4 ' " _ f;‘ V.., -‘o r ‘ ‘ .- - . - 5 I A _ A - -“* Li; ' ‘ ‘3 - . ‘_ - A -:-—-_"$"‘9~ _ _ >_ .- _ ~— " \ _ -‘.— ..—'_1.l"—— ____.—_:,r_:_:'. 7;;253- —_;._ -.——i ‘A 4 ‘w -.—A- :vrr I III II “I III I JIIIIJ " AH. II IIIIIII . 4? f ‘5- ' III I:« -‘ »- II .I- If I . 5 ‘ 3 ”I ”II II It. I . '“ -' I I II II I I . ‘. . " MN? " " .2 ‘ . ‘ I I I . n ‘a-.I I :i“ i‘y I I III'III I " I - I I - ..n 35*“ 31;“ I; II I I' I .I' I2)" ’ 3:. ‘5' . Ihu I IHI bf‘;xifi‘q II“ .I '5 I II I III I III I I; III M- -;..____.__::"= :W W M“: _,:__.__.- ._._—. —-——— w- _.—...-- 4—3. _.._ __:;.._ ——:~—.-“ d. M“...- H __..._.. ___.__1._._ b4‘ “- ”*— M __,___.,. W -M—‘v _ " ‘ ' ' ' :-———~--—~ ,_ ,..,.~_.— M ‘— is? M 3):: — H 3—“... En; _._... ' F... “ M w, _.~ ._ ”— _. q _. —-‘~ .— ~— ~— -——:~_-"_:__:.:.- ."I I II III III III III IIHIIII I I III I III I IIII IIIIII JIIIIIIIIII “III” II IIIIIIIIIIII I I» "I: I . -. III” I I I. ' 1:11! II IIIIII' I I III, I I,. III, IIIIIII IIIIII I I . :I'I'III‘ .rzafi'I I ". I" A I I 3‘ "I . “ ‘I‘IIII IIIIII II II" I.» é'iIII‘“ ‘I III" I", .I‘IV'I 3f II I I, I ”I, I: J, III.- - d' 1;}; :" I= . ' 3;; ‘IIIII II IIurII“. HE~IIIII\II' II 1*. IEI'IIIIIIII ' IIIILR' I: I III III. I‘I’III III; III I I III IL I '32-";E :=E§=; ._,9 é..4: “' - III Will ll llllllifllll’lll 31293m004010048209 L :2? f I 5 i? with: ~ *1 334“ Unive s FEW (a?) is» w s-q This is to certify that the thesis entitled Prostaglandin Metabolism In Papillary Collecting Tubule Cells From Rabbit Kidney presented by Frank Charles Grenier has been accepted towards fulfillment of the requirements for degree in _Bj.o.ch.em_Ls t ry Doctor of Philosophy Major professor Date nag/5431703) /5 /r./ 5/ 0-7639 \ 2 9 1998 OVERDUE FINES: 25¢ per day per item RETURNING LIBRARY MATERIALS: Place in book return to move charge from circulation records PROSTAGLANDIN METABOLISM IN PAPILLARY COLLECTING TUBULE CELLS FROM RABBIT KIDNEY By Frank Charles Grenier A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Department of Biochemistry 1981 a I h A :V .1 I . .. at.“ N a ,t'; av". L, Lw/vr r' rm ABSTRACT PROSTAGLANDIN METABOLISM IN PAPILLARY COLLECTING TUBULE CELLS FROM RABBIT KIDNEY By Frank Charles Grenier Homogeneous (>97%) populations of renal papillary collecting tubule (RPCT) cells were isolated from the rabbit kidney. RPCT cells were characterized as being derived from the collecting tubule on the basis of anatomical source, size, enzyme histochemistry, and cyclooxygenase antigenicity; in addition RPCT cells synthesized 3',5'-cyclic AMP (CAMP) in response to arginine vasopressin, formed hemicysts when grown to confluency and adhered with morphological asymmetry to Millipore filters. Homogenates of RPCT cells when incubated with [3HJ-arachidonic acid formed 6-keto-PGF1G, PGFZQ, PGEz and PGDZ. At arachidonic acid concentrations below 2 E! the major product formed was 6-keto-PGF1a; at higher concentrations PGEZ was the major radioactive prostaglandin formed. A series of hormones were tested for their influence on the release of immunoreactive prostaglandins (iPG) by intact RPCT cells grown in monolayer culture; the major product (ca. 75%) under both bassal and stimulated conditions was iPGEZ. At very low concentrations (1 10-10.!) bradykinin, lysyl-bradykinin and methionyl-lysylbrady- kinin all caused 3-5 fold increases in iPGEZ formation. Significantly, neither arginine vasopressin (AVP) (10'7_M) nor desamino-AVP (10-7.!) caused prostaglandin release by RPCT cells. These results indicate that kinins can act directly on the collecting tubule to elicit PGE2 formation; furthermore, the effect of kinins may be natriuretic since PGEZ has been shown by others to inhibit Na+ resorption by the medullary collecting tubule. RPCT cells synthesized cAMP in response to AVP, parathyroid hormone and glucagon but not to adrenergic agents. In addition, exogenous PGEZ and P612, increased intracellular cAMP concentrations in RPCT cells. The effects of PGE2 and P612 on AVP-induced cAMP production by RPCT cells were investigated. At 10‘7 M AVP and 10'5 M PGEZ or P612, the increase in both total and intracellular cAMP levels was less than that expected for an additive response of AVP plus prostaglandin. This suggests that prostaglandins can partially block AVP-induced cAMP accumulation in the collecting tubule. TO KEARSTIE ii ACKNOWLEDGEMENTS I would like to thank Dr. "Wild Bill" Smith for his advice, encouragement and financial support. His kindness was only superceded by his generosity and good looks. I would also like to thank all the members of the laboratory and secretarial staff for many hours of enjoyment in and out of the laboratory. I would like to express my gratitude to Thomas Rollins for doing the electron microscopy reported in this thesis. A special thanks goes out to Arlyn Garcia-Perez for being social director, and to Rick Huslig and Dave DeWitt both of whom began this journey with me. whether chasing sheep through petrified muck during December or floating through "ozone trips", we've shared innunerable indignities. And most importantly I would like to thank my wife Kearstie. She stood by her man. Page LIST OF TABLES ....................................................... v LIST OF FIGURES ...................................... . .............. vi INTRODUCTION ......................................................... 1 CHAPTER I. LITERATURE REVIEW.... ........................................ 3 Prostaglandin Metabolism ..................................... 3 Kidney Function .............................................. 8 Localization of Prostaglandin Synthesis ..................... 11 Prostaglandin Function in the Kidney ........................ 13 Prostaglandins and Water Resorption..... .................... 14 The Kallikrein-Kinin system and Prostaglandins .............. 16 II. PROSTAGLANDIN SYNTHESIS BY RPCT CELLS ISOLATED BY TRYPSIN DISSOCIATION . ................ . ..................... 20 Materials and Methods ....................................... 21 Results and Discussion ...................................... 27 III. KINlN-INDUCED PROSTAGLANDIN SYNTHESIS BY RPCT CELLS IN CULTURE.... ...... .................... ..... . .............. 49 Materials and Methods ...... . ............. .. ............... ..50 Results ..................................................... 61 Discussion .................................................. 89 IV. INTERRELATIONSHIPS AMONG BRADYKININ, PGEZ, VASOPRESSIN AND CAMP IN RPCT CELLS. O O O O O O I O O O O O ....... O O O O O O O O O O O ....... 94 Materials and Methods ....................................... 95 Results .............. . ...................................... 99 Discussion.. .................. . ............................ 112 SUMMARY DISCUSSION ........ . ...... .... .............................. 118 BIBLIOGRAPHY ....................................................... 122 TABLE OF CONTENTS iv LIST OF TABLES Table Page I Prostaglandin products formed from arachidonic acid by cell p0pulations isolated from rabbit renal papillae ......................... . ................... 41 II Characterization of 6-keto-PGF1G (Rf values) ................ 42 III Cross-reactivities of prostaglandin anti-sera with various prostaglandins......................... ............. 54 IV Formation of 3',5'-cyclic AMP by RPCT cells in response to hormonal effectors..................... ......... 71 V Synthesis of iPGEz by RPCT cells in response to hormonal effectors ................... ...... ................. 79 VI Characterization of prostaglandins formed by RPCT cells in response to hormonal effectors ..................... 81 Figure Page 1 Overview of prostaglandin metabolism ........................ ....5 2 Schematic representation of the sites of cyclooxygenase in the kidney.... ......... ... ........ ........... ............... IO 3 Trypsin-isolated RPCT cells in suspension. ..... .. .............. 29 4 Fluorescent photomicrographs of RPCT cells treated with and without anti-cyclooxygenase serum............ .............. 32 5 [14CJ-glucose oxidation by isolated RPCT cells ................. 34 6 [14CJ-leucine incorporation by isolated RPCT cells ............. 36 7 A radiochromatogram of the products formed upon incubation of [3 H]- arachidonic acid ................ . ...................... 4O 8 Time course for the biosynthesis of various prostaglandin products by a RPCT cell homogenate ................. . ........... 45 9 Prostaglandin biosynthesis by RPCT cell homogenates at different initial concentrations of [3H]-arachidonic acid ......................................... 48 10 Phase photomicrograph of RPCT cells grown eight days in monolayer culture ......... ................ .................. 64 11 Electron micrographs of (A) intact collecting tubule from rabbit, (B) isolated RPCT cells grown on Millipore filters, and (C) junctional complex of RPCT cell grown on Millipore filter...... ....... . .............. 66 12 Hemicysts present in culture of RPCT cells....... .............. 69 13 Release of cAMP into media by RPCT cells incubated with AVP, oxytocin and PTH....... ...... .......... .............. 73 14 Time course of intracellular and extracellular cAMP by RPCT cells in response to 10'7 M AVP ......... .............. 75 15 RPCT cell growth in monolayer culture ..... ....... .............. 78 LIST OF FIGURES vi Figure 16 17 18 19 20 21 22 23 Time course of iPGEz synthesis by RPCT cells in monolayer culture... ...... . ........ .. ...... . ..... . ......... Dependence of iPGEZ synthesis on kinin concentration ....... Dependence of intracellular cAMP formation on prostaglandin concentration.................... ........... . Time course for intracellular cAMP formation caused by PGE2 and bradykinin................................... .......... Effect of stirring on intracellular cAMP formation caused by PGEZ and bradykinin ........ ..... ......... ............... Time course of the effect of PGE2 on intracellular cAMP formation caused by AVP .................................... Effect of AVP, PGEZ and bradykinin on intracellular cAMP synthesis by RPCT cells..................... ............... Model illustrating the effects of prostaglandins, bradykinin, cAMP and AVP in RPCT cells ..................... ....85 ....87 ...101 ...103 ...106 ...109 ...111 ...114 INTRODUCTION Various experimental approaches have been used to investigate how prostaglandins are involved with kidney processes including 1) whole animal clearance methods 2) perfusion of isolated kidneys, 3) thicropuncture and stopped-flow techniques and 4) techniques using renal :slices and renal homogenates (38,57). A drawback of all these methods, f1owever, is that in each case many different kidney cells may influence aarry measurements made using these systems. It is virtually impossible t:c> interpret such data on the individual cell level. Nevertheless, these methods have been useful in defining both the prostaglandin b‘i osynthetic capacity of the kidney and the hormones which cause prostaglandin biosynthesis by the kidney (57). Frustrated by the relative indirectness of these methods, Burg 5232 ad, developed a technique for microperfusing individual segments of kridney tubules (42). The tremendous advantage of this technique was its liack of complexity. Only one tubular cell type was being studied and therefore the interpretation of the data was greatly simplified. Using perfused collecting tubules, for example, Grantham gt _a_l_. were able to (demonstrate that PGE1 inhibits arginine vaSOpressin (AVP)-stimulated vvater resorption in the collecting tubule (61). This information was lJnattainable prior to Burg's advancement. The major limitation of Burg's method, however, is the difficulty ‘in making biochemical measurements with isolated tubule segments. 1 2 Dissection of tubule segments is laborious and yields segments only 1-2 mm in length (42). It would require several hundred tubule segments (106 cells) to make many biochemical measurements (e.g. PGEZ and 3',5'-cyclic AMP radioimmunoassays) that could be made easily with one partially confluent 24-well culture dish. Renal medullary interstitial cells, for instance, have been used in culture to make a series of biochemical measurements pertaining to prostaglandin metabolism (1). As with tubule segments though, the data obtained with cultured cells can also be cell specific. Our interest in investigating prostaglandin metabolism in isolated collecting tubule cells was based on two observations. The first was that immunohistochemical localization of the prostaglandin-forming enyzme, cyclooxygenase, demonstrated that the collecting tubule cell was the only tubular cell type that stained for the enzyme (52). The second was Grantham's observation that PGE1 inhibits water resorption in kidney (61). Taken together, these data suggested that prostaglandins synthesized by the collecting tubule may be closely involved with kidney function. In order to test our hypothesis biochemically, we chose to isolate a population of collecting tubule cells from the rabbit kidney and to study prostaglandin metabolism in these cells. LITERATURE REVIEW Prostaglandin Metabolism. The pathways for the biosynthesis of various prostaglandins are illustrated in Fig. 1. The first step in this "arachidonate cascade" is the liberation of arachidonic acid from an esterified precursor in reSponse to an exogenous stimulus. The precursor is most commonly a phospholipid. Triglycerides, even in triglyceride-rich cells such as renal medullary interstitial cells, are apparently not a primary source of arachidonic acid (1). The release of arachidonic acid from phospholipids may occur via two different mechanisms. The first pathway involves the straightforward action of a phospholipase A2 on a 2-arachidonyl-phosphoglyceride to yield free arachidonic acid. Bills et al. have shown that platelets contain a phospholipase A2 activity which will preferentially catalyze the release of fatty acids from phOSphatidylcholine containing arachidonic acid at the 2-position (2,3). A second pathway for arachidonate release also exists in platelets. A cytosolic phospholipase C specific for phosphatidylinositol catalyzes the cleavage of phosphatidylinositol, yielding diglyceride and inositol phosphate (4,5,6) and a diglyceride lipase then catalyzes the release of arachidonic acid from the diglyceride. This phospholipase C- diglyceride lipase pathway provides for the specific release of arachidonic acid because approximately 90% of the phosphatidylinositol in platelets contains arachidonic acid esterified at the 2-position Figure 1. Overview of prostaglandin biosynthesis. wizcrcm El 3: 2...... >01 10 u- Dissociation of papillae by mincing and subsequent treatment with trypsin yielded mixtures of both large RPCT cells and contaminating small cell types. All small cells disappeared when the isotonic preparative media was diluted 1:2 vri th water and incubated for 3 min. Apparently, the small cells were preferentially lysed by this treatment while the number and morphology of the large cells was unaffected. The size and shape of the large cells (Fig. 3) were similar to collecting tubule cells observed in histological sections of renal Papillae (111). Two or three RPCT cells were often seen adhered along their long axis forming a slight arc. Strings of RPCT cells 10-15 cells T n length were also observed. Both the RPCT cells in histological sections and the isolated large CeTls stained positively for NADH-diaphorase activity. None of the Small cells isolated were prominently stained for this enzyme. These observations are consistent with the distribution of NADH-diaphorase in 27 28 Figure 3. Phase contrast photomicrograph of trypsin-isolated RPCT cells in suspension. Magnification, x250. 29 3D the renal medulla of the rabbit and serve to further confirm the identity of the isolated large cells as RPCT cells (109). Isolated RPCT cells were subjected to immunohistofluorescence staining with rabbit anti-cyclooxygenase serum and rabbit pre-immune serum, respectively (Fig. 4). The RPCT cells stained much more intensely with the immune serum in agreement with staining seen in tissue sections (110,112). Metabolic characterization of RPCT cells. The isolated RPCT cells excluded both trypan blue and erythrosine red dyes. A maximum of 106 RPCT cells were obtained per g of papillae by our method although the average yield was approximately half of that value. Figs. 5 and 6 illustrate the metabolism of [14C]-glucose and [3HJ-leucine, respectively, by isolated cells. RPCT cells both oxidized [14CJ-glucose to 14C02 and incorporated [3HJ-leucine into trichloroacetic acid-precipitable material in a time-dependent fashion for about 2 h. Cells preincubated for 30 min with NaN3 (0.02%) showed a 75-90% reduction in [14C]-glucose oxidation. Incorporation of [3H]-leucine into tricholoracetic acid-precipitable radioactivity was inhibited at least 80% when cells were preincubated with cycloheximide (1 ug/ml) for 30 min. In contrast, streptomycin, a prokaryotic protein synthesis inhibitor, at a concentration of 100 ug/ml did not block [3HJ-leucine incorporation. Isolated RPCT cells adhered to both glass and plastic petri dishes. Approximately 40% of the isolated cells became attached within a few hours when incubated in Dulbecco's modified Eagle media supplemented with 10% fetal calf serum, 2.9M glutamine and antibiotic-antimycotic. Figure 4. 31 Fluorescence photomicrographs of isolated RPCT cells treated with (A) anti- -c clooxygenase or (B) rabbit perimmune serumthen FIT labe ed goat anti- rabbit IgG. he rounding of the cells as compared to those in Fig. 3 was caused by chloroform/methanol fixation prior to staining. Magnification, x 250. 32 33 Figure 5. [14CJ-glucose oxidation by isolated RPCT cells. _ RPET cells were incubated for the indifiated times with [ CJ-glucose] and the formation of [ C]-C02 measured as described in the text. 34 I J M N 'l (91199.,01 x Z/cpt xde) wsnoavuw asoonio-o” TlME(hr) 35 Figure 6. [3HJ-leucine incorporation into trichloroacetic acid precipitable-products by isolated RPCT cells. RPCT cells were incubated for the indicated times with [ H]-leucine and the formation of trichloroacetic acid precipitable-radioactivity measured as described in the ext. 36 l N a Q Fl ($1163,01x5/,_01x undo) wsnoauaw amonzi'I-I-te TIME (hr) 37 The attached cells could be removed from these surfaces by treatment with trypsin (0.05% in PBS for 5 min) and subcultured on other petri dishes where they retained distinctive RPCT cell morphology and continued to exclude vital dyes. Subcultured RPCT cells when grown in a 10% C02 atmosphere still excluded vital dyes after 10-14 days during which time only the culture media was exchanged. No increase in cell number occurred during this time. Attempts were also made to isolate RPCT cells following tissue dissociation in which EDTA (0.001-0.01%) was substituted for trypsin. EDTA treatment of minced papillae did provide relatively high yields of RPCT cells (6-10 x 106 cells per g papillae). The EDTA-isolated cells also had the same capacity as trypsin-isolated cells both to oxidize glucose and to convert exogenous leucine into protein. However, the EDTA-isolated cells were permeable to vital dyes, were lysed rapidly by treatment with trypsin and were unable to repair their permeability defect during a 48 hr incubation in culture media. "Small" cell isolation. Relatively large numbers of small cells were obtained (5-8 x 106 cells per g of papillae) and were contaminated with only 1-5% CT cells. The small cells were Spherical and approximately 6p in diameter. None of these cells stained with vital dyes or for NADH-diaphorase activity. I made no attempt to distinguish between vascular endothelial cells, medullary interstitial cells and the epithelial cells of the thin loop of Henle. Characterization of arachidonic acid metabolites formed by RPCT cells. Fig. 7 shows a radiochromatogram of the products synthesized 38 from [3H]-arachidonic acid by CT cell homogenates. The major products synthesized by homogenates of both CT cells and small cells chromatographed with authentic PGDZ, PGEZ and PGFZQ standards as expected (49,113). The identity of radioactivity chromatographing with P602 and PGE2 in solvent system A was verified by chromatography in solvent systems B and C and by reduction with NaBH4 and chromatography in solvent system B. The two peaks of radioactivity seen in the PGan region of the thin-layer plate (Fig. 7) were seldom resolved clearly. However, as described below, further examination indicated that 50% of the radioactivity migrating with PGFZa was actually [3HJ-6-keto-PGF10 while only 30% was found to be due to [3HJ-PGF2a. Tsz was not synthesized in significant quantities ((3%). When cell homogenates were incubated with [3HJ-arachidonic acid in the presence of Fluriprofen (10'4M) only unreacted arachi- donate was recovered indicating that all products were derived from the activity of the prostaglandin-forming cyclooxygenase and not a lipoxy- genase. Thus, the small amount of radioactivity chromatographing in the monohydroxy acid region of the thin-layer plate (Fig. 7) is likely HHT and not a 20 carbon product (114). Data derived from incubations with five different RPCT homogenates are summarized in Table 1. Material with Rf = 0.17-0.19 in solvent system A was eluted from the silica gel G with chloroform/methanol (1/1, v/v). The presence of both radioactive 6-keto-PGF1Q and PGan was indicated by comparing the chromatographic properties of the eluted material with those of various other prostaglandin derivatives in solvent systems 8 and C (Table 2). Treatment of the radioactive material with ethereal diazomethane converted 40% of the radioactivity into a product which Figure 7. 39 A radiochromatogram of the products formed upon incubation of [ HJ-arachidonic acid (100 EU) with a RPCT cell homogenate. Products were separated by chromatography in solvent 5 stem A. Radioscanning was performed as described in t e text. AA-arachidon1c acid; RC-ricinoleic acid. 40 .5. 2.2.5 ”...: :2... 325m... mu 3 m _ . _ _ <<_ “3...» O N Q (v.01 x wdo 118) umuovonovu 49 41 .cmeLm552m mew mucme_cwaxm mumcmqmm m eo mu—zmmc wee .Azm No.0v neon uwcou_;umcmimzmw warm: umeLoecma mew: mcopuummmm gm-~ 8m.-o. ae~-N. eem-.e em.-m. meeeeeGOEOe ..ee =__e5m= a~-. Rm-m www-m. gee-om Rom-o meeeeemeEe; ..ee he umxe Nome meme e.eoe-eeex-e eweoe eee___eee .eeee e.eeem seed emuwpomH meowpm_:qoa _Pmu >3 vwo< owcouwzomc< Eocd custom muozuoce cwccmchumoca .H mpnmp 42 .mEmumzm ucm>Pom pcmscmmnsm we» a. amdwe Lo efidoaiopmxim saw: mgamcmoquocsu sows: < Empmxm ucm>Pom :_ mH.o-~H.o u we ;u_z xuw>wpumovumc mo mmmpcmucma we“ wumuwucw mmmwzucmcma cw mmmmucmucmem Amcmpmm Fxcumev Agemvem.o .m.o .e.o Aeoev.e.o me.o-oe.o u eeemxm eee>.em .eeeeeee exmezv .wmmvwm.o .m.o mm.o .$.mvwe.o ee.o-me.o m Sachem bee>_em .gmmvm..o mm.o em.o Asmevm~.o e~.o-mm.o u seemxm uee>_em Ammmvmm.o ow.o we.o Axemvwe.o ee.o-me.o m seemam eee>_em .Roo.v...o .e.o wN.o .eoo.vmw.o m..o-e..o q Seemxm peas—em memeue New. Nmoe me.eoe-x-e we._eeepez .mee.e> eev eHeoe-eeex-e .0 ee_ee~.eeeeeeeeo .N epeep 43 also chromatographed with the methyl ester prepared from 6-keto-PGF1G. NaBH4 treatment of the radioactive metabolite and authentic 6-keto-PGF1G failed to alter the chromatographic properties of either compound in agreement with the finding of Pace-Asciak (115), although PGDZ and PGE2 were reduced to PGF isomers by NaBH4 (>90%) under these conditions. We performed two control experiments which verified that 6-keto-PGF1a formation was actually being catalyzed by the biosynthetic machinery originating from the intact RPCT cells and not by enzymes derived from cellular debris which might possibly have remained after hypotonic lysis of small papillary cells. In the first experiment, we examined the supernatant obtained from the final wash of RPCT cells, but found no prostaglandin biosynthetic activity. In the second experiment, we compared 6-keto-PGFla synthesis by homogenates of two different preparations of RPCT cells: one in which trypsin (0.5 mg/ml) was included and the other in which trypsin was excluded during the hypotonic lysis step. We reasoned that trypsin would inactivate any prostaglandin biosynthetic enzymes not sequestered within the intact plasma membrane. We found no differences in the amounts of the different radioactive products formed per cell by homogenates of RPCT cells which had been isolated by each of these two procedures. Fig. 8 shows a time course for the production of various prostaglandin derivatives when isolated RPCT cell homogenates were incubated with 2.5 EM_arachidonic acid. PGEZ and 6-keto-PGF1a were synthesized at initial rates of 150 pmol/min/IO6 cells. The reactions were complete within 10 min and prior to the complete Figure 8. 44 Time course for the biosynthesis of various prostaglandin pr ducts by a RPCT cell homogenate incubated with [ HJ—arachidonic acid (2.5 uM). Reactions were performed for the indicated-times and the products analyzed as described in the text. .5522: m e - — it... -~ (IMMUNOILVWHOi lOnGOUd . \. -2: 1 OGN 1 00m 46 utilization of substrate, apparently reflecting the self-catalyzed destruction of the cyclooxygenase (116). Fig. 9 compares the amounts of different prostaglandins synthesized at different initial concentrations of arachidonate. At concentrations of arachidonic acid of less than 2 EM, 6-keto-PGF1a was the major product formed while at higher arachidonate concentrations, PGE2 was the major product. Vane and coworkers (117) and Sun et al. (118) have made similar observations of the effect of substrate concentrations on the prostaglandin product distribution in other tissues. Apparently, the Km of the PGIZ synthase is much lower than that of the isomerase catalyzing PGEZ formation. We suSpect that the fall in overall 6-keto-PGF1Q production noted at high concentrations of arachidonic acid (e.g. 100 EU) may have resulted from the presence of small but inhibitory levels of hydroperoxide contaminants (119) in the substrate solution. Although PGIZ is known to be formed by the renal cortex (120,121), apparently by renal vascular endothelial cells (122), the renal medulla has previously been reported to form only the classical prostaglandin derivatives (1,49,113,123,124). Thus, it was somewhat surprising to find that isolated collecting tubule cells as well as populations enriched in medullary interstitial cells have the capacity to produce significant quantities of PGIZ. Since this work was reported, Dunn (124) and Silberbauer (125) have also reported that PGIZ can be produced by the renal medulla. 47 Figure 9. Prostaglandin biosynthesis by RPCT cell homogenates at different initial concentrations of [3HJ-arachidonic acid. Incubations were performed for 15 min and the radioactive products measured as described in the text. 48 whoancmm z_nz<._6 10%) of nonpolygonal fibroblast-like cells had occurred; and second, unlike MDCK cells, RPCT cells do not fonn a stable confluent monolayer, but instead begin to detach and die upon reaching confluency (see below, Figure 15). 63 Figure 10. Phase photomigraph of RPCT cells grown eight days in monolayer culture. Magnification, x 250. Nucleus (N). 64 65 Figure 11. Electron micrographs of (A) intact collecting tubule from rabbit renal papillae (magnification, X4,500), (B) isolated RPCT cells grown in Millipore filters (magnification, X12,250) and (C) junctional complex of RPCT cells grown on Millipore filter (magnification X20,000). Nucleus (N), mitochondria (M), basement membrane (BM), oil dr0plets (0), rough endOplasmic reticulum (RER), basolateral surface (8), apical surface (AS), Millipore filter (MF), desmosome (D), tight junction (TJ), and gap junction (Gd). 66 \ 67 68 Figure 12. Hemicysts present in culture of RPCT cells grown to confluence without changing the growth medium. Magnification, X80. 69 70 The RPCT cells gave intense and uniformly positive staining for both NADH diaphorase and a-glycerophosphate dehydrogenase activities but did not stain for succinate dehydrogenase activity. This pattern of staining distinguishes collecting tubules from other epithelial cells of the papillae and inner medulla (133). Biochemical Characteristics of RPCT Cells. We had previously identified isolated RPCT cells in suspension as being derived from the collecting tubules on the basis of tissue source, cell diameter and immunohistochemical pr0perties. To determine if monolayer cultures of RPCT cells retained differentiated biochemical characteristics expected for collecting tubules, we measured the ability of one day old RPCT cells to synthesize 3',5'-cyclic AMP in response to a variety of effectors (Table 4). Agents capable of increasing 3',5'-cAMP levels at relatively high concentrations (:10'7 M) were AVP, dD-AVP, lysine vaSOpressin, cholera toxin, oxytocin, glucagon and, unexpectedly, parathyroid hormone; in contrast, adrenergic agents, calcitonin and histamine were ineffective. Fig. 13 compares the concentration dependence of the AVP-, parathyroid hormone- and oxytocin-induced increases in extracellular 3',5'-cyclic AMP. As anticipated, the most active agent on a molar basis was AVP. The concentration dependence observed with AVP is similar to that seen with intact tubules dissected from the outer medulla (134). Fig. 14 shows the time course of the AVP-induced appearance of both intracellular and extracellular 3',5'-cyclic AMP. There was an approximately linear increase in extracellular 3',5'-cyclic AMP during the 60 min incubation period whereas intracellular 3',5'-cyclic AMP 71 .ucmEymmcumea Levee new meowmn mEmm we» we: mcouumwem cmcym ow mmcoammc mzu ”Acmpzppmumcuxmv cowumELow az< uwpoxo1.m.m cw mmmmcocw upov N miemmamo A: ~1oH .Nmua gov cwcwxxvmcn .pcmEpmmcpmcg mesa mo mucmmnw we“ cw newcwamm :5 .o zuwz L; H Low mFPmo Hume mo unusummcumca mcwzo_pom vwcweLmumu we: :wcwxxumcn cu mmco mm; web a 1. .pxmu we» cw umawcummu we cmxmmmm ez< uw—u»01.m.m ecu A: eiofiv x H we mucmmmcq use cw a m an ewe on Low emacoecwa mew: mcoaomwwm ;u_3 meowumnaucm Amo.ov m mmzpm> _ocucoo soc» m cmso pcmuwewcmwm m mumuPucw hev xmwemamm cm sue: cmxcme mmosu ”sum w memes pcmmwcamc mm=Pm> w 1. «N.m.hmm. 5-0. cameo: m 1. tm.m~w~m. Rio. cwmmmcgome> mcwm _ o.~m~. .m.e.wm~. .-o. e.eee»xe 1. .e...nwm. .-o. ceases; e.ec»eeeeee 1. o.mA~e Niofi HH :wmcmuowmcm 1. ~.ehmm m-o. ee.Eeem.e 1. ..ehum ~-o. e.eee.e_ee I. m.mmwo m1o~ Focmcmuocaom? 1. N...wwm m-o. ee.eeeee_ee e.omm m.~.wme m-o. ee.eeeee.eeeee m.~he. w..~.wmo. .-o. e.xee acepeee a..em.m .e.e.mme. “-0. e><-ae .m.mn.e w~.e.moow ~-o. a>< m..w~. e..awmm e-o. e.e.x»eace m.owm ..ehwe e1o. ..eepeeev xzm. e.onm m.ewm~ --- eeez Acwmuoca 1. ._mu m:\mm—oEev Acwwuocn P—mu m:\mm—OEWV sz ez< e..e»e-.m..m axe e._e»e-.m.m ee.eaeceeeeeu Educate“ Lapsppmomcucfim Lw—s—qumcuxmm mcouomwmm _mcoELo; op mmcoammc cw m_Pmo poem an ez< ow_oxot.m..m eo co_uoeLom .e m4m mew: vmcwmuao mm=10> .co11001mu 10 011511 as» 301mg mo: me11 .umccov m:_c:01mmumogg 1010» we mommucmoema «10 mwmmgpcmema c1 mem251: 11oeucoo mg“ 5011 “cocmmw10 .1m023: 010:3 ummgomc as» ou nmvcaoe .axmu exp :1 000110000 m0 10mmmoczes1owv01 x0 umumuwucmau mcwucm_m01moga ucm :15 cm 10» mgouumvww mo mcowumcucmocoo umwwowucw mg» :11: umumnzocp mew; m—qu human 00 101 0 1010101 1011 01 1011 00 1-01 11 01000101000 1001 101-01 111111 1010-00 1011-101 1.01 101111 001 100 0 101 0 1011 m1 111v 00 0-01 0011100010010: 10 101 0 1111-01 1011 N1 1011 10 0-01 00111000100 00 101 0 111 0 1111 01 1010 10 1-01 1><-00 11 101 0 10V 0 1011 11 1010 00 1-01 1>< -001 101-11 111-11 1011-11 1111-101 1-01 0111110111 01 101 0 1mv 0 1010 11 1010 m0 --- 0002 10101 e1101-0101-0 N001 e1101 1100 1:1 covumgucmucou Louummwm 1:15 om\:_wuoga 11mo m:\mm_oE+v mumoga m>wuo0mcocses10 110101 .mgouuwwww _acoELo; o» mmcoamwe :1 m110o 101m an umEco1 mcwvcmpmwpmoca mo cowum~wgwuumgmzu .w m1m<\ . 00m (uleimd "as brl/saiouu) dWV3 WTIBOVHLNI 110 Figure 22. Effect of AVP (10-7 M), Poi-:2 (10-5 M), and bradykinin (10"7 M) on intracellular cAMP synthesis by RPCT cells. Incubations were performed for 10 min at 37°C and cAMP assayed as described in the text. 'omed »d in 111 lJ alt- Cl> I l l p, 8 (almond llao btvulw ovsalouu) dwva uv1n'l‘laovaml NI slow-lo M09) )3 MN >18+dAV dAV >18 calm) +dAV 339a + dAv dAV 339d “ICELLNOO DISCUSSION A model illustrating the interrelationships of prostaglandins, bradykinin, cAMP and AVP in RPCT cells is illustrated in Fig. 23. Bradykinin, but not AVP, caused iPGEZ biosynthesis by RPCT cells. Receptors for PGEZ on RPCT cells are envisioned to be present both exogenously and on the serosal surface of RPCT cells. Stokes observed that only PGEZ added to the serosal surface of cortical collecting tubules caused natriuresis; lumenal PGEZ had no effect (37). In addition, our data suggests that PGEZ synthesized due to bradykinin exits the cell and acts exogenously. While no known functions of prostaglandins synthesized due to bradykinin currently exist in RPCT cells, two potential functions are to mediate bradykinin—induced natriuresis and diuresis. The reported natriuretic effect of bradykinin in the kidney is probably due to an effect on renal hemodynamics and an effect on tubular sodium transport (37,56,104). Stokes and Kokko have shown that exogenous PGEZ inhibits sodium backflux, but not sodium efflux in the perfused rabbit cortical collecting tubule (37). Bradykinin-stimulated PGE2 formation may cause a similar effect in the papillary collecting tubule. The means by which PGEZ causes natriuresis in the cortical collecting tubule, however, is unknown. cAMP synthesized due to PGE2 by RPCT cells may be involved in this process. PGEZ caused 112 113 Figure 23. Model illustrating the effects of prostaglandins, bradykinin (BK), cAMP and AVP in RPCT cells. 114 URINE BLOOD \ COLLECTING weuua CELL GOMPARTMENT all ANTIDIURESIS <——-— cAW< COMPARTMENT 1"2 Pol-i2 <——zo:4 <———— BK PGEZ % 9PGEz NATRlLRESlS 7