of

CD3ε + cells were calculated based on comparison to secondary antibody control per each day

(as seen above).

Page 94: Edwardsiella Ictaluri Is Capable of Persisting in Channel

85

Results

Because WT and ΔeseK did not show significant differences between one another during

the course of this experiment, the focus will be on the WT versus the uninfected catfish. Graphs

still include the ΔeseK treatment for reference.

Head Kidney: CD74/MIF (Figure 4.2). CD74 was downregulated compared to the

Sham from days 2 through 7 in HK samples treated with WT. The inverse of this pattern was

observed in MIF expression of HK samples treated with WT, where upregulation was observed

from days 2 through 7.

Figure 4.2: CD74 and MIF expression in HK per treatment graphed by the log2 transformation of

normalized RNA counts per day. Letters denote significant differences among treatments within

each day, identified by two-way ANOVA with a CI of 95%.

T Cell Markers (Figure 4.3). In HK samples treated with WT, CD3 was downregulated

compared to the Sham on day 7. This same trend of latent downregulation was observed in

CD4_1 expression, where HK samples treated with WT were downregulated compared to the

Sham on day 7. More significantly, CD4_2 expression in HK samples treated with WT was

downregulated on days 4 through 7.

Page 95: Edwardsiella Ictaluri Is Capable of Persisting in Channel

86

CD8α_1 followed a similar pattern to CD3 and CD4, where downregulation was

observed on days 6 and 7 in WT treated HK samples compared to the Sham. However, CD8α_2

was downregulated on day 7 in WT treated HK samples compared to the Sham.

CD40L followed a similar pattern to the other T cell markers, where downregulation was

observed on days 6 and 7 in both WT treated HK samples compared to the Sham.

Cytokine Indicators (Figure 4.4). IL-8_2 was upregulated in HK samples infected with

WT compared to Sham on days 2 through 5, but no difference in IL-8_1 expression was

observed between treatments. IL-8_3 did not amplify.

In HK samples treated with WT, IFN-γ was upregulated compared to the Sham on days 2

and 5, and TGF-β1 was downregulated on days 2 and 5 in WT treated HK samples compared to

the Sham.

Similarly, IL-1β was upregulated in HK samples infected with WT compared to Sham on

days 2 and 3. In addition, IL-6 was upregulated in HK samples infected with WT compared to

Sham on day 2. No differences in the expression of IL-10, TNF-α, or IL-17A were observed

between WT and Sham.

Page 96: Edwardsiella Ictaluri Is Capable of Persisting in Channel

87

Figure 4.3: Expression of T cell Markers in HK per treatment graphed by the log2 transformation of normalized RNA counts per day.

Letters denote significant differences among treatments within each day, identified by two-way ANOVA with a CI of 95%.

Page 97: Edwardsiella Ictaluri Is Capable of Persisting in Channel

88

Figure 4.4: Expression of other cytokine indicators in HK per treatment graphed by the log2 transformation of normalized RNA

counts per day. Letters denote significant differences among treatments within each day, identified by two-way ANOVA with a CI of

95%.

Page 98: Edwardsiella Ictaluri Is Capable of Persisting in Channel

89

PBL: CD74/MIF (Figure 4.5). CD74 was downregulated compared to the Sham on days

2, and 5 through 7 in PBL samples treated with WT. The inverse of this pattern was observed in

MIF expression of PBL samples treated with WT, where upregulation was observed from days 3

through 7

Figure 4.5: CD74 and MIF expression in PBL per treatment graphed by the log2 transformation

of normalized RNA counts per day. Letters denote significant differences among treatments

within each day, identified by two-way ANOVA with a CI of 95%.

T-Cell Markers (Figure 4.6). In PBL samples, no difference in CD3 expression was

observed between among any treatment. In contrast to HK expression, no difference in CD4_2

expression was observed between treatments. CD4_1 did not amplify.

However, CD8α_1 upregulation was observed on day 5 in WT treated PBL samples

compared to the Sham. Similarly, CD8α_1 was upregulated on day 6 in WT treated HK samples

compared to the Sham. Following the pattern observed in HK, CD40L expression was

downregulation on days 0, and 5 through 7 in WT treated PBL samples compared to the Sham.

Page 99: Edwardsiella Ictaluri Is Capable of Persisting in Channel

90

Figure 4.6: Expression of T cell markers in PBL per treatment graphed by the log2 transformation of normalized RNA counts per day.

Letters denote significant differences among treatments within each day, identified by two-way ANOVA with a CI of 95%.

Page 100: Edwardsiella Ictaluri Is Capable of Persisting in Channel

91

Cytokine Indicators (Figure 4.7). No difference was observed in IL-8_1 expression

between WT and Sham. Following the same regulation patterns in HK, IL-8_2 was upregulated

in PBL samples infected with WT compared to Sham on day 7.

TGF-β1 downregulation was observed on days 6 and 7 in WT infected PBL samples

compared to the Sham. IL-10 expression in PBL samples treated with WT, were upregulated

compared to the Sham on days 2 and 7. IL-17A expression in PBL samples treated with WT,

were upregulated compared to the Sham on days 2 and 5. TNF-α expression in PBL samples

treated with WT, were downregulated compared to the Sham on day 4.

No difference in the expression of IL-6 or IFN-γ was observed between WT and Sham in

PBL samples.

Flow Cytometry. CD3ε detection in PBL populations from WT, ΔeseK, and uninfected

fish did not show any statistically significant changes in population number among treatments

per day, suggesting that overall T cell populations are not proliferating (Figure 4.8, Table 4.2).

Discussion

This work characterized cell-mediated responses to WT infection. We examined

components of the catfish immune system to gain insight into naïve catfish immune responses

generated to E. ictaluri infection, with a special interest in cell mediated responses. The

secondary goal focused on determining if these responses were specific to hematopoietic tissue

such as the HK, or if they were also present as part of a systemic response. The results showed

that CD74 and CD40L were significantly downregulated across multiple days in all bacterial

samples in all tissues compared to the Sham. Similarly, MIF was upregulated in all infected

tissue compared to the Sham, although this regulation was less pronounced in ΔeseK infected

PBL tissue. In addition, expression of some genes were robust but only present in one

Page 101: Edwardsiella Ictaluri Is Capable of Persisting in Channel

92

Figure 4.7: Expression of other cytokine indicators in PBL per treatment graphed by the log2 transformation of normalized RNA

counts per day. Letters denote significant differences among treatments within each day, identified by two-way ANOVA with a CI of

95%.

Page 102: Edwardsiella Ictaluri Is Capable of Persisting in Channel

93

Figure 4.8: Percentage of CD3e+ lymphocytes grouped by treatment with SEM. Percentages

were calculated by gating CD3 populations based on F1 scatter and subtracted from respective

secondary per respective day.

tissue type, such as a decrease in the expression of T cell related transcripts and IL-8 in HK.

Likewise, TGF-β1 also exhibited strong downregulation in PBL samples due to infection. One

of the targets that we observed, CD74, was consistently downregulated in the bacterial strains in

both sample sets. It is interesting that EseK does not heavily affect the downregulation of CD74

compared to the WT, even though it was shown to bind with CD74. It is likely that the effect of

EseK on CD74 is at the level of the protein and this will require further testing. CD74

downregulation in the presence of E. ictaluri could infer several roles in bacterial regulation of

the cell, because CD74 is involved in numerous functions in the cell, especially APCs (30).

Some well-documented functions include aiding assembly, as well as the timing of peptide

Treatment Tank Day 0 Day 1 Day 2 Day 3 Day 4 Day 5 Day 6 Day 7 Day 8 Day 9 Day 10

Uninf. t34 59.3 23.4 27.0 57.3 47.9 57.9 69.0 63.7 67.2 83.6 28.5

Uninf. t37 44.2 14.8 32.6 48.2 58.2 58.8 63.9 58.3 49.9 78.3 33.6

Uninf. t40 31.2 16.4 23.1 49.2 34.3 41.5 64.4 58.9 47.2 68.4 32.6

WT t36 68.6 12.8 48.2 44.0 26.2 53.6 65.4 48.7 71.0 71.9 50.4

WT t42 40.1 15.5 32.6 38.3 66.3 53.5 62.2 49.4 46.2 73.0 41.0

WT t45 51.1 11.4 24.0 25.3 42.8 48.3 52.7 61.3 33.4 47.5 37.9

ΔeseK t35 85.4 21.4 25.0 54.5 53.9 62.1 69.2 68.4 60.4 52.2 27.9

ΔeseK t39 44.2 25.4 22.6 48.7 35.6 72.0 50.9 45.6 37.4 50.7 20.6

ΔeseK t44 24.3 18.7 16.7 57.9 34.7 57.5 66.4 81.2 28.9 77.7 55.1

0 1 2 3 4 5 6 7 8 9 10

0

20

40

60

80

100

CD3+ Lymphocytes

Day

Perc

en

t

Uninfected

ΔeseK

WT

Page 103: Edwardsiella Ictaluri Is Capable of Persisting in Channel

94

binding to MHC class II, acting as a binding ligand for MIF, and cross-presentation to MHC

Class I (31). Any one of these functions could have a massive impact on the ability of APC’s to

mount a proper cell-mediated adaptive immune response, ranging from lack of MHC class II

assembly, MIF binding, or cross-presentation to MHC class I. Unfortunately, our present study

did not classify the regulation of MHC class II, so we can only speculate as to its regulation.

Beyond the major role in downregulation of T cell-mediated responses, CD74 is implied

in other roles through varying levels of experimentation in mammalian species: potentially

modifying the proteolytic activity of endosomes, thereby preserving epitopes for antigen

presentation (32); carrying out regulatory functions affecting B cells and dendritic cells in mice,

using the cytosolic of the protein (33, 34); utilizing that same domain to facilitate endosomal

fusions, thereby accelerating the speed at which antigens are trafficked through the endocytic

pathway (32, 35). Furthermore, CD74 may also be involved in the migration of dendritic cells

and B cells through an interaction with myosin II (4). CD74 was reported in soluble forms in

humans able to form complexes with circulating MIF (36). If any of these things are also able to

occur in catfish, it could explain some of the expression results we found.

In this study, MIF accumulation was observed in the bacterial strains compared to the

Sham in both sample sets, although it was not as robust in PBL samples. As stated, CD74 acts as

a binding ligand for MIF and could form complexes with MIF in circulation (PBL samples). As a

consequence, this interaction could be suppressing MIF’s ability to enhance the pro-

inflammatory effect of cytokines, such as TNF-α (37). Finally, CD40L is typically expressed on

CD4+ T cells in mammals, and is an indicator of T cell activation (8). Studies conducted in

salmon and zebrafish with CD40L suggest the conservation of CD40-CD40L roles in fish and

mammals (38). CD40L also allows for the upregulation of cytokines, such as IL-1β, IL-6, IL-8,

Page 104: Edwardsiella Ictaluri Is Capable of Persisting in Channel

95

IL-12, TNF-α, and IL-10, as well as type I IFNs (8). CD40L interaction also drives subset

differentiation towards a Th1 phenotype (39, 40). A similar role in the catfish could suggest that

loss of CD40L after E. ictaluri infection could indicate loss in activation and subset

differentiation of CD4 T cells and a reduced antibody response (8).

Head Kidney. The three previously mentioned T cell markers CD3, CD4 and CD8α, as

well as IL-8_2, showed significant differences in regulation for several consecutive days or for

day 7 in HK samples in response to E. ictaluri. Several other cytokines, such as IFNγ, IL-1β and

TGF-β1, also showed a difference in regulation, but these were all isolated to one or two non-

consecutive days, with the exception of IL-1β, which spiked on day 2 and 3 in both strains.

As with mammals, CD3 is present on all fish T cells, and we observed a downregulation

of CD3 expression in HK tissue on day 7 (41). This indicated that E. ictaluri suppresses T cell

populations towards the later days of infection in the HK. Downregulation of CD4 and CD8α in

HK samples in the latter days of this experiment gives additional support to the response

observed with CD3, but also implies a suppression of Th subset and CTL responses. CD4 and

CD8 also behave similarly in teleosts as their mammalian counterparts (42-47). However, one

catfish study suggests that catfish may have CD4+ CTLs in addition to CD8 + CTLs (48), and if

this is the case then the downregulation we observed in CD4 could suggest an additional role in

expression of CTL populations. This knowledge may explain the differences in expression seen

in the two CD4 variants. A study looking at CD4 and CD8 in the HK found an upregulation in

these two genes, an observation in contrast to this study. Differences in experimental design,

such as the length of time fish were allowed to acclimate before infection may explain the

discrepancies (49).

Page 105: Edwardsiella Ictaluri Is Capable of Persisting in Channel

96

While the responses in T cells and CMI pathways are most interesting, we also saw

upregulation in IL-8 midway in this experiment, which was also observed in other studies (49,

50). The structure of catfish IL-8 likely allows attraction and recruitment of neutrophils, as in

mammals (13, 51). This would suggest that neutrophils are recruited to the HK shortly after

infection.

Additional indicators for the type of response seen in HK may be elucidated using IFN-γ,

IL-1β, and TGF-β1, which also exhibited changes in expression on more than one day, although

TGF-β1 was highly significant in PBL samples. Previous reports indicate an upregulation in

IFN-γ in response to E. ictaluri, a result also seen in this experiment (49). IFN-γ aids in an

antiviral state and drives CD4 T cells towards a cell-mediated Th1 response (52, 53). IFN-γ in

teleosts also acts on monocytes and macrophages to increase the production of nitric oxide and

reactive oxygen intermediates (ROI), as well as phagocyte activity in several species. Studies

show IFN-γ is capable of inducing expression of more than 2 dozen genes, which include STAT1

and TLR3 in catfish, and MHC class II β chain and respiratory burst in trout (52-54). IL-1β was

also significantly upregulated in WT compared to Sham in HK, as seen in other studies (49, 55).

Studies of these cytokines showed an ability to increase the expression of bactericidal and

phagocytic effects of HK leukocytes (51, 56, 57).

Our data shows that the upregulation of IFN-γ and IL-1β is not continuous in infected HK

samples. This suggests that there is a tendency to lean towards CMI responses in response to

infection of an intracellular pathogen, as expected. It appears that E. ictaluri keeps these

responses in check, possibly due to reduced activation of T cell related pathways by APC’s.

Peripheral Blood Leukocytes. In the PBL samples, the robust changes in T cell markers

were not as apparent as in the HK, but CD74, CD40L, and MIF followed the same trends.

Page 106: Edwardsiella Ictaluri Is Capable of Persisting in Channel

97

Additionally TGF-β1 was downregulated. Pointed differences in regulation were also observed

in several genes, particularly IL-10 and IL-17. No change in CD3 transcripts or cell number was

observed in PBL samples, suggesting that T cell abundance is not affected in PBL samples

during the first several days of infection. Our flow cytometry data did not detect any changes

between CD3ε among treatments in PBL samples. This is in general agreement with the

nCounter® data, but it is important to note that the CD3ε antibody used in the flow cytometry

assay did not show the same shift in positive CD3ε populations for most of the experimental

samples as observed in the preliminary optimization experiments, leading to questions of this

particular antibodies binding efficiency to catfish CD3ε. This observation in overall T cell

populations may be supported by a recent study that found that CTL populations do not appear to

proliferate in the presence of E. ictaluri infected HKDM (58).

Consistent downregulation of TGF-β1 occurred in PBL samples. This occurrence in the

center of the experiment overlaps with other cytokines, such as IL-10 and IL-17, and could be

indicative of an attempt to regulate T cell leaning. TGF-β1 reduces cytotoxic responses and

acute phase responses of TNF-α induction of IL-1Β, IL-6 and IL-8 in fish (59, 60). TGF-β1

provides an interesting paradigm because, on its own, it suppresses immune responses secreted

by regulatory T cells, but in combination with IL-6, it allows Th17 differentiation (15). Because

TGF-β1 is so heavily downregulated in PBL samples, and IFN-γ is not differentially regulated,

this indicates that E. ictaluri is suppressing most Th subsets including CMI and T regulatory

responses. However, IL-10 and IL-17 were upregulated on approximately day 2 and day 5. IL-10

functions similarly in fish as in mammalian counterparts by suppressing inflammatory responses

(15). IL-17 is important in several tissues of catfish infected with E. ictaluri, such as gill, skin,

and intestine (61). The results seen with IL-10 and IL-17 could indicate an attempt to produce a

Page 107: Edwardsiella Ictaluri Is Capable of Persisting in Channel

98

Th17 or Th2 subset response, but the strong downregulation of TGF-β1 and CD40L gives

credence to the idea that all T cell responses are being suppressed and that these minor

upregulations are knocked down by E. ictaluri in an attempt to control T cell responses.

The data suggests an influence of E. ictaluri on catfish to control T cell and acute phase

responses. This influence is seen in the HK as a reduction in T cell related markers likely leading

to a decrease in T cell activation and response, and also in PBL samples by a reduction in TGF-

β1 and general stabilization in cytokines known to play a part in differentiation and productivity

of other T cell subsets. This is further evidenced by the suppression of CD74, vital to initiation of

immune responses on APC and the reduction of CD40L, a marker of T cell activation. Taken

together this suggests the ability of E. ictaluri to persist in naïve catfish by suppressing CMI

responses.

Literature Cited

1. J. P. Hawke RMD, R. L. Thune, A, C. Camus. 1998. ESC- Enteric Septecemia of Catfish.

2. USDA. 2010. USDA Catfish 2010: Part III: Changes in Catfish Health and Production

Practices in the United States, 2002-2009.

3. Abul K. Abbas AHHL, Shic Pillai. 2012. Basic Immunology: Functions and Disorders of

the Immune System, 4 ed.

4. Schroder B. 2016. The multifaceted roles of the invariant chain CD74-more than just a

chaperone. Biochim Biophys Acta 1863:1269-81.

5. Uribe C, Folch H, Enríquez R, Moran G. 2011. Innate and adaptive immunity in teleost

fish: a review. Veterinarni Medicina 56:486-503.

6. Edholm ES, Bengten E, Stafford JL, Sahoo M, Taylor EB, Miller NW, Wilson M. 2010.

Identification of two IgD+ B cell populations in channel catfish, Ictalurus punctatus. J

Immunol 185:4082-94.

Page 108: Edwardsiella Ictaluri Is Capable of Persisting in Channel

99

7. Iliev DB, Roach JC, Mackenzie S, Planas JV, Goetz FW. 2005. Endotoxin recognition: in

fish or not in fish? FEBS Lett 579:6519-28.

8. Lagos LX, Iliev DB, Helland R, Rosemblatt M, Jorgensen JB. 2012. CD40L--a

costimulatory molecule involved in the maturation of antigen presenting cells in Atlantic

salmon (Salmo salar). Dev Comp Immunol 38:416-30.

9. Moore LJ, Dijkstra JM, Koppang EO, Hordvik I. 2009. CD4 homologues in Atlantic

salmon. Fish Shellfish Immunol 26:10-8.

10. Takizawa F, Koppang EO, Ohtani M, Nakanishi T, Hashimoto K, Fischer U, Dijkstra JM.

2011. Constitutive high expression of interleukin-4/13A and GATA-3 in gill and skin of

salmonid fishes suggests that these tissues form Th2-skewed immune environments. Mol

Immunol 48:1360-8.

11. Zou J, Secombes CJ. 2011. Teleost fish interferons and their role in immunity. Dev Comp

Immunol 35:1376-87.

12. Gudding R, Lillehaug A, Evensen Ø. 2014. Fish vaccination. Chichester, West Sussex ;

Hoboken, NJ : Wiley Blackwell, 2014.

13. Zaccone G. 2009. Fish Defenses Volume 1: Immunology, vol 1. Science Publishers.

14. Bird S, Tafalla C. 2015. Teleost chemokines and their receptors. Biology (basel) 4:756-

84.

15. Zou J, Secombes CJ. 2016. The function of fish cytokines. Biology 5.

16. Dubytska LP, Rogge ML, Thune RL. 2016. Identification and Characterization of

Putative Translocated Effector Proteins of the Edwardsiella ictaluri Type III Secretion

System. mSphere 1.

17. Beswick EJ, Reyes VE. 2009. CD74 in antigen presentation, inflammation, and cancers

of the gastrointestinal tract. J Gastroenterol 15:2855-61.

18. Lapaque N, Hutchinson JL, Jones DC, Meresse S, Holden DW, Trowsdale J, Kelly AP.

2009. Salmonella regulates polyubiquitination and surface expression of MHC class II

antigens. Proceedings of the National Academy of Sciences of the United States of

America 106:14052-7.

Page 109: Edwardsiella Ictaluri Is Capable of Persisting in Channel

100

19. Basha G, Omilusik K, Chavez-Steenbock A, Reinicke AT, Lack N, Choi KB, Jefferies

WA. 2012. A CD74-dependent MHC class I endolysosomal cross-presentation pathway.

Nat Immunol 13:237.

20. Bouzas ML, Oliveira JR, Queiroz A, Fukutani KF, Barral A, Rector A, Wollants E,

Keyaerts E, Van der Gucht W, Van Ranst M, Beuselinck K, de Oliveira CI, Van

Weyenbergh J, Nascimento-Carvalho CM. 2018. Diagnostic accuracy of digital RNA

quantification versus real-time PCR for the detection of respiratory syncytial virus in

nasopharyngeal aspirates from children with acute respiratory infection. J Clin Virol

106:34-40.

21. Chen X, Deane NG, Lewis KB, Li J, Zhu J, Washington MK, Beauchamp RD. 2016.

Comparison of nanostring nCounter® data on FFPE colon cancer samples and affymetrix

microarray data on matched frozen tissues. PLoS One 11:e0153784-e0153784.

22. Geiss GK, Bumgarner RE, Birditt B, Dahl T, Dowidar N, Dunaway DL, Fell HP, Ferree

S, George RD, Grogan T, James JJ, Maysuria M, Mitton JD, Oliveri P, Osborn JL, Peng

T, Ratcliffe AL, Webster PJ, Davidson EH, Hood L, Dimitrov K. 2008. Direct

multiplexed measurement of gene expression with color-coded probe pairs. Nat

Biotechnol 26:317-325.

23. Reis PP, Waldron L, Goswami RS, Xu W, Xuan Y, Perez-Ordonez B, Gullane P, Irish J,

Jurisica I, Kamel-Reid S. 2011. mRNA transcript quantification in archival samples using

multiplexed, color-coded probes. BMC Biotechnol 11:46.

24. Speranza E, Altamura LA, Kulcsar K, Bixler SL, Rossi CA, Schoepp RJ, Nagle E,

Aguilar W, Douglas CE, Delp KL, Minogue TD, Palacios G, Goff AJ, Connor JH. 2017.

Comparison of transcriptomic platforms for analysis of whole blood from Ebola-infected

Cynomolgus Macaques. Sci Rep 7:14756.

25. Rogge M. 2009. Expression of the Edwardsiella ictaluri type III secretion system and its

relationship to the native plasmids. Doctor of Philosophy. Louisiana State University and

Agricultural and Mechanical College

26. Booth NJ, Beekman JB, Thune RL. 2009. Edwardsiella ictaluri encodes an acid-

activated urease that is required for intracellular replication in channel catfish (Ictalurus

punctatus) macrophages. Appl Environ Microbiol 75:6712-20.

27. Miller NW, Rycyzyn MA, Wilson MR, Warr GW, Naftel JP, Clem LW. 1994.

Development and characterization of channel catfish long term B cell lines. J Immunol

152:2180-9.

Page 110: Edwardsiella Ictaluri Is Capable of Persisting in Channel

101

28. Pierrard M-A, Roland K, Kestemont P, Dieu M, Raes M, Silvestre F. 2012. Fish

peripheral blood mononuclear cells preparation for future monitoring applications. Anal

Biochem 426:153-165.

29. Fukutani KF, Nascimento-Carvalho CM, Van der Gucht W, Wollants E, Khouri R,

Dierckx T, Van Ranst M, Houspie L, Bouzas ML, Oliveira JR, Barral A, Van

Weyenbergh J, de Oliveira CI. 2015. Pathogen transcriptional profile in nasopharyngeal

aspirates of children with acute respiratory tract infection. J Clin Virol 69:190-196.

30. Momburg F, Koch N, Möller P, Moldenhauer G, Butcher GW, Hämmerling G. 1986.

Differential expression of Ia and Ia-associated invariant chain in mouse tissues after in

vivo treatment with IFN-gamma. Immunology 136:940-948.

31. Leddon SA, Sant AJ. 2010. Generation of MHC class II-peptide ligands for CD4 T-cell

allorecognition of MHC class II molecules. Curr Opin Organ Transplant 15:505-11.

32. Gregers TF, Nordeng TW, Birkeland HC, Sandlie I, Bakke O. 2003. The cytoplasmic tail

of invariant chain modulates antigen processing and presentation. Eur J Immunol 33:277-

286.

33. Annaert WG, Saftig P. Regulated intramembrane proteolysis—a story about sheddases

and I-CliPs, p 125. In (ed),

34. Wolfe MS. 2009. Intramembrane proteolysis. Chem Rev 109:1599-1612.

35. Nordeng TW, Gregers TF, Kongsvik TL, Méresse S, Gorvel J-P, Jourdan F, Motta A,

Bakke O. 2002. The cytoplasmic tail of invariant chain regulates endosome fusion and

morphology. Mol Biol Cell 13:1846-1856.

36. Stoppe C, Rex S, Goetzenich A, Kraemer S, Emontzpohl C, Soppert J, Averdunk L, Sun

Y, Rossaint R, Lue H. 2015. Interaction of MIF family proteins in myocardial

ischemia/reperfusion damage and their influence on clinical outcome of cardiac surgery

patients. Antioxid Redox Signal 23:865-879.

37. Calandra T, Roger T. 2003. Macrophage migration inhibitory factor: a regulator of innate

immunity. Nat Rev Immunol 3:791-800.

38. Dickerson HW, Findly RC. 2017. Vertebrate adaptive immunity-comparative insights

from a Teleost model. Front Immunol 8:1379.

Page 111: Edwardsiella Ictaluri Is Capable of Persisting in Channel

102

39. Mackey MF, Barth Jr RJ, Noelle RJ. 1998. The role of CD40/CD154 interactions in the

priming, differentiation, and effector function of helper and cytotoxic T cells. J

Leukocyte Biol 63:418-428.

40. Pinchuk LM, Polacino PS, Agy MB, Klaus SJ, Clark EA. 1994. The role of CD40 and

CD80 accessory cell molecules in dendritic cell-dependent HIV-1 infection. Immunity

1:317-325.

41. Laing KJ, Hansen JD. 2011. Fish T cells: recent advances through genomics. Dev Comp

Immunol 35:1282-95.

42. Sun X-F, Shang N, Hu W, Wang Y-P, Guo Q-L. 2007. Molecular cloning and

characterization of carp (Cyprinus carpio L.) CD8beta and CD4-like genes. Fish

Shellfish Immunol 23:1242-1255.

43. Somamoto T, Yoshiura Y, Sato A, Nakao M, Nakanishi T, Okamoto N, Ototake M. 2006.

Expression profiles of TCRbeta and CD8alpha mRNA correlate with virus-specific cell-

mediated cytotoxic activity in ginbuna crucian carp. Virology 348:370-7.

44. Patel S, Overgård A-C, Nerland AH. 2008. CD8alpha and CD8beta in Atlantic halibut,

Hippoglossus hippoglossus: cloning, characterization and gene expression during viral

and bacterial infection. Fish Shellfish Immunol 25:570-580.

45. Moore LJ, Somamoto T, Lie KK, Dijkstra JM, Hordvik I. 2005. Characterisation of

salmon and trout CD8alpha and CD8beta. Mol Immunol 42:1225-34.

46. Hansen JD, Strassburger P. 2000. Description of an ectothermic TCR coreceptor, CD8α,

in rainbow trout. J Immunol 164:3132-3139.

47. Buonocore F, Randelli E, Bird S, Secombes CJ, Costantini S, Facchiano A, Mazzini M,

Scapigliati G. 2006. The CD8α from sea bass (Dicentrarchus labrax L.): cloning,

expression and 3D modelling. Fish Shellfish Immunol 20:637-646.

48. Edholm ES, Stafford JL, Quiniou SM, Waldbieser G, Miller NW, Bengten E, Wilson M.

2007. Channel catfish, Ictalurus punctatus, CD4-like molecules. Dev Comp Immunol

31:172-87.

49. Kordon AO, Abdelhamed H, Ahmed H, Baumgartner W, Karsi A, Pinchuk LM. 2019.

Assessment of the live attenuated and wild-type Edwardsiella ictaluri-induced immune

Page 112: Edwardsiella Ictaluri Is Capable of Persisting in Channel

103

gene expression and langerhans-like cell profiles in the immune-related organs of catfish.

Front Immunol 10:392.

50. Chen L, He C, Baoprasertkul P, Xu P, Li P, Serapion J, Waldbieser G, Wolters W, Liu Z.

2005. Analysis of a catfish gene resembling interleukin-8: cDNA cloning, gene structure,

and expression after infection with Edwardsiella ictaluri. Dev Comp Immunol 29:135-

42.

51. Zou J, Peddie S, Scapigliati G, Zhang Y, Bols NC, Ellis AE, Secombes CJ. 2003.

Functional characterisation of the recombinant tumor necrosis factors in rainbow trout,

Oncorhynchus mykiss. Dev Comp Immunol 27:813-22.

52. Milev-Milovanovic I, Long S, Wilson M, Bengten E, Miller NW, Chinchar VG. 2006.

Identification and expression analysis of interferon gamma genes in channel catfish.

Immunogenetics 58:70-80.

53. Milev-Milovanovic I, Majji S, Thodima V, Deng Y, Hanson L, Arnizaut A, Waldbieser

G, Chinchar VG. 2009. Identification and expression analyses of poly [I:C]-stimulated

genes in channel catfish (Ictalurus punctatus). Fish Shellfish Immunol 26:811-20.

54. Schroder K, Hertzog PJ, Ravasi T, Hume DA. 2004. Interferon-gamma: an overview of

signals, mechanisms and functions. J Leukoc Biol 75:163-89.

55. Wang Y, Wang Q, Baoprasertkul P, Peatman E, Liu Z. 2006. Genomic organization, gene

duplication, and expression analysis of interleukin-1beta in channel catfish (Ictalurus

punctatus). Mol Immunol 43:1653-64.

56. Hong S, Zou J, Crampe M, Peddie S, Scapigliati G, Bols N, Cunningham C, Secombes

CJ. 2001. The production and bioactivity of rainbow trout (Oncorhynchus mykiss)

recombinant IL-1 beta. Vet Immunol Immunopathol 81:1-14.

57. Peddie S, Zou J, Secombes CJ. 2002. A biologically active IL-1beta derived peptide

stimulates phagocytosis and bactericidal activity in rainbow trout, Oncorhynchus mykiss

(Walbaum), head kidney leucocytes in vitro. J Fish Dis 25:351-360.

58. Blackmon LE, Quiniou SMA, Wilson M, Bengten E. 2020. Catfish lymphocytes

expressing CC41-reactive leukocyte immune-type receptors (LITRs) proliferate in

response to Edwardsiella ictaluri infection in vitro. Dev Comp Immunol 106:103610.

Page 113: Edwardsiella Ictaluri Is Capable of Persisting in Channel

104

59. Haddad G, Hanington PC, Wilson EC, Grayfer L, Belosevic M. 2008. Molecular and

functional characterization of goldfish (Carassius auratus L.) transforming growth factor

beta. Dev Comp Immunol 32:654-63.

60. Wei H, Yin L, Feng S, Wang X, Yang K, Zhang A, Zhou H. 2015. Dual-parallel

inhibition of IL-10 and TGF-β1 controls LPS-induced inflammatory response via NF-κB

signaling in grass carp monocytes/macrophages. Fish Shellfish Immunol 44:445-452.

61. Wang X, Li C, Thongda W, Luo Y, Beck B, Peatman E. 2014. Characterization and

mucosal responses of interleukin 17 family ligand and receptor genes in channel catfish

Ictalurus punctatus. Fish Shellfish Immunol 38:47-55.

Page 114: Edwardsiella Ictaluri Is Capable of Persisting in Channel

105

CHAPTER 5. GENERAL CONCLUSIONS

The scope of this work focused on better characterizing the immune response generated

by channel catfish to wild-type Edwardsiella ictaluri (WT). This was done ex vivo in head-

kidney-derived-macrophages (HKDM) and in vivo in the head-kidney (HK) and peripheral blood

leukocyte (PBL) samples. Additionally, the scope of this work was to characterize one of the

Type III Secretion System (T3SS) effectors, EseK, as it relates to virulence using the same

parameters tested for the WT. The hypotheses of this work were first, that the WT is specifically

capable of persisting in HKDM, and subsequently the host, by limiting cell-mediated immune

(CMI) responses. Additionally, we speculated that EseK, which is relevant in HKDM

replication, may be pertinent for the evasion of CMI responses.

The first goal of this work was to determine if EseK could be of interest to pursue further

characterization. The studies in Chapter 2 focused on confirming EseK’s host-binding protein

and determining the potential for an attenuated EseK mutant (ΔeseK) to provide protection

against E. ictaluri infection. EseK was fused to a 3XFLAG sequence for specific detection in a

mix of other highly homologous E. ictaluri leucine-rich-repeat (LRR) T3SS effectors using

specific FLAG antibody. This tagged protein can restore the WT phenotype, so it was used to

carry out a co-immunoprecipitation (Co-IP) using the tagged EseK and a HKDM whole-cell

lysate. As a follow up to a yeast-2-hybrid assay performed outside the scope of this work, Co-IP

was coupled with mass-spectrometry analysis to confirm an interaction of EseK with CD74 in

HKDM. As previously mentioned, both strategies have very low rates of false positives, so the

two items together are strong proof of the interaction between EseK and CD74 in HKDMs. The

interaction of EseK with CD74 is of interest because CD74 is highly pleiotropic in other

Page 115: Edwardsiella Ictaluri Is Capable of Persisting in Channel

106

organisms, especially in cell-mediated immune pathways, as its expression is strongest in antigen

presenting cells (APC)s (1).

After confirming that CD74 is the binding partner of EseK, and that this bond could be of

practical benefit, ΔeseK was used to infect fish to determine its virulence relative to the WT and

if immersion exposure could protect against subsequent WT challenge. Vaccination is a critical

strategy in fish health, and in catfish oral or immersion vaccination with live attenuated E.

ictaluri strains is the common strategy to protect against infection with E. ictaluri (2-4). The

results showed a high degree of similarity in virulence and subsequent protection between the

WT and ΔeseK at the highest dose. The ΔeseK strain showed a high level of protection to

subsequent WT exposures at all tested doses 4 weeks post-vaccination (PV) in both trials, and

later exposure at 8 weeks also showed protection, although one trial had a drastically reduced

survival rate, with the lowest dose only providing 28% protection. Reasons ΔeseK provides

protection in a short window but not consistently for a longer duration could have to do with the

strength of the adaptive immune responses generated, specifically T cell-dependent antibody

generation and CMI responses. However, the ΔeseK strain is promising, because it does provide

a short-term protective response, with some extended protection observed in several doses,

suggesting that EseK is involved in regulation of the cell-mediated pathway. A vaccine using an

EseK deletion would be improved by finding and adding the component(s) that are aiding E.

ictaluri’s evasion of CMI responses.

The studies carried out in chapters 3 and 4 aimed to better characterize non-activated

HKDM and naïve fish in response to E. ictaluri infection with the hypothesis that naïve fish do

not mount robust CMI responses to E. ictaluri infection. Contrary to expectations, results did not

give additional indications of EseK’s role in CMI relative to the WT. While the expression data

Page 116: Edwardsiella Ictaluri Is Capable of Persisting in Channel

107

was exciting because it showed that E. ictaluri clearly has a role in downregulating CD74, it was

also highly surprising in that the binding partner, EseK, seemingly is not highly pertinent to this

response, at least not at the transcriptional level. Because ΔeseK did not exhibit significant

change compared to WT, the remainder of this discussion will focus on the WT relative to non-

infected control samples.

The RNA-sequencing (RNA-seq) data from chapter 3 led to several immediate

observations of E. ictaluri’s interaction with non-activated HKDMs. The ingenuity pathway

analysis (IPA) predicted 180 pathways with significant regulating differences between WT

infected and uninfected HKDM. Among those pathways, the regulation of TNFR1/2 pathways

(5), the P13K/AKT pathway (6), the NF-κB virus signaling pathway, and the peroxisome

proliferator receptor (PPAR) signaling pathway indicate an M1 polarization (7). This was further

supported by the regulation of several other pathways, such as the IL-1 signaling pathway, the

acute phase response pathway, and the iNOS signaling pathway (8). In addition, previous

discoveries in the lab showed the ability of E. ictaluri to survive and replicate in Edwardsiella-

containing vacuoles (ECV) within HKDMs (9). This, in addition to the regulation of several

pathways seen in this work, such as the NF-κB viral and P13K/AKT pathways, the Tec Kinase

pathway, and the death receptor pathway, indicate that E. ictaluri infection facilitates a strong

inclination towards HKDM autophagy. While it is possible that the host eventually overcomes

bacterial evasion of autophagy, it appears more likely that E. ictaluri is largely unhindered by the

autophagic environment of the HKDM, like many other bacterial pathogens (10).

After analyzing the data from chapter 4, we found that the most striking results from the

RNA-seq data is the indication of E. ictaluri regulation of the CD40 pathway in HKDMs. The

CD40 pathway is downregulated, which suggests a limited ability to fully mature and activate T

Page 117: Edwardsiella Ictaluri Is Capable of Persisting in Channel

108

cells, in the presence of E. ictaluri (11, 12). In chapter 4, we determined the naïve catfish

response to E. ictaluri. Our study focused on responses generated in the HK, a hematopoietic

tissue, and PBL’s, a potential indicator of systemic responses. The data showed a strong

downregulation of CD74 and CD40L, and upregulation of MIF in both HK and PBL samples in

response to E. ictaluri infection. CD74 was downregulated from day two, which is likely

indicative of E. ictaluri throttling T cell activation and responses, which include the CMI

involved T cell sets, CD8+ CTLs and the CD4+ Th1 subset. The downregulation of CD40L was

also of interest because CD40L allows for the expression of cytokines such as IL-1β, IL-6, IL-8,

IL-12, TNF-α, and IL-10, as well as type I IFNs (12). Additionally, other notable observations in

HK tissues showed a trend of downregulation towards the end of the experiment in T cell

markers for CD4 and CD8, and an upregulation of IL-8. In addition, TGF-β1 was highly

downregulated in PBL samples, possibly suggesting a reduction in several T cell subsets,

including regulatory T cells. TGF-β1 provides an interesting paradigm because, on its own, it

suppresses immune responses secreted by regulatory T cells, but in combination with IL-6, it

allows Th17 differentiation (13). The potential for Th17 differentiation is interesting because this

T cell subtype is more involved with neutrophil recruitment and controlling extracellular

bacteria, and E. ictaluri does not do either of these things (14). TGF-β1 also reduces cytotoxic

responses (nitric oxide), and acute phase responses in fish (15, 16).

The observation that the CD40 pathway is downregulated in HKDMs, and CD40L is also

downregulated in vivo supports the theory that E. ictaluri seemingly evades T cell dependent

responses by inhibiting the CD40-CD40L pathway. This would aid E. ictaluri’s ability to persist

in the catfish. Further research will need to be conducted to both confirm and better understand

exactly how E. ictaluri is downregulating the CD40 pathway.

Page 118: Edwardsiella Ictaluri Is Capable of Persisting in Channel

109

In conclusion, the binding partner of EseK was determined to be CD74, confirming a

previous yeast-two-hybrid assay (data unpublished). A protection assay showed that exposure to

the ΔeseK strain has a protective effect on subsequent exposure to WT. Next, RNA-seq carried

out on infected HKDM showed several pathways differentially regulated compared to uninfected

HKDM, leading to M1 polarization. Interestingly, the pathway analysis also indicated the E.

ictaluri downregulated pathways leading to programmed cell death and instead favors host cell

survival and replication. Finally, immune responses were determined in vivo, which found that

EseK has a minor effect on virulence, but WT drastically modulates immune responses, likely

aiding in persistence in the host. The strongest observation from the combined data suggests that

this is occurring by a downregulation of the critical T cell activating pathway, CD40-CD40L.

Figure 5.1 Macrophage and T cell activation with A) CD74 production allowing for MHC class

II (MHC II) to migrate with antigen to the cell surface to be recognized by T cells, in turn

allowing CD40-CD40L activation to occur, and B) in the absence of CD74, CD40 and CD40L.

Created with BioRender.com.

Page 119: Edwardsiella Ictaluri Is Capable of Persisting in Channel

110

Literature Cited

1. Momburg F, Koch N, Möller P, Moldenhauer G, Butcher GW, Hämmerling G. 1986.

Differential expression of Ia and Ia-associated invariant chain in mouse tissues after in

vivo treatment with IFN-gamma. Immunology 136:940-948.

2. Wise DJ, Greenway TE, Byars TS, Griffin MJ, Khoo LH. 2015. Oral vaccination of

channel catfish against Enteric Septicemia of Catfish using a live attenuated Edwardsiella

ictaluri isolate. J Aquat Anim Health 27:135-43.

3. Thune RL, Fernandez DH, Battista JR. 1999. An aroA mutant of Edwardsiella ictaluri is

safe and efficacious as a live, attenuated vaccine. J Aquat Anim Health 11:358-372.

4. Abdelhamed H, Lu J, Shaheen A, Abbass A, Lawrence ML, Karsi A. 2013. Construction

and evaluation of an Edwardsiella ictaluri fhuC mutant. Vet Microbiol 162:858-865.

5. Wajant H, Siegmund D. 2019. TNFR1 and TNFR2 in the control of the life and death

balance of macrophages. Front Cell Dev Biol 7:91-91.

6. Zhou YY, Li Y, Jiang WQ, Zhou LF. 2015. MAPK/JNK signalling: a potential

autophagy regulation pathway. Biosci Rep 35.

7. Evans RM, Mangelsdorf DJ. 2014. Nuclear receptors, RXR, and the big bang. Cell

157:255-266.

8. Kaler P, Godasi BN, Augenlicht L, Klampfer L. 2009. The NF-κB/AKT-dependent

induction of wnt signaling in colon cancer cells by macrophages and IL-1β. Cancer

Microenviron 2:69.

9. Booth NJ, Elkamel A, Thune RL. 2006. Intracellular replication of Edwardsiella ictaluri

in channel catfish macrophages. J Aquat Anim Health 18:101-108.

10. Kimmey JM, Stallings CL. 2016. Bacterial pathogens versus autophagy: Implications for

therapeutic interventions. Trends Mol Med 22:1060-1076.

11. Mackey MF, Barth Jr RJ, Noelle RJ. 1998. The role of CD40/CD154 interactions in the

priming, differentiation, and effector function of helper and cytotoxic T cells. J

Leukocyte Biol 63:418-428.

Page 120: Edwardsiella Ictaluri Is Capable of Persisting in Channel

111

12. Lagos LX, Iliev DB, Helland R, Rosemblatt M, Jorgensen JB. 2012. CD40L--a

costimulatory molecule involved in the maturation of antigen presenting cells in Atlantic

salmon (Salmo salar). Dev Comp Immunol 38:416-30.

13. Zou J, Secombes CJ. 2016. The function of fish cytokines. Biology 5.

14. Aujla SJ, Dubin PJ, Kolls JK. 2007. Th17 cells and mucosal host defense. Semin

Immunol 19:377-382.

15. Haddad G, Hanington PC, Wilson EC, Grayfer L, Belosevic M. 2008. Molecular and

functional characterization of goldfish (Carassius auratus L.) transforming growth factor

beta. Dev Comp Immunol 32:654-63.

16. Wei H, Yin L, Feng S, Wang X, Yang K, Zhang A, Zhou H. 2015. Dual-parallel

inhibition of IL-10 and TGF-β1 controls LPS-induced inflammatory response via NF-κB

signaling in grass carp monocytes/macrophages. Fish Shellfish Immunol 44:445-452.

Page 121: Edwardsiella Ictaluri Is Capable of Persisting in Channel

112

APPENDIX I: ABBREVIATIONS COMMONLY USED IN THIS

DISSERTATION

ANOVA- Analysis of variance

APC- antigen presenting cell

BA- Blood Agar

BHI- Brain Heart Infusion

CCMM- channel catfish macrophage media

CCO- channel catfish ovary

CD- cluster of differentiation

CFU- colony forming units

CMI- cell-mediated immunity

Co-IP- Co-immunoprecipitation

DNA-deoxyribonucleic acid

ECV- Edwardsiella-containing vacuole

ESC- enteric septicemia of channel catfish

HK- Head Kidney

HKDM- Head-Kidney Derived Macrophage

IFN- interferon

IL- interleukin

JAK- janus kinase

LRR- Leucine rich repeat

MHC- major histocompatibility complex

MIF- migration inhibitory factor

Page 122: Edwardsiella Ictaluri Is Capable of Persisting in Channel

113

MLSA- multi-locus sequence analysis

O/N- overnight

PBL- Peripheral blood leukocytes

PCR- polymerase chain reaction

PV- post-vaccination

qRT-PCR- quantitative reverse transcriptase PCR

RNA- Ribonucleic acid

SCV- Salmonella-containing vacuole

SPI- Salmonella Pathogenicity Island

STAT- signal transduces and activators of transcription

T3SS- Type III Secretion System

T4SS- Type IV Secretion System

T6SS- Type VI Secretion System

TGF- transformation growth factor

Th- T helper

TNF- Tumor necrosis factor

WT- wild type

Page 123: Edwardsiella Ictaluri Is Capable of Persisting in Channel

114

APPENDIX II: CHAPTER 1 SUPPLEMENTAL MATERIAL

eseK Nucleic Acid Sequence

ATGCCACTTTATGTCGGCTCCGGGTATTTCCCCGCCACCATCAGTAATAACCGTATTC

AGCGCATCGTCAGTGCAAATATCACTCCGGATATGAGCGTTCGGGAGAATGTTCATG

AGTATTTCAACTCAACACATAAGTCTGAAGCCCTGACCTGTATCCAGGAAATTTGTC

ATCCCCCAGCCGGAACGACACGGGACGCTGTAGTGGGTAGATTTGGGCGACTCAGG

GGACTCGCCTATCCTGGATTCGCGGATAAGATCAAGTCTGGCGACAAGGACAATGG

AGATAACCGCCTCTGTATCCTGGATGAAAACGGACAGGAAATGTTATCAGTCATCCT

TGGTGATGACGAAAAATATACCGTCATCTATCCAAATGGCAGGGTAACGCATGACC

TCAGCCTGCCCGCATCACAAGGCGATGAGGGTCTCCTGGCATCATGTACTTTCGCCA

CCGCGGGCCCACAAACAGCGGATGAGTATGAGGCTGTTTGGTTAGCCTGGGAAAGG

GCTGCGCCGCCAGCAGAGGCAGAATGCCGAACCCAGACAGTGCAGAAGCTACGTCA

ATGCATGCAGAATGACACCCGGCTGGATGTCTCTCACACCCCACTAACCAGTCTGCC

GCCGCTGCCCACCGGACTGCAGAGTCTGGACATCTCTTACACTTCACTGACCAAGCT

GCCGCCGCTGCCCGACGGACTGCAGAGTCTGGACGTCTCTCACACCTCACTGACCAA

GCTACCGACGCTGCCCCACGGACTACAGAGTCTGAACGTCTCTCACACTTCACTGAT

AGGCTTACCGCTGCTGCCCCACGGACTGCAGAGTCTGGACGGCTCTCACACCTCACT

GACAGGCTTACCGCCGCTGCCCCAGAGACTGCAGAGTCTGGACGTCTCTCACACCTC

ACTGACAGGCCTACCGCCGCTGCTCCACGGACTGCAGAGTCTGGACGTCTCTCACAC

TTCACTGACCCAGCTGCCGACGCTGCCCCAAGGGTTGCAAAGTCTCAACATTTCTCA

CACTTCACTGACCGACCTGCAGACGCTGCCCTCCAAACTGCAGAGGTTGGATGCCTC

TCACACCTCACTGGCCAAACTGCAGACGCTGCCCTCCAAACTGCAGAAGTTGGATGT

CTCTCACACCTCACTGACCGAGCTACCGAAGATGCCATCCAGACTGGAAACTCTGAA

TGTCTCAGGCACCAAAATGACCAAACTATCGAGTTTGCCCTACGGTCTAAAGACACT

GAATGCCTCAAATACCCTACTGACAAGAATGCCAAGGCTGTACAACGAACTGCAAA

ATCTGGACCTCTCTAACACCACAATAGACTTCATAGGCCCATTTGAACTTCCGTATG

GACTGCAAACGCTGGATGTCTCAGGCACCCGACTGATCAGACTGCCACAACTGCCC

ACCGGACTGCAGTCGCTGAATATGTCTCACTTACCCTTAAAAGCCCTACCCCATATG

CCATCCGGACTGCAGACGCTGAATATGTCTCACATCCAAAAACTGATCAACATTACG

CATCTTCCCACCGAACTGCAGACGCTGAATGTCTCTCAGACCCCACTGCTCAACCTG

TCACAGCTCCCCACCGGACTGCAGTCGCTGAATATCTCTGACACCCCACTGTACAGA

CTGCCGTCGCTGCCCCCTGTACTGCAAATGCTGGATGTCTCTGGCACCAACCTGATC

AACCTGCCAGAAAGCCTAGCCAGGCTATCGCGGGATACAACTGTCCTCATGCAGAA

TAATCACATGCCAGATAGTACGATTCAGTTATTACGAAACAGGGTCAATGAACCCG

ACTATCAGGGACCCAGGATAGAGTTTGATAGGGGAGCCTCTTCAGGCGTTCAGGAA

ACCAATGCCTTGCACTTGCTGATGGCTAACTGGATGACAATAGAGGAAGCTTCAGAT

GATAGATGGAAAACCTTCTGGAGTGTAGACGATGCCCGCGCCTTCAGCATGTTTCTC

AATCAGTTGAAAGAGACGAAAAATAATAAGTCAGCCGATTTTCGCAAACAGATAGC

CACCTGGCTAACCCTGCTGGCTAATGATGATGAGCTAAGAGCCAAAACCTTTGCCAT

GGCGACAGAGTCAACCTCAAGCTGTGAGGACCGGGTCACGCTGGCGATGAACAGTA

Page 124: Edwardsiella Ictaluri Is Capable of Persisting in Channel

115

TGAAGAGCGTGCAACTGCTCCATAATGCTGAAAAAGGGAAATTCGACAACGATATC

CCGGGGCTCGTGTCGGTAGGTCGTGAAATGTTCCGCCTGGAGAAGCTGGAGCTGATT

GCCCGCGAGAAGATAAAAACGCTACGCGATGAGGTCTATGGGGGCGATGAAGATAA

AGTCGATGAGATTGAGGTTTTTCTTGGCTATCAGAACCAATTGAAAAAATCACTTGA

ACTGACCGGGGCCACTGACGAAATGCGATATTTTGACGTATCCGACATTACCGAGTC

AGACCTTCAGGCTGCTGAAATTCAGGTGAAAACCGCCGAGGACAGCCAGTTCGGGG

AGTGGATACTGCAGTGGGAGCCGTTGCACAAAGTACTGATACGTAAAAATAAGGAC

GACTGGGATGCGCTCATTGACAAGAAAATAGAGGATTATGAGCGTGAATACCAAGA

ATTATATGACACAGAGTTGGAGCCGGCAGGTCTGGTCGGCGACATCGAGTCTGAGC

GAACTATCGGCGCAAGAGCGATGGCGAGCACAGAAAAGTTATTTCAGCAAGGCCTA

CGCCCGCTGGCAGAGAAGTTGCTGGGCAAGCATCTGGGAGCCCGATGGTTGCTAGA

TGCGTCCCCTGTATAG

EseK Amino Acid Sequence

MPLYVGSGYFPATISNNRIQRIVSANITPDMSVRENVHEYFNSTHKSEALTCIQEICHPPA

GTTRDAVVGRFGRLRGLAYPGFADKIKSGDKDNGDNRLCILDENGQEMLSVILGDDEK

YTVIYPNGRVTHDLSLPASQGDEGLLASCTFATAGPQTADEYEAVWLAWERAAPPAEA

ECRTQTVQKLRQCMQNDTRLDVSHTPLTSLPPLPTGLQSLDISYTSLTKLPPLPDGLQSL

DVSHTSLTKLPTLPHGLQSLNVSHTSLIGLPLLPHGLQSLDGSHTSLTGLPPLPQRLQSLD

VSHTSLTGLPPLLHGLQSLDVSHTSLTQLPTLPQGLQSLNISHTSLTDLQTLPSKLQRLDA

SHTSLAKLQTLPSKLQKLDVSHTSLTELPKMPSRLETLNVSGTKMTKLSSLPYGLKTLNA

SNTLLTRMPRLYNELQNLDLSNTTIDFIGPFELPYGLQTLDVSGTRLIRLPQLPTGLQSLN

MSHLPLKALPHMPSGLQTLNMSHIQKLINITHLPTELQTLNVSQTPLLNLSQLPTGLQSLN

ISDTPLYRLPSLPPVLQMLDVSGTNLINLPESLARLSRDTTVLMQNNHMPDSTIQLLRNR

VNEPDYQGPRIEFDRGASSGVQETNALHLLMANWMTIEEASDDRWKTFWSVDDARAF

SMFLNQLKETKNNKSADFRKQIATWLTLLANDDELRAKTFAMATESTSSCEDRVTLAM

NSMKSVQLLHNAEKGKFDNDIPGLVSVGREMFRLEKLELIAREKIKTLRDEVYGGDEDK

VDEIEVFLGYQNQLKKSLELTGATDEMRYFDVSDITESDLQAAEIQVKTAEDSQFGEWIL

QWEPLHKVLIRKNKDDWDALIDKKIEDYEREYQELYDTELEPAGLVGDIESERTIGARA

MASTEKLFQQGLRPLAEKLLGKHLGARWLLDASPV(Stop)

Page 125: Edwardsiella Ictaluri Is Capable of Persisting in Channel

116

Figure II.1: ΔeseK creation. Primer (P) 190 and P 197 were used to amplify a sequence directly upstream of eseK and P191 and P192

amplified the 3’ tail of the eseK gene. Overlap PCR was implemented via reverse complemented sequences embedded into the P197

and P191 primers by amplifying the two fragments using P190 and P192. Yellow line indicates translation region. Figure generated

using SnapGene® software (from GSL Biotech; available at snapgene.com)

Page 126: Edwardsiella Ictaluri Is Capable of Persisting in Channel

117

Figure II.2: ΔeseK ligation to pRE107 plasmid. A) digest pRE107and ΔeseK using SacI and

XbaI, and B) Ligation of pRE107and ΔeseK. Figure generated using SnapGene® software (from

GSL Biotech; available at snapgene.com)

Page 127: Edwardsiella Ictaluri Is Capable of Persisting in Channel

118

Figure II.3: ΔeseK on pRE107 for E. ictaluri genome insertion. The pRE107 plasmid carries a cis-acting oriT that allows the plasmid

to be transferred to the E. ictaluri chromosome. As the sacB gene carried on the plasmid is translated, it accumulates a polymer

sucrose in the periplasmic space and prevents bacterial growth. Figure generated using SnapGene® software (from GSL Biotech;

available at snapgene.com)

Page 128: Edwardsiella Ictaluri Is Capable of Persisting in Channel

119

Figure II.4. EseK::3XFLAG creation A) digest pBBR1-MCS4 and eseK::3XFLAG (previously

amplified using OL1-3XFLAG primer) using SacI and XbaI, and B) Ligation of pBBR1-MCS4

to EseK::3XFLAG. Figure generated using SnapGene® software (from GSL Biotech; available

at snapgene.com)

Page 129: Edwardsiella Ictaluri Is Capable of Persisting in Channel

APPENDIX II: CHAPTER 4 SUPPLEMENTAL MATERIAL

Appendix II, Table 1: Probe Targets for nCounter®

Gene

Target Accession Position Target Sequence # Hit

Isoforms Hit by

Probe

β2M NM_00120

0072.1

243-342 AATGGCGAGGTGATTCCAAATGCAGAACAGACCGACC

TGGCCTTTGAGAAGGGATGGAAGTTTCATCTGACCAAG

AGCGTCTCCTTCACTCCCACCAGCA

1 NM_001200072.1

ACTβ XM_01745

4668.1 1338-

1437 CTCAGGATCTAAAAACTGGAACGGTGAAGGTGACGGC

AATGTTTTTTGGCAAATAAGCATCCCCGAAGTTCTACA

ATGCATCTGAGGACTCAAAGTACTT

1 XM_017454668.1

EEF1A XM_01745

4316.1

1454-

1553

GGCTGCTGGTGCTGGCAAGGTCACGAAGTCTGCACAG

AAGGCTGCCAAGACCAAGTGAATTCTCATTCCAGATGT

GTTAAAGGTGGTGGGGCATCCTCCC

1 XM_017454316.1

MIF NM_00120

0304.1

264-363 TCGGGAAGATCGGAGGCTCGCAGAATAAACAGTATTC

CAAACTGCTCATGGGTGTGCTTCATAAACACCTGGGCA

TTTCACCTGACAGGATCTACATTAA

1 NM_001200304.1

CD74 XM_01748

4954.1

589-688 ACCTCGGCGCTTCCTGCTTTCGTGTTGGAGTCTAAGTGT

AAGATTGAGGCTGGGCAGGTCAAACCAGGCTTCTTCG

AGCCACAGTGTGATGAGGAGGGCC

2 XM_017484954.1;

XM_017484955.1

CD3ε XM_01749

1706.1

215-314 ATGTGAGTATCTCGGGTACCACAGTCACACTGACCTGT

CCAGCTGATCCGGAGGACACCATACAATGGTTCAAAT

ATGCACAAACAGACCCACTCTCACT

4 XM_017491706.1;

XM_017491707.1;

XM_017491709.1;

XM_017491708.1

CD4 NM_00120

0226.1

1333-

1432

AGACAGATGATGAGGTATAGATGCCGTAAGGGGAGAG

TCTGCTGCTGTAAGAATCCCAAGCCCAAAGGTTTCTAC

AAGACCTGAATGAAGCCTCTTCTTG

2 NM_001200226.1;

XM_017474004.1

(table cont’d)

Page 130: Edwardsiella Ictaluri Is Capable of Persisting in Channel

121

Appendix II, Table 1 continued

Gene

Target Accession Position Target Sequence # Hit

Isoforms Hit by

Probe

CD4_2 NM_00120

0227.1

616-715 GTTGTTGGTCCTTTGAACACACCAAGGGAAGTTAAGAC

TCATGAAGGGGGCAGTGCAGTGCTCCCGTGTTTTCTAC

CTACCAAGAGCCAACTGCCTATAA

1 NM_001200227.1

CD40L XM_01747

3417.1 455-554 AGCGCTCCCATCAAACACACCATTAGTCAAGTTCTTGT

CGGTGCTGCTGCTGCTGCTAATGATACAGACGTTTGGA

GGGTTTCTCTACCTGTTCCATGCA

1 XM_017473417.1

CD8α

NM_00120

0331.1

113-212 ACAAGAATTATCCATCGACAGTCTTTTGGCTTCGACTG

AAAGAAAACGGCCAGGGCTTTGAGTACATTGCGTCGT

ACAGCAAGACTAAGAAATCAGGCAA

1 NM_001200331.1

CD8α_

2

XM_01746

6933.1 605-704 GCATCATTAGCTTTACTGCTGATTATTGCCTGCCTCACA

TCTGCCATGTGCTTTATATACAGAGGTCGCTGCGTGAG

ACCAAAACTAAAGACGGATCGCC

1 XM_017466933.1

IFN-γ NM_00120

0302.1 420-519 ACTCAGGATGAAACGCTGAAGAATCACTTGCACGAAG

TGAAAGACCAAATGAACAAGCTGAAAGAACACTACTT

CTCCGGCAAACATGCAGACATCAAGA

2 NM_001200302.1;

XM_017493646.1

IL-10 XM_01745

0800.1

888-987 AGAATTTCAGAACGCCCATCGACACCATCGGAAGCTTA

TTTCAAGAGCTCAAGAGGGAGCTGGTCAAATGTAGGA

GTTACTTCACATGCCGGAAACCTTT

1 XM_017450800.1

IL-12α XM_01747

2329.1

303-402 TACTGGTCAACCTTTAAATCTTACCGTGATCCAGATCG

AATCCTGGAACAGTCAGTGCTCAGAAGCATAGAGAAC

CTCATGCAGGACTGCTTTTCTTCCA

1 XM_017472329.1

IL-17A XM_01747

5825.1 354-453 CCGATATACAGCACCATTCTGGTGCTCACGCAGGACTC

AACAAACAGGAAGTGCTTCAGTGTGAAGTTCCAGAGA

GTCACCGTTGGCTGCACGTGTGTGT

1 XM_017475825.1

(table cont’d)

Page 131: Edwardsiella Ictaluri Is Capable of Persisting in Channel

122

Appendix II, Table 1 continued

Gene

Target Accession Position Target Sequence # Hit

Isoforms Hit by

Probe

IL-1β NM_00120

0220.1

678-777 AGAGACTGAAGTCCATCCAGGAGAATGATGGCATGGA

ACGCTTCCTTTTCTTCAGAAACGGCACTGGTGACTCCC

TTAACACCTTCGAGTCGGTCAAATA

2 NM_001200220.1;

NM_001200219.1

IL-6 XM_01745

5306.1 607-706 GGGCGATCAATTACATGAAGACTACGAAATTAATCCA

ACACATGGCGAAGGAACACGGGTGGCCCTCCAAACAC

GAGCAATGAATTTCCTGATGTTGTAT

1 XM_017455306.1

IL-8_1 XM_01747

2609.1

288-387 CTGCAAGAACACAGAGGTCATTGCTGGCTTGGTGAGTG

GAGAAAAGATTTGCCTGAATCCTCGGACTGCATGGGTT

AAGAAACTTATCCAGTTCATTGAG

1 XM_017472609.1

IL-8_2 NM_00120

0175.1 299-398 AAATTCCAAGGGAAGAAGGGGAAAGAAGCAGAAGGA

GATCAAACGGCAAAACTGAAGTTTACAGCTGGAGAAT

TCTACACATATATGATTGCTCAACCTT

3 NM_001200175.1;

XM_017451175.1;

XM_017451174.1

IL-8_3 XM_01747

2610.1 385-484 TCTTGTGGAGGAGGATGTGTGTTATGCAATCTTTCAGT

TTCACTGGAGGCAGAATGGCTTTAAAAGGACCCTAAG

CTCAGCTTCTTAACTGAGAACATCT

1 XM_017472610.1

TGF-

β1

XM_01749

3613.1

1107-

1206

GTTGGAAATGGATACACCAACCCTCCGGCTATTATGCA

AACTACTGTATCGGCTCCTGTTCGTTTGTCTGGAACAC

GGAAAATAAGTACTCACAGGTGAT

3 XM_017493613.1;

XM_017493612.1;

XM_017493614.1

TNF-α NM_00120

0172.1

361-460 CCCGCAGGTTTCGAGCGTATCTATGCAGTGGTTCGACA

ACGCAGACCAGTCCTTCTCTTCAGGCTTGAAACTGGAG

GACAACGAGATCAAGATTCTCCGC

1 NM_001200172.1

Page 132: Edwardsiella Ictaluri Is Capable of Persisting in Channel

APPENDIX III: FAILED AND ONGOING EXPERIMENTS

IFA and PLA. Two experiments that could help further this work would be determining

where in the macrophage EseK interacts with CD74, via a PLA, and a ubiquitination assay.

While EseK and CD74 interact (both) yeast-2-hybrid and Co-IP assays are highly artificial;

therefore, it would be helpful to determine if this interaction is occurring in a more native

situation, such as in the HKDM. This could give insight into if this interaction actually occurs in

HKDM, and if so, if it is localized to certain locations on and/or in the cell, as well as the degree

and frequency that this interaction takes place.

An experiment utilizing EseK::3XFLAG with anti-FLAG antibody in conjunction with

goat anti-CD74 antibody has not yielded successful PLA results (or IFA). It is possible that full

optimization with these two antibodies has not yet occurred, or it is also possible that the 3X

FLAG system is not useful in this particular system even though it was used in other similar

experiments in Salmonella (seen in chapter 2). While Salmonella is closely related to E. ictaluri,

it is possible that the difference in species and cell targets used in other experiments are enough

to account for this inability to replicate similar experiments in this system. If that is the case, then

this could potentially be resolved by switching the tag or transfecting the HKDM cells with EseK

attached to an expression plasmid.

Ubiquitination Assay. EseK contains an E3 ubiquitin ligase motif and a translocation

sequence. The translocation sequence was previously shown to prevent secretion through the

T3SS when deleted, but it is also of interest to know if the E3 ubiquitin ligase sequence is

providing some function. The proteasome is found in two primary locations in the cell: the

cytosol and the nucleus and can degrade protein after the protein is ubiquitinated, which is the

Page 133: Edwardsiella Ictaluri Is Capable of Persisting in Channel

124

role of an E3 ubiquitin ligase motif. If EseK’s E3 ubiquitin ligase motif is functional, EseK may

be interacting with CD74 to ubiquitinate it, thus targeting it for destruction or modifying it to

carry out some other function. This would effectively prevent the formation of MHC class II and

might also limit MHC class I activity from cross-presentation, or various other functions CD74 is

involved with, discussed previously in this work. An attempt to observe this interaction was

carried out twice in HKDM. It is possible that the ubiquitin motif is not functional in EseK, or

that this interaction is taking place at a very particular time during infection that is outside the

scope of the present experimental design.

TNP-KLH Assay. An ongoing experiment looking at T independent antibody production

should aid in the in vivo studies. This will help determine what the antibody response is to WT

relative to uninfected cells, as well as ΔeseK. The goal of this study is to collect blood samples

from infected fish at a two-week interval over 11 weeks. Fish were injected with the bacterial

strain of interest as well as TNP-KLH, a T cell-dependent hapten-carrier system that will aid in

the isolation of T cell-dependent antibody responses. The purpose of injection was to control the

efficiency of infection by by-passing the typical entry routes. This also allowed us to infect the

fish at a much lower dosage known to produce antibody responses, thus giving us a better

probability that a significant number of fish would survive to the conclusion of the study in each

treatment. After sample collection, an ELISA will be conducted using plates coated with anti-

TNP to determine antibody responses. Separate ELISA’s (or WB’s) will also be conducted to

verify ESC infection. Based on the present work, we would expect to see a reduction in T cell-

dependent antibody responses in the WT, and ΔeseK relative to 65ST, the T3SS mutant.

Page 134: Edwardsiella Ictaluri Is Capable of Persisting in Channel

125

Page 135: Edwardsiella Ictaluri Is Capable of Persisting in Channel

126

LIST OF REFERENCES

Abayneh, T., Colquhoun, D. J., & Sorum, H. (2012). Multi-locus Sequence Analysis (MLSA) of

Edwardsiella tarda isolates from fish. Veterinary Microbiology, 158(3-4), 367-375.

doi:10.1016/j.vetmic.2012.03.006

Abdelhamed, H., Lu, J., Shaheen, A., Abbass, A., Lawrence, M. L., & Karsi, A. (2013).

Construction and evaluation of an Edwardsiella ictaluri fhuC mutant. Veterinary

Microbiology, 162(2), 858-865. doi:https://doi.org/10.1016/j.vetmic.2012.11.006

Abul K. Abbas, A. H. H. L., Shic Pillai. (2012). Basic Immunology: Functions and Disorders of

the Immune System (4 ed.).

Acharya, M., Maiti, N. K., Mohanty, S., Mishra, P., & Samanta, M. (2007). Genotyping of

Edwardsiella tarda isolated from freshwater fish culture system. Comp Immunol

Microbiol Infect Dis, 30(1), 33-40. doi:10.1016/j.cimid.2006.10.003

Ainsworth, A. J., & Chen, D. X. (1990). Differences in the phagocytosis of four bacteria by

channel catfish neutrophils. Developmental & Comparative Immunology, 14(2), 201-209.

doi:10.1016/0145-305x(90)90091-r

Akgul, A., Akgul, A., Lawrence, M. L., & Karsi, A. (2018). Stress-related genes promote

Edwardsiella ictaluri pathogenesis. PLoS One, 13(3), e0194669.

doi:10.1371/journal.pone.0194669

Annaert, W. G., & Saftig, P. (2009). Regulated intramembrane proteolysis—a story about

sheddases and I-CliPs. Paper presented at the Seminars in Cell and Developmental

Biology.

Ashida, H., Mimuro, H., Ogawa, M., Kobayashi, T., Sanada, T., Kim, M., & Sasakawa, C.

(2011). Cell death and infection: a double-edged sword for host and pathogen survival.

Journal of Cell Biology, 195(6), 931-942. doi:10.1083/jcb.201108081

Aujla, S. J., Dubin, P. J., & Kolls, J. K. (2007). Th17 cells and mucosal host defense. Seminars

in Immunology, 19(6), 377-382. doi:https://doi.org/10.1016/j.smim.2007.10.009

Baldwin Jr, A. S. (1996). The NF-κB and IκB proteins: new discoveries and insights. Annual

Review of Immunology, 14(1), 649-681.

Page 136: Edwardsiella Ictaluri Is Capable of Persisting in Channel

127

Baldwin, T. J. (1992). Edwardsiella Ictaluri: pathogenic mechanisms and antigens recognized

by infected channel catfish. (Doctor of Philosophy ), Louisiana State University

Baldwin, T. J., & Newton, J. C. (1993). Pathogenesis of Enteric Septicemia of Channel Catfish,

caused by Edwardsiella ictaluri: bacteriologic and light and electron microscopic

findings. Journal of Aquatic Animal Health, 5(3), 189-198. doi:10.1577/1548-

8667(1993)005<0189:Poesoc>2.3.Co;2

Basha, G., Omilusik, K., Chavez-Steenbock, A., Reinicke, A. T., Lack, N., Choi, K. B., &

Jefferies, W. A. (2012). A CD74-dependent MHC class I endolysosomal cross-

presentation pathway. Nature Immunology, 13(3), 237.

Baumgartner, W. A., Dubytska, L., Rogge, M. L., Mottram, P. J., & Thune, R. L. (2014).

Modulation of vacuolar pH is required for replication of Edwardsiella ictaluri in channel

catfish macrophages. Infection and Immunity, 82(6), 2329-2336. doi:10.1128/IAI.01616-

13

Beswick, E. J., & Reyes, V. E. (2009). CD74 in antigen presentation, inflammation, and cancers

of the gastrointestinal tract. J Gastroenterol, 15(23), 2855-2861.

doi:10.3748/wjg.15.2855

Bhavsar, A. P., Brown, N. F., Stoepel, J., Wiermer, M., Martin, D. D., Hsu, K. J., . . . Finlay, B.

B. (2013). The Salmonella type III effector SspH2 specifically exploits the NLR co-

chaperone activity of SGT1 to subvert immunity. PLoS Pathogens, 9(7), e1003518.

doi:10.1371/journal.ppat.1003518

Bird, S., & Tafalla, C. (2015). Teleost chemokines and their receptors. Biology (basel) 4(4), 756-

784. doi:10.3390/biology4040756

Blackmon, L. E., Quiniou, S. M. A., Wilson, M., & Bengten, E. (2020). Catfish lymphocytes

expressing CC41-reactive leukocyte immune-type receptors (LITRs) proliferate in

response to Edwardsiella ictaluri infection in vitro. Developmental & Comparative

Immunology, 106, 103610. doi:10.1016/j.dci.2020.103610

Bockemuhl, J., Pan-Urai, R., & Burkhardt, F. (1971). Edwardsiella tarda associated with human

disease. Pathol Microbiol, 37(5), 393-401.

Bogdan, C., Röllinghoff, M., & Diefenbach, A. (2000). The role of nitric oxide in innate

immunity. Immunological Reviews, 173, 17-26.

Page 137: Edwardsiella Ictaluri Is Capable of Persisting in Channel

128

Booth, N. J., Beekman, J. B., & Thune, R. L. (2009). Edwardsiella ictaluri encodes an acid-

activated urease that is required for intracellular replication in channel catfish (Ictalurus

punctatus) macrophages. Applied and Environmental Microbiology, 75(21), 6712-6720.

doi:10.1128/AEM.01670-09

Booth, N. J., Elkamel, A., & Thune, R. L. (2006). Intracellular replication of Edwardsiella

ictaluri in channel catfish macrophages. Journal of Aquatic Animal Health, 18(2), 101-

108. doi:10.1577/H05-025.1

Bouzas, M. L., Oliveira, J. R., Queiroz, A., Fukutani, K. F., Barral, A., Rector, A., . . .

Nascimento-Carvalho, C. M. (2018). Diagnostic accuracy of digital RNA quantification

versus real-time PCR for the detection of respiratory syncytial virus in nasopharyngeal

aspirates from children with acute respiratory infection. Journal of Clinical Virology,

106, 34-40. doi:https://doi.org/10.1016/j.jcv.2018.07.003

Brumell, J. H., Marcus, S. L., & Finlay, B. B. (2000). N‐terminal conservation of putative type

III secreted effectors of Salmonella typhimurium. Molecular Microbiology, 36(3), 773-

774.

Bujan, N., Mohammed, H., Balboa, S., Romalde, J. L., Toranzo, A. E., Arias, C. R., &

Magarinos, B. (2018). Genetic studies to re-affiliate Edwardsiella tarda fish isolates to

Edwardsiella piscicida and Edwardsiella anguillarum species. Systematic and Applied

Microbiology, 41(1), 30-37. doi:10.1016/j.syapm.2017.09.004

Buonocore, F., Randelli, E., Bird, S., Secombes, C. J., Costantini, S., Facchiano, A., Scapigliati,

G. (2006). The CD8α from sea bass (Dicentrarchus labrax L.): cloning, expression and

3D modelling. Fish & Shellfish Immunology, 20(4), 637-646.

Burne, R. A., & Chen, Y.-Y. M. (2000). Bacterial ureases in infectious diseases. Microbes and

Infection, 2(5), 533-542.

Calandra, T., & Roger, T. (2003). Macrophage migration inhibitory factor: a regulator of innate

immunity. Nature Reviews: Immunology, 3(10), 791-800.

Castro, N., Toranzo, A. E., Bastardo, A., Barja, J. L., & Magarinos, B. (2011). Intraspecific

genetic variability of Edwardsiella tarda strains from cultured turbot. Diseases of Aquatic

Organisms, 95(3), 253-258. doi:10.3354/dao02363

Chang, C.-I., Liao, J. C., & Kuo, L. (1998). Arginase modulates nitric oxide production in

activated macrophages. American Journal of Physiology: Heart and Circulatory

Physiology, 274(1), H342-H348.

Page 138: Edwardsiella Ictaluri Is Capable of Persisting in Channel

129

Chen, L., He, C., Baoprasertkul, P., Xu, P., Li, P., Serapion, J., Liu, Z. (2005). Analysis of a

catfish gene resembling interleukin-8: cDNA cloning, gene structure, and expression after

infection with Edwardsiella ictaluri. Developmental & Comparative Immunology, 29(2),

135-142. doi:10.1016/j.dci.2004.06.011

Chen, X., Deane, N. G., Lewis, K. B., Li, J., Zhu, J., Washington, M. K., & Beauchamp, R. D.

(2016). Comparison of nanostring nCounter® data on FFPE colon cancer samples and

affymetrix microarray data on matched frozen tissues. PLoS One, 11(5), e0153784-

e0153784. doi:10.1371/journal.pone.0153784

Collins, L. A., & Thune, R. L. (1996). Development of a defined minimal medium for the growth

of Edwardsiella ictaluri. Applied and Environmental Microbiology, 62(3), 848-852.

Cooper, J. E., & Feil, E. J. (2004). Multilocus sequence typing-what is resolved? Trends in

Microbiology, 12(8), 373-377. doi:10.1016/j.tim.2004.06.003

de Lorenzo, V., Herrero, M., Jakubzik, U., & Timmis, K. N. (1990). Mini-Tn5 transposon

derivatives for insertion mutagenesis, promoter probing, and chromosomal insertion of

cloned DNA in gram-negative eubacteria. Journal of Bacteriology, 172(11), 6568-6572.

doi:10.1128/jb.172.11.6568-6572.1990

Dickerson, H. W., & Findly, R. C. (2017). Vertebrate adaptive immunity-comparative insights

from a Teleost model. Front Immunol, 8, 1379. doi:10.3389/fimmu.2017.01379

Dubytska, L. P., Rogge, M. L., & Thune, R. L. (2016). Identification and characterization of

putative translocated effector proteins of the Edwardsiella ictaluri type III secretion

system. mSphere, 1(3), e00039-00016. doi:10.1128/mSphere.00039-16

Dubytska, L. P., Rogge, M. L., & Thune, R. L. (2016). Identification and Characterization of

Putative Translocated Effector Proteins of the Edwardsiella ictaluri Type III Secretion

System. mSphere, 1(3).

Dubytska, L. P., & Thune, R. L. (2018). Edwardsiella ictaluri type III secretion system (T3SS)

effector EseN is a phosphothreonine lyase that inactivates ERK1/2. Diseases of Aquatic

Organisms, 130(2), 117-129.

Edholm, E. S., Bengten, E., Stafford, J. L., Sahoo, M., Taylor, E. B., Miller, N. W., & Wilson,

M. (2010). Identification of two IgD+ B cell populations in channel catfish, Ictalurus

punctatus. Journal of Immunology, 185(7), 4082-4094. doi:10.4049/jimmunol.1000631

Page 139: Edwardsiella Ictaluri Is Capable of Persisting in Channel

130

Edholm, E. S., Stafford, J. L., Quiniou, S. M., Waldbieser, G., Miller, N. W., Bengten, E., &

Wilson, M. (2007). Channel catfish, Ictalurus punctatus, CD4-like molecules.

Developmental & Comparative Immunology, 31(2), 172-187.

doi:10.1016/j.dci.2006.05.012

Edwards, R. A., Keller, L. H., & Schifferli, D. M. (1998). Improved allelic exchange vectors and

their use to analyze 987P fimbria gene expression. Gene, 207(2), 149-157. doi:Doi

10.1016/S0378-1119(97)00619-7

Elmore, S. (2007). Apoptosis: a review of programmed cell death. Toxicologic Pathology, 35(4),

495-516. doi:10.1080/01926230701320337

Evans, R. M., & Mangelsdorf, D. J. (2014). Nuclear receptors, RXR, and the big bang. Cell,

157(1), 255-266.

FAO. (2011-2012). Fisheries and aquaculture software. FishStatJ. Rome.

FAO. (2018). The State of World Fisheries and Aquaculture 2018: Meeting the Sustainable

Development Goals (978-92-5-130562-1). Retrieved from Rome:

Ferguson, H. W., Turnbull, J. F., Shinn, A., Thompson, K., Dung, T. T., & Crumlish, M. (2001).

Bacillary necrosis in farmed Pangasius hypophthalmus (Sauvage) from the Mekong

Delta, Vietnam. Journal of Fish Diseases, 24(9), 509-513. doi:DOI 10.1046/j.1365-

2761.2001.00308.x

Fernandez, D. H., Pittman-Cooley, L., & Thune, R. L. (2001). Sequencing and analysis of the

Edwardsiella ictaluri plasmids. Plasmid, 45(1), 52-56. doi:10.1006/plas.2000.1499

Finn, C. E., Chong, A., Cooper, K. G., Starr, T., & Steele-Mortimer, O. (2017). A second wave

of Salmonella T3SS1 activity prolongs the lifespan of infected epithelial cells. PLoS

Pathogens, 13(4), e1006354. doi:10.1371/journal.ppat.1006354

Francis-Floyd, R., Beleau, M. H., Waterstrat, P. R., & Bowser, P. R. (1987). Effect of water

temperature on the clinical outcome of infection with Edwardsiella ictaluri in channel

catfish. Journal of the American Veterinary Medical Association, 191(11), 1413-1416.

Fukutani, K. F., Nascimento-Carvalho, C. M., Van der Gucht, W., Wollants, E., Khouri, R.,

Dierckx, T., de Oliveira, C. I. (2015). Pathogen transcriptional profile in nasopharyngeal

aspirates of children with acute respiratory tract infection. Journal of Clinical Virology,

69, 190-196. doi:https://doi.org/10.1016/j.jcv.2015.06.005

Page 140: Edwardsiella Ictaluri Is Capable of Persisting in Channel

131

Geiss, G. K., Bumgarner, R. E., Birditt, B., Dahl, T., Dowidar, N., Dunaway, D. L., . . .

Dimitrov, K. (2008). Direct multiplexed measurement of gene expression with color-

coded probe pairs. Nature Biotechnology, 26(3), 317-325. doi:10.1038/nbt1385

Gregers, T. F., Nordeng, T. W., Birkeland, H. C., Sandlie, I., & Bakke, O. (2003). The

cytoplasmic tail of invariant chain modulates antigen processing and presentation.

European Journal of Immunology, 33(2), 277-286.

Griffin, M. J., Quiniou, S. M., Cody, T., Tabuchi, M., Ware, C., Cipriano, R. C., . . . Soto, E.

(2013). Comparative analysis of Edwardsiella isolates from fish in the eastern United

States identifies two distinct genetic taxa amongst organisms phenotypically classified as

E. tarda. Veterinary Microbiology, 165(3-4), 358-372. doi:10.1016/j.vetmic.2013.03.027

Griffin, M. J., Reichley, S. R., Greenway, T. E., Quiniou, S. M., Ware, C., Gao, D. X., . . . Soto,

E. (2016). Comparison of Edwardsiella ictaluri isolates from different hosts and

geographic origins. Journal of Fish Diseases, 39(8), 947-969. doi:10.1111/jfd.12431

Griffin, M. J., Ware, C., Quiniou, S. M., Steadman, J. M., Gaunt, P. S., Khoo, L. H., & Soto, E.

(2014). Edwardsiella piscicida identified in the Southeastern USA by gyrB sequence,

species-specific and repetitive sequence-mediated PCR. Diseases of Aquatic Organisms,

108(1), 23-35. doi:10.3354/dao02687

Grimont, P., Grimont, F., Richard, C., & Sakazaki, R. (1980). Edwardsiella hoshinae, a new

species of Enterobacteriaceae (Vol. 4).

Gudding, R., Lillehaug, A., & Evensen, Ø. (2014). Fish vaccination: Chichester, West Sussex ;

Hoboken, NJ : Wiley Blackwell, 2014.

H. Ewing, W., C. Mcwhorter, A., R. Escobar, M., & H. Lubin, A. (1965). Edwardsiella, a new

genus of Enterobacteriaceae based on a new species, E. tarda. International Journal of

Systematic and Evolutionary Microbiology, 15. doi:10.1099/00207713-15-1-33

Haddad, G., Hanington, P. C., Wilson, E. C., Grayfer, L., & Belosevic, M. (2008). Molecular and

functional characterization of goldfish (Carassius auratus L.) transforming growth factor

beta. Developmental & Comparative Immunology, 32(6), 654-663.

doi:10.1016/j.dci.2007.10.003

Hansen, J. D., & Strassburger, P. (2000). Description of an ectothermic TCR coreceptor, CD8α,

in rainbow trout. Journal of Immunology, 164(6), 3132-3139.

Page 141: Edwardsiella Ictaluri Is Capable of Persisting in Channel

132

Hawke, J., McWhorter, A., Steigerwalt, A. G., & Brenner, D. (1981). Edwardsiella ictaluri sp.

nov., the causative agent of Enteric Septicemia of Catfish. International Journal of

Systematic Bacteriology, 31, 396-400. doi:10.1099/00207713-31-4-396

Hawke, J. P. (1979). A Bacterium Associated with Disease of Pond Cultured Channel Catfish,

Ictalurus punctatus. Canadian Journal of Fisheries and Aquatic Sciences, 36(12), 1508-

1512. doi:10.1139/f79-219

Hensel, M., Shea, J. E., Waterman, S. R., Mundy, R., Nikolaus, T., Banks, G., . . . Holden, D. W.

(1998). Genes encoding putative effector proteins of the type III secretion system of

Salmonella pathogenicity island 2 are required for bacterial virulence and proliferation in

macrophages. Molecular Microbiology, 30(1), 163-174. doi:10.1046/j.1365-

2958.1998.01047.x

Herrero, M., de Lorenzo, V., & Timmis, K. N. (1990). Transposon vectors containing non-

antibiotic resistance selection markers for cloning and stable chromosomal insertion of

foreign genes in gram-negative bacteria. Journal of Bacteriology, 172(11), 6557-6567.

doi:10.1128/jb.172.11.6557-6567.1990

Hong, S., Zou, J., Crampe, M., Peddie, S., Scapigliati, G., Bols, N., . . . Secombes, C. J. (2001).

The production and bioactivity of rainbow trout (Oncorhynchus mykiss) recombinant IL-

1 beta. Veterinary Immunology and Immunopathology, 81(1-2), 1-14. doi:10.1016/s0165-

2427(01)00328-2

Iliev, D. B., Roach, J. C., Mackenzie, S., Planas, J. V., & Goetz, F. W. (2005). Endotoxin

recognition: in fish or not in fish? FEBS Letters, 579(29), 6519-6528.

doi:10.1016/j.febslet.2005.10.061

Israël, A. (2010). The IKK complex, a central regulator of NF-κB activation. Cold Spring Harb

Perspect Med, 2(3), a000158.

J. P. Hawke, R. M. D., R. L. Thune, A, C. Camus. (1998). ESC- Enteric Septecemia of Catfish.

Janda, J. M., Abbott, S. L., Kroske-Bystrom, S., Cheung, W. K., Powers, C., Kokka, R. P., &

Tamura, K. (1991). Pathogenic properties of Edwardsiella species. Journal of Clinical

Microbiology, 29(9), 1997-2001.

Jordan, G. W., & Hadley, W. K. (1969). Human infection with Edwardsiella tarda. Ann. intern.,

70(2), 283-288. doi:10.7326/0003-4819-70-2-283

Page 142: Edwardsiella Ictaluri Is Capable of Persisting in Channel

133

Kaler, P., Godasi, B. N., Augenlicht, L., & Klampfer, L. (2009). The NF-κB/AKT-dependent

induction of wnt signaling in colon cancer cells by macrophages and IL-1β. Cancer

Microenviron, 2(1), 69. doi:10.1007/s12307-009-0030-y

Kasornchandra, J., Rogers, W. A., & Plumb, J. A. (1987). Edwardsiella-Ictaluri from Walking

Catfish, Clarias-Batrachus L, in Thailand. Journal of Fish Diseases, 10(2), 137-138.

doi:DOI 10.1111/j.1365-2761.1987.tb00729.x

Kaufmann, S. H. (1993). Immunity to intracellular bacteria. Annual Review of Immunology,

11(1), 129-163.

Kelley, L. A., Mezulis, S., Yates, C. M., Wass, M. N., & Sternberg, M. J. E. (2015). The Phyre2

web portal for protein modeling, prediction and analysis. Nat Protoc, 10(6), 845-858.

doi:10.1038/nprot.2015.053

Kent, M., & Lyons, J. (1982). Edwardsiella ictaluri in the green knifefish, Eigemannia virescens.

Fish Health News.

Keskin, O., Secer, S. U., Izgur, M., Turkyilmaz, S., & Mkakosya, R. S. (2004). Edwardsiella

ictaluri infection in rainbow trout (Oncorhynchus mykiss). Turk J Vet Anim Sci, 28(4),

649-653.

Kimmey, J. M., & Stallings, C. L. (2016). Bacterial pathogens versus autophagy: Implications

for therapeutic interventions. Trends in Molecular Medicine, 22(12), 1060-1076.

doi:10.1016/j.molmed.2016.10.008

Klesius, P. H., & Sealey, W. M. (1995). Characteristics of serum antibody in Enteric Septicemia

of Catfish. Journal of Aquatic Animal Health, 7(3), 205-210. doi:10.1577/1548-

8667(1995)007<0205:Cosaie>2.3.Co;2

Kobe, B., & Kajava, A. V. (2001). The leucine-rich repeat as a protein recognition motif.

Current Opinion in Structural Biology, 11(6), 725-732.

Kordon, A. O., Abdelhamed, H., Ahmed, H., Baumgartner, W., Karsi, A., & Pinchuk, L. M.

(2019). Assessment of the live attenuated and wild-type Edwardsiella ictaluri-induced

immune gene expression and langerhans-like cell profiles in the immune-related organs

of catfish. Front Immunol, 10, 392. doi:10.3389/fimmu.2019.00392

Kordon, A. O., Abdelhamed, H., Ahmed, H., Park, J. Y., Karsi, A., & Pinchuk, L. M. (2017).

Phagocytic and bactericidal properties of channel catfish peritoneal macrophages exposed

Page 143: Edwardsiella Ictaluri Is Capable of Persisting in Channel

134

to Edwardsiella ictaluri live attenuated vaccine and wild-type strains. Front Microbiol,

8(2638), 2638. doi:10.3389/fmicb.2017.02638

Kordon, A. O., Kalindamar, S., Majors, K., Abdelhamed, H., Tan, W., Karsi, A., & Pinchuk, L.

M. (2019). Effects of live attenuated vaccine and wild type strains of Edwardsiella

ictaluri on phagocytosis, bacterial killing, and survival of catfish B cells. Front Immunol,

10, 2383. doi:10.3389/fimmu.2019.02383

Kovach, M. E., Elzer, P. H., Hill, D. S., Robertson, G. T., Farris, M. A., Roop, R. M., 2nd, &

Peterson, K. M. (1995). Four new derivatives of the broad-host-range cloning vector

pBBR1MCS, carrying different antibiotic-resistance cassettes. Gene, 166(1), 175-176.

doi:10.1016/0378-1119(95)00584-1

Lagos, L. X., Iliev, D. B., Helland, R., Rosemblatt, M., & Jorgensen, J. B. (2012). CD40L--a

costimulatory molecule involved in the maturation of antigen presenting cells in Atlantic

salmon (Salmo salar). Developmental & Comparative Immunology, 38(3), 416-430.

doi:10.1016/j.dci.2012.07.011

Laing, K. J., & Hansen, J. D. (2011). Fish T cells: recent advances through genomics.

Developmental & Comparative Immunology, 35(12), 1282-1295.

doi:10.1016/j.dci.2011.03.004

Lapaque, N., Hutchinson, J. L., Jones, D. C., Meresse, S., Holden, D. W., Trowsdale, J., & Kelly,

A. P. (2009). Salmonella regulates polyubiquitination and surface expression of MHC

class II antigens. PNAS, 106(33), 14052-14057. doi:10.1073/pnas.0906735106

Leddon, S. A., & Sant, A. J. (2010). Generation of MHC class II-peptide ligands for CD4 T-cell

allorecognition of MHC class II molecules. Curr Opin Organ Transplant, 15(4), 505-

511. doi:10.1097/MOT.0b013e32833bfc5c

Leng, N., Dawson, J. A., Thomson, J. A., Ruotti, V., Rissman, A. I., Smits, B. M., Kendziorski,

C. (2013). EBSeq: an empirical Bayes hierarchical model for inference in RNA-seq

experiments. Bioinformatics, 29(8), 1035-1043.

Li, C., Zhang, Y., Wang, R., Lu, J., Nandi, S., Mohanty, S., . . . Peatman, E. (2012). RNA-seq

analysis of mucosal immune responses reveals signatures of intestinal barrier disruption

and pathogen entry following Edwardsiella ictaluri infection in channel catfish, Ictalurus

punctatus. Fish & Shellfish Immunology, 32(5), 816-827.

Liu, T., Zhang, L., Joo, D., & Sun, S.-C. (2017). NF-κB signaling in inflammation. Signal

Transduct Target Ther, 2, 17023. doi:10.1038/sigtrans.2017.23

Page 144: Edwardsiella Ictaluri Is Capable of Persisting in Channel

135

Liu, Z., Liu, S., Yao, J., Bao, L., Zhang, J., Li, Y., Waldbieser, G. C. (2016). The channel catfish

genome sequence provides insights into the evolution of scale formation in teleosts. Nat

Commun, 7, 11757. doi:10.1038/ncomms11757

Mackey, M. F., Barth Jr, R. J., & Noelle, R. J. (1998). The role of CD40/CD154 interactions in

the priming, differentiation, and effector function of helper and cytotoxic T cells. Journal

of Leukocyte Biology, 63(4), 418-428.

Maiti, N. K., Mandal, A., Mohanty, S., & Mandal, R. N. (2009). Phenotypic and genetic

characterization of Edwardsiella tarda isolated from pond sediments. Comp Immunol

Microbiol Infect Dis, 32(1), 1-8. doi:10.1016/j.cimid.2007.11.001

Mariño, G., Niso-Santano, M., Baehrecke, E. H., & Kroemer, G. (2014). Self-consumption: the

interplay of autophagy and apoptosis. Nature Reviews: Molecular Cell Biology, 15(2),

81-94. doi:10.1038/nrm3735

Matsuyama, T., Kamaishi, T., Ooseko, N., Kurohara, K., & Iida, T. (2005). Pathogenicity of

motile and non-motile Edwardsiella tarda to some marine fish. Fish Pathology, 40(3),

133-135. doi:DOI 10.3147/jsfp.40.133

McShan, A. C., & De Guzman, R. N. (2015). The bacterial type III secretion system as a target

for developing new antibiotics. Chem Biol Drug Des, 85(1), 30-42.

doi:10.1111/cbdd.12422

Melcher, M., Unger, B., Schmidt, U., Rajantie, I. A., Alitalo, K., & Ellmeier, W. (2008).

Essential roles for the Tec family kinases Tec and Btk in M-CSF receptor signaling

pathways that regulate macrophage survival. Journal of Immunology, 180(12), 8048-

8056. doi:10.4049/jimmunol.180.12.8048

Meyer, F. P., & Bullock, G. L. (1973). Edwardsiella tarda, a new pathogen of channel catfish

(Ictalurus punctatus). Appl Microbiol, 25(1), 155-156.

Miao, E. A., & Miller, S. I. (2000). A conserved amino acid sequence directing intracellular type

III secretion by Salmonella typhimurium. PNAS, 97(13), 7539-7544.

Milev-Milovanovic, I., Long, S., Wilson, M., Bengten, E., Miller, N. W., & Chinchar, V. G.

(2006). Identification and expression analysis of interferon gamma genes in channel

catfish. Immunogenetics, 58(1), 70-80. doi:10.1007/s00251-006-0081-x

Page 145: Edwardsiella Ictaluri Is Capable of Persisting in Channel

136

Milev-Milovanovic, I., Majji, S., Thodima, V., Deng, Y., Hanson, L., Arnizaut, A., Chinchar, V.

G. (2009). Identification and expression analyses of poly [I:C]-stimulated genes in

channel catfish (Ictalurus punctatus). Fish & Shellfish Immunology, 26(5), 811-820.

doi:10.1016/j.fsi.2009.03.006

Miller, N. W., Rycyzyn, M. A., Wilson, M. R., Warr, G. W., Naftel, J. P., & Clem, L. W. (1994).

Development and characterization of channel catfish long term B cell lines. Journal of

Immunology, 152(5), 2180-2189.

Miyazaki, T., & Plumb, J. A. (1985). Histopathology of Edwardsiella-Ictaluri in channel catfish,

Ictalurus-Punctatus (Rafinesque). Journal of Fish Diseases, 8(4), 389-392. doi:DOI

10.1111/j.1365-2761.1985.tb00961.x

Mohanty, B. R., & Sahoo, P. K. (2007). Edwardsiellosis in fish: a brief review. Biosci, 32(7),

1331-1344. doi:10.1007/s12038-007-0143-8

Momburg, F., Koch, N., Möller, P., Moldenhauer, G., Butcher, G. W., & Hämmerling, G.

(1986). Differential expression of Ia and Ia-associated invariant chain in mouse tissues

after in vivo treatment with IFN-gamma. Immunology, 136(3), 940-948.

Moore, L. J., Dijkstra, J. M., Koppang, E. O., & Hordvik, I. (2009). CD4 homologues in Atlantic

salmon. Fish & Shellfish Immunology, 26(1), 10-18. doi:10.1016/j.fsi.2008.09.019

Moore, L. J., Somamoto, T., Lie, K. K., Dijkstra, J. M., & Hordvik, I. (2005). Characterisation of

salmon and trout CD8alpha and CD8beta. Molecular Immunology, 42(10), 1225-1234.

doi:10.1016/j.molimm.2004.11.017

Moresco, J. J., Carvalho, P. C., & Yates, J. R., 3rd. (2010). Identifying components of protein

complexes in C. elegans using co-immunoprecipitation and mass spectrometry. J

Proteomics, 73(11), 2198-2204. doi:10.1016/j.jprot.2010.05.008

Mori, M. (2007). Regulation of nitric oxide synthesis and apoptosis by arginase and arginine

recycling. Journal of Nutrition, 137(6), 1616S-1620S.

Mqolomba, T. N., & Plumb, J. A. (1992). Longevity of Edwardsiella-Ictaluri in the organs of

experimentally infected channel catfish, Ictalurus-Punctatus. Aquaculture, 101(1-2), 1-6.

doi:Doi 10.1016/0044-8486(92)90226-B

Nagai, T., Iwamoto, E., Sakai, T., Arima, T., Tensha, K., Iida, Y., . . . Nakai, T. (2008).

Characterization of Edwardsiella ictaluri Isolated from Wild Ayu Plecoglossus altivelis

in Japan. Fish Pathology, 43(4), 158-163. doi:DOI 10.3147/jsfp.43.158

Page 146: Edwardsiella Ictaluri Is Capable of Persisting in Channel

137

National Library of Medicine (US), N. C. f. B. I. (1988). National Center for Biotechnology

Information (NCBI) [Internet]. Retrieved from https://www.ncbi.nlm.nih.gov/

Newton, J., Wolfe, L., Grizzle, J., & Plumb, J. (1989). Pathology of experimental enteric

septicaemia in channel catfish, Ictalurus punctatus (rafinesque), following immersion‐

exposure to Edwardsiella ictaluri. Journal of Fish Diseases, 12(4), 335-347.

Nho, S. W., Abdelhamed, H., Karsi, A., & Lawrence, M. L. (2017). Improving safety of a live

attenuated Edwardsiella ictaluri vaccine against enteric septicemia of catfish and

evaluation of efficacy. Veterinary Microbiology, 210, 83-90.

doi:10.1016/j.vetmic.2017.09.004

Nordeng, T. W., Gregers, T. F., Kongsvik, T. L., Méresse, S., Gorvel, J.-P., Jourdan, F., Bakke,

O. (2002). The cytoplasmic tail of invariant chain regulates endosome fusion and

morphology. Mol Biol Cell, 13(6), 1846-1856.

Orecchioni, M., Ghosheh, Y., Pramod, A. B., & Ley, K. (2019). Macrophage polarization:

different gene signatures in M1(LPS+) vs. classically and M2(LPS–) vs. alternatively

activated macrophages. Front Immunol, 10(1084). doi:10.3389/fimmu.2019.01084

Pallen, M. J., Beatson, S. A., & Bailey, C. M. (2005). Bioinformatics, genomics and evolution of

non-flagellar type-III secretion systems: a Darwinian perspective. FEMS Microbiology

Reviews, 29(2), 201-229. doi:10.1016/j.femsre.2005.01.001

Panangala, V. S., Shoemaker, C. A., McNulty, S. T., Arias, C. R., & Klesius, P. H. (2006). Intra‐

and interspecific phenotypic characteristics of fish‐pathogenic Edwardsiella ictaluri and

E. tarda. Aquaculture Research, 37(1), 49-60.

Park, S.-I., Wakabayashi, H., & Watanabe, Y. (1983). Serotype and virulence of Edwardsiella

tarda isolated from eel and their environment. Fish Pathology, 18(2), 85-89.

Patel, S., Overgård, A.-C., & Nerland, A. H. (2008). CD8alpha and CD8beta in Atlantic halibut,

Hippoglossus hippoglossus: cloning, characterization and gene expression during viral

and bacterial infection. Fish & Shellfish Immunology, 25(5), 570-580.

doi:10.1016/j.fsi.2008.08.007

Paul, G. (2012). Aquaculture in the United States of America: present status and future

opportunities. Bull. Fish. Res. Agen. No, 35, 7-13.

Page 147: Edwardsiella Ictaluri Is Capable of Persisting in Channel

138

Peatman, E., Terhune, J., Baoprasertkul, P., Xu, P., Nandi, S., Wang, S., Liu, Z. (2008).

Microarray analysis of gene expression in the blue catfish liver reveals early activation of

the MHC class I pathway after infection with Edwardsiella ictaluri. Molecular

Immunology, 45(2), 553-566. doi:https://doi.org/10.1016/j.molimm.2007.05.012

Peddie, S., Zou, J., & Secombes, C. J. (2002). A biologically active IL-1beta derived peptide

stimulates phagocytosis and bactericidal activity in rainbow trout, Oncorhynchus mykiss

(Walbaum), head kidney leucocytes in vitro. Journal of Fish Diseases, 25(6), 351-360.

doi:10.1046/j.1365-2761.2002.00380.x

Pierrard, M.-A., Roland, K., Kestemont, P., Dieu, M., Raes, M., & Silvestre, F. (2012). Fish

peripheral blood mononuclear cells preparation for future monitoring applications.

Analytical Biochemistry, 426(2), 153-165. doi:https://doi.org/10.1016/j.ab.2012.04.009

Pinchuk, L. M., Polacino, P. S., Agy, M. B., Klaus, S. J., & Clark, E. A. (1994). The role of

CD40 and CD80 accessory cell molecules in dendritic cell-dependent HIV-1 infection.

Immunity, 1(4), 317-325.

Plumb, J. A., & Quinlan, E. E. (1986). Survival of Edwardsiella ictaluri in pond water and

bottom mud. Progressive Fish-Culturist, 48(3), 212-214. doi:Doi 10.1577/1548-

8640(1986)48<212:Soeiip>2.0.Co;2

Pohlenz, C., Buentello, A., Mwangi, W., & Gatlin, D. M., 3rd. (2012). Arginine and glutamine

supplementation to culture media improves the performance of various channel catfish

immune cells. Fish & Shellfish Immunology, 32(5), 762-768.

doi:10.1016/j.fsi.2012.01.029

Ramona, T. S., & Thomas, J. B. (2002). Edwardsiella ictaluri invasion of IEC-6, Henle 407,

fathead minnow and channel catfish enteric epithelial cells. Diseases of Aquatic

Organisms, 51(3), 161-167.

Reis, P. P., Waldron, L., Goswami, R. S., Xu, W., Xuan, Y., Perez-Ordonez, B., Kamel-Reid, S.

(2011). mRNA transcript quantification in archival samples using multiplexed, color-

coded probes. BMC Biotechnology, 11(1), 46.

Rogge, M. (2009). Expression of the Edwardsiella ictaluri type III secretion system and its

relationship to the native plasmids. (Doctor of Philosophy), Louisiana State University

and Agricultural and Mechanical College

Page 148: Edwardsiella Ictaluri Is Capable of Persisting in Channel

139

Rogge, M. L., & Thune, R. L. (2011). Regulation of the Edwardsiella ictaluri type III secretion

system by pH and phosphate concentration through EsrA, EsrB, and EsrC. Applied and

Environmental Microbiology, 77(13), 4293-4302. doi:10.1128/AEM.00195-11

Russo, R., Shoemaker, C. A., Panangala, V. S., & Klesius, P. H. (2009). In vitro and in vivo

interaction of macrophages from vaccinated and non-vaccinated channel catfish

(Ictalurus punctatus) to Edwardsiella ictaluri. Fish & Shellfish Immunology, 26(3), 543-

552. doi:10.1016/j.fsi.2009.02.011

Sakai, T., Yuasa, K., Sano, M., & Iida, T. (2009). Identification of Edwardsiella ictaluri and E.

tarda by species-specific polymerase chain reaction targeted to the upstream region of the

fimbrial gene. Journal of Aquatic Animal Health, 21(2), 124-132. doi:10.1577/H08-061.1

Santander, J., Martin, T., Loh, A., Pohlenz, C., Gatlin III, D. M., & Curtiss III, R. (2013).

Mechanisms of intrinsic resistance to antimicrobial peptides of Edwardsiella ictaluri and

its influence on fish gut inflammation and virulence. Microbiology, 159(Pt 7), 1471.

Schroder, B. (2016). The multifaceted roles of the invariant chain CD74-more than just a

chaperone. Biochimica et Biophysica Acta (BBA) - Bioenergetics, 1863(6 Pt A), 1269-

1281. doi:10.1016/j.bbamcr.2016.03.026

Schroder, K., Hertzog, P. J., Ravasi, T., & Hume, D. A. (2004). Interferon-gamma: an overview

of signals, mechanisms and functions. J Leukoc Biol, 75(2), 163-189.

doi:10.1189/jlb.0603252

Shao, S., Lai, Q., Liu, Q., Wu, H., Xiao, J., Shao, Z., Zhang, Y. (2015). Phylogenomics

characterization of a highly virulent Edwardsiella strain ET080813(T) encoding two

distinct T3SS and three T6SS gene clusters: propose a novel species as Edwardsiella

anguillarum sp. nov. Systematic and Applied Microbiology, 38(1), 36-47.

doi:10.1016/j.syapm.2014.10.008

Shotts, E., Blazer, V., & Waltman, W. (1986). Pathogenesis of experimental Edwardsiella

ictaluri infections in channel catfish (Ictalurus punctatus). Canadian Journal of Fisheries

and Aquatic Sciences, 43(1), 36-42.

Silverman, J. M., Brunet, Y. R., Cascales, E., & Mougous, J. D. (2012). Structure and Regulation

of the Type VI Secretion System. Annual Review of Microbiology, 66, 453-472.

doi:10.1146/annurev-micro-121809-151619

Page 149: Edwardsiella Ictaluri Is Capable of Persisting in Channel

140

Somamoto, T., Yoshiura, Y., Sato, A., Nakao, M., Nakanishi, T., Okamoto, N., & Ototake, M.

(2006). Expression profiles of TCRbeta and CD8alpha mRNA correlate with virus-

specific cell-mediated cytotoxic activity in ginbuna crucian carp. Virology, 348(2), 370-

377. doi:10.1016/j.virol.2006.01.019

Spencer, D. A., Quiniou, S. M. A., Crider, J., Musungu, B., Bengten, E., & Wilson, M. (2019).

Insights into the dynamics of memory, effector and apoptotic cytotoxic T lymphocytes in

channel catfish, Ictalurus punctatus. Developmental & Comparative Immunology, 92,

116-128. doi:10.1016/j.dci.2018.11.001

Speranza, E., Altamura, L. A., Kulcsar, K., Bixler, S. L., Rossi, C. A., Schoepp, R. J., Connor, J.

H. (2017). Comparison of transcriptomic platforms for analysis of whole blood from

Ebola-infected Cynomolgus Macaques. Sci Rep, 7(1), 14756. doi:10.1038/s41598-017-

15145-7

Srinivasa Rao, P., Yamada, Y., Tan, Y. P., & Leung, K. Y. (2004). Use of proteomics to identify

novel virulence determinants that are required for Edwardsiella tarda pathogenesis.

Molecular Microbiology, 53(2), 573-586.

Stoppe, C., Rex, S., Goetzenich, A., Kraemer, S., Emontzpohl, C., Soppert, J., . . . Lue, H.

(2015). Interaction of MIF family proteins in myocardial ischemia/reperfusion damage

and their influence on clinical outcome of cardiac surgery patients. Antioxidants and

Redox Signaling, 23(11), 865-879.

Sun, F., Peatman, E., Li, C., Liu, S., Jiang, Y., Zhou, Z., & Liu, Z. (2012). Transcriptomic

signatures of attachment, NF-κB suppression and IFN stimulation in the catfish gill

following columnaris bacterial infection. Developmental & Comparative Immunology,

38(1), 169-180. doi:https://doi.org/10.1016/j.dci.2012.05.006

Sun, X.-F., Shang, N., Hu, W., Wang, Y.-P., & Guo, Q.-L. (2007). Molecular cloning and

characterization of carp (Cyprinus carpio L.) CD8beta and CD4-like genes. Fish &

Shellfish Immunology, 23(6), 1242-1255. doi:10.1016/j.fsi.2007.06.004

Takizawa, F., Koppang, E. O., Ohtani, M., Nakanishi, T., Hashimoto, K., Fischer, U., & Dijkstra,

J. M. (2011). Constitutive high expression of interleukin-4/13A and GATA-3 in gill and

skin of salmonid fishes suggests that these tissues form Th2-skewed immune

environments. Molecular Immunology, 48(12-13), 1360-1368.

doi:10.1016/j.molimm.2011.02.014

Tamura, K., Sakazaki, R., McWhorter, A. C., & Kosako, Y. (1988). Edwardsiella tarda

serotyping scheme for international use. Journal of Clinical Microbiology, 26(11), 2343-

2346.

Page 150: Edwardsiella Ictaluri Is Capable of Persisting in Channel

141

Tan, S., Wang, W., Tian, C., Niu, D., Zhou, T., Yang, Y., Liu, Z. (2019). Post-transcriptional

regulation through alternative splicing after infection with Flavobacterium columnare in

channel catfish (Ictalurus punctatus). Fish & Shellfish Immunology, 91, 188-193.

doi:https://doi.org/10.1016/j.fsi.2019.05.008

Tan, S., Wang, W., Zhou, T., Yang, Y., Gao, D., Dunham, R., & Liu, Z. (2019). Polyadenylation

sites and their characteristics in the genome of channel catfish (Ictalurus punctatus) as

revealed by using RNA-Seq data. Comp Biochem Physiol D: Genomics Proteomics, 30,

248-255. doi:https://doi.org/10.1016/j.cbd.2019.03.008

Tekedar, H. C., Blom, J., Kalindamar, S., Nho, S., Karsi, A., & Lawrence, M. L. (2020).

Comparative genomics of the fish pathogens Edwardsiella ictaluri 93-146 and

Edwardsiella piscicida C07-087. Microb Genom, 6(2). doi:10.1099/mgen.0.000322

Thune, R., A. Stanley, L., & Cooper, R. (1993). Pathogenesis of Gram-negative bacterial

infection in warm water fish. Annual Review of Fish Diseases, 3, 37-68.

doi:10.1016/0959-8030(93)90028-A

Thune, R. L., Fernandez, D. H., & Battista, J. R. (1999). An aroA mutant of Edwardsiella

ictaluri is safe and efficacious as a live, attenuated vaccine. Journal of Aquatic Animal

Health, 11(4), 358-372. doi:10.1577/1548-8667(1999)011<0358:Aamoei>2.0.Co;2

Thune, R. L., Fernandez, D. H., Benoit, J. L., Kelly-Smith, M., Rogge, M. L., Booth, N. J., . . .

Bologna, R. A. (2007). Signature-Tagged Mutagenesis of Edwardsiella ictaluri Identifies

Virulence-Related Genes, Including a Salmonella Pathogenicity Island 2 Class of Type

III Secretion Systems. Applied and Environmental Microbiology, 73(24), 7934-7946.

doi:10.1128/aem.01115-07

Thune, R. L., Fernandez, D. H., Benoit, J. L., Kelly-Smith, M., Rogge, M. L., Booth, N. J., . . .

Bologna, R. A. (2007). Signature-tagged mutagenesis of Edwardsiella ictaluri identifies

virulence-related genes, including a salmonella pathogenicity island 2 class of type III

secretion systems. Applied and Environmental Microbiology, 73(24), 7934-7946.

doi:10.1128/AEM.01115-07

Tucker, C. C., & Robinson, E. H. (1990). Channel catfish farming handbook: Springer Science

& Business Media.

Ucko, M., Colorni, A., Dubytska, L., & Thune, R. L. (2016). Edwardsiella piscicida-like

pathogen in cultured grouper. Diseases of Aquatic Organisms, 121(2), 141-148.

doi:10.3354/dao03051

Page 151: Edwardsiella Ictaluri Is Capable of Persisting in Channel

142

Uribe, C., Folch, H., Enríquez, R., & Moran, G. (2011). Innate and adaptive immunity in teleost

fish: a review. Veterinarni Medicina, 56(10), 486-503.

USDA. (2010). USDA Catfish 2010: Part III: Changes in Catfish Health and Production

Practices in the United States, 2002-2009. Retrieved from

Vergadi, E., Ieronymaki, E., Lyroni, K., Vaporidi, K., & Tsatsanis, C. (2017). Akt signaling

pathway in macrophage activation and M1/M2 polarization. J Immunol, 198(3), 1006-

1014. doi:10.4049/jimmunol.1601515

Victor, S. P., Craig, A. S., Vicky, L. v. S., Kevin, D., & Phillip, H. K. (2007). Multiplex-PCR for

simultaneous detection of 3 bacterial fish pathogens, Flavobacterium columnare,

Edwardsiella ictaluri, and Aeromonas hydrophila. Diseases of Aquatic Organisms, 74(3),

199-208.

Vinitnantharat, S., & Plumb, J. A. (1993). Protection of channel catfish Ictalurus punctatus

following natural exposure to Edwardsiella ictaluri and effects of feeding antigen on

antibody titer. Diseases of Aquatic Organisms, 15, 31-34. doi:10.3354/dao015031

Vishwakarma, V., Pati, N. B., Chandel, H. S., Sahoo, S. S., Saha, B., & Suar, M. (2012).

Evaluation of Salmonella enterica serovar Typhimurium TTSS-2 deficient fur mutant as

safe live-attenuated vaccine candidate for immunocompromised mice. PLoS One, 7(12),

e52043. doi:10.1371/journal.pone.0052043

Wajant, H., & Siegmund, D. (2019). TNFR1 and TNFR2 in the control of the life and death

balance of macrophages. Front Cell Dev Biol, 7, 91-91. doi:10.3389/fcell.2019.00091

Wakabayashi, H. (1973). Edwardsiella tarda (Paracolobacterium anguillimortiferum) associated

with pond-cultured eel diseases. Bul Japan Soc Sci Fish, 39, 931-939.

Wang, X., Li, C., Thongda, W., Luo, Y., Beck, B., & Peatman, E. (2014). Characterization and

mucosal responses of interleukin 17 family ligand and receptor genes in channel catfish

Ictalurus punctatus. Fish & Shellfish Immunology, 38(1), 47-55.

doi:10.1016/j.fsi.2014.02.020

Wang, X., Liu, S., Yang, Y., Fu, Q., Abebe, A., & Liu, Z. (2017). Identification of NF-kappaB

related genes in channel catfish and their expression profiles in mucosal tissues after

columnaris bacterial infection. Developmental & Comparative Immunology, 70, 27-38.

doi:10.1016/j.dci.2017.01.003

Page 152: Edwardsiella Ictaluri Is Capable of Persisting in Channel

143

Wang, Y., Wang, Q., Baoprasertkul, P., Peatman, E., & Liu, Z. (2006). Genomic organization,

gene duplication, and expression analysis of interleukin-1beta in channel catfish

(Ictalurus punctatus). Molecular Immunology, 43(10), 1653-1664.

doi:10.1016/j.molimm.2005.09.024

Waterstrat, P. R., Ainsworth, A. J., & Capley, G. (1991). In vitro responses of channel catfish,

Ictalurus punctatus, neutrophils to Edwardsiella ictaluri. Developmental & Comparative

Immunology, 15(1-2), 53-63. doi:10.1016/0145-305x(91)90047-3

Wei, H., Yin, L., Feng, S., Wang, X., Yang, K., Zhang, A., & Zhou, H. (2015). Dual-parallel

inhibition of IL-10 and TGF-β1 controls LPS-induced inflammatory response via NF-κB

signaling in grass carp monocytes/macrophages. Fish & Shellfish Immunology, 44(2),

445-452.

Wei, L., & Musa, N. (2008). Phenotyping, genotyping and whole cell protein profiling of

Edwardsiella tarda isolated from cultured and natural habitat freshwater fish. Am

Eurasian J Agric Environ Sci, 3.

Wilson, B. A., Salyers, A. A., Whitt, D. D., & Winkler, M. E. (2011). Bacterial pathogenesis: a

molecular approach: American Society for Microbiology (ASM).

Wise, D. J., Greenway, T. E., Byars, T. S., Griffin, M. J., & Khoo, L. H. (2015). Oral vaccination

of channel catfish against Enteric Septicemia of Catfish using a live attenuated

Edwardsiella ictaluri isolate. Journal of Aquatic Animal Health, 27(2), 135-143.

doi:10.1080/08997659.2015.1032440

Wolfe, M. S. (2009). Intramembrane proteolysis. Chem Rev, 109(4), 1599-1612.

Wu, M.-Y., & Lu, J.-H. (2020). Autophagy and macrophage functions: inflammatory response

and phagocytosis. Cells, 9(1), 70.

Xie, H. X., Yu, H. B., Zheng, J., Nie, P., Foster, L. J., Mok, Y.-K., Leung, K. Y. (2010). EseG,

an effector of the type III secretion system of Edwardsiella tarda, triggers microtubule

destabilization. Infection and Immunity, 78(12), 5011-5021.

Xu, Z., Chen, J., Li, X., Ge, J., Pan, J., & Xu, X. (2013). Identification and characterization of

microRNAs in channel catfish (Ictalurus punctatus) by using Solexa sequencing

technology. PloS One, 8(1), e54174-e54174. doi:10.1371/journal.pone.0054174

Xue, Q., Yan, Y., Zhang, R., & Xiong, H. (2018). Regulation of iNOS on immune cells and its

role in diseases. Int J Mol Sci, 19(12), 3805. doi:10.3390/ijms19123805

Page 153: Edwardsiella Ictaluri Is Capable of Persisting in Channel

144

Yang, M., Lv, Y., Xiao, J., Wu, H., Zheng, H., Liu, Q., Wang, Q. (2012). Edwardsiella

comparative phylogenomics reveal the new intra/inter-species taxonomic relationships,

virulence evolution and niche adaptation mechanisms. PLoS One, 7(5), e36987.

doi:10.1371/journal.pone.0036987

Yang, Q., Pan, Y. L., Wang, K. Y., Wang, J., He, Y., Wang, E. L., Huang, X. L. (2016). OmpN,

outer membrane proteins of Edwardsiella ictaluri are potential vaccine candidates for

channel catfish (Ictalurus punctatus). Molecular Immunology, 78, 1-8.

doi:10.1016/j.molimm.2016.08.011

Yasunaga, N. (1982). Characteristics of the fish pathogen Edwardsiella isolated from several

species of cultured marine fish. Bulletin of Nagasaki Prefectural Institute of Fisheries, 8,

67-73.

Yuasa, K., Kholidin, E. B., Panigoro, N., & Hatai, K. (2003). First isolation of Edwardsiella

ictaluri from cultured striped catfish Pangasius hypophthalmus in Indonesia. Fish

Pathology, 38(4), 181-183. doi:DOI 10.3147/jsfp.38.181

Yunna, C., Mengru, H., Lei, W., & Weidong, C. (2020). Macrophage M1/M2 polarization.

European Journal of Pharmacology, 877, 173090. doi:10.1016/j.ejphar.2020.173090

Zaccone, G. (2009). Fish Defenses Volume 1: Immunology (G. Zaccone Ed. Vol. 1): Science

Publishers.

Zhang, A., Chen, D., Wei, H., Du, L., Zhao, T., Wang, X., & Zhou, H. (2012). Functional

characterization of TNF-α in grass carp head kidney leukocytes: induction and

involvement in the regulation of NF-κB signaling. Fish & Shellfish Immunology, 33(5),

1123-1132.

Zheng, J., Li, N., Tan, Y. P., Sivaraman, J., Mok, Y.-K., Mo, Z. L., & Leung, K. Y. (2007). EscC

is a chaperone for the Edwardsiella tarda type III secretion system putative translocon

components EseB and EseD. Microbiology, 153(6), 1953-1962.

Zhou, T., Yuan, Z., Tan, S., Jin, Y., Yang, Y., Shi, H., . . . Liu, Z. (2018). A review of molecular

responses of catfish to bacterial diseases and abiotic stresses. Front Physiol, 9(1113),

1113. doi:10.3389/fphys.2018.01113

Zhou, Y. Y., Li, Y., Jiang, W. Q., & Zhou, L. F. (2015). MAPK/JNK signalling: a potential

autophagy regulation pathway. Biosci Rep, 35(3). doi:10.1042/BSR20140141

Page 154: Edwardsiella Ictaluri Is Capable of Persisting in Channel

145

Zou, J., Peddie, S., Scapigliati, G., Zhang, Y., Bols, N. C., Ellis, A. E., & Secombes, C. J. (2003).

Functional characterisation of the recombinant tumor necrosis factors in rainbow trout,

Oncorhynchus mykiss. Developmental & Comparative Immunology, 27(9), 813-822.

doi:10.1016/s0145-305x(03)00077-6

Zou, J., & Secombes, C. J. (2011). Teleost fish interferons and their role in immunity.

Developmental & Comparative Immunology, 35(12), 1376-1387.

doi:10.1016/j.dci.2011.07.001

Zou, J., & Secombes, C. J. (2016). The function of fish cytokines. Biology, 5(2).

doi:10.3390/biology5020023

Page 155: Edwardsiella Ictaluri Is Capable of Persisting in Channel

146

VITA

Elizabeth Griggs was born in Baton Rouge, Louisiana in December, 1988. She lived with her

parents, Thomas and Cathie Griggs, and her older sister Catherine through her early years. She

attended St. Alphonsus School during elementary and middle school, followed by high school at

St. Joseph’s Academy. While in high school she developed an interest in volunteer service and

was honored with an award for service in her senior year. After graduation, Elizabeth left Baton

Rouge to attend Louisiana Tech University in the much smaller city of Ruston, Louisiana. She

was studying Environmental Science when she happened to enroll in a class in microbiology

decided that she had found her calling. After receiving her bachelor’s degree in Environmental

Science, and with Dr. Giorno-McConnell’s urging, she enrolled in a master’s program in

Microbiology, and two years later received her master’s degree. During her years at Louisiana

Tech, she discovered a deep appreciation for the outdoors, and ever since then has enjoyed

outdoor activities such as camping and obstacle course races. After graduation Elizabeth moved

back home to attend the school several generations of her family have matriculated through,

Louisiana State University.