overview of gut flora and probiotics.pdf

17
International Journal of Food Microbiology 41 (1998) 85–101 Overview of gut flora and probiotics a, a b a * Wilhelm H. Holzapfel , Petra Haberer , Johannes Snel , Ulrich Schillinger , b Jos H.J. Huis in’t Veld a Institute of Hygiene and Toxicology, Federal Research Centre for Nutrition, Engesserstr. 20, D-76131 Karlsruhe, Germany b Department of the Science of Food of Animal Origin, Faculty of Veterinary Medicine, Utrecht University, P .O. Box 80175, Yalelaan 2, NL-3508 TD Utrecht, The Netherlands Accepted 6 March 1998 Abstract Scientific developments in recent years have opened new frontiers and enable a better understanding of the gastro- intestinal tract (GIT) as a complex and delicately balanced ecosystem. This paper focuses on more recent information related to the microbial population of the GIT and its functional role in human physiology and health. Special attention is also given to modern approaches for improving or stabilising the intestinal system and its functioning by the deliberate application of viable microbial cultures, so-called ‘probiotics’, selected for special functional properties. 1998 Elsevier Science B.V. Keywords: Bifidobacteria; Functional properties; Gastro-intestinal tract; Lactobacilli; Probiotics 1. Introduction attempts have been made, especially during the last two to three decades, to improve the health status by There is general agreement on the important role modulating the indigenous intestinal flora by live of the gastro-intestinal (GI) microflora in the health microbial adjuncts, now called ‘probiotics’. Although status of men and animals. The importance of a number of definitions have been proposed to lactobacilli for human health and longevity was first describe probiotics, an appropriate one was sug- hypothesized by Metchnikoff at the beginning of this gested by Havenaar et al. (1992), according to which century. He, however, considered the gut microbes in probiotics are defined as ‘‘mono- or mixed cultures total as detrimental rather than beneficial, and sug- of live microorganisms which, when applied to gested that desirable effects might only be expected animal or man, beneficially affect the host by from their substitution by yogurt bacteria. Since then, improving the properties of the indigenous microfl- ora’’. This definition has certain advantages com- pared to others, e.g.: * Corresponding author. IHT / BFE, Engesserstr. 20, D-76131 Karlsruhe, Germany. Tel.: 1 49 721 6625 115; fax: 1 49 721 6625 142; e-mail: [email protected] it does not restrict ‘probiotic’ activities to the 0168-1605 / 98 / $19.00 1998 Elsevier Science B.V. All rights reserved. PII: S0168-1605(98)00044-0

Upload: milu1312

Post on 15-Apr-2016

39 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Overview of gut flora and probiotics.pdf

International Journal of Food Microbiology 41 (1998) 85–101

Overview of gut flora and probiotics

a , a b a*Wilhelm H. Holzapfel , Petra Haberer , Johannes Snel , Ulrich Schillinger ,bJos H.J. Huis in’t Veld

aInstitute of Hygiene and Toxicology, Federal Research Centre for Nutrition, Engesserstr. 20, D-76131 Karlsruhe, GermanybDepartment of the Science of Food of Animal Origin, Faculty of Veterinary Medicine, Utrecht University, P.O. Box 80175, Yalelaan 2,

NL-3508 TD Utrecht, The Netherlands

Accepted 6 March 1998

Abstract

Scientific developments in recent years have opened new frontiers and enable a better understanding of the gastro-intestinal tract (GIT) as a complex and delicately balanced ecosystem. This paper focuses on more recent information relatedto the microbial population of the GIT and its functional role in human physiology and health. Special attention is also givento modern approaches for improving or stabilising the intestinal system and its functioning by the deliberate application ofviable microbial cultures, so-called ‘probiotics’, selected for special functional properties. 1998 Elsevier Science B.V.

Keywords: Bifidobacteria; Functional properties; Gastro-intestinal tract; Lactobacilli; Probiotics

1. Introduction attempts have been made, especially during the lasttwo to three decades, to improve the health status by

There is general agreement on the important role modulating the indigenous intestinal flora by liveof the gastro-intestinal (GI) microflora in the health microbial adjuncts, now called ‘probiotics’. Althoughstatus of men and animals. The importance of a number of definitions have been proposed tolactobacilli for human health and longevity was first describe probiotics, an appropriate one was sug-hypothesized by Metchnikoff at the beginning of this gested by Havenaar et al. (1992), according to whichcentury. He, however, considered the gut microbes in probiotics are defined as ‘‘mono- or mixed culturestotal as detrimental rather than beneficial, and sug- of live microorganisms which, when applied togested that desirable effects might only be expected animal or man, beneficially affect the host byfrom their substitution by yogurt bacteria. Since then, improving the properties of the indigenous microfl-

ora’’. This definition has certain advantages com-pared to others, e.g.:*Corresponding author. IHT/BFE, Engesserstr. 20, D-76131

Karlsruhe, Germany. Tel.: 1 49 721 6625 115; fax: 1 49 7216625 142; e-mail: [email protected] • it does not restrict ‘probiotic’ activities to the

0168-1605/98/$19.00 1998 Elsevier Science B.V. All rights reserved.PI I : S0168-1605( 98 )00044-0

Page 2: Overview of gut flora and probiotics.pdf

86 W.H. Holzapfel et al. / International Journal of Food Microbiology 41 (1998) 85 –101

intestinal microflora, but also to microbial com- 2. Development, composition and function of themunities at other sites of the body; microbial population in the intestinal system

• the ‘probiotic’ might consist of more than onebacterial species; and 2.1. The ecosystem

• it can be applied to both man and animals.The bacterial flora of humans is the most intimate

Several studies have focused on the phar- portion of their biological environment and mediatesmacokinetics of different probiotics in humans, and many interactions with the chemical environment. Inour knowledge, especially of the complex mecha- particular, the gastro-intestinal microflora representsnisms behind these effects, is constantly increasing. an ecosystem of the highest complexity and ourToday we know that certain lactic acid bacteria understanding of this system and its interactions is(LAB) can induce specific immune regulators as a still limited (Berg, 1996). This complex microbialresult of interaction with mononuclear phagocytes population may be considered as ‘‘an open eco-and endothelial cells of the host (Brassart and system comprising a group of microbial populationsSchiffrin, 1997). Furthermore, it has been observed coexisting in an equilibrium in a spatiotemporallythat particular strains of LAB showed adjuvant defined region’’ (Ducluzeau, 1989).

2properties by stimulation of a specific antibody Compared to the ca. 2 m skin surface of our bodyresponse after infection with (attenuated) pathogenic the GI tract (GIT) represents a much larger contactmicroorganisms (Pouwels et al., 1996). Another area with the environment (Van Dijk, 1997). Asintriguing development is the observation that certain shown in Fig. 1, the mucosal surface of the smallLAB can strengthen the gut mucosal barrier and intestine is increased in several ways, e.g.: three-foldthereby influence gut mucosal permeability and by forming circular folds, 7–10-fold by folding ofpossible diarrhoea. This paper will evaluate the the epithelium (intestinal villi) and 15–40-fold bypresent status of probiotics and discuss recent de- the formation of microvilli in the enterocyte resorp-velopments in this area. tive luminal membrane. The resulting surface of the

Fig. 1. Increase in the mucosal surface by folding (modified after Waldeck, 1990).

Page 3: Overview of gut flora and probiotics.pdf

W.H. Holzapfel et al. / International Journal of Food Microbiology 41 (1998) 85 –101 87

2GI system is calculated to be 150–200 m (Waldeck, explained only by numbers of leading species, and1990), thereby providing the necessary space for their definition therefore has limitations. Further-interactions during the digestive process and for more, studies on sampled material do not necessarilyadhesion to the mucosal wall and concomitant reflect intimate microbial interactions and in vivocolonisation. The GIT of an adult human is estimated growth kinetics.

14to harbour about 10 viable bacteria (Luckey andFloch, 1972), i.e. 10 times the total number of 2.2. Microbial numbers and the ecosystemeukaryotic cells in all tissues of man’s body. Theimportance of these bacteria in the GIT was neg- Both the variety and the overall sum of microbiallected for a long time, while the focus was merely numbers are determined by an array of complexplaced on enteric pathogens and other factors leading factors, intrinsic to the respective GI sections, andto gastro-intestinal ‘disorders’. ‘extrinsic’ related to, for example, diet, stress, drugs,

The intact intestinal epithelium with an optimal etc. After the more or less neutral pH of the oralintestinal flora represents a barrier to the invasion or cavity, the low pH of the stomach (ranging from 2.5uptake of pathogenic microorganisms, antigens and to 3.5) is destructive to most microbes. The popula-

3 21harmful compounds from the gut lumen. In addition tion averages 10 bacteria g , and is dominated byto the barrier function, the intestinal mucosa is Gram-positive bacteria such as streptococci andefficient in assimilating antigens. Specialised antigen lactobacilli, and by yeasts (see Fig. 2). Due to thetransport mechanisms present in the villus epithelium aggressive intestinal fluids (e.g., bile, pancreaticand Peyer’s patches are essential for evoking specific juices) and the short transit time, the duodenum alsoimmune responses (Heyman et al., 1982). In healthy represents a hostile environment and contains rela-individuals this barrier is stable, ensuring host tively low numbers of merely transient microbes. Inprotection and providing normal intestinal function the early 1960s, Reuter and coworkers revealed newand immunological resistance. Protection against and carefully elaborated data on the microbial popu-infective agents is complemented intrinsically by the lation of the different sections of the human GITbarriers of the gut-associated lymphoid tissues (Lerche and Reuter, 1962; Reuter, 1965a,b, 1969).(GALT), considered to be the largest ‘immune Further comprehensive data of such an extent haveorgan’ in the human body. Per meter of small bowel, not been added to our knowledge yet. Research in

10approximately 80% (10 ) of all immunoglobulin the following two decades concentrated on the easilyproducing cells are found here (Shanahan, 1994). available fecal flora and, so far, no comprehensiveThe gut flora itself is essential for mucosal immune evaluation has been made using up-to-date mi-stimulation (activation) and amplification of im-munocompetent cells.

A number of terms are used in the literature todescribe either the ‘state of balance’ within themicrobial population or the status of particularmicrobial groups within the GIT. The balance,considered to be maintained by sensitive interactionsbetween living and abiotic compounds in this ‘en-clave of external environment’ (Ducluzeau, 1989),can be called ‘eubiosis’. The opposite situation istermed ‘dysbiosis’ (Haenel and Bendig, 1975;Gedek, 1993). Although not precisely defined, thisunstable state refers to qualitative and quantitativechanges in the intestinal flora, their metabolic activi-ty and their local distribution. Metabolic activity andcertain turnover rates may be more important thanactual numbers of particular bacterial species. The Fig. 2. Microbial colonisation of the human gastro-intestinal tractsituations ‘eubiosis’ and ‘dysbiosis’ may not be (modified after Simon and Gorbach, 1982).

Page 4: Overview of gut flora and probiotics.pdf

88 W.H. Holzapfel et al. / International Journal of Food Microbiology 41 (1998) 85 –101

crobiological means of sampling and identification. the composition of these populations may remainStill, the relatively sparse data available on the relatively constant for some individuals and mayhuman jejunum and ileum suggest a continuous fluctuate considerably for others.

8 21increase in the numbers (up to 10 g ) and variety Minor groups of pathogenic and opportunisticof the flora towards the distal regions (Lerche and organisms, the so-called ‘residual flora’ according toReuter, 1962; Reuter, 1965b; Bhat et al., 1989; Gedek (1993), are always present in low numbers. InBernhardt and Knoke, 1980; Nielsen et al., 1994). In a healthy state, the quantity even of toxic metabolitesaddition to the LAB groups, with increasing numbers seems insufficient to act detrimentally on the host.of bifidobacteria towards the more distal regions, Ducluzeau (1989) proposed the action on the host toGram-negative facultative aerobic groups such as be considerable only when the number of particular

7 21Enterobacteriaceae, and the obligatory anaerobic bacteria exceeds 5 3 10 g . This may be the casegenera such as Bacteroides and Fusobacterium, also for the colon, but lower numbers may be required toappear. Strict anaerobes are present in increasing dominate in other regions and to promote significantnumbers even above the ileo-caecal valve. Beyond pharmacokinetic interactions, e.g. in the jejunum. Inthe valve, however, the strict anaerobes outnumber these regions the concentration of metabolites is hostthe facultative anaerobes in the lumen by 100 to mediated by active resorption pathways.1000 times, and bacterial numbers typically exceed

11 2110 g . 2.3. Colonisation, succession and influencingIt is estimated that the colon of healthy adults factors

harbours about 300–400 different cultivable speciesbelonging to more than 190 genera. A significant During delivery, the new-born becomes contami-additional proportion of the colon microflora is, nated with microorganisms from the birth canal ofhowever, not cultivable by existing techniques (see the mother and the environment. At first EscherichiaSection 5.2). Among the known colonic microbial coli and Streptococcus predominate, but in breast fedflora only a few major groups (‘main flora’, accord- infants there is a sharp increase in the numbers of

10ing to Gedek, 1993) dominate at levels around 10 – Bifidobacterium with a concomitant decrease of E.11 2110 g , all of which are strict anaerobes such as coli and Streptococcus, whilst Clostridium is low or

Bacteroides, Eubacterium, Bifidobacterium and Pep- absent. In formula fed babies this shift in com-tostreptococcus (Fig. 2). Facultative aerobes are position is not observed, and their GI flora becomesconsidered to belong to the sub-dominant (inter- rather complex with relatively high numbers ofmediate) flora or ‘satellite flora’ (Gedek, 1993), Bacteroides, Clostridium and Streptococcus.constituting Enterobacteriaceae, streptococci and lac- Bifidobacterium is present but not predominating.tobacilli. This is a strong indication that the diet can influence

Compared to the colonic flora, the fecal flora the ratio between the microbial species and strains ofundergoes distinct quantitative variations and seems the intestinal flora. When breast fed infants areto be a good qualitative indicator of the distal colonic provided with food supplements, clostridia, strep-microflora. It does not reflect, however, the intestinal tococci and E. coli attain higher levels and the floraflora and most definitely not those of the small becomes similar to formula fed infants. Now alsointestine. Furthermore, our current knowledge on the Bacteroides and Gram-positive cocci appear in pro-stability of the strains, species and even genera gressively higher numbers. After weaning, a conver-relationships is still extremely limited. While stability sion to the normal adult flora occurs. Generally,in species composition may be a feature of the streptococci and E. coli populations decrease and by‘normal’ microflora, stability of bacterial strains the second year of life the intestinal microflora iswithin the population may be less common (McCar- similar to that of the adult (Mevissen-Verhage et al.,tney et al., 1996). Genetic fingerprinting techniques 1985; Bullen et al., 1977).indicated the presence of a collection of Bifidobac- The intestinal physiology and host defense mecha-terium and Lactobacillus strains ‘‘unique of each nisms play an important role in preventing over-human’’ (Tannock, 1997). It was also suggested that growth of the microflora and in determining the final

Page 5: Overview of gut flora and probiotics.pdf

W.H. Holzapfel et al. / International Journal of Food Microbiology 41 (1998) 85 –101 89

composition and distribution of the flora throughout 2.4. Functions of the intestinal florathe gastro-intestinal tract. The major factors influenc-ing the composition of the microflora are summa- By a number of physiological functions the intesti-rised in Table 1. These factors may be related to nal flora contribute to overall health. Disturbance ofchanges in physiological conditions of the host the ecological balance in the gastro-intestinal system(aging, stress, health status, ethnical environment), may therefore be detrimental to health. Bacteriacomposition of the diet and environmental circum- typical of the ‘normal’ intestinal flora may possess astances (e.g., contamination with pathogens, use of range of beneficial features, and are (e.g.) able tomedicines). In this way the conditions underlying degrade certain food components, produce certain Bdigestion (e.g., pH, substrate availability, redox vitamins, stimulate the immune system and producepotential, transit time, flow of enteric fluid, IgA digestive and protective enzymes. The normal florasecretion, etc.) may be modulated. This could result is also involved in the metabolism of some poten-in a decline of the beneficial bacteria and in an tially carcinogenic substances and may play a role inincrease in potentially harmful bacteria. Changes in drug efficacy. These effects can be either beneficialdiet or climate, aging, medication, illness, stress or or detrimental to health. Furthermore, the coloninfection generally lead to an increase in anaerobes mucosa is dependent on short chain fatty acidsand E. coli in the small intestine and to an increase (SCFA) produced by the colonic microflora. Prod-of Enterobacteriaceae and streptococci in the colon ucts of polysaccharide metabolism SCFA are pas-concomitantly with a decrease of bifidobacteria sively absorbed by the enterocytes (Hoverstad,(Mitsuoka, 1990, 1992). Implicit interactions of 1989). Roediger (1982) estimated that 40–50% oftypical intestinal bacteria may also contribute to the required energy has to be provided by the colonicstabilisation or destabilisation, e.g. by the production microflora, suggesting one form of establishedof H O , acids and bacteriocins. mutualism in a region where SCFA are also partially2 2

responsible for bowel motility and circulation (Kvie-tys and Granger, 1981).

Table 1 The role of the gut flora as a barrier againstFactors affecting the microflora of the gastro-intestinal tract pathogenic and opportunistic microorganisms is sur-1. Host mediated factors prisingly effective, considering the large amounts ofpH, secretions such as immunoglobulins, bile, salts, enzymes allochthonous (‘non-resident’) bacteria entering theMotility, e.g. speed, peristalsis GIT. Most of them have no chance to establishPhysiology, e.g. compartmentalisation

within the system. One approach to understandingExfoliated cells, mucins, tissue exudateaspects of microbial interactions related to in situ

2. Microbial factors barrier effects was to study restoration effects inAdhesion

simplified two-strain models, using a continuousMotilityflow system (Du Toit et al., 1998a). It could beNutritional flexibility

Spores, capsules, enzymes, antimicrobial components shown that the probiotic Enterocccus faecium SF68Generation time promoted restoration of sublethally damaged Lac-

3. Microbial interactions tobacillus reuteri, a representative of the autochthon-Synergy ous gut flora. More complex in vitro models and inMetabolic cooperation vivo experiments need to be conducted for furtherGrowth factors and vitamin excretion

elucidating the complex microbial interactions andChanges to E , pH, O tensionh 2host dependent factors involved in restoration. TheseAntagonism/stimulation

Short-chain fatty acids, amines aspects will be addressed briefly in Section 5 (FutureChanges to E , pH, O tensionh 2 developments).Antimicrobial components, siderophores Increased interest exists in possibilities of man-Nutritional requirements, etc.

ipulating the composition of the gut microflora by4. Diet foods or food ingredients. The aim is to increase theComposition, non-digestible fibres, drugs, etc. numbers and activities of those microorganisms

Page 6: Overview of gut flora and probiotics.pdf

90 W.H. Holzapfel et al. / International Journal of Food Microbiology 41 (1998) 85 –101

suggested to possess health promoting properties e.g. L. casei strain Shirota, is responsible for thesuch as Bifidobacterium and Lactobacillus species. fermentation.) Probiotic strains for these products areThe question now arises what scientific evidence is generally derived from the GIT of the adult humanavailable to support the postulated mechanisms host. This is exemplified, for example, by the highbehind these beneficial effects of probiotics. frequency in which strains of autochthonous Lac-

tobacillus spp., associated with the human gut, areapplied in these products, Lactobacillus salivarius

3. Present status of probiotics being the only exception (see Table 4). Especially L.casei and the so-called ‘acidophilus’ group appear to

Different product types or supplements containing have special value as probiotic agents. Formerlyviable microorganisms with probiotic properties are considered as one species, L. acidophilus was showncommercially available either in lyophilised form or to be heterogeneous, first by Lerche and Reuteras fermented food commodities. Strains of L. (1962), and later, on the basis of DNA homologyacidophilus and L. casei strain Shirota probably have groups, by Lauer et al. (1980) and Johnson et al.the longest history among known bacterial strains for (1980). This information is summarised in Table 5,application on account of their health benefits. In in which the present species status of these homol-present-day commercial probiotic products, Lac- ogy groups, their typical hosts, and some phenotypictobacillus spp. are well represented, followed by features are given.Bifidobacterium spp., some other LAB genera andeven a few non-lactics (see Table 2). These non-lactics, however, are rarely used in dairy or otherfood commodities, but find application rather as 4. How do probiotics work?lyophilised or encapsulated ‘pharmaceutical’ prepa-rations. Although probiotic microorganisms are considered

With the emphasis mainly on ‘novel-type’ fer- to promote health, the actual mechanisms involvedmented dairy products, a steadily increasing range of have not yet been fully elucidated. In addition toyogurt-like products is available on the European desirable technical features, factors related to healthmarket (see Table 3). Streptococcus thermophilus, promotion or health sustaining, serve as importantalthough generally associated with these ‘mild’ criteria for strain selection. Three categories of keyyogurt types, is applied for technical reasons and criteria have been defined as desirable for probioticsustains the low acid fermentation typical of these bacteria (Havenaar et al., 1992) and are brieflynovel yogurt types. (In some cases, only one strain, discussed below.

Table 2Microorganisms applied in probiotic products

Lactobacillus species Bifidobacterium species Other LAB Non-lacticsa a,dL. acidophilus B. adolescentis Ent. faecalis Bacillus cereus (‘toyoi’)

dL. casei B. animalis Ent. faecium Escherichia coli (‘Nissle 1917’)cL. crispatus B. bifidum Lactoc. lactis

a c a,dL. gallinarum B. breve Leuc. mesenteroides Propionibacterium freudenreichiicL. gasseri B. infantis Ped. acidilactici

b a dL. johnsonii B. lactis Sporolactobacillus inulinus Saccharomyces cerevisiae (‘boulardii’)(L. paracasei) B. longum Strep. thermophilusL. plantarumL. reuteriL. rhamnosusaMainly used for animals.bProbably synonymous with B. animalis.cLittle known about probiotic properties.dMainly as pharmaceutical preparations.

Page 7: Overview of gut flora and probiotics.pdf

W.H. Holzapfel et al. / International Journal of Food Microbiology 41 (1998) 85 –101 91

Table 3‘Probiotic’ lactobacilli detected in novel-type yoghurts (data combined from own results and from Reuter, 1997a)

Manufacturer Origin Lactobacillus viable Lactobacilli indicated by Lactobacilli identified by21counts (log CFU g ) the manufacturer DNA–DNA hybridization

bA Germany 7.9–8.9 L. casei Shirota L. paracasei (casei)B Germany 6.4–8.1 L. acidophilus LA-1 L. johnsoniiC Germany 8.0–8.4 Lactobacillus casei GG L. rhamnosus

6.3–7.8 L. acidophilusD Germany 5.4–6.4 BactoLab cultures L. acidophilusE Germany 7.4–8.1 L. casei Actimell L. paracasei (casei)F Germany 6.8–8.2 L. acidophilus L. acidophilus

6.2–7.8 L. casei L. paracasei (casei)G Germany 3.9–5.7 L. acidophilus LA-7 L. acidophilusH Germany 4.7–6.2 L. acidophilus LA7 L. acidophilusI Germany 5.2–5.9 Not indicated L. acidophilusJ Germany 5.5–6.8 L. acidophilus NDK Germany 7.1–7.8 BIOGARDE cultures L. johnsoniiL Germany 8.6–8.7 L. casei L. paracasei (casei)M Germany 8.1–8.4 L. acidophilus LA-H3 ND

4.7–5.3 L. casei LC-H2aC Netherlands 6.8 L. acidophilus Gilliland L. crispatusa a6.4 L. casei L. paracasei (casei)

N Sweden 9.2 L. acidophilus L. acidophilusaO Germany 7.3 L. acidophilus

a aP Switzerland 7.6 L. acidophilus L. acidophilusa a9.0 L. casei L. paracasei (casei)a a5.2 L. reuteri L. reuteria aQ Switzerland 8.3 L. casei L. rhamnosusa aR France 8.3 L. casei L. paracasei (casei)

aAccording to Reuter (1997a).bL. casei suggested for L. paracasei by Dicks et al. (1997).

Table 4Autochthonous lactobacilli associated with the human host (according to Reuter, 1997b)

aI. Homofermentative‘L. acidophilus’L. acidophilus sensu strictoL. gasseriL. crispatusL. johnsonii

‘L. salivarius’L. salivariussubsp. salivariussubsp. salicinius

‘L. casei’L. caseisubsp. caseisubsp. tolerans

L. rhamnosus

bII. HeterofermentativeL. reuteri (formerly L. fermentum II) including L. oris and L. vaginalis

aPredominantly lactic acid and no production of gas from glucose.bProduction of both lactic acid and other organic acids (acetic and formic acid) and of CO from glucose.2

Page 8: Overview of gut flora and probiotics.pdf

92 W.H. Holzapfel et al. / International Journal of Food Microbiology 41 (1998) 85 –101

Table 5Typical features of the species of the ‘acidophilus-group’ (modified after Mitsuoka, 1992; Reuter, personal communication)

aSpecies Habitat mol% G 1 C in the DNA ‘Biotypes’ according to: DNA homology groups according to:

Lerche and Reuter (1962) Lauer et al. (1980) Johnson et al. (1980)

L. acidophilus All? 32–37 I,II Ia A-1L. amylovorus P/C 40 IV (III) Ib A-3L. crispatus H/W 35–38 III Ic A-2L. gallinarum W 33–36 Id A-4L. gasseri H/C 33–35 I IIa B-1L. johnsonii H/P/W 32–38 I,II IIb B-2aH, humans; P, pig; C, cattle; W, poultry.

4.1. General microbiological criteria

These aspects refer to safety (nonpathogenicity),survival of the defense system located in the upperregions of the human GIT (saliva, gastric and bilejuice), presumable human origin, and genetic stabili-ty. Most difficult appears to be the safety assessmentof a probiotic strain. Whilst this issue seems un-problematic for commercial strains with some ‘safetyrecord’, future approval of new strains may requireback-up by sound scientific data. Guidelines arepresently in discussion and will probably take ac-count of intrinsic properties of a strain, its interac-tions in vivo with the host, and its pharmacokinetics(Marteau et al., 1993; Pelletier et al., 1996; Saxelin,1996). Pronounced differences have been observedin the survival rate of probiotic strains passing

Fig. 3. Inactivation of three different commercial LAB strainsthrough the stomach and upper intestinal tract (Mar-associated with probiotic products, using a simulated stomach–

teau et al., 1993). In vitro model studies, taking duodenum passage. The values represent the average of duplicatesaccount of the low pH of 2.5 in the stomach, and the for each of three separate experiments conducted over time. Straintoxic effects of bile salts, show marked differences in 1 (Lactobacillus johnsonii) exhibited good acid and bile tolerance

and survived these conditions at a high rate. Strain 2 (Lac-the sensitivity of commercial strains, as is shown intobacillus acidophilus), also labelled as ‘probiotic’, survived acidFig. 3. Safety assessment of new strains may alsostress at levels presumably insufficient to exert significant prob-

take into account their acute toxicity, which has been iotic effects. Strain 3 (Lactobacillus delbrueckii spp. bulgaricus)21shown to exceed an LD of 6 g kg body weight, represents a classical yoghurt strain and served as a ‘non-prob-50

for strains of L. fermentum, Enterococcus faecium, L. iotic’ negative control.

helveticus and Lactobacillus GG (Donohue andSalminen, 1996) (see also Section 6).

colour, taste, aroma and texture. Probiotic strains4.2. Technological aspects claimed to be present in a product should remain

viable in sufficiently high numbers and retain meta-The possibility of cultivating the organism on an bolic activity even beyond the ‘use-before’ date.

industrial scale has most important practical conse-quences for the manufacturer. Strains should be 4.3. Functional effects and underlying mechanismsadapted to a suitable carrier or fermentable substrate(e.g., milk), and the final product should have an Numerous beneficial effects have been suggestedacceptable shelf-life and sensory attributes such as to result from probiotic activities in the gut. Sound

Page 9: Overview of gut flora and probiotics.pdf

W.H. Holzapfel et al. / International Journal of Food Microbiology 41 (1998) 85 –101 93

scientific evidence in support of health claims is supplemented with yoghurt containing live LAB wasbased either on a limited number of in vivo studies found to enhance host resistance against Salmonellaor on deductions from well-founded in vitro ‘model’ typhimurium (DeSimone et al., 1988; Paubert-studies. Some of the most important functional Braquet et al., 1995) or persistent diarrhoea (Boud-effects, backed up by scientific evidence, have been raa et al., 1990). Fermented milk containing L.summarised by Salminen et al. (1996), and include acidophilus La1 and bifidobacteria was shown toaspects such as immune modulation and strengthen- induce changes in the intestinal flora and to modulateing the gut mucosal barrier, due to: (1) gut mi- the immune response in humans (Link-Amster et al.,croflora modification, (2) adherence to the intestinal 1994; Majamaa and Isolauri, 1997). Several prob-mucosa with capacity to prevent pathogen adherence iotic strains have been proven effective in theor pathogen activation, (3) modification of dietary treatment of different types of intestinal disorders.proteins by the intestinal microflora, (4) modification Significant reduction in either the duration or severe-of bacterial enzyme capacity especially of those ness of gastroenteritis was achieved when differentsuggested to be related to tumour induction, and (5) LAB strains (Kaila et al., 1992; Saavedra et al.,influence on gut mucosal permeability. Examples 1994), Lactobacillus GG (Isolauri et al., 1994; Razareferring to well-characterised probiotic strains are et al., 1995), L. casei Shirota (Sugita and Tagawa,shown in Table 6 (see also Salminen et al., 1996). 1994) or Bifidobacterium bifidum were administered.These observations are predominantly based on These effects were suggested to result from immunestudies either with lyophilised bacterial strains or response promoted by particular LAB strains (Kailafermented milks. In contrast to probiotic foods which et al., 1992; Majamaa et al., 1995). Acute enteritisare aimed at the ‘normal’ (healthy) population, most was successfully treated in double-blind controlledstudies have been conducted on adults and children trials with Enterococcus faecium (Camarri et al.,with intestinal disorders (Salminen et al., 1996). 1981) and Lactobacillus GG (Isolauri et al., 1991).Probiotic products applied as therapeutics and aimed The latter strain was also found effective in theat curing a disorder may therefore be considered as treatment of recurrent Clostridium difficile colitispharmaceuticals (‘probiotic drugs’). The issue of (Biller et al., 1995). In addition, live L. acidophilushealth claims connected with probiotic foods is cultures were found to support the intestinal integritypresently heavily debated. Authorities will probably during radiotherapy (Salminen et al., 1988).rely on judicial considerations for solving present Although application of probiotics shows promis-discrepancies. ing results on immunomodulation, the underlying

Referring to early hypotheses, numerous studies mechanisms are not yet well understood. There arehave convincingly shown the stabilising influence on indications that specific cell wall components orthe gut ecosystem of either probiotic cultures or surface layers may be involved. Apparently somefermented milks containing such strains. A diet similarity exists with Gram-negative bacteria, e.g.

Table 6Successful probiotic bacteria and their reported effects (as reported by Salminen et al., 1996)

Strain Reported effects in clinical studies

Lactobacillus acidophilus LA1 Immune enhancer, adjuvant, adherent to human intestinal cells, balances intestinal microflora

Lactobacillus acidophilus NCFB 1748 Lowering faecal enzyme activity, decreased faecal mutagenicity, prevention of radiotherapy related diarrhoea, treatment of constipation

Lactobacillus GG (ATCC 53013) Prevention of antibiotic associated diarrhoea, treatment and prevention of rotavirus diarrhoea, treatment of relapsing Clostridium difficile

diarrhoea, prevention of acute diarrhoea, Crohn’s disease, antagonistic against cariogenic bacteria, vaccine adjuvant

Lactobacillus casei Shirota Prevention of intestinal disturbances, treatment of rotavirus diarrhoea, balancing intestinal bacteria, lowering faecal enzyme activities,

positive effects in the treatment of superficial bladder cancer, immune enhancer

in early colon cancer, immune enhancement

Streptococcus thermophilus; No effect on rotavirus diarrhoea, no immune enhancing effect during rotavirus diarrhoea, no effect on faecal enzymes

Lactobacillus bulgaricus

Bifidobacterium bifidum Treatment of rotavirus diarrhoea, balancing intestinal microflora, treatment of viral diarrhoea

Lactobacillus gasseri (ADH) Faecal enzyme reduction, survival in the intestinal tract

Lactobacillus reuteri Colonising the intestinal tract, mainly animal studies so far, possibly an emerging human probiotic

Page 10: Overview of gut flora and probiotics.pdf

94 W.H. Holzapfel et al. / International Journal of Food Microbiology 41 (1998) 85 –101

Bacteroides spp. or Enterobacteriaceae, where it has 1995), although consumption of LAB with the dietbeen shown that peptidoglycan and lipopolysac- seems to reduce the excretion of mutagenic activitycharide components of the cell wall not only express in faeces and urine (Lidbeck et al., 1992; Hayatsudetrimental effects but, under certain conditions, also and Hayatsu, 1993).induce a large spectrum of activities which may be Adhesion is considered an important property ofconsidered advantageous for the host (Hamann et al., probiotic LAB able to colonise the GIT. This is1998, this issue). These activities include induction mediated either non-specifically by physico-chemicalof the non-specific resistance to microbial infections factors or, specifically, by adhesive bacterial surfaceand irradiation, as well as necrosis of certain tumour molecules and epithelial receptor molecules. Humantypes. It has been clearly shown that the effects of cell lines such as Caco-2 or HT 29 cells may serve aspeptidoglycan–lipopolysaccharide complexes, inde- in vitro models for assessment of adherence ability.pendent of whether they are toxic or beneficial, These and faecal recovery studies have shown (e.g.)represent mediated phenomena. They are the result that traditional ‘dairy’ strains do not possess theseof interaction with mononuclear phagocytes and properties (Elo et al., 1991; Saxelin et al., 1995;endothelial cells of the host, the production of Salminen et al., 1996).endogenous mediators such as tumour necrosis factor The postulated ability of some gut microflora toor other interleukines, and the action of such medi- reduce the serum cholesterol level is still a matter ofators on the host. It is very intriguing to realise that dispute. It is however an established fact that choles-both Gram-positive and Gram-negative resident bac- terol and bile salt metabolism are closely linked. Bileteria of the intestinal tract may exert similar effects, salts may be deconjugated by the enzyme bile saltprovided that the subtle equilibrium in the microbial hydrolase (BSH) (E.C. 3.5.1.24), typical of some gutcomposition of the intestinal flora is maintained. bacteria. According to the ‘BSH hypothesis’ the freeInteractions between host epithelial cells, the gut bile salts are excreted more readily and may thusassociated lymphoid tissue and the intestinal mi- contribute to reducing serum cholesterol levelscroflora with or without probiotic bacteria are pres- (Chikai et al., 1987). This hypothesis is, however,ently under study in detail (Farstad et al., 1997). The not undisputed and is not supported by currentoverall challenge still remains to link effects of knowledge on the passive absorption kinetics of freeprobiotics with health related responses of the host. bile acids in the GIT (Aldini et al., 1996; Marteau etValidated disease models might be valuable for this al., 1995). Enhanced faecal loss of bile acids,purpose since several animal studies have shown however, may result in an increased requirement forhigher survival rates after gut flora modification with cholesterol as precursor for the de novo synthesis ofprobiotic bacteria. bile salts, thereby reducing cholesterol levels. Recent

Of special significance are observations on protec- observations based on pig and minipig feedingtive effects of particular LAB against carcinogenesis. experiments with highly BSH active lactobacilli,Reported as antimutagenic (Pool-Zobel et al., 1993a) strongly suggest a cholesterol lowering effect byor antigenotoxic (Pool-Zobel et al., 1993b) effects, such strains (De Smet et al., 1995; Du Toit et al.,these phenomena are probably related to the ability 1998b). The fact that a number of commercialof such strains to prevent or reduce DNA damage as probiotic strains exhibit high BSH activities shouldthe early event in the process of carcinogenesis. The be taken into consideration, also in view of theincidence of colon tumours in rats was significantly formation of potentially toxic levels of secondaryreduced, for example, when L. acidophilus was bile acids in the gut, but also with regard to theadministered (Goldin et al., 1980), whilst B. longum question of the desirability or not of this property forwas shown to prevent the induction of colon, liver probiotic strains.and mammary tumours by the cooked food car-cinogen IQ (Reddy and Rivenson, 1993). However,only limited convincing evidence, both scientific and 5. Future developmentsepidemiologic (De Vrese, 1996), for anticarcinogeniceffects of LAB in humans is available (Aso and In spite of considerable progress in ‘probiotic’Akazan, 1992; Aso et al., 1995). Some observations research over the last five years, not all probioticeven appear to be contradictory (Moore and Moore, bacteria available on the market have a solid sci-

Page 11: Overview of gut flora and probiotics.pdf

W.H. Holzapfel et al. / International Journal of Food Microbiology 41 (1998) 85 –101 95

entific record. If nutritional and health benefits are to addition, the growth conditions may vitally influencebe derived from products containing probiotic bac- the expression of bacterial surface structures. Theteria, it is imperative that we understand the mecha- cell surface, for example of an overnight culture, isnisms underlying these benefits. The ‘probiotic con- quite different from that of bacteria which justcept’ will only be accepted by regulatory bodies and passed the stomach and small intestine and haveauthorities if these mechanisms are elucidated and been stressed by gastric acid, bile and pancreaticappropriate selection criteria for probiotic micro- juice. The use of microorganisms surviving theseorganisms are defined. It is clear that the selection of conditions and the use of mucus secreting cell lines,strains for probiotic use must be based on criteria such as HT-29MTX (Lesuffleur et al., 1991), maywhich are coherent with the claim the probiotic is represent the in vivo situation more closely. Finally,used for. although there are differences in in vitro adherence

Rational selection and validation of promising between different probiotic strains, no evidence hasmicrobial strains should be based on evidence ob- thus far been found for any significant differences intained in in vitro models with a reliable predicted colonisation under in vivo conditions. As yet, nonevalue or function, and followed by studies in of the well known probiotic strains has been shownhumans. Acceptance of the probiotic concept by both to colonise the GI tract permanently.the scientific world and regulatory bodies must be In vitro methods are suitable for assessment ofbased on evidence obtained from fundamental re- some of the above-mentioned criteria. However, it issearch with respect to the three M’s: Mechanisms to important to note that results from such studies mayverify, Models to certify and Methods to quantify not be predictive of the actual in vivo situation. Forspecially controlled studies in humans. The first M, example, the survival of microorganisms at low pHpossible mechanisms, has been discussed above and or in the presence of bile in test tubes is not asome scientific evidence has been presented. The reflection of interactions in the stomach and thequestion is, how to proceed and how to select models intestine, where more complex physiological con-that would enable reliable elucidation of the underly- ditions exist. Therefore, a dynamic model thating mechanisms. mimics the successive in vivo gastro-intestinal con-

ditions as closely as possible, would present a major5.1. In vitro models advantage to conventional in vitro methods. Such a

dynamic model of the stomach and small intestineIt is generally believed that adherence to mucosal has been developed at TNO Nutrition and Food

surfaces contributes to the efficacy of a probiotic Research at Zeist, The Netherlands, and was de-strain since adherent strains could confer a competi- scribed in detail by Minekus et al. (1995). Recently,tive advantage, important for bacterial maintenance a new type of large intestinal model has beenin the gastro-intestinal tract. Furthermore, by block- developed (Minekus et al., personal communication).ing the attachment sites, probiotics might contribute It is designed to be complementary to the dynamicto the prevention of infection by pathogenic micro- multi-compartmental model of the stomach and smallorganisms. The non-mucus secreting enterocyte-like intestine. This colon model combines removal ofCaco-2 cell line displays typical features of en- metabolites and water, necessary to obtain physio-terocytic intestinal cells. By using this cell line as in logical concentrations of microorganisms, dry mattervitro model, it was shown that L. acidophilus and L. and microbial metabolites. High densities of micro-rhamnosus strains adhere in relatively high numbers organisms, comparable to that found in the colon inand are able to prevent attachment of pathogenic vivo, were achieved by absorption of water andmicroorganisms such as Salmonella typhimurium, dialysis of metabolites through hollow fibres insideYersinia enterocolitica and enteropathogenic E. coli the compartments. Incubation with faecal flora re-(Coconnier et al., 1993; Bernet et al., 1994; Hudault sulted in total anaerobic bacterial counts of

10 21et al., 1997). The value of these observations can, . 10 CFU ml with stable physiological levels ofhowever, be questioned. First of all, only a three-fold Bifidobacterium, Lactobacillus, Enterobacteriaceaedifference between ‘good’ and ‘poor’ adherent and Clostridium. The dry matter content was approx-strains was detected (Lehto and Salminen, 1996), imately 10%, while the total short chain fatty acidswhich microbiologically is not highly significant. In concentration was maintained at physiological levels

Page 12: Overview of gut flora and probiotics.pdf

96 W.H. Holzapfel et al. / International Journal of Food Microbiology 41 (1998) 85 –101

with similar molar ratios of acetic acid, propionic bacterium (Angert et al., 1993). Difficulties in cultur-acid and butyric acid as measured in vivo. The ing techniques have led to the development of novelvalidation studies, so far, have shown that this model molecular methods based on DNA technology byaccurately reproduces the in vivo conditions and that which bacteria can be identified without cultivationthe results can be extrapolated to the human situa- (Amann et al., 1995).tion.Validation and application of this in vitro gastro- Presently, ribosomal RNA (rRNA) is commonlyintestinal model in relation to the survival of ingested used as a tool to study microbial populations. Duringlactic acid bacteria (Marteau et al., 1997) and other the last two decades the gene encoding for 16Sresearch topics have been published (Havenaar and rRNA of Escherichia coli was sequenced, and manyMinekus, 1996). Obviously, disadvantages of such more 16S rRNA sequences of other bacteria havemodels should also be considered when planning an been determined. The polymerase chain reactionexperiment, for example with regard to epithelium (PCR) and improved sequencing technology haveand immune response. However, combined with greatly facilitated retrieval of new sequences. Atother in vitro models the TNO model offers a range present, several thousands of 16S rRNA sequencesof additional possibilities for probiotic research. of different bacteria are available in genetic data-Furthermore, such combined models can be used for bases, even from intestinal bacterial species thatstudying bioconversions by the intestinal flora of cannot be cultured in vitro (Angert et al., 1993; Sneldrugs, toxic components or (pro)carcinogens. et al., 1994). The 16S rRNA gene is approximately

1540 bases long, and includes several variable5.2. Methods for studying gut flora composition regions while the overall structure is highly con-

served. This makes it possible to use this gene toA drawback in studying the role of probiotics in study phylogenetic relationships between micro-

the modification of the intestinal microflora is the organisms (Woese, 1987).lack of appropriate techniques to sample or identify An approach to analyse the genetic diversity ofchanges in the endogenous microflora. Practically all complex microbial populations is denaturing gradientresults are based on faecal samples, which obviously gel electrophoresis (DGGE) or temperature gradientare a poor description of the situation in the terminal gel electrophoresis (TGGE). This technique is basedileum or the ascending colon. Furthermore, our on the separation of PCR amplified fragments ofmicrobiological techniques are hardly sufficient to genes coding for 16S rRNA, all having the sameeven isolate or identify the predominant microbial length (Muyzer et al., 1993). This results in uniquespecies in the GIT. separation patterns for different microbial popula-

In order to study the effect of probiotics on the tions, and will contribute to the description ofcomposition of the microflora, monitoring at certain changes or differences in the microflora compositionintervals before, during and after administration of of (uncharacterised) microbial populations.the probiotic is essential. The use of selective media From sequence data, rRNA-targeted oligonucleo-for growth of different groups of bacteria is widely tide probes can be developed that are species- orused. However, in several cases, bacteria cannot group-specific. For analysis of intestinal microflora,correctly be enumerated by plate counting since such probes labeled with a radio-active marker havethese viable counts do not correlate with micro- been used for the first time to study ruminal micro-scopical counts (Langendijk et al., 1995). In addi- bial ecology (Stahl et al., 1988). In this study,tion, many species cannot be cultured in vitro at all. hybridisation was quantified in a dot-blot procedure.It is speculated that at present less than 20% of all Later, for similar studies, oligonucleotide probesmicroorganisms can be cultured (Ward et al., 1990), were labeled with radio-active or fluorescent markerswhereas only a fraction of the total culturable and a and used for in situ hybridisation, which enablesfew unculturable species are known. Examples of specific identification of individual cells (DeLong etknown unculturable intestinal bacteria are segmented al., 1989; Amann et al., 1990, 1995). Techniquesfilamentous bacteria in mice (Snel et al., 1994), or based on 16S rRNA detection have recently beenEpulopiscium fishelsoni, an intestinal inhabitant of used to detect probiotic Bifidobacterium species inthe surgeon fish and considered as the largest infant faeces (Kok et al., 1996).

Page 13: Overview of gut flora and probiotics.pdf

W.H. Holzapfel et al. / International Journal of Food Microbiology 41 (1998) 85 –101 97

Restriction enzyme analysis has been applied for Recent papers by Bry et al. (1996), Matsumoto etprobiotic lactobacilli (Charteris et al., 1997) and al. (1992) and Umesaki et al. (1993), (1995), (1997)bifidobacteria (McCartney et al., 1996; Roy et al., report that host epithelial cells in the small intestine1996). This method also served to characterise express fucosylated glycoconjugates in response tostrains within the L. acidophilus group (Roussel et the presence of specific strictly anaerobic bacteriaal., 1993), of L. reuteri (Stahl and Molin, 1994) and (Bacteroides thetaiomicron and a Segmented Fila-L. casei (Ferrero et al., 1996). Likewise, oligo- mentous Bacterium (‘SFB’), respectively). The ob-nucleotide probes (Langendijk et al., 1995; Charteris servation that one species can induce epithelialet al., 1997), RAPD-PCR (Du Plessis and Dicks, surface structures such as fucosylated glycoconju-1995) and ribotyping (McCartney et al., 1996) have gates, by which the attachment of other bacteria isshown promising results for species and even strain- influenced, has significance for the use of (e.g.) Cacolevel differentiation of intestinal LAB. cell lines or gnotobiotic animals as model system, to

Further new techniques for studying bacterial study either adherence or infectious diseases as wellcolonisation of bacteria are described by Contag et as for strategies to prevent and to treat gastro-al. (1995). They marked strains of Salmonella intestinal diseases. To induce and sustain fucosylatedtyphimurium with bioluminescence through trans- glycoconjugate production, it is thought that bacteriaformation with a plasmid conferring constitutive should reach a critical population density. This mayexpression of bacterial luciferase. They were able to be an indication for a soluble bacterial factor thatdetect photons transmitted through tissues of animals produces a concentration-dependent response in theinfected with these bioluminescent strains. This epithelium. Another (theoretical) possibility might beenables in vivo detection of intestinal bacteria with- a density-dependent change in the metabolic prop-out invasive procedures. erties of the bacteria that affects production of a

signalling molecule, a process known as ‘quorumsensing’ (Kaiser, 1996).

5.3. Interactions between the intestinal microflora It is not fully known whether these changes inand epithelial cells fucosylation observed in the small intestine are also

present in the large bowel or the stomach and, if so,Although attachment is thought to be an important which microorganism might bring about such

factor, mechanisms that allow certain species to changes. Especially for the stomach this informationestablish at specific parts of the intestinal tract are would be important, because certain glycoconjugateslargely unknown. Highly significant, however, is the have been identified as receptors for the attachmentrecent observation that the intestinal microflora can of Helicobacter pylori.influence the expression of epithelial glycoconjugateswhich may serve as receptor for attachment of(pathogenic) microorganisms. As discussed before,the interface between a mammalian host and the 6. Safetymicroflora in the lumen is the mucous gel layer andthe underlying cell coat (glycocalix) which consist of The use of LAB in food products has a long safetyglycoconjugates on the apical surface of the epi- record. Strains used in probiotic dairy products arethelium. It is hypothesized that integration of the also considered to be non-pathogenic for humans. Inecosystem into the host may be achieved, at least in several clinical and epidemiological studies LAB andpart, through dynamic host–microbe interactions that bifidobacteria have been examined for their role inallow microbes to modify cellular differentiation health (Donohue and Salminen, 1996). In addition,programs and thus create favorable niches (Bry et while it has been suggested that probiotic strainsal., 1996). This hypothesis was supported by experi- have been implicated in some cases of patients withments in which one signalling bacterial genetic locus bacteraemia, it is important to note that none of thefrom indigenous Bacteroides thetaiotaomicron and pathogenic strains isolated in such cases was relatedone associated responding mammalian gene were to lactobacilli used in probiotic food productsstudied. (Donohue and Salminen, 1996; Saxelin et al., 1996).

Page 14: Overview of gut flora and probiotics.pdf

98 W.H. Holzapfel et al. / International Journal of Food Microbiology 41 (1998) 85 –101

Aldini, R., Montagnani, M., Roda, A., Hrelia, S., Biagi, P.L.,There is general consensus that the consumptionRoda, E., 1996. Intestinal absorption of bile acids in the rabbit:of probiotics, even in dosages as high asDifferent transport rates in jejunum and ileum. Gastroenterolo-12 2110 cfu d , failed to exhibit any toxicity. Neverthe- gy 110, 459–468.

less, future approval of ‘new’ probiotic strains Amann, R.I., Binder, B.J., Olsen, R.J., Chrisholm, S.W., Devereux,should also take into account safety aspects. A R., Stahl, D.A., 1990. Combination of 16S rRNA-targeted

oligonucleotide probes with flow cytometry for analyzingscheme proposed by Donohue and Salminen (1996)mixed microbial populations. Appl. Environ. Microbiol. 56,for safety assessment also takes intrinsic properties1919–1925.of probiotic strains into consideration, in addition to

Amann, R.I., Ludwig, W., Schleifer, K.H., 1995. Phylogeneticmetabolic products, toxicity, mucosal effects, dose– identification and in situ detection of individual microbial cellsresponse effects, clinical assessment and epi- without cultivation. Microbiol. Rev. 59, 143–169.

Angert, E.R., Clements, K.D., Pace, N.R., 1993. The largestdemiological studies. In view of their frequentbacterium. Nature 362, 239–241.association with human infection and possible easy

Aso, Y., Akazan, H., 1992. Prophylactic effect of Lactobacillusacquisition of (glycopeptide) antibiotic resistance,casei preparation on the recurrence of superficial bladder

the use of enterococci as probiotics is still a matter of cancer. BLP study group. Urol. Int. 49, 125–129.some concern (Adams and Marteau, 1995; Farrag et Aso, Y., Akaza, H., Kotake, T., Tsukamoto, T., Imai, K., Naito, S.,al., 1996; Bonten et al., 1996). 1995. Preventive effect of a Lactobacillus casei preparation on

the recurrence of superficial bladder cancer in a double-blindtrial. The BLP study group. Eur. Urol. 27, 104–109.

Berg, R.D., 1996. The indigenous gastrointestinal microflora.7. Conclusions Trends Microbiol. 4, 430–435.

Bernet, M.F., Brassart, D., Neeser, J.R., Servin, A.L., 1994.Increasing knowledge underlines the important Lactobacillus acidophilus LA1 binds to cultured human

intestinal cell lines and inhibits cell attachment and cellrole of the intestinal flora for maintaining health andinvasion by enterovirulent bacteria. Gut 35, 483–489.in the prevention of disease. Evidence is emerging

Bernhardt, H., Knoke, M., 1980. Zur Charakterisierung derthat the intestinal flora does not exist as an entity by¨Mikroflora des Dunndarms. Zentralbl. Bakt. Hyg. I. Abt. Orig.

itself, but is constantly interacting (‘communicating’) A 246, 379–392.with the environment, the central nervous system, the Bhat, P., Albert, M.J., Rajan, D., Ponniah, J., Mathan, H., Baker,

S.J., 1989. Bacterial flora of the jejunum: A comparison ofendocrine system and the immune system (Bry et al.,luminal aspirate and mucosal biopsy. J. Med. Microbiol. 13,1996; Shanahan, 1997; Umesaki et al., 1997; Wang247–256.et al., 1997). Disturbance of this delicate balance

Biller, J.A., Katz, A.J., Flores, A.F., Buie, T.M., Gorbach, S.T.,may lead to other disorders and thus facilitate 1995. Treatment of recurrent Clostridium difficile colitis withestablishing a state of disease. Lactobacilus GG. J. Pediatr. Gastroenterol. Nutr. 21, 224–226.

Probiotics offer dietary means to support the Bonten, M.J., Hayden, M.K., Nathan, C., van Voorhis, J.,Matushek, M., Slaughter, S., Rice, T., Weinstein, R.A., 1996.balance of intestinal flora. They may be used toEpidemiology of colonisation of patients and environmentcounteract local immunological dysfunctions, towith vancomycin-resistant enterococci. Lancet 348, 1615–stabilise the gut mucosal barrier function, to prevent1619.

infectious succession of pathogenic microorganisms Boudraa, G., Touhani, M., Pochart, P., Soltana, R., Mary, J.Y.,or to influence intestinal metabolism. Many of the Desjeux, J.F., 1990. Effect of feeding yoghurt versus milk in

children with persistent diarrhoea. J. Pediatr. Gastroenterol.proposed mechanisms have to be validated in con-Nutr. 11, 509–512.trolled clinical trials in humans. Novel methods are

Brassart, D., Schiffrin, E.J., 1997. The use of probiotics tourgently needed to monitor changes in the com-reinforce mucosal defence mechanisms. Trends Food Sci.

position of the intestinal flora and their mutual Technol. 8, 321–326.interaction with the host’s immunological functions Bry, L., Falk, P.G., Midtvedt, T., Gordon, J.I., 1996. A model ofand metabolism. host–microbial interactions in an open mammalian ecosystem.

Science 273, 1380–1383.Bullen, C.L., Tearle, P.V., Stewart, M.G., 1977. The effect of

‘humanised’ milks and supplemented breastfeeding on theReferences faecal flora of infants. J. Med. Microbiol. 10, 403–413.

Camarri, E., Belvisi, A., Guidoni, G., Marini, G., Frigerio, G.,Adams, M.R., Marteau, P., 1995. On the safety of lactic acid 1981. A double-blind comparison of two different treatments

bacteria. Int. J. Food Microbiol. 27, 263–264. for acute enteritis in adults. Chemotherapy 27, 466–470.

Page 15: Overview of gut flora and probiotics.pdf

W.H. Holzapfel et al. / International Journal of Food Microbiology 41 (1998) 85 –101 99

Charteris, W.P., Kelly, P.M., Morelli, L., Collins, J.K., 1997. Farrag, N., Eltringham, I., Liddy, H., 1996.Vancomycin-dependentSelective detection, enumeration and identification of poten- Enterococcus faecalis. Lancet 348, 1581–1582.tially probiotic Lactobacillus and Bifidobacterium species in Farstad, I.N., Norstein, J., Brandtzaeg, P., 1997. Phenotype of Bmixed microbial populations. Int. J. Food Microbiol. 35, 1–27. and T cells in human intestinal and mesenteric lymph.

Chikai, T., Nakao, H., Uchida, K., 1987. Deconjugation of bile Gastroenterology 112, 163–173.acids by human intestinal bacteria implanted in germ-free rats. Ferrero, M., Cesena, C., Morelli, L., Scolari, G., Vescovo, M.,Lipids 22, 669–671. 1996. Molecular characterisation of Lactobacillus casei

´ `Coconnier, M.H., Bernet, M.F., Kerneis, S., Chauviere, G., strains. FEMS Microbiol. Lett. 140, 215–219.¨Fourniat, J., Servin, A.L., 1993. Inhibition of adhesion of Gedek, B., 1993. Darmflora—Physiologie und Okologie.

enteroinvasive pathogens to human intestinal Caco-2 cells by Chemother. J., Suppl. 1, 2–6.Lactobacillus acidophilus strain LB decreases bacterial inva- Goldin, B.R., Swenson, L., Dwyer, J., Sexton, M., Gorbach, S.L.,sion. FEMS Microbiol. Lett. 110, 299–305. 1980. Effect of the diet and Lactobacillus acidophilus supple-

Contag, C.H., Contag, P.R., Mullins, J.I., Spilman, S.D., Steven- ments on human faecal bacterial enzymes. J. Natl. Cancer Inst.son, D.K., Benaron, D.A., 1995. Photonic detection of bacteri- 64, 255–261.al pathogens in living hosts. Mol. Microbiol. 18, 593–603. Haenel, H., Bendig, J., 1975. Intestinal flora in health and disease.

Dicks, L.M.T., Du Plessis, E.M., Dellaglio, F., Lauer, E., 1997. Prog. Food Nutr. Sci. 1, 21–64.Reclassification of Lactobacillus rhamnosus ATCC 15820 as Hamann, L., El-Samalouti, V., Ulmer, A.J., Flad, H.-D., Rietschel,Lactobacillus zeae nom. rev., designation of ATCC 334 as E.Th., 1998. Components of gut bacteria as immuno-neotype of L. casei subsp. casei, and rejection of the name modulators. Int. J. Food Microbiol. (this issue)Lactobacillus paracasei. Int. J. Syst. Bacteriol. 46, 337–340. Havenaar, R., Ten Brink, B., Huis in ’t Veld, J.H.J., 1992. In:

DeLong, E.F., Wickham, G.S., Pace, N.R., 1989. Phylogenetic Fuller, R. (Ed.), Probiotics, The Scientific Basis. Chapmann &strains: Ribosomal RNA-based probes for the identification of Hall, London, pp. 209–224.single cells. Science 243, 1360–1363. Havenaar, R., Minekus, M., 1996. Simulated assimilation. Dairy

DeSimone, C., Tzantzoglou, S., Baldinelli, L., Di Fabio, S., Ind. Int. 61, 17–23.Bianchi-Salvatori, B., Jirilli, E.,Vesely, R., 1988. Enhancement Hayatsu, H., Hayatsu, T., 1993. Suppressing effect of Lactobacil-of host resistance against Salmonella typhimurium infection by lus casei administration on the urinary mutagenicity arisinga diet supplemented with yoghurt. Immunopharmacol. Im- from ingestion of fried ground beef in the human. Cancer Lett.munotoxicol. 10, 399–415. 73, 173–179.

De Smet, I.,Van Hoorde, L.,Van de Woestyne, M., Christiaens, H.,Heyman, M., Ducroc, R., Desjeux, J.F., Morgat, J.L., 1982.

Verstraete, W., 1995. Significance of bile salt hydrolyticHorseradish peroxidase transport across adult rabbit jejunum

activities of lactobacilli. J. Appl. Bacteriol. 29, 292–301.in vitro. Am. J. Physiol. 242, G558–G564.

De Vrese, M., 1996. Probiotische Milchprodukte. Dtsch. Milch-Hoverstad, T., 1989. The normal microflora and short-chain fatty

wirtschaft 18, 798–801.acids. In: Grubb, R., Midtvedt, T., Norin, E. (Eds.), The

Donohue, D., Salminen, S., 1996. Safety of probiotic bacteria.Regulatory and Protective Role of the Normal Microflora.

Asia Pac. J. Clin. Nutr. 5, 25–28.MacMillan, London, pp. 89–108.

Ducluzeau, R., 1989. Role of experimental ecology in gastroen-Hudault, S., Lievin, V., Bernet-Camard, M.F., Servin, A., 1997.

terology. In: Bergogne-Berezin, E. (Ed.), Microbial EcologyAntagonistic activity exerted in vitro and in vivo by Lac-

and Intestinal Infections. Springer, Paris, pp. 1–5.tobacillus casei (strain GG) against Salmonella typhimurium

Du Plessis, E., Dicks, L.M.T., 1995. Evaluation of randomC5 infection. Appl. Environ. Microbiol. 63, 513–518.

polymorphic DNA (RAPD)-PCR as a method to differentiateIsolauri, E., Juntunen, M., Rautanen, T., Sillanaukee, P., Koivula,Lactobacillus acidophilus, Lactobacillus crispatus, Lactobacil-

T., 1991. A human Lactobacillus strain (Lactobacillus GG)lus amylovorus, Lactobacillus gallinarum, Lactobacillus gas-promotes recovery from acute diarrhea in children. Pediatricsseri, and Lactobacillus johnsonii. Curr. Microbiol. 31, 114–88, 90–97.118.

Isolauri, E., Kaila, M., Mykkanen, H., Ling, W.H., Salminen, S.,¨ ¨Du Toit, M., Knopfel, S., Skowronek, F.A., Specht, I., Bockeler,1994. Oral bacteriotherapy for viral gastroenteritis. Dig. Dis.W., Holzapfel, W.H., 1998a. A modified continuous flowSci. 39, 2595–2600.system for studying microbial interactions related to the

Johnson, J.L., Phelps, C.F., Cummins, C.S., London, J., Gasser,gastro-intestinal tract: Application of the probiotic organismF., 1980. Taxonomy of the Lactobacillus acidophilus group.Enterococcus faecium SF68. Biosci. Microflora (in press).Int. J. Syst. Bacteriol. 30, 53–68.Du Toit, M., Franz, C.M.A.P., Dicks, L.M.T., Schillinger, U.,

Kaila, M., Isolauri, E., Soppi, E., Virta nen, E., Laine, S.,Haberer, P., Warlies, B., Ahrens, F., Holzapfel, W.H., 1998.Arvilommi, H., 1992. Enhancement of the circulating antibodyCharacterisation and selection of probiotic lactobacilli for asecreting cell response in human diarrhea by a human Lac-preliminary minipig feeding trial and their effect on serumtobacillus strain. Pediatr. Res. 32, 141–144.cholesterol levels, faeces pH and faeces moisture content. Int.

Kaiser, D., 1996. Bacteria also vote. Science 272, 1598–1599.J. Food Microbiol. 40, 93–104.Kok, R.G., de Waal, A., Schut, F., Welling, G.W., Weenk, G.,Elo, S., Saxelin, M., Salminen, S., 1991. Attachment of Lac-

Hellingwerf, K.J., 1996. Specific detection and analysis of atobacillus casei strain GG to human colon carcinoma cell lineprobiotic Bifidobacterium strain in infant faeces. Appl. En-Caco-2: Comparison with other dairy strains. Lett. Appl.viron. Microbiol. 62, 3668–3672.Microbiol. 13, 154–156.

Page 16: Overview of gut flora and probiotics.pdf

100 W.H. Holzapfel et al. / International Journal of Food Microbiology 41 (1998) 85 –101

Kvietys, P.R., Granger, D.N., 1981. Effect of volatile fatty acids tobacillus microflora of humans. Appl. Environ. Microbiol. 62,on blood flow and oxygen uptake by the dog colon. Gastroen- 4608–4613.terology 80, 962–969. Mevissen-Verhage, E.A.E., Marcelis, J.H., Harmsen-van

Langendijk, P.S., Schut, F., Jansen, G.J., Raangs, G.C., Kamphuis, Amerongen, W.C.M., De Vos, N.M., Berkel, J., Verhoef, J.,G.R., Wilkinson, M.H.F., Welling, G.W., 1995. Quantitative 1985. Effect of iron on development of the neonatal gut florafluorescence in situ hybridization of Bifidobacterium spp. with during the first week of life. Eur. J. Clin. Microbiol. 4, 14–18.genus-specific 16S rRNA-targeted probes and its application in Minekus, M., Marteau, P., Havenaar, R., Huis in ’t Veld, J.H.J.,fecal samples. Appl. Environ. Microbiol. 61, 2069–3075. 1995. A multicompartmental dynamic computer-controlled

Lauer, E., Helming, C., Kandler, D., 1980. Heterogeneity of the model simulating the stomach and small intestine. ATLA 23,species Lactobacillus acidophilus (Moro) Hansen and Moquot 197–209.as revealed by biochemical characteristics and DNA-DNA Mitsuoka, T., 1990. Bifidobacteria and their role in human health.hybridisation. Zbl. Bakt. Abt. Orig. C1, 150–168. J. Ind. Microbiol. 6, 263–268.

Lehto, E.M., Salminen, S.J., 1996. Adhesion of twelve different Mitsuoka, T., 1992. Intestinal flora and aging. Nutr. Rev. 50,Lactobacillus strains to Caco-2 cell cultures. Nutr. Today 31, 438–446.49S–50S. Moore, W.E.C., Moore, L.H., 1995. Intestinal floras of populations

Lerche, M., Reuter, G., 1962. Das Vorkommen aerob wachsender that have a high risk of colon cancer. Appl. Environ. Mi-¨grampositiver Stabchen des Genus Lactobacillus Beijerinck im crobiol. 61, 3202–3207.

Darminhalt erwachsener Menschen. Zentralbl. Bakt. Hyg., I. Muyzer, G., de Waal, E.C., Uitterlinden, A.G., 1993. Profiling ofAbt. Orig. A 185, 446–481. complex microbial populations by denaturing gradient gel

Lesuffleur, T., Barbat, A., Luccioni, C., Beaumatin, J., Clair, M., electrophoresis analysis of polymerase chain reaction-am-Kornowski, A., Dussaulx, E., Dutrillaux, B., Zweibaum, A., plified genes coding for 16S rRNA. Appl. Environ. Microbiol.1991. Dihydrofolate reductase gene amplification-associated 59, 695–700.shift of differentiation in methotrexate-adapted HT-29 cells. J. Nielsen, O.H., Jorgensen, S., Pedersen, K., Justesen, T., 1994.Cell Biol. 115, 1409–1418. Microbial evaluation of jejunal aspirates and faecal samples

Lidbeck, A., Overvick, E., Rafter, J., Nord, C.E., Gustafsson, J.A., after oral administration of bifidobacteria and lactic acid1992. Effect of Lactobacillus acidophilus supplements on bacteria. J. Appl. Bacteriol. 76, 469–474.mutagen excretion in faeces and urine in humans. Microb. Paubert-Braquet, M., Gan, X.H., Gaudichon, C., Hedef, N.,Ecol. Health Dis. 5, 59–62. Serikoff, A., Bouly, C., Bonavida, B., Braquet, P., 1995.

Link-Amster, H., Rochat, F., Saudan, K.Y., Mignot, O., Aesch- Enhancement of host resistance against Salmonellalimann, J.M., 1994. Modulation of a specific humoral immune typhimurium in mice fed a diet supplemented with yogurt orresponse and changes in intestinal flora mediated through milks fermented with various Lactobacillus casei strains. Int.fermented milk intake. FEMS Immunol. Med. Microbiol. 10, J. Immunother. XI, 153–161.55–64. Pelletier, C., Bouley, C., Bourliov, P., Carbon, C., 1996. Evalua-

Luckey, T.D., Floch, M.H., 1972. Introduction to intestinal tion of safety properties of Lactobacillus strains by using anmicroecology. Am. J. Clin. Nutr. 25, 1291–1295. experimental model of endocarditis in rabbit. Abstract,

Majamaa, H., Isolauri, E., 1997. Probiotics: A novel approach in SOMED Meeting, Paris, 1996.¨the management of food allergy. J. Allergy Clin. Immunol. 99, Pool-Zobel, B.L., Munzner, R., Holzapfel, W.H., 1993. An-

179–185. tigenotoxic properties of lactic acid bacteria in the SalmonellaMajamaa, H., Isolauri, E., Saxelin, M., Vesikari, T., 1995. Lactic typhimurium mutagenicity assay. Nutr. Cancer 20, 261–270.

acid bacteria in the treatment of acute rotavirus gastroenteritis. Pool-Zobel, B.L., Bertram, B., Knoll, M., Lambertz, R.,J. Pediatr. Gastroenterol. Nutr. 20, 333–338. Neudecker, C., Schillinger, U., Schmezer, P., Holzapfel, W.H.,

Marteau, P., Pochart, P., Bouhnik, Y., Rambaud, J.C., 1993. Fate 1993. Antigenotoxic properties of lactic acid bacteria in vivoand effects of some transiting microorganisms in the human in the gastro-intestinal tract of rats. Nutr. Cancer 20, 271–281.gastrointestinal tract. World Rev. Nutr. Diet 74, 1–21. Pouwels, P.H., Leer, R.J., Boersma, W.J.A., 1996. The potential of

Marteau, P., Gerhardt, M.F., Myara, A., Bouvier, E., Trvin, F., Lactobacillus as a carrier for oral immunization: DevelopmentRambaud, J.C., 1995. Bifidobacteria and lactobacilli ingested and preliminary characterization of vector systems for targetedin fermented dairy products can metabolise bile salts in the delivery of antigens. J. Biotechnol. 44, 183–192.human small intestine. Microbiol. Ecol. Health Dis. 8, 151– Raza, S., Graham, S.M., Allen, S.J., Sultana, S., Cuevas, L., Hart,157. C.A., 1995. Lactobacillus GG promotes recovery from acute

Marteau, P., Havenaar, R., Huis in ’t Veld, J.H.J., 1997. Survival non-bloody diarrhea in Pakistan. Pediatr. Infect. Dis. J. 14,of lactic acid bacteria in a dynamic model of the stomach and 107–111.small intestine: Validation and the effect of bile. J. Dairy Sci. Reddy, B.S., Rivenson, A., 1993. Inhibitory effect of Bifidobac-(in press). terium longum on colon, mammary and livery carcinogenesis

Matsumoto, S., Setoyama, H., Umesaki, Y., 1992. Differential induced by 2-amino-3-methylimidazo[4,5-f]quinoline, a foodinduction of major histocompatibility complex molecules on mutagen. Cancer Res. 53, 3914–3918.mouse intestine by bacterial colonization. Gastroenterology Reuter, G., 1965. Das Vorkommen von Laktobazillen in Leben-103, 1777–1782. smitteln und ihr Verhalten im menschlichen Intestinaltrakt.

McCartney, A.L., Wenzhi, W., Tannock, G.W., 1996. Molecular Zentralbl. Bakt. Hyg. I. Abt. Orig. A 197, 468–487.¨analysis of the composition of the Bifidobacterial and Lac- Reuter, G., 1965. Untersuchungen uber die Zusammensetzung und

Page 17: Overview of gut flora and probiotics.pdf

W.H. Holzapfel et al. / International Journal of Food Microbiology 41 (1998) 85 –101 101

Beeinflußbarkeit der menschlichen Magen- und Darmflora Shanahan, F., 1997. A gut reaction: Lymphoepithelial communica-¨ ¨unter besonderer Berucksichtigung der Laktobazillen. Ernah- tion in the intestine. Science 275, 1897–1898.

rungsforsch. 10, 429–435. Simon, G.L., Gorbach, S.L., 1982. Intestinal microflora. Ned.Reuter, G., 1969. Zusammensetzung und Anwendung von Bak- Clin. North Am. 66, 557–574.

¨terienkulturen fur therapeutische Zwecke. Arzneim.-Forsch. Snel, J., Blok, H.J., Kengen, H.M.P., Ludwig, W., Poelma, F.G.J.,Drug. Res. 50, 951–954. Koopman, J.P., Akkermans, A.D.L., 1994. Phylogenetic

Reuter, G., 1997. Present and future of probiotics in Germany and characterization of Clostridium related segmented filamentousin Central Europe. Biosci. Microflora 16, 43–51. bacteria in mice based on 16S ribosomal RNA analysis. Syst.

¨Reuter, G., 1997b. Was ist unter Doderlein-Bakterien zu ver- Appl. Microbiol. 17, 172–179.¨stehen? 12. Int. Tagung: Infektionen in der Gynakologie, Stahl, M., Molin, G., 1994. Classification of Lactobacillus reuteri

¨Geburtshilfe und Urologie, Munchen, Klinikum Großhadern, by restriction endonuclease analysis of chromosomal DNA.15.02.1997. Int. J. Syst. Bacteriol. 44, 9–14.

Roediger, W.E.W., 1982. The effect of bacterial metabolites on Stahl, D.A., Flesher, B., Mansfield, H.R., Montgomery, L., 1988.nutrition and function of the colonic mucosa. Symbiosis Use of phylogenetically based hybridization probes for studiesbetween men and bacteria. In: Kasper, H., Goebell, H. (Eds.), of ruminal microbial ecology. Appl. Environ. Microbiol. 54,Colon and Nutrition. Proceedings of the Falk Symposium, 1079–1084.Titisee. MTP Press, Lancaster, PA, pp. 11–24. Sugita, T., Tagawa, M., 1994. Efficacy of Lactobacillus prepara-

Roussel, Y., Colmin, C., Simonet, J.M., Decaris, B., 1993. Strain tion Biolactis powder in children with rotavirus enteritis. Jpn.characterisation, genome size and plasmid content in the J. Pediatr. 47, 2755–2762.Lactobacillus acidophilus group (Hansen and Mocquot). J.

Tannock, G.W., 1997. Probiotic properties of lactic acid bacteria:Appl. Bacteriol. 74, 549–556.

Plenty of scope for fundamental R&D. Trends Biotechnol. 15,Roy, D., Ward, P., Champagne, G., 1996. Differentiation of

270–274.bifidobacteria by use of pulsed-field gel electrophoresis and

Umesaki, Y., Setoyama, H., Matsumoto, S., Okada, Y., 1993.polymerase chain reaction. Int. J. Food Microbiol. 29, 11–29.

Expansion of T-cell receptor-bearing intestinal intraepithelialSaavedra, J.M., Bauman, N.A., Oung, I., Perman, J.A., Yolken,

lymphocytes after microbial colonization in germ-free miceR.H., 1994. Feeding of Bifidobacterium bifidum and Strep-

and its independence from thymus. Immunology 79, 32–37.tococcus thermophilus to infants in hospital for prevention of

Umesaki, Y., Okada, Y., Matsumoto, S., Imaoka, A., Setoyama, H.,diahrroea and shedding of rotavirus. Lancet 344, 1046–1049.1995. Segmented filamentous bacteria are indigenous intestinalSalminen, E., Elomaa, I., Minkkinen, J., Vapaatalo, H., Salminen,bacteria that activate intraepithelial lymphocytes and induceS., 1988. Preservation of intestinal integrity duringMHC class II molecules and fucosyl asialo GM1 glycolipidsradiotherapy using live Lactobacillus acidophilus cultures.on the small intestinal epithelial cells in the ex-germ-freeClin. Radiol. 39, 435–437.mouse. Microbiol. Immunol. 39, 555–562.Salminen, S., Isolauri, E., Salminen, E., 1996. Clinical uses of

Umesaki, Y., Okada, Y., Imaoka, A., Setoyama, H., Matsumoto, S.,probiotics for stabilizing the gut mucosal barrier: Successful1997. Interactions between epithelial cells and bacteria, normalstrains for future challenges. Antonie Van Leeuwenhoek 70,and pathogenic. Science 276, 964–965.347–358.

Van Dijk, J.E., 1997. Morphology of the gut barrier. Eur. J.Saxelin, M., 1996. Colonisation of the human gastrointestinal tractCompar. Gastroenterol. 2, 23–27.by probiotic bacteria. Nutr. Today 31, 5S–8S.

Waldeck, F., 1990. Funktionen des Magen-Darm-Kanals. In:Saxelin, M., Pessi, T., Salminen, S., 1995. Fecal recovery follow-Schmidt, R.F., Thews, G. (Eds.), Physiologie des Menschen,ing oral administration of Lactobacillus strain GG (ATCC24. Aufl. Springer, Berlin.53103) in gelatine capsules to healthy volunteers. Int. J. Food

Wang, J., Whetsell, M., Klein, J.R., 1997. Local hormone net-Microbiol. 25, 199–203.works and intestinal T cell homeostasis. Science 275, 1937–¨ ¨Saxelin, M., Rautelin, H., Salminen, S., Makela, H., 1996. The1939.safety of commercial products with viable Lactobacillus

Ward, D.M., Weller, R., Bateson, M.M., 1990. 16S rRNA se-strains. Infect. Dis. Clin. Pract. 5, 331–335.quences reveal numerous uncultured microorganisms in aShanahan, F., 1994. The intestinal immune system. In: Johnson,natural community. Nature 345, 63–65.L.R., Christensen, J., Jacobsen, E., Walsh, J.H. (Eds.), Physi-

ology of the Gastrointestinal Tract, 3rd ed. Raven Press, New Woese, C.R., 1987. Bacterial evolution. Microbiol. Rev. 51, 221–York, pp. 643–684. 271.