insulin uptake across the luminal membrane of the rat ...€¦ · innovative methodology insulin...

11
Innovative Methodology Insulin uptake across the luminal membrane of the rat proximal tubule in vivo and in vitro Pavel Kolman, 1,6 * Angelo Pica, 2,4 * Nicolas Carvou, 5 * Alan Boyde, 3,6 Shamshad Cockcroft, 5 Andrew Loesch, 6 Arnold Pizzey, 7 Mariadelina Simeoni, 2,4 Giovambattista Capasso, 2 and Robert J. Unwin 4,5 1 Institute of Physical Engineering, Brno University of Technology, Brno, Czech Republic; 2 Department of Internal Medicine, Second University of Naples, Naples, Italy; and 3 Barts and The London School of Medicine and Dentistry, 4 Centre for Nephrology, 5 Department of Physiology, 6 Department of Anatomy and Developmental Biology, and 7 Department of Haematology, University College London, London, United Kingdom Submitted 8 June 2008; accepted in final form 2 March 2009 Kolman P, Pica A, Carvou N, Boyde A, Cockcroft S, Loesch A, Pizzey A, Simeoni M, Capasso G, Unwin RJ. Insulin uptake across the luminal membrane of the rat proximal tubule in vivo and in vitro. Am J Physiol Renal Physiol 296: F1227–F1237, 2009. First published March 4, 2009; doi:10.1152/ajprenal.90351.2008.—We visualized insulin uptake in vivo across the apical membrane of the rat proximal tubule (PT) by confocal microscopy; we compared it with in vitro findings in a rat PT cell line (WKPT) using fluorescence microscopy and flow cytometry. Surface tubules were observed in vivo with a 633-nm single laser-illuminated real-time video-rate confocal scan- ning microscope in upright configuration for optical sectioning below the renal capsule. Fields were selected containing proximal and distal tubules; Cy5-labeled insulin was injected twice (the second time after 140 min) into the right jugular vein, and the fluorescence signal (at 650 – 670 nm) was recorded. Fluorescence was detected almost im- mediately at the brush-border membrane (BBM) of PT cells only, moving inside cells within 30 – 40 min. As a measure of insulin uptake, the ratio of the fluorescence signal after the second injection to the first doubled (ratio: 2.11 0.26, mean SE, n 10), indicating a “priming,” or stimulating, effect of insulin on its uptake mechanism at the BBM. This effect did not occur after pretreatment with intravenous lysine (ratio: 1.03 0.07, n 6; P 0.01). Cy2- or Cy3-labeled insulin uptake in a PT cell line in vitro was monitored by 488-nm excitation fluorescence microscopy using an inverted microscope. Insulin localized toward the apical membrane of these cells. Semiquantitative analysis of insulin uptake by flow cytometry also demonstrated a priming effect (upregulation) on insulin internal- ization in the presence of increasing amounts of insulin, as was observed in vivo; moreover, this effect was not seen with, or affected by, the similarly endocytosed ligand 2-glycoprotein. intravital confocal microscopy; fluorescence; kidney RENAL HANDLING OF INSULIN has been described previously using radiolabeled insulin combined with histochemical studies after timed injections in vivo (2) or in isolated proximal tubule (PT) perfusion studies in vivo (10) and in vitro (21, 20, 17) and in cultured cells (24). Insulin uptake in rabbit PT has also been monitored by electron microscopy (EM) after perfusion of gold-labeled insulin and subsequent cross-linking to its receptor with disuccinimidylsuberate (18). These studies have shown that insulin binding and uptake occur predominantly at the brush- border membrane (BBM) of PT cells (2, 22), where it is then translocated to endocytic vesicles and vacuoles, and even- tually degraded in lysosomes; a small portion (0.5%) is directly transcytosed to the basolateral membrane (19). However, unlike many other filtered peptides (34), insulin seems not to be degraded by brush-border enzymes (11). The receptors for insulin binding are recycled to the plasma membrane via dense apical tubules, either from small ves- icles or from larger endocytic vacuoles; although the early mechanisms involved in insulin binding and internalization in the PT are still unclear. Higher levels of insulin receptors are expressed on the basolateral membrane of PT cells than on the apical membrane (9, 29), and it is the basolateral receptors, present at other nephron sites (4), that are believed to mediate insulin’s bio- logical effects (32). Thus it was originally proposed that nonspecific charge interactions were responsible for the initial binding of insulin to the BBM (28). However, later studies reported a high-capacity, low-affinity receptor for insulin in the PT (15), which was eventually thought to be the endocytic receptor megalin. Orlando et al. (23), using chemical cross- linking of 125 I-labeled insulin to a renal microvillar membrane preparation, identified megalin as the binding protein, which required a 1,000-fold excess of unlabeled insulin to compete for binding; it was also competed with by glucagon. In con- trast, RAP (receptor-associated protein), which inhibits binding of other endocytosed ligands to megalin, such as retinol bind- ing protein (RBP) (13), transthyretin (27), and 2 -glycoprotein (2gpI) (16), did not inhibit binding. In the same study (23), uptake of FITC-labeled insulin in L2 yolk sac-derived cells (which are rich in megalin) was inhibited by a 50-fold excess of unlabeled insulin and by anti-megalin IgG, but again not by RAP. In the present study, we describe a method we have devel- oped for quantitative assessment of the disposition of fluores- cently labeled substances filtered at the kidney glomerulus using conventional confocal, rather than multiphoton (8), mi- croscopy in vivo. We chose to investigate insulin because of the available literature for comparison and as a prototypical filtered peptide hormone. We found that insulin’s visible up- take in vivo and in real time is confined to the PT and that it occurs apically, and we made the novel observation that insulin uptake is self-priming and upregulated. This finding was also confirmed in vitro in cultured PT cells and contrasted with the similarly endocytosed ligand 2gpI. * P. Kolman, A. Pica, and N. Carvou contributed equally to this work. Address for reprint requests and other correspondence: R. Unwin, Centre for Nephrology (London Epithelial Group), Univ. College London, Royal Free Campus, Rowland Hill St., London NW3 2PK, UK (e-mail: robert.unwin @ucl.ac.uk). Am J Physiol Renal Physiol 296: F1227–F1237, 2009. First published March 4, 2009; doi:10.1152/ajprenal.90351.2008. 0363-6127/09 $8.00 Copyright © 2009 the American Physiological Society http://www.ajprenal.org F1227 by 10.220.33.6 on November 6, 2017 http://ajprenal.physiology.org/ Downloaded from

Upload: others

Post on 30-Apr-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Insulin uptake across the luminal membrane of the rat ...€¦ · Innovative Methodology Insulin uptake across the luminal membrane of the rat proximal tubule in vivo and in vitro

Innovative Methodology

Insulin uptake across the luminal membrane of the rat proximal tubule in vivoand in vitro

Pavel Kolman,1,6* Angelo Pica,2,4* Nicolas Carvou,5* Alan Boyde,3,6 Shamshad Cockcroft,5

Andrew Loesch,6 Arnold Pizzey,7 Mariadelina Simeoni,2,4 Giovambattista Capasso,2

and Robert J. Unwin4,5

1Institute of Physical Engineering, Brno University of Technology, Brno, Czech Republic; 2Department of Internal Medicine,Second University of Naples, Naples, Italy; and 3Barts and The London School of Medicine and Dentistry, 4Centrefor Nephrology, 5Department of Physiology, 6Department of Anatomy and Developmental Biology, and 7Departmentof Haematology, University College London, London, United Kingdom

Submitted 8 June 2008; accepted in final form 2 March 2009

Kolman P, Pica A, Carvou N, Boyde A, Cockcroft S, Loesch A,Pizzey A, Simeoni M, Capasso G, Unwin RJ. Insulin uptake acrossthe luminal membrane of the rat proximal tubule in vivo and in vitro.Am J Physiol Renal Physiol 296: F1227–F1237, 2009. First publishedMarch 4, 2009; doi:10.1152/ajprenal.90351.2008.—We visualizedinsulin uptake in vivo across the apical membrane of the rat proximaltubule (PT) by confocal microscopy; we compared it with in vitrofindings in a rat PT cell line (WKPT) using fluorescence microscopyand flow cytometry. Surface tubules were observed in vivo with a633-nm single laser-illuminated real-time video-rate confocal scan-ning microscope in upright configuration for optical sectioning belowthe renal capsule. Fields were selected containing proximal and distaltubules; Cy5-labeled insulin was injected twice (the second time after�140 min) into the right jugular vein, and the fluorescence signal (at650–670 nm) was recorded. Fluorescence was detected almost im-mediately at the brush-border membrane (BBM) of PT cells only,moving inside cells within 30–40 min. As a measure of insulinuptake, the ratio of the fluorescence signal after the second injectionto the first doubled (ratio: 2.11 � 0.26, mean � SE, n � 10),indicating a “priming,” or stimulating, effect of insulin on its uptakemechanism at the BBM. This effect did not occur after pretreatmentwith intravenous lysine (ratio: 1.03 � 0.07, n � 6; P � 0.01). Cy2-or Cy3-labeled insulin uptake in a PT cell line in vitro was monitoredby 488-nm excitation fluorescence microscopy using an invertedmicroscope. Insulin localized toward the apical membrane of thesecells. Semiquantitative analysis of insulin uptake by flow cytometryalso demonstrated a priming effect (upregulation) on insulin internal-ization in the presence of increasing amounts of insulin, as wasobserved in vivo; moreover, this effect was not seen with, or affectedby, the similarly endocytosed ligand �2-glycoprotein.

intravital confocal microscopy; fluorescence; kidney

RENAL HANDLING OF INSULIN has been described previously usingradiolabeled insulin combined with histochemical studies aftertimed injections in vivo (2) or in isolated proximal tubule (PT)perfusion studies in vivo (10) and in vitro (21, 20, 17) and incultured cells (24). Insulin uptake in rabbit PT has also beenmonitored by electron microscopy (EM) after perfusion ofgold-labeled insulin and subsequent cross-linking to its receptorwith disuccinimidylsuberate (18). These studies have shown thatinsulin binding and uptake occur predominantly at the brush-

border membrane (BBM) of PT cells (2, 22), where it is thentranslocated to endocytic vesicles and vacuoles, and even-tually degraded in lysosomes; a small portion (�0.5%) isdirectly transcytosed to the basolateral membrane (19).However, unlike many other filtered peptides (34), insulinseems not to be degraded by brush-border enzymes (11).The receptors for insulin binding are recycled to the plasmamembrane via dense apical tubules, either from small ves-icles or from larger endocytic vacuoles; although the earlymechanisms involved in insulin binding and internalizationin the PT are still unclear.

Higher levels of insulin receptors are expressed on thebasolateral membrane of PT cells than on the apical membrane(9, 29), and it is the basolateral receptors, present at othernephron sites (4), that are believed to mediate insulin’s bio-logical effects (32). Thus it was originally proposed thatnonspecific charge interactions were responsible for the initialbinding of insulin to the BBM (28). However, later studiesreported a high-capacity, low-affinity receptor for insulin in thePT (15), which was eventually thought to be the endocyticreceptor megalin. Orlando et al. (23), using chemical cross-linking of 125I-labeled insulin to a renal microvillar membranepreparation, identified megalin as the binding protein, whichrequired a 1,000-fold excess of unlabeled insulin to competefor binding; it was also competed with by glucagon. In con-trast, RAP (receptor-associated protein), which inhibits bindingof other endocytosed ligands to megalin, such as retinol bind-ing protein (RBP) (13), transthyretin (27), and �2-glycoprotein(�2gpI) (16), did not inhibit binding. In the same study (23),uptake of FITC-labeled insulin in L2 yolk sac-derived cells(which are rich in megalin) was inhibited by a 50-foldexcess of unlabeled insulin and by anti-megalin IgG, butagain not by RAP.

In the present study, we describe a method we have devel-oped for quantitative assessment of the disposition of fluores-cently labeled substances filtered at the kidney glomerulususing conventional confocal, rather than multiphoton (8), mi-croscopy in vivo. We chose to investigate insulin because ofthe available literature for comparison and as a prototypicalfiltered peptide hormone. We found that insulin’s visible up-take in vivo and in real time is confined to the PT and that itoccurs apically, and we made the novel observation that insulinuptake is self-priming and upregulated. This finding was alsoconfirmed in vitro in cultured PT cells and contrasted with thesimilarly endocytosed ligand �2gpI.

* P. Kolman, A. Pica, and N. Carvou contributed equally to this work.Address for reprint requests and other correspondence: R. Unwin, Centre for

Nephrology (London Epithelial Group), Univ. College London, Royal FreeCampus, Rowland Hill St., London NW3 2PK, UK (e-mail: [email protected]).

Am J Physiol Renal Physiol 296: F1227–F1237, 2009.First published March 4, 2009; doi:10.1152/ajprenal.90351.2008.

0363-6127/09 $8.00 Copyright © 2009 the American Physiological Societyhttp://www.ajprenal.org F1227

by 10.220.33.6 on Novem

ber 6, 2017http://ajprenal.physiology.org/

Dow

nloaded from

Page 2: Insulin uptake across the luminal membrane of the rat ...€¦ · Innovative Methodology Insulin uptake across the luminal membrane of the rat proximal tubule in vivo and in vitro

METHODS

Insulin Labeling

Insulin was purchased from Sigma, and �2gpI was provided byScipac (Sittingbourne, UK). For the in vitro cell culture experiments,insulin was prepared and labeled with Cy2 or Cy3, as previouslydescribed (6). For the in vivo study, Cy5 was chosen as a suitablefluorophor, because we were using a 633-nm HeNe laser to curtailmitochondrial autofluorescence (3) (Cy5 is a far red fluorescent dye)and to improve reflection mode imaging. Insulin was labeled with Cy5according to the manufacturer’s instructions as follows: 1 mg ofinsulin was mixed with 75 �g of Cy5, and the labeled insulin wasseparated by size exclusion using a Sephadex G-15 column and theneluted with PBS. Labeling efficiency was estimated to be �30%.

Confocal Microscopy In Vivo

Male Sprague-Dawley rats (n � 4) weighing �200 g were main-tained on a standard laboratory diet and handled using approvedprotocols in accordance with institutional and Home Office licenseregulations. They were anesthetized with sodium thiopentone (100mg/kg ip; Link Pharmaceuticals, Horsham, UK) and prepared surgi-cally as for micropuncture of the left kidney (5). Rats were infusedintravenously via a right jugular venous catheter with a 0.154 M NaClsolution (“normal saline”) at a rate of 2 ml/h. During each experiment,the rat was placed beneath the objective of a confocal microscope(Noran Odyssey video-rate laser scanning confocal unit with a633-nm laser on a Nikon Optiphot upright microscope) on a speciallyadapted and thermoregulated stage. We used a 40/1.0 oil-immersionobjective lens with glass coverslips sandwiched to a thickness of 425�m to fill the space between the kidney surface and the objective lens;this also stabilized the kidney surface and minimized transmittedmovement from respiration and arterial pulsation. The confocal slit forthe fluorescence signal was set at 100 �m. In those experiments inwhich the rats were preinfused with lysine, a 10% solution in 0.154 MNaCl was started �30 min before the first insulin injection andcontinued throughout the experiment at dose of 200 mg/kg body wt,according to the protocol of Tucker et al. (33).

Image recording and analysis. Insulin uptake was rapid and suit-able for imaging renal tubules at video rate. The Noran Odysseyvideo-rate laser scanning confocal unit has standard video outputs,one for reflection mode, another for fluorescence, and one for themixed signal (not used in this study). We were not able to record bothchannels simultaneously. The signal was recorded using a SONYdigital camcorder. We used a microswitch controlled by the left kneeof the operator to select the reflection or fluorescence imaging modes.The time delay between the captured reflection and fluorescenceimages was 1 or 2/25 of a second. The image proved stable enough totreat the two frames as coming from the same place.

Cy5-insulin was injected into the jugular vein catheter (0.1 mg/kgbody wt) in a volume of 0.1 ml (although we also did some experi-ments using a femoral artery cannula in the aorta and placed justabove the left renal artery). Two consecutive injections were given atan interval of �140 min. The reflection and fluorescence images(Fig. 1, A and C, respectively) were “grabbed” from the short videosequences using Pinnacle Studio software (Pinnacle Systems; AvidTechnology, Daly City, CA). The average time interval betweenrepeated recording sequences was �10 min; the format of the grabbedimage frames was 640 � 480 pixels at 8 bits/pixel. Recordings weremade from several tubules (in different fields) during each experi-ment. Once we had captured the images, we used JASC Paint ShopPro 5 (Corel, Ottawa, ON, Canada), with its freehand smart edgeselection tool, to select from the reflection image (Fig. 1A) the tubulesegment to be analyzed, filling in the cells, including their whitereflective (negative black in Fig. 1, A–D) brush border, to create abinary mask. (The images shown in Fig. 1, A–D, are cropped free ofartifacts.)

For further analysis, we wrote a procedure in MATLAB software(The MathWorks, Natick, MA). The fluorescence intensity image(Fig. 1C) was transformed to an insulin pseudoconcentration map withthe use of a calibration function determined from calibration measure-ments. The mask was then divided into equal-width strips of �0.85�m (Fig. 1B); it was applied to monitor the spatial distribution ofinsulin in the tubular cells of the selected PT segment by calculatingthe averaged concentration in each strip separately. The concentration

Fig. 1. Real-time reflection and fluorescencemicroscopy images of rat kidney in vivo. Theimages are presented in negative (inverted)contrast, and a black border has been added tomake the comparison between images morereliable. A: reflection mode, a single videoframe used to define the area of interest foranalysis. A distal tubule (DT) is visible at leftside of the field of view (without the moreintense black brush border). PT, proximaltubule. B: contour map derived from the bi-nary mask that defines the linear strips withinwhich fluorescence was quantified to estimatethe change in mean fluorescence signal inten-sity within and across (apical to basolateral)PT cells. C: fluorescence mode, a single videoframe from which intensity is calculated.D: fluorescence mode, with an average of 5video frames to show morphology moreclearly.

Innovative Methodology

F1228 PROXIMAL TUBULE INSULIN UPTAKE IN VIVO AND IN VITRO

AJP-Renal Physiol • VOL 296 • MAY 2009 • www.ajprenal.org

by 10.220.33.6 on Novem

ber 6, 2017http://ajprenal.physiology.org/

Dow

nloaded from

Page 3: Insulin uptake across the luminal membrane of the rat ...€¦ · Innovative Methodology Insulin uptake across the luminal membrane of the rat proximal tubule in vivo and in vitro

values were corrected by using another calibration function to com-pensate for the effect of photobleaching.

Figure 2 illustrates the output of the MATLAB program in arepresentative control and lysine experiment. Figure 2, Ai and Bi,shows the temporospatial concentration of insulin inside proximaltubular cells. The origin of the spatial axis corresponds to the innerborder of the tubule (the apical brush border). Figure 2, Aii–Aiii andBii–Biii, shows the linear uptake and rate of uptake, respectively, afterthe first and second injections of labeled insulin; insulin values are inpseudoconcentration units (see calibration below).

Calibration of the fluorescence signal. We found that the intensitydistribution in fluorescence mode across the field of view was notconstant and that a calibration procedure had to be performed tocorrect for this, again using MATLAB. Calibration of Cy5-labeledinsulin concentration was performed in vitro with Cy5. We used aspecial bacteriological chamber with a depth of 10 �m (WeberScientific, Trenton, NJ). A series of solutions with different Cy5-insulin concentrations was then measured. The image data taken as ashort video sequence were averaged using MATLAB software, andthe resulting image was smoothed in Paint Shop Pro 5 using aGaussian blur filter (radius 3). However, we found that the intensity offluorescence for calibration measurements of high concentrations(�0.26 mg/ml) of Cy5-insulin varied by as much as 50%, probablybecause of significant evaporation from a small sample volume on arelatively large surface area. Therefore, it was not possible to calibrateand transform the intensity values recorded into real concentrationvalues for insulin. The intensity values were in the range of 0 to 255,with a value of 16 for the blank (water). Thus pseudoconcentrationswere derived by assuming a linear relationship between a given mean

fluorescence intensity in the image (I) and Cy5-insulin concentration(C) equal to 1/255 � (I � 16).

Correction for bleaching. After each Cy5-insulin injection, tubuleswere imaged approximately every 10 min, and the duration of irradi-ation in every measurement was �10 s. This gave a total of �5 minof irradiation in any one region so that the effect of photobleachingcould not be ignored. We determined experimentally how the fluo-rescence intensity dropped on bleaching the labeled insulin in renaltubular cells. The concentration values calculated from the fluores-cence images were corrected using these data (see Fig. 3).

Insulin Localization by EM in Kidney Tissue at �1 and �45 MinAfter Insulin Injection

Tissue preparation. For ultrastructural study, kidneys were per-fused through the renal artery at 1 min and 30–45 min after the insulininjection with 5 ml of normal saline, followed by 10 ml of fixative,consisting of 4% paraformaldehyde and 0.2% glutaraldehyde in 0.1 Mphosphate buffer (pH 7.4). The kidneys were then dissected out andimmersed in the same fixative for 5 h (at 4°C), followed by washingwith phosphate buffer and 0.1 M Tris-buffered saline (TBS; pH 7.6).Sections (60–70 �m thick) were cut through the kidney on a vi-bratome and collected in TBS. These were then processed with insulinantibody by using a preembedding immunogold-silver labeling tech-nique for EM (see below).

Immunogold-silver labeling. To improve immunoreactivity, thevibratome sections were infiltrated for 45 min with cryoprotectant,consisting of 25% sucrose and 10% glycerol in phosphate buffer,before being immersed in liquid nitrogen-cooled isopentane and then

Fig. 2. Insulin uptake in vivo in control (A) and lysine (B) experiments. i: insulin pseudoconcentration as a function of time and position within PT cells in therat kidney in vivo estimated from the fluorescence of Cy5-labeled insulin (Cy5-insulin). ii: uptake curve showing mean insulin pseudoconcentration vs. time afterinjection, comparing the 1st (F) and 2nd (�) insulin injections. iii: logarithmic plot of insulin pseudoconcentration vs. time after injection. The slope of eachcurve is a measure of the rate of insulin uptake after the 1st (F) and 2nd (�) injections. Insets in Ai and Bi are planar views of the 3-dimensional gray-scale mapsof pseudoconcentration.

Innovative Methodology

F1229PROXIMAL TUBULE INSULIN UPTAKE IN VIVO AND IN VITRO

AJP-Renal Physiol • VOL 296 • MAY 2009 • www.ajprenal.org

by 10.220.33.6 on Novem

ber 6, 2017http://ajprenal.physiology.org/

Dow

nloaded from

Page 4: Insulin uptake across the luminal membrane of the rat ...€¦ · Innovative Methodology Insulin uptake across the luminal membrane of the rat proximal tubule in vivo and in vitro

in liquid nitrogen for �10 s and were then thawed in cryoprotectantbefore being washed in TBS and placed for 1 h at room temperaturein 10% nonimmune normal goat serum (British BioCell International,Cardiff, UK) diluted in TBS containing 0.1% sodium azide. Sectionswere then 1) exposed for 20 h at 4°C to a mouse monoclonal insulin

antibody (see below for details) at a concentration of 0.3 �g/ml TBSwith 0.1% sodium azide, 2) washed 3 times for 10 min each in TBS,3) incubated for 20 h at 4°C with a goat anti-mouse IgG:1-nm goldconjugate (GAM1; British BioCell International) diluted 1:100 inTBS with sodium azide, 4) washed in TBS and then in distilled water,5) fixed for 10 min with 1% glutaraldehyde, 6) washed 10 times for5 min with deionized distilled water, and 7) subjected to augmentedgold labeling for 10 min with a silver enhancement kit (BritishBioCell International). The specimens were then washed with distilledwater and sodium cacodylate buffer (0.1 M), treated with 1% osmiumtetroxide for 10 min, and dehydrated in progressively higher concen-trations of ethanol followed by propylene oxide. The specimens wereembedded flat in Araldite. Ultrathin sections were stained with uranylacetate and lead citrate, and subsequently examined with a JEM-1010transmission electron microscope (TEM).

Immunocytochemical controls. The insulin antibody (2D11-H5)used in this study was a mouse monoclonal IgG1 antibody correspond-ing to amino acids 1–84, representing full-length porcine insulin(Santa Cruz Biotechnology, Santa Cruz, CA). According to thesupplier, this antibody reacts with insulin of human, porcine, andbovine origin by immunohistochemistry, immunoprecipitation, andELISA. In the present study, the specificity of immunolabeling wasinvestigated routinely by omission of the insulin antibody and IgGsteps, as well as by replacement of primary antibody with nonimmunenormal goat serum.

Fig. 3. Derivation of the bleaching correction. The curve shows thepseudoconcentration of Cy5-insulin as a function of time under constantconditions of laser irradiation (see text for details). C, change in concen-tration (increment); T, duration of irradiation.

Fig. 4. Electron micrograph of insulin bind-ing and uptake into cells of the PT detectedby an immunogold-silver labeling method at1–2 min (A) and 30–45 min (B) after insulininjection and under control conditions (C).A: at 1–2 min, insulin immunogold-silvergrains (arrows) localize to the brush border.N, cell nucleus; Mv, brush-border microvilli;Cap, a peritubular capillary. B: insulin im-munogold-silver grains are localized in thecytoplasm (arrows) and in an apical vacuole/multivesicular body (mvb) (at �45 minpostinjection). C: no insulin immunogold-silver grains were seen when the primaryantibody step was omitted or in control tis-sue (not shown). Bar, 1 �m.

Innovative Methodology

F1230 PROXIMAL TUBULE INSULIN UPTAKE IN VIVO AND IN VITRO

AJP-Renal Physiol • VOL 296 • MAY 2009 • www.ajprenal.org

by 10.220.33.6 on Novem

ber 6, 2017http://ajprenal.physiology.org/

Dow

nloaded from

Page 5: Insulin uptake across the luminal membrane of the rat ...€¦ · Innovative Methodology Insulin uptake across the luminal membrane of the rat proximal tubule in vivo and in vitro

Cell Culture

The WKPT 9302 Cl.2 cell line was a gift from Prof. F. Thevenod(Witten, Germany) and cultured as described previously (31). Thisepithelial cell line is derived from the early segment (S1) of the PTof normotensive Wistar-Kyoto rats and transduced with the onco-gene SV40 large T-antigen (35); it forms confluent, electricallyresistive polarized monolayers expressing apical microvilli, tightjunctions, and convolutions of the basolateral plasma membrane.Cells were maintained in Dulbecco’s modified Eagle’s medium(DMEM)/Ham’s nutrient mixture F-12 (1:1; Sigma) supplementedwith 10% bovine calf serum, 1.2 mg/ml NaHCO3, 5 �g/ml insulin,5 �g/ml apotransferrin, 4 �g/ml dexamethasone, 10 ng/ml EGF,and 100 U/ml penicillin and streptomycin. Cells were grown in75-cm2 flasks at 37°C with 5.0% CO2 and split 1/10 or 1/20 threetimes a week.

In Vitro Endocytosis Assay

WKPT cells were plated onto 13-mm-diameter glass coverslipsplaced in 16-mm-diameter wells (24-well plate) and washed twice inwarm HEPES buffer (20 mM HEPES, 137 mM NaCl, 3.0 mM KCl,2.0 mM MgCl2, 1.0 mM CaCl2, and 1 mg/ml glucose, 37°C). TheCy-labeled ligand (1–5 �M) was added to the cells (250 �l/well) andincubated for 2 h at 37°C. The cells were incubated in 0.2 M aceticacid and 0.5 M NaCl for 5 min at room temperature to remove anyresidual extracellular fluorescence, washed four times with PBS, andeither fixed in 4.0% formaldehyde for 10 min at room temperature foranalysis by confocal microscopy or detached in trypsin-EDTA foranalysis by flow cytometry. In competition experiments, the cellswere preincubated with unlabeled insulin or �2gpI for 10 min at 37°Cbefore the Cy2-labeled ligand was added. In priming experiments, thecells were incubated at 37°C for 60 min with unlabeled insulin orunlabeled �2gpI and washed four times with HEPES buffer beforeincubation with labeled ligand.

Confocal Microscopy In Vitro

After the fixed cells were washed three times in PBS, the coverslipswere mounted onto a 10-�l drop of Mowiol overnight at 4°C. Cellfluorescence was recorded by excitation at 488 nm with an Omnichromeseries 43 (PerkinElmer, Milan, Italy) argon ion laser system using anOlympus IX70 microscope fitted with a �100 oil-immersion objec-tive, unless stated otherwise. Images were acquired with a charge-coupled device camera (PerkinElmer) cooled to �35°C and controlledwith the Ultraview 4.0 software (PerkinElmer).

Analysis by Flow Cytometry

After 2 h of incubation with fluorescent ligand, the WKPT cells werewashed four times with ice-cold HEPES buffer supplemented with 1 mMEDTA. Cells were then incubated on ice with HEPES buffer plus 10 mMEDTA for 10 min and detached with trypsin-EDTA at 37°C for 5–15min. The trypsin was then quenched by addition of WKPT culturemedium, and the cells were pelleted by centrifugation at 1,300 rpm for 5min. The cell pellets were resuspended in ice-cold HEPES buffer plus 1mM EDTA and kept on ice until ready for analysis by flow cytometry. Atleast 10,000 cells per data point were analyzed for fluorescence on anEPICS Elite Flow cytometer (Beckman-Coulter, High Wycombe, UK).A gating strategy was employed that excluded cell aggregates andhigh-scatter cell events judged to be nonviable cells. The mean cellfluorescence was recorded for each data point. The data were thenprocessed with Summit v3.1 software (Cytomation, Fort Collins, CO).Histogram charts were produced for each sample, and the mean fluores-cence was resolved.

Statistics

All statistical comparisons were made using nonparametricKruskal-Wallis one-way ANOVA and/or unpaired Mann-Whitneytests; P � 0.05 was considered significant.

Fig. 5. Absence of competition between insulin and �2-glycoprotein (�2gpI) uptake. WKPT cells were incubated with 1 �M Cy2-labeled �2gpI (A, a–c) orCy2-labeled insulin (B, d–f ) for 2 h at 37°C in the presence of either 10 �M unlabeled �2gpI (b and e) or 10 �M unlabeled insulin (c and f ). For the control(a and d ), no unlabeled ligand was added. Uptake of insulin and �2gpI was assessed by confocal microscopy.

Innovative Methodology

F1231PROXIMAL TUBULE INSULIN UPTAKE IN VIVO AND IN VITRO

AJP-Renal Physiol • VOL 296 • MAY 2009 • www.ajprenal.org

by 10.220.33.6 on Novem

ber 6, 2017http://ajprenal.physiology.org/

Dow

nloaded from

Page 6: Insulin uptake across the luminal membrane of the rat ...€¦ · Innovative Methodology Insulin uptake across the luminal membrane of the rat proximal tubule in vivo and in vitro

RESULTS

In Vivo Confocal Microscopy

Descriptive analysis. As we (3) reported previously usingthe Noran Odyssey confocal microscope, the reflection modeimage showed details of capillary blood flow in identifiable anddistinguishable proximal and distal tubules: PTs were recog-nized by their intensely bright (light scattering) brush border,especially early PT segments (Fig. 1A; see Supplemental Video);erythrocytes were strongly reflective, and white cells were rec-ognizable by their slower motion, adhering to capillary wallsand some even marginating (3, 26). (Supplemental data for thisarticle is available online at the American Journal of Physiol-ogy-Renal Physiology website.) After the injection of free Cy5,fluorescence was evident in the lumen of distal tubules within1 min, but no uptake of fluorescence was seen in either theproximal or distal tubules (not shown).

After injection of Cy5-insulin, fluorescence appeared inperitubular capillaries within 0.5 min; soon afterward, flu-orescence was seen at the PT brush border, although fluo-rescence intensity and distribution were variable (see Sup-plemental Video). By �20 –30 min, there was an increase inintracellular fluorescence with a punctate pattern distributedtoward the basolateral membrane (Fig. 1, C and D; seeSupplemental Video). No fluorescence was detectable in the

distal tubules (Fig. 1, C and D; see Supplemental Video). Ineach recording in fluorescence mode, before injection ofCy5-insulin, there was no tubular autofluorescence; afterCy-5 insulin, there were no changes in the reflection modeimages.

Quantitative analysis. Quantitative analysis of the intra-cellular fluorescence signal after Cy5-insulin injection (Fig.2) showed an increase in intracellular fluorescence intensityup to �20 min, when it had almost reached a steady state.However, after the second injection of insulin, uptake ofinsulin seemed to be enhanced in magnitude (Fig. 2Aii) andrate (Fig. 2Aiii). The mean ratio for the change in concen-tration, as a measure of insulin uptake, after the secondinjection (second/first) was 2.11 � 0.26 (mean � SE). Afterpretreatment with lysine, this enhancement, or priming ef-fect, was abolished with a ratio of 1.03 � 0.07 (Fig. 2, Biiand Biii). These ratios were significantly different whencompared using the nonparametric Mann-Whitney test (two-tailed exact test, P � 0.01) with median values of 2.03 vs.0.98, respectively.

EM of insulin immunolabeling. EM examination of ratkidney tissue after injection of insulin showed localization ofinsulin at 1 and 45 min. Insulin immunolabeling wasevident in PT cells: 1 min after insulin injection, label wasdetected predominantly along microvilli of the brush border of

Fig. 6. Analysis of insulin uptake by flow cytometry. WKPT cells were incubated with 5 �M Cy2-labeled insulin for 2 h at 37°C in the presence of 0, 20, 50,100, or 200 �M unlabeled insulin. A: flow cytometry results. The shaded area shows the distribution for the no-ligand negative control (�ve ctrl), and the hatchedarea shows the distribution for the sample. B: mean fluorescence of the cells analyzed by flow cytometry. Values are means � SE (n � 3).

Innovative Methodology

F1232 PROXIMAL TUBULE INSULIN UPTAKE IN VIVO AND IN VITRO

AJP-Renal Physiol • VOL 296 • MAY 2009 • www.ajprenal.org

by 10.220.33.6 on Novem

ber 6, 2017http://ajprenal.physiology.org/

Dow

nloaded from

Page 7: Insulin uptake across the luminal membrane of the rat ...€¦ · Innovative Methodology Insulin uptake across the luminal membrane of the rat proximal tubule in vivo and in vitro

proximal tubular cells (Fig. 4A); after 45 min, there was mainlyintracellular localization (Fig. 4B). Label was present through-out the cytoplasm and also in endocytic vesicles and/or mul-tivesicular bodies (Fig. 4B). No immunolabeling was detected

in control sections (Fig. 4C), when no insulin was injected orinsulin was injected without incubation with anti-insulin anti-body, or when the primary antibody step was omitted insections from insulin-injected animals.

β β

ββ

Fig. 7. Analysis of �2gpI uptake by flow cytometry. WKPT cells were incubated with 1 �M Cy2-labeled �2gpI for 2 h at 37°C in the presence of 0, 4, 10, or20 �M unlabeled �2gpI. A: fluorescence-activated cell sorting (FACS) results. The shaded area shows the distribution for the no-ligand negative control, andthe hatched area shows the distribution for the sample. B: mean fluorescence of the cells analyzed by FACS. Values are means � SE (n � 3).

Fig. 8. L-Lysine does not affect insulin uptake in vitro. Confluent WKPT cells were incubated with 5 �M Cy2-insulin for 2 h at 37°C in presence of 40 �Munlabeled insulin, 40 �M �2gpI, or 40 �M L-lysine. Uptake of Cy2-insulin was assessed by confocal microscopy. A: confocal visualization of endocytosed Cy2-insulin(�40 objective). ve ctrl, positive control. B: histogram showing calculated mean fluorescence for 5 random fields. Values are means � SE (n � 5).

Innovative Methodology

F1233PROXIMAL TUBULE INSULIN UPTAKE IN VIVO AND IN VITRO

AJP-Renal Physiol • VOL 296 • MAY 2009 • www.ajprenal.org

by 10.220.33.6 on Novem

ber 6, 2017http://ajprenal.physiology.org/

Dow

nloaded from

Page 8: Insulin uptake across the luminal membrane of the rat ...€¦ · Innovative Methodology Insulin uptake across the luminal membrane of the rat proximal tubule in vivo and in vitro

In Vitro Cell Line Experiments

We (6) have shown previously that WKPT cells endocytoseinsulin and �2gpI, that uptake is temperature dependent, andthat these two proteins do not colocalize. To confirm thatinsulin and �2gpI do not compete for uptake, we incubatedCy2-�2gpI in the presence of excess unlabeled insulin or�2gpI (Fig. 5A, a–c). A 10-fold molar excess of �2gpI blockedCy2-�2gpI endocytosis (Fig. 5Ab), whereas the same concen-tration of insulin did not affect Cy2-�2gpI uptake (Fig. 5Ac). Inthe reciprocal experiment, a 10-fold molar excess of �2gpI didnot block Cy2-insulin uptake (Fig. 5B, d vs. e). A 10-foldmolar excess of insulin did not block uptake of Cy2-insulin,but appeared to enhance it (Fig. 5B, d vs. f). These resultsindicate that insulin and �2gpI are endocytosed independentlyand bind to different sites on the surface of WKPT cells. Inseparate experiments, we had examined whether their endo-cytic compartments could be differentiated based on stainingwith EEA1, a marker for early endosomes. We found thatEEA1 partially colocalizes with Cy2-�2gpI but not with Cy2-insulin (6).

Analysis by flow cytometry was used to quantify the uptakeof 5 �M Cy2-insulin in the presence of increasing amounts ofunlabeled insulin (Fig. 6). The mean fluorescence of the sortedcells increased in a linear manner and nearly doubled from 23.8 �1.19 arbitrary units (AU) when no unlabeled insulin was added, to51.0 � 2.44 AU with 100 �M unlabeled insulin, before reachinga plateau with 200 �M unlabeled insulin (50.5 � 0.93 AU). The

opposite trend was observed with �2gpI (Fig. 7): in cells exposedto 1 �M Cy2-�2gpI for 2 h, the mean fluorescence decreasedfrom 13.3 � 0.03 AU to the control value (4.6 � 0.47 AU) in thepresence of 10 �M �2gpI.

L-Lysine did not seem to affect insulin uptake in vivo(although it did prevent the priming effect), so we tested itseffect in vitro (Fig. 8). Again, addition of 40 �M unlabeledinsulin significantly enhanced Cy2-insulin uptake comparedwith the positive control; however, neither 40 �M �2gpI (asexpected) nor 40 �M L-lysine affected insulin uptake (althoughwe were unable to test the priming effect in the same way wedid in vivo).

In another set of experiments, WKPT cells were primedwith increasing amounts of insulin for 1 h, washed, and thenincubated with 5 �M Cy2-insulin. After 2 h of incubation,insulin uptake was measured in the insulin-primed cells:Cy2-insulin uptake was enhanced (Fig. 9). Together, theseresults suggest that endocytosis of insulin is regulated by apositive feedback mechanism, possibly involving the upregu-lation of its endocytic receptor. This is in contrast to �2gpIuptake, where excess �2gpI competes effectively with labeled�2gpI, indicating that the uptake system is saturable.

In L2 cells, which express high levels of megalin, �2gpIbinds to megalin and RAP is known to inhibit its uptake (16).Insulin internalization is also mediated by megalin, but RAPhas no effect on insulin uptake in L2 cells (13). To study theeffect of RAP on insulin and �2gpI endocytosis in WKPT

Fig. 9. Priming of insulin uptake in vitro. Confluent WKPT cells were incubated with unlabeled insulin (0–100 �M) for 1 h at 37°C. The cells were then washed4 times and incubated with 5 �M Cy2-labeled insulin for 2 h at 37°C. A: confocal visualization of endocytosed Cy2-insulin. B: histogram showing the calculatedmean fluorescence for 10 random fields. Values are means � SE (n � 10). *P � 0.05; **P � 0.01 vs. 0 �M insulin (Kruskal-Wallis 1-way ANOVA, followedby unpaired Mann-Whitney test).

Innovative Methodology

F1234 PROXIMAL TUBULE INSULIN UPTAKE IN VIVO AND IN VITRO

AJP-Renal Physiol • VOL 296 • MAY 2009 • www.ajprenal.org

by 10.220.33.6 on Novem

ber 6, 2017http://ajprenal.physiology.org/

Dow

nloaded from

Page 9: Insulin uptake across the luminal membrane of the rat ...€¦ · Innovative Methodology Insulin uptake across the luminal membrane of the rat proximal tubule in vivo and in vitro

cells, we monitored the uptake of Cy2-insulin or Cy2-�2gpI inthe presence of various concentrations of RAP (Fig. 10). RAPintegrity was tested by SDS-PAGE (Fig. 10A), and becauseBSA was present in the RAP buffer, a BSA control was usedin the uptake assay (Fig. 10, B and C, BSA). RAP did notinhibit insulin uptake (Fig. 10B) or �2gpI internalization,which is at odds with findings in L2 cells and suggests that inWKPT cells, �2gpI internalization is not mediated by megalin.

DISCUSSION

In Vivo Experiments

An important aim of our study was to devise a method forquantifying labeled insulin uptake in renal tubules in vivo byusing conventional single-laser confocal microscopy and to seewhether we could inhibit it and/or detect any change in uptake.Video-rate, long-wavelength confocal microscopy is a power-ful technique for this purpose because of its optical sectioningproperty at increased depth within the intact functioning kidneyin both reflection and fluorescence modes. Renal tubule mor-phology is readily discriminated in reflection mode, and insulinlabeled with a fluorescent dye is observable in fluorescencemode. When capturing a set of images and analyzing them bycomputer, we can get a spatiotemporal distribution of insulinmolecules in renal tubular cells.

In previous work exploiting the advantages of real-timevideo-rate confocal scanning light microscopy, we used aNoran Odyssey scanner coupled to an upright Nikon micro-scope to study subcapsular layers in the rat kidney (3, 26). Thepreparation of the animal for kidney imaging is as for renalmicropuncture experiments. In the original configuration (3)using an argon ion laser and the dominant 488-nm laser line,

we identified proximal and distal tubule segments by theirmorphology, and we imaged capillary flow in reflection modeand mitochondrial autofluorescence in the yellow-orange spec-tral range. A number of fluorescent substances were used tovisualize capillary plasma, highlighting red cells in negativecontrast, prelabeled white cells, and passage into and transitthrough the tubular lumen (3). The strong mitochondrial autofluo-rescence in the yellow-orange spectral range limited the utility ofthe fluorescence approach with dyes excited by the 488-nm laserline (3). Therefore we modified the instrument by fitting a633-nm HeNe laser with the expectation that the reduction inlight scattering at the longer wavelength would give a usefulimprovement in reflection mode imaging, which we used tostudy diuretic-induced changes in renal tubular diameter (26).A similar in vivo approach also has been described, thoughusing dual-photon microscopy (8), which has some advantages(particularly the ability to use more than one fluorophor),although it is more costly and less widely available. This tech-nique has also been widely described (25), and usually withoutparallel or complementary in vitro studies, although attempts havebeen made to make more quantitative analyses of glomerularfiltration and permeability (12, 36).

In the present study, we have shown that we can visualizenative Cy5 and Cy5-labeled substances introduced into thecirculation and monitor their immediate binding, rapid uptake,and passage through the renal tubule in real time, in vivo.Indeed, this pattern in the PT was confirmed on immuno-EM ofkidney sections taken at early and late time points after insulininjection. However, we were faced with the challenge ofquantifying the fluorescence signal to measure uptake of fluo-rescent label. Difficulties arose because of 1) the nonunifor-

β I

Fig. 10. Receptor-associated protein (RAP)does not inhibit uptake of either insulin or�2gpI. A: analysis of recombinant RAP (0.5,1.0, and 2.0 �g) by SDS-PAGE. BSA sam-ples (0.5, 1.0, 2.0, and 3.0 �g) were loaded asa control, since BSA was present in the RAP.RAP (40 kDa) is indicated on the gel. B andC: confluent WKPT cells were preincubatedwith 0.2, 0.5, or 1.0 �M RAP for 1 h at 37°C,and 5 �M Cy2-�2gpI (B) or Cy2-insulin (C)was subsequently added to the cells and in-cubated for 2 h at 37°C. The fluorescence ofthe cells was measured by confocal micros-copy. For each condition, the mean fluores-cence for 10 random fields was quantified.Values are means � SE (n � 10).

Innovative Methodology

F1235PROXIMAL TUBULE INSULIN UPTAKE IN VIVO AND IN VITRO

AJP-Renal Physiol • VOL 296 • MAY 2009 • www.ajprenal.org

by 10.220.33.6 on Novem

ber 6, 2017http://ajprenal.physiology.org/

Dow

nloaded from

Page 10: Insulin uptake across the luminal membrane of the rat ...€¦ · Innovative Methodology Insulin uptake across the luminal membrane of the rat proximal tubule in vivo and in vitro

mity of fluorescence excitation within the field of view, whichwe solved by calibrating every pixel address separately; and2) a longer-term bleaching effect for bound Cy5 label, solved byknowing the irradiation time for each field and an empiricallydetermined bleaching rate, and by applying an appropriate cor-rection. On the basis of our solutions, we have been able to showthat the method can be used for relative quantification of physi-ological measurement of fluorescent label uptake in vivo.

Previous studies have shown that L-lysine can inhibit tubularreabsorption of filtered peptides (14), including albumin (33);lysine was also recently shown to alter endocytic receptortrafficking (30). We tested lysine’s effect on insulin uptake byinfusing L-lysine intravenously for 90 min, followed immedi-ately by a first injection of labeled insulin; the second injectionfollowed after 140 min. In control experiments, we found thatthe first injection of insulin stimulated its uptake following asecond injection, a priming effect, and that this effect wasblunted after lysine pretreatment.

In one experiment, we did try to combine PT micropuncturewith confocal microscopy to visualize the uptake of insulininjected directly into the tubule lumen. Although this approachallowed more accurate dosage, as well as localized delivery ofinsulin (or any other fluorescently labeled substance), it provedtechnically demanding because of the short working distanceof the high-aperture objective lenses needed for confocal mi-croscopy, which made insertion of the micropipette under theobjective difficult (see Supplemental Video).

In Vitro Experiments

We have also confirmed in vitro that insulin and �2gpI(which was not tested in vivo) are endocytosed by two path-ways in WKPT cells: insulin and �2gpI do not compete forbinding and do not colocalize in the same intracellular com-partment after uptake; �2gpI endocytosis is constitutive andcan be blocked with excess �2gpI, whereas insulin uptake isagain upregulated (primed) in the presence of increasingamounts of insulin.

We have shown recently that the uptake of both insulin and�2gpI is mediated by clathrin and that insulin internalizationoccurs via the adaptor complex AP-2, although AP-2 expres-sion is not required for �2gpI uptake; moreover, uptake of�2gpI is widespread and insulin uptake is predominantly apical(6). The lack of interference of insulin endocytosis by insulinwas previously noted in 1980 when 125I-labeled insulin wasinjected in the whole animal: quantitative autoradiographyshowed extensive labeling over proximal convoluted tubules,which was not depressed by coinjection of unlabeled insulin(1). Our finding of the effect of insulin appears to relate to theincrease in its binding sites: once cells have been sensitized toinsulin, insulin can be removed, and the cells still showenhanced uptake. This suggests that insulin can stimulatesignal transducing pathways that can mobilize sequesteredproteins from the interior of the cell to the plasma membranefor the enhanced uptake of insulin.

Two previous studies in yolk sac L2 cells (which expresshigh levels of the endocytic receptor megalin) reported that�2gpI and insulin were endocytosed by binding to megalin.However, whereas �2gpI uptake was Ca2 dependent andcompletely blocked by RAP (16), insulin binding to megalindid not require Ca2, and insulin uptake was not inhibited by

the megalin binding protein RAP (23). In the WKPT cell line,the expression level of megalin is low and similar to that in theLLCPK cell line. RAP did not inhibit endocytosis of insulin or�2gpI, which suggests that uptake of �2gpI and insulin doesnot depend on megalin. The observation that megalin-depen-dent uptake is mainly apical, whereas we observed that �2gpIuptake is more widespread, suggests an alternative receptor.Although �2gpI uptake is clathrin mediated, the adaptor isunlikely to be AP-2, since depletion of the �2-subunit of AP2,AP-50, had no effect on endocytosis; in contrast, insulin uptakeis apical and is inhibited by depletion of AP-50 protein (6).

Retinol binding protein (RBP) uptake in vivo is also medi-ated almost entirely by megalin, as suggested by the absence ofRBP-containing granules in the PT of megalin knockout mice(7). In immortalized rat renal proximal tubule (IRPT) cells,RBP transcytosis is also mediated by megalin, but RBP trans-port to the lysosome and subsequent degradation (55% ofinternalized RBP) are independent of megalin (13). Therefore,in the IRPT model, other megalin-independent mechanisms ofendocytosis must account for RBP degradation: other non-megalin receptors, or fluid-phase pinocytosis, has been sug-gested.

Finally, we have shown that it is not only possible to imagethe distribution of filtered fluorescently labeled substances inreal time but also that it is feasible to develop methods forrelative quantification of the fluorescence signal in vivo; more-over, in the case of insulin, there is a novel priming effect onits uptake in the early PT following initial exposure, althoughthe mechanism of this phenomenon is not apparent from ourexperiments.

Several questions need to be addressed in future studies.What is the mechanism underlying the priming effect observedwith insulin in vivo and in vitro, and does it have any func-tional significance for the effects of insulin? Does it extend toother filtered proteins or peptides? Does it have any implicationfor protein handling by the proximal tubule?

ACKNOWLEDGMENTS

We thank Frank Thevenod for providing the WKPT cell line. We thankHarvey McMahon, George Banting, and Mark Marsh for reagents. �-2 Gly-coprotein-I was a generous gift from Scipac (Sittingbourne, UK). The real-timeconfocal Cy5 fluorescence studies were conducted in the Department ofAnatomy and Developmental Biology, University College London.

GRANTS

We thank the National Kidney Research Fund and St Peter’s Trust forfunding support. The Odyssey confocal microscope was funded by the Well-come Trust, and its upgrade to red laser operation was funded by the RoyalSociety of London.

REFERENCES

1. Bergeron JJ, Rachubinski R, Searle N, Borts D, Sikstrom R, PosnerBI. Polypeptide hormone receptors in vivo: demonstration of insulinbinding to adrenal gland and gastrointestinal epithelium by quantitativeradioautography. J Histochem Cytochem 28: 824–835, 1980.

2. Bourdeau JE, Chen ER, Carone FA. Insulin uptake in the renal proxi-mal tubule. Am J Physiol 225: 1399–1404, 1973.

3. Boyde A, Capasso G, Unwin RJ. Conventional and confocal epi-reflec-tion and fluorescence microscopy of the rat kidney in vivo. Exp Nephrol6: 398–408, 1998.

4. Butlen D, Vadrot S, Roseau S, Morel F. Insulin receptors along the ratnephron: [125I] insulin binding in microdissected glomeruli and tubules.Pflugers Arch 412: 604–612, 1988.

Innovative Methodology

F1236 PROXIMAL TUBULE INSULIN UPTAKE IN VIVO AND IN VITRO

AJP-Renal Physiol • VOL 296 • MAY 2009 • www.ajprenal.org

by 10.220.33.6 on Novem

ber 6, 2017http://ajprenal.physiology.org/

Dow

nloaded from

Page 11: Insulin uptake across the luminal membrane of the rat ...€¦ · Innovative Methodology Insulin uptake across the luminal membrane of the rat proximal tubule in vivo and in vitro

5. Capasso G, Unwin R, Agulian S, Giebisch G. Bicarbonate transportalong the loop of Henle. I. Microperfusion studies of load and inhibitorsensitivity. J Clin Invest 88: 430–437, 1991.

6. Carvou N, Norden AG, Unwin RJ, Cockcroft S. Signalling throughphospholipase C interferes with clathrin-mediated endocytosis. Cell Signal19: 42–51, 2007.

7. Christensen EI, Moskaug JO, Vorum H, Jacobsen C, Gundersen TE,Nykjaer A, Blomhoff R, Willnow TE, Moestrup SK. Evidence for anessential role of megalin in transepithelial transport of retinol. J Am SocNephrol 10: 685–695, 1999.

8. Dunn KW, Sandoval RM, Kelly KJ, Dagher PC, Tanner GA, Atkin-son SJ, Bacallao RL, Molitoris BA. Functional studies of the kidney ofliving animals using multicolor two-photon microscopy. Am J Physiol CellPhysiol 283: C905–C916, 2002.

9. Hammerman MR. Interaction of insulin with the renal proximal tubularcell. Am J Physiol Renal Fluid Electrolyte Physiol 249: F1–F11, 1985.

10. Hellfritzsch M, Christensen EI, Sonne O. Luminal uptake and intracel-lular transport of insulin in renal proximal tubules. Kidney Int 29: 983–988, 1986.

11. Hjelle JT, Oparil S, Peterson DR. Subcellular sites of insulin hydrolysisin renal proximal tubules. Am J Physiol Renal Fluid Electrolyte Physiol246: F409–F416, 1984.

12. Kang JJ, Toma I, Sipos A, McCulloch F, Peti-Peterdi J. Quantitativeimaging of basic functions in renal (patho)physiology. Am J Physiol RenalPhysiol 291: F495–F502, 2006.

13. Marino M, Andrews D, Brown D, McCluskey RT. Transcytosis ofretinol-binding protein across renal proximal tubule cells after megalin (gp330)-mediated endocytosis. J Am Soc Nephrol 12: 637–648, 2001.

14. Mazanti I, Hermann KL, Nielsen AH, Poulsen K. Ultrafiltration ofrenin in the mouse kidney studied by inhibition of tubular protein reab-sorption with lysine. Clin Sci (Lond) 75: 331–336, 1988.

15. Meezan E, Pillion DJ, Elgavish A. Binding and degradation of 125I-insulin by isolated rat renal brush border membranes: evidence for lowaffinity, high capacity insulin recognition sites. J Membr Biol 105: 113–129, 1988.

16. Moestrup SK, Schousboe I, Jacobsen C, Leheste JR, Christensen EI,Willnow TE. Beta2-glycoprotein-I (apolipoprotein H) and beta2-glyco-protein-I-phospholipid complex harbor a recognition site for the endocyticreceptor megalin. J Clin Invest 102: 902–909, 1998.

17. Nielsen S. Time course and kinetics of proximal tubular processing ofinsulin. Am J Physiol Renal Fluid Electrolyte Physiol 262: F813–F822,1992.

18. Nielsen S. Endocytosis in proximal tubule cells involves a two-phasemembrane-recycling pathway. Am J Physiol Cell Physiol 264: C823–C835, 1993.

19. Nielsen S. Sorting and recycling efficiency of apical insulin binding sitesduring endocytosis in proximal tubule cells. Am J Physiol Cell Physiol264: C810–C822, 1993.

20. Nielsen S, Christensen EI. Insulin absorption in renal proximal tubules:a quantitative immunocytochemical study. J Ultrastruct Mol Struct Res102: 205–220, 1989.

21. Nielsen S, Nielsen JT. Influence of flow rate and perfused load on insulinabsorption in isolated proximal tubules. Am J Physiol Renal Fluid Elec-trolyte Physiol 254: F802–F812, 1988.

22. Nielsen S, Nielsen JT, Christensen EI. Luminal and basolateral uptake ofinsulin in isolated, perfused, proximal tubules. Am J Physiol Renal FluidElectrolyte Physiol 253: F857–F867, 1987.

23. Orlando RA, Rader K, Authier F, Yamazaki H, Posner BI, BergeronJJ, Farquhar MG. Megalin is an endocytic receptor for insulin. J Am SocNephrol 9: 1759–1766, 1998.

24. Rabkin R, Hamik A, Yagil C, Hamel FG, Duckworth WC, Fawcett J.Processing of 125I-insulin by polarized cultured kidney cells. Exp Cell Res224: 136–142, 1996.

25. Sandoval RM, Kennedy MD, Low PS, Molitoris BA. Uptake andtrafficking of fluorescent conjugates of folic acid in intact kidney deter-mined using intravital two-photon microscopy. Am J Physiol Cell Physiol287: C517–C526, 2004.

26. Simeoni M, Boyde A, Shirley DG, Capasso G, Unwin RJ. Applicationof red laser video-rate scanning confocal microscopy to in vivo assessmentof tubular function in the rat: selective action of diuretics on tubulardiameter. Exp Physiol 89: 181–185, 2004.

27. Sousa MM, Saraiva MJ. Internalization of transthyretin. Evidence of anovel yet unidentified receptor-associated protein (RAP)-sensitive recep-tor. J Biol Chem 276: 14420–14425, 2001.

28. Sumpio BE, Maack T. Kinetics, competition, and selectivity of tubularabsorption of proteins. Am J Physiol Renal Fluid Electrolyte Physiol 243:F379–F392, 1982.

29. Talor Z, Emmanouel DS, Katz AI. Insulin binding and degradation byluminal and basolateral tubular membranes from rabbit kidney. J ClinInvest 69: 1136–1146, 1982.

30. Thelle K, Christensen EI, Vorum H, Orskov H, Birn H. Characteriza-tion of proteinuria and tubular protein uptake in a new model of oralL-lysine administration in rats. Kidney Int 69: 1333–1340, 2006.

31. Thevenod F, Friedmann JM. Cadmium-mediated oxidative stress inkidney proximal tubule cells induces degradation of Na/K-ATPasethrough proteasomal and endo-/lysosomal proteolytic pathways. FASEB J13: 1751–1761, 1999.

32. Tiwari S, Sharma N, Gill PS, Igarashi P, Kahn CR, Wade JB,Ecelbarger CM. Impaired sodium excretion and increased blood pressurein mice with targeted deletion of renal epithelial insulin receptor. ProcNatl Acad Sci USA 105: 6469–6474, 2008.

33. Tucker BJ, Rasch R, Blantz RC. Glomerular filtration and tubularreabsorption of albumin in preproteinuric and proteinuric diabetic rats.J Clin Invest 92: 686–694, 1993.

34. Wilkins MR, Unwin RJ, Kenny AJ. Endopeptidase-24.11 and its inhib-itors: potential therapeutic agents for edematous disorders and hyperten-sion. Kidney Int 43: 273–285, 1993.

35. Woost PG, Orosz DE, Jin W, Frisa PS, Jacobberger JW, Douglas JG,Hopfer U. Immortalization and characterization of proximal tubule cellsderived from kidneys of spontaneously hypertensive and normotensiverats. Kidney Int 50: 125–134, 1996.

36. Yu W, Sandoval RM, Molitoris BA. Quantitative intravital microscopyusing a Generalized Polarity concept for kidney studies. Am J Physiol CellPhysiol 289: C1197–C1208, 2005.

Innovative Methodology

F1237PROXIMAL TUBULE INSULIN UPTAKE IN VIVO AND IN VITRO

AJP-Renal Physiol • VOL 296 • MAY 2009 • www.ajprenal.org

by 10.220.33.6 on Novem

ber 6, 2017http://ajprenal.physiology.org/

Dow

nloaded from