Literature
首页医源资料库在线期刊美国生理学杂志2007年第290卷第1期

Renal prostaglandin E 2 receptor (EP) expression profile is altered in streptozotocin and B6-Ins2 Akita type I diabetic mice

来源:《美国生理学杂志》
摘要:【摘要】ThehomeostaticfunctionofprostaglandinE2(PGE2)isdependentonabalanceofEPreceptor-mediatedevents。AlthoughPGE2excretioniselevatedindiabetes,theexpressionofspecificEPreceptorsubtypeshasnotbeenstudiedinthediabetickidney。Therefore,thepurposeof......

点击显示 收起

【摘要】  The homeostatic function of prostaglandin E 2 (PGE 2 ) is dependent on a balance of EP receptor-mediated events. A disruption in this balance may contribute to the progression of renal injury. Although PGE 2 excretion is elevated in diabetes, the expression of specific EP receptor subtypes has not been studied in the diabetic kidney. Therefore, the purpose of this study was to characterize the expression profile of four EP receptor subtypes (EP 1-4 ) in 16-wk streptozotocin (STZ) and B6-Ins2 Akita type I diabetic mice. In diabetic mice, the ratio of kidney weight to body weight was increased twofold compared with controls, blood glucose was elevated, but urine albumin was only increased in B6-Ins2 Akita mice. The excretion of PGE 2 and its metabolite was augmented two- to fourfold as determined by enzyme immunoassay. Accordingly, renal cyclooxygenases were also increased in diabetic mice, with isoform-specific and regional differences in each model. Finally, there was altered EP 1-4 receptor expression in diabetic kidneys, with significant differences between STZ and B6-Ins2 Akita mice (fold-control). In STZ mice, cortical EP 1 increased by 1.6, EP 3 increased by 2.3, and EP 4 decreased by 0.63; yet in B6-Ins2 Akita mice, cortical EP 1 increased by 2.4, but there was a general decrease in the remaining subtypes. Similarly, in the STZ medulla EP 3 increased by 3.6, but both EP 1 and EP 3 increased by 5.5 and 1.95, respectively, in B6-Ins2 Akita mice. Therefore, knowing the pattern of change in relative EP receptor expression in the kidney could be useful in identifying specific EP targets for the prevention of various components of diabetic kidney disease.

【关键词】  cyclooxygenase realtime reverse transcriptase polymerasechain reaction renal EP receptors streptozotocindiabetic mice


DIABETIC NEPHROPATHY (DN) is a leading cause of chronic kidney disease resulting in end-stage renal disease (ESRD). The renal changes associated with DN consist of glomerular, vascular, and tubular events that result in altered renal hemodynamics, growth responses, matrix accumulation, and tubular transport processes. To date, the most common therapy for management of the disease, aimed at slowing the development of diabetic complications and ESRD, consists of angiotensin-converting enzyme (ACE) inhibitors and angiotensin II receptor antagonists, which reduce blood pressure and delay the progressive loss of renal function ( 4 ). Despite this intervention, diabetes is threatening to reach epidemic proportions, and current modes of therapy are ineffective in preventing this course. While glucose is the main determinant of the changes associated with the pathogenesis of DN, the induction of numerous downstream effectors results in a vicious cycle of events, all uniting to perpetuate the extent of renal injury. It is therefore imperative to clarify the specific disturbances underlying the injury to the kidney, to find alternative targets for more specific therapeutic intervention.


Prostaglandin E 2 (PGE 2 ) is by far the most predominant arachidonic acid metabolite produced in the kidney ( 6 ), particularly in the glomerular regions and the inner medulla. Its synthesis is dependent on the activity of two cyclooxygenase isoforms (COX-1 and COX-2) that are inhibited by nonsteroidal anti-inflammatory drugs (NSAIDS). PGE 2 is important in various aspects of renal physiology, hemodynamics, renin release, and tubular transport processes. Its biological effects are mediated by four distinct G protein-coupled receptors, EP 1-4 ( 32 ): the EP 1 receptor stimulates intracellular calcium and activates protein kinase C via the G q protein family; the EP 2 receptor stimulates adenylate cyclase via G s protein, and to date it remains unclear whether this receptor is expressed in renal tubules and what role it plays in renal physiology; the EP 3 receptor inhibits adenylate cyclase via pertussis toxin-sensitive G i protein, and by this mechanism PGE 2 antagonizes arginine vasopressin (AVP) responses; and the EP 4 receptor stimulates adenylate cyclase via G s protein. The existence of various PGE 2 /EP receptor pathways in renal cells provides an interesting example of homeostatic processes, with redundant functions as well as opposing actions. This is exemplified by the fact that no obvious renal pathology or disturbance in function has been observed in any of the EP knockout mice ( 14, 23, 36, 41 ).


The past decade has led to many advances in the study of the effects of PGE 2 through EP receptors in the kidney, but little is known about the pathophysiological role of each EP subtype in renal disorders and what effect a change in the relative responses in different EP pathways would have in a disease setting. While alterations in PG levels have been implicated in the pathogenesis of diabetic nephropathy ( 20, 27 ), resulting in hemodynamic changes and structural variations ( 13 ), the individual contribution of COX-1 and COX-2, as well as specific PG receptor pathways, to distinct aspects (altered H 2 O and Na + balance, growth responses, matrix expansion, etc.) of diabetic kidney disease is lacking.


Therefore, we hypothesize that 1 ) an imbalance in the PGE 2 /EP system may be evident in diabetic kidneys and 2 ) this may contribute to renal injury in diabetes. Thus the purpose of this work addresses the first part of the hypothesis and examines the expression of four EP receptor subtypes in the different regions of the kidney in 16-wk streptozotocin (STZ)-diabetic and B6-Ins2 Akita mice, two mouse models of type 1 diabetes. Identifying the relative expression of EP receptors should shed light on the usefulness of specifically targeting EP receptors to rectify an imbalance that may influence the evolution of diabetic nephropathy.


MATERIALS AND METHODS


Diabetic mouse models. Two separate studies were carried out using chemically induced-diabetic as well as genetically diabetic mice.


STZ-induced type I diabetes is the most widespread method of inducing type I diabetes in rodents by destroying the pancreatic -cells ( 40 ). We administered 65 mg/kg of STZ/Na-citrate buffer (Sigma) three times daily intramuscularly, which induces diabetes in C57BL/6 mice within 1 wk after the infusion. Vehicle-treated controls were used for comparison. The major drawback with this model is the lack of similarity to human diabetes ( 15 ), with very mild changes reported in the kidney but also the known cellular toxicity of STZ ( 5 ). For this reason, a spontaneous type I diabetes model, the B6-Ins2 Akita mouse, was also studied.


The B6-Ins2 Akita model of spontaneous type I diabetes is a relatively new model of nonobese insulin-dependent diabetes, characterized by early-age onset and autosomal dominant inheritance. B6-Ins2 Akita mice were purchased from Jackson Laboratories and were derived from C57BL/6 mice, allowing for better comparisons in our studies, given the recognized strain differences in development of diabetic kidney changes ( 15 ). A mutation in the insulin 2-gene (Cys96Tyr) is responsible for their phenotype, showing progressive diabetes characterized by hyperglycemia ( 3 ) and notable pancreatic -cell dysfunction. In a recent review by Breyer et al. ( 7 ) for the Animal Models of Diabetes Complications Consortium (AMDCC), the B6-Ins2 Akita model is reported as the optimal substitute for the well-established STZ diabetes model, to avoid issues of nonspecific cell toxicity and because it is commercially available through Jackson Laboratories. The homozygous mice die within 2 mo of age, but the male heterozygotes display diabetic symptoms including hyperglycemia, hypoinsulinemia, polydipsia, and polyuria by 3-4 wk of age. The female mice are less susceptible to development of diabetic features, with milder hyperglycemia ( 15 ). Therefore, the male heterozygote B6-Ins2 Akita mice were used for our studies at 16 wk of age, along with their wild-type littermates as the control.


Standard protocols were utilized for comparative studies to characterize the diabetic state of the mice, including kidney and body weight measurements, weekly determination of blood glucose levels using a blood glucose meter (Ascensia Elite, Bayer), systolic blood pressure measured by tail-cuff plethysmography (BP-2000, Visitech Systems), as well as urine albumin levels determined by enzyme-linked immunosorbent assay (Albuwell M competitive ELISA, Cedarlane Labs) and normalized by urine creatinine determination (colorimetric assay, Oxford Biomedical Research).


Western blotting. Protein lysates from cortex and medullary regions were prepared by homogenizing the tissue in RIPA buffer containing 1% NP-40, 1% sodium deoxycholate, 0.1% SDS (wt/vol), 4.5 mM NaCl, 2.5 mM Tris (pH 7.4), 8 µM EDTA, 0.2 mM sodium phosphate (pH 7.2), and freshly added 0.5 mM PMSF, 1:100 protease inhibitor cocktail (Sigma), 1 mM sodium pyrophosphate, 10 mM sodium fluoride, and 100 µM sodium orthovanadate. Twenty-five micrograms of each sample were resolved by SDS-PAGE on a polyacrylamide gel and transferred to a nitrocellulose membrane. After blocking for 2 h in 10% milk/TBS-T, the membranes were incubated overnight with either anti-COX-1 (Santa Cruz) or anti-COX-2 (Cayman) polyclonal antibodies. Following incubation with a horseradish peroxidase-conjugated anti-rabbit IgG secondary antibody, enhanced chemiluminescence was used to visualize the signals. A single band of 65 or 72 kDa was obtained for COX-1 and COX-2, respectively. The samples were then normalized with detection of -actin, and a densitometric analysis was performed using Kodak Digital Science 1D Image Analysis software (Eastman Kodak).


Enzyme immunoassays. Urine was collected from wild-type and diabetic mice at 16 wk of diabetes. The amount of PGE 2 and its metabolite 11-deoxy-13,14-dihydro-15-keto-11, 16 -cyclo-PGE 2 (PGEM) was determined by competitive enzyme immunoassays (Cayman Chemical) following the manufacturer?s instructions. Briefly, the assay is based on a competitive binding of PGE 2 or PGEM and their respective acetylcholinesterase conjugate (tracer) for a limited amount of monoclonal antibody. Since the tracer concentration is held constant, the amount of tracer bound to the antibody will be inversely proportional to the amount of PG in the sample. Detection is based on a colorimetric reaction using Ellman?s Reagent, which contains the substrate to acetylcholinesterase. The intensity is then determined by spectrophotometry. A colorimetric assay of urinary creatinine was performed for each sample to normalize the amount of PGE 2 or PGEM.


Real-time RT-PCR. Total RNA was isolated using TRIzol (GIBCO) from different preparations of cortex and medulla from wild-type and diabetic mice at 16 wk of diabetes. The relative quantity of each target nucleic acid in different samples was determined by analyzing the cycle-to-cycle change in fluorescence signal as a result of amplification during a PCR. To quantify the amount of RNA in each sample, a relative standard curve was prepared by diluting a stock of control RNA. Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) mRNA is detected as an internal control to standardize the amount of sample RNA added to a reaction. RT-PCR was performed using a Taq Man PCR Core Reagents Kit and GAPDH Control Reagents Kit providing a VIC-labeled GAPDH probe. The following parameters were employed: 48°C for 30 min then 95°C for 10 min, followed by 40 cycles of 95°C for 30 s and 60°C for 1 min. Primers and probes were selected and obtained from the Custom Oligonucleotide Synthesis Service of Applied Bio-Systems. The upstream and downstream primers as well as fluorescent probes for EP receptors and GAPDH are listed in Table 1. Computer analysis of data was performed using the ABI Prism 7000 sequence detection system, and data are expressed as the ratio of EP mRNA to GAPDH mRNA.


Table 1. Probes and primers used for real-time RT-PCR


Statistics. GraphPad Prism software for Windows, version 4.02 (May 17, 2004), was used to analyze data. Results are expressed as means ± SE. An unpaired t -test was used to assess the statistical significance between data points, and a P value <0.05 was considered statistically significant.


RESULTS


Comparison of diabetic characteristics in 16 wk STZ and B6-Ins2 Akita mice. At 16 wk of age, STZ and B6-Ins2 Akita mice are hyperglycemic (see Tables 2 and 3 ), with significantly elevated blood glucose levels in the range of 25-30 mM. It has previously been reported that hyperglycemia is evident in both models at 4 wk of age ( 3, 28 ). Since renal hypertrophy is an important feature of diabetic kidneys, we observed an increase in kidney weight-to-body weight ratios in both groups of diabetic mice as expected. Urine albumin levels were elevated 3.8-fold in 16-wk B6-Ins2 Akita mice ( Table 3 ). However, our work shows no differences in urine albumin in STZ mice (see Table 2 ), similar to recent reports by Gurley et al. ( 15 ). To the best of our knowledge, there have been no reports of changes in blood pressure in the STZ-diabetic mice, although we did not expect hypertension in these mice as early as 16 wk of age ( Table 2 ); as shown in Table 3, we also did not detect any differences in blood pressures in B6-Ins2 Akita mice.


Table 2. Summary of characteristics of 16-wk STZ-diabetic mice


Table 3. Summary of characteristics of 16-wk B6-Ins2 Akita mice


Renal COX-1 and COX-2 and urinary PGE 2 are increased in diabetic mice. Our group and others have previously demonstrated that COX enzymes are elevated in diabetic kidneys ( 27, 33 - 35 ). As shown in Figs. 1 and 2, Western blot analysis confirms that COX isoforms are altered in both mouse models of type I diabetes. However, the following notable differences were observed. 1 ) In STZ mouse cortex, both COX-1 and COX-2 are increased 3.6- and 3-fold, respectively, but in B6-Ins2 Akita mice, a 2-fold increase is only seen for COX-1. 2 ) In the medulla, COX-1 is unchanged in the STZ mice and decreases to 0.14-fold of control in B6-Ins2 Akita mice; however, COX-2 increases 2- and 2.75-fold in STZ and B6-Ins2 Akita mice, respectively. Consistent with increased renal COX expression, PGE 2 excretion is elevated in both STZ and B6-Ins2 Akita mouse urine. We show a 1.9-fold increase in PGE 2 in STZ mice ( Fig. 3 A ), and both PGE 2 and PGEM are elevated 3.2- and 4.3-fold, respectively in B6-Ins2 Akita mice ( Fig. 3, A and B ).


Fig. 1. Renal cyclooxygenase (COX) levels are altered in 16-wk streptozotocin (STZ)-induced diabetic mice. Protein was isolated from the cortex ( A ) and medulla ( B ) of control and 16-wk STZ-diabetic mice. COX-1 and -2 levels were determined by Western blotting. Densitometric analysis of COX is shown. Detection of -actin was done to normalize samples. The ratio of COX to -actin is presented as means ± SE (arbitrary units); n = 3-6. * P < 0.05.


Fig. 2. Renal COX levels are altered in 16-wk B6-Ins2 Akita mice. Protein was isolated from the cortex ( A ) and medulla ( B ) of control and 16-wk B6-Ins2 Akita diabetic mice. COX-1 and -2 levels were determined by Western blotting. Densitometric analysis of COX is shown. Detection of -actin was done to normalize samples. The ratio of COX to -actin is presented as means ± SE (arbitrary units); n = 3-6. * P < 0.05.


Fig. 3. Urinary PGE 2 ( A ) and PGEM ( B ) levels are increased in 16-wk STZ- and B6-Ins2 Akita -diabetic mice. Urine samples were obtained from mice after 16 wk of diabetes. PGE 2 and PGEM levels were assayed by competitive enzyme immunoassay. Urine creatinine levels were assayed to normalize the samples. The ratio of prostaglandin to creatinine is presented as means ± SE; n = 6. * P < 0.05. ** P < 0.001.


An imbalance between EP 2 /EP 4 and EP 1 /EP 3 receptors is seen in diabetes. In this study, real-time RT-PCR analysis was utilized to measure the relative expression of EP receptor subtype mRNA in cortical and medullary regions of the kidney of STZ and B6-Ins2 Akita mice. Table 4 lists the ratio of EP mRNA to GAPDH mRNA for each receptor subtype, in the cortex and medulla of each diabetic model and their respective controls. As shown in Fig. 4 A, there is an increase in cortical EP 1 and EP 3 receptors in STZ mice by 1.6- and 2.3-fold, respectively. Conversely, EP 4 receptors are diminished 0.63-fold. On the other hand, medullary EP 1 receptors are diminished in STZ mice, while EP 3 receptors are increased 3.6-fold ( Fig. 4 B ). Similarly, in B6-Ins2 Akita mice cortical EP 1 receptors are increased 2.4-fold ( Fig. 5 A ); however, the remaining EP receptor subtypes are significantly diminished 0.26-, 0.38-, and 0.47-fold for EP 2, EP 3, and EP 4, respectively. In the medullary region of B6-Ins2 Akita mice ( Fig. 5 B ), both EP 1 and EP 3 are increased 5.5- and 1.95-fold, respectively, but EP 2 and EP 4 are unchanged.


Table 4. Summary of real-time RT-PCR analysis of EP receptor expression in 16-wk control and diabetic mice


Fig. 4. Renal EP mRNA expression is altered in 16-wk STZ-diabetic mice. Real-time RT-PCR detection of EP 1-4 receptor mRNA was performed on total RNA samples isolated from the cortex ( A ) and medulla ( B ) of control and 16-wk STZ-diabetic mice using specific probes and primers for each EP receptor subtype. GAPDH mRNA was detected as an internal control. The ratio of EP mRNA to GAPDH mRNA is presented as means ± SE ( n = 3-5) expressed as fold-control (control = 1). * P < 0.05.


Fig. 5. Renal EP mRNA expression is altered in 16-wk B6-Ins2 Akita mice. Real-time RT-PCR detection of EP 1-4 receptor mRNA was performed on total RNA samples isolated from the cortex ( A ) and medulla ( B ) of control and 16-wk B6-Ins2 Akita diabetic mice using specific probes and primers for each EP receptor subtype. GAPDH mRNA was detected as an internal control. The ratio of EP mRNA to GAPDH mRNA is presented as means ± SE ( n = 3-5) expressed as fold-control (control = 1). * P < 0.05.


DISCUSSION


Diabetes is a leading cause of chronic kidney disease ( 46 ). COX-derived PGs have been directly or indirectly implicated in diabetic kidney disease, initiating diabetic features or antagonizing other pathophysiological agents such as the renin-angiotensin system. EP 1 receptor antagonists ( 29 ), prostacyclin receptor (IP) agonists ( 25, 37, 38 ), and thromboxane A 2 synthase inhibitors ( 43 ) have proven to be beneficial, but the underlying mechanisms of PG involvement remain uncertain.


In the current study, we examined the levels of renal COX, urinary PGE 2, and the expression of EP receptor subtypes in two recognized mouse models of type I diabetes, in the early stages of diabetic nephropathy before major changes in glomerular filtration rate (GFR). In both diabetic models, we observed an increase in the excretion of PGE 2 as well as its metabolite in the B6-Ins2 Akita mice. However, notable differences were observed in both models with respect to the expression of COX isoforms. While both COX-1 and COX-2 are increased in the cortex of STZ mice, only COX-1 increases in the cortex of B6-Ins2 Akita mice. On the other hand, in the medulla, COX-1 is unchanged in the STZ mice and decreases in B6-Ins2 Akita mice, but COX-2 increases in both models. Previously, we reported increased medullary levels of both COX isoforms in STZ-diabetic rats ( 35 ) as well as increased COX-2 levels in response to high glucose in rat mesangial cells ( 33, 34 ) and cultured inner medullary cells ( 35 ). Interestingly, in 4-wk STZ rats, Komers et al. ( 27 ) reported increased COX-2 and not COX-1 in the renal cortex and in a recent study found that only COX-2 is elevated in the obese Zucker fatty rat model of type II diabetes ( 26 ). The importance of elevated renal COX in diabetes is highlighted by the use of NSAIDS to inhibit COX activity and PG synthesis. Only a few studies have reported the effects of NSAIDS on renal function in diabetes, but these are controversial due to differences in the type of NSAID studied and the duration of treatment ( 1, 10, 27, 31 ). Taken together, they indicate that COX-derived PGs contribute to diabetic alterations in the kidney and that the increase in COX-2 may play a significant role in the pathogenesis of diabetic kidney disease.


Another major finding in our current study is that there is altered EP receptor expression in diabetic mice kidneys, such that EP 1 and/or EP 3 receptors are increased in the renal cortex and/or medulla, but the EP 4 and/or EP 2 receptors are either decreased or unchanged. Of importance, however, are the obvious discrepancies in both diabetic models. For instance, in the renal cortex of STZ mice we showed increased EP 1 and EP 3 receptors and diminished EP 4 receptors, whereas in B6-Ins2 Akita mice, cortical EP 1 receptors are increased, and the remaining EP receptor subtypes are significantly diminished. Similarly, in the medulla, in STZ mice EP 1 receptors are diminished and EP 3 receptors are increased, but in B6-Ins2 Akita mice both EP 1 and EP 3 are increased. Whether these differences are attributable to actual diversity in the diabetic state of the mice, or are related to the use of STZ, remains to be determined. Nonetheless, a disturbance in EP-mediated responses could surely influence the course of diabetic kidney disease. Since we have shown that renal PGE 2 is elevated, this could have an impact on glomerular and collecting duct function, two major sites of renal PGE 2 synthesis and action. For instance, within the kidney, a majority of EP 1 mRNA is found in the collecting duct ( 8 ), and in the present study we show the largest increase in EP 1 receptor mRNA relative to other EP receptors. Defective PGE 2 /EP receptor signaling could interfere with the fine-tuning of salt and water transport, and these abnormalities could contribute to edema, hypertension, and vascular changes associated with diabetic nephropathy. To further support this idea, the significance of PGE 2 to the maintenance of salt and water homeostasis is clearly demonstrated by the undesirable renal effects such as sodium and potassium retention ( 39 ) associated with the use of NSAIDS, which inhibit the production of PGs. Therefore the increase in EP 1 receptors reported in this paper would surely contribute to renal salt and H 2 O alterations seen in diabetes. Future studies in our laboratory will target EP 1 receptors with pharmacological or molecular interventions at various stages of diabetic nephropathy to clarify the role of EP 1 in the development of diabetic complications.


In our study, we also observed an increase in EP 3 receptors, which could play a part in diabetic changes in the kidney. A similar deregulation of cortical EP 3 responses is proposed to play a role in the progression of kidney disease in rats with passive Heymann nephritis ( 45 ). Our group and others have demonstrated an important role for PGE 2 in the collecting duct to limit AVP responses ( 17 - 19 ); thus the elevated EP 3 receptor levels reported here could serve to oppose AVP-mediated H 2 O reabsorption and reduce volume expansion in diabetes ( 2 ).


The highest intrarenal expression of the EP 4 receptor is reported in the cortex ( 8, 21, 42 ). In contrast, in our study we show higher expression of EP 4 in the renal medulla. Furthermore, our work indicates that EP 4 receptor mRNA is either diminished or unchanged in diabetes, which could also affect the progression of diabetic change in the tubule and glomeruli. For example, it is clear that PGE 2 activates EP 4 receptors located on collecting duct principal cells to stimulate H 2 O reabsorption via aquaporin-2 ( 24, 44 ). Therefore, the significance of diminished EP 4 receptors in our study could be to prevent excessive H 2 O reabsorption in the diabetic collecting duct that would otherwise lead to volume expansion and further supports the diuretic role of PGE 2 in diabetes. Additionally, a defect in EP 4 receptors could influence glomerular function. For example, in podocytes EP 4 receptors are important in preventing the morphological changes required for podocytes to adapt to mechanical stretch in vitro, which could contribute to proteinuria in hypertensive patients, for example ( 30 ). Also, our group and others showed that in mesangial cells cAMP-stimulating PGs alter cell proliferation and matrix turnover ( 20, 33, 34 ), thus contributing to diabetic glomerular disease. Since EP 2 receptors are mainly found in the renal vasculature and interstitial cells, the reduction in cortical EP 2 receptors found in our study could influence vascular and interstitial cell function in diabetes.


It is noteworthy to recognize the discrepancies between the two diabetic models presented in this paper considering the known toxicity of STZ and that B6-Ins2 Akita mice develop more severe diabetic complications in the kidney ( 14 ). Interestingly, we did not detect any changes in blood pressure in both models, and a study by Kakoki et al. ( 22 ) indicates that blood pressure in 6-mo B6-Ins2 Akita mice is not significantly different from that in controls. In contrast, a recent study by Gurley et al. ( 15 ) reports a significant increase in systolic blood pressure in 16-wk B6-Ins2 Akita mice. The reason for this discrepancy is not clear at this time.


Also, the alterations in the EP receptor expression profile in these mice are noted before any major disturbances in renal function or structural changes due to diabetes, although the kidneys are enlarged in both models relative to body weight, and in the B6-Ins2 Akita mice we did observe an increase in urine albumin. Our work suggests that the defect in EP (2 +/or 4) receptors relative to EP (1 +/or 3) pathways reported here may be important in the initiation of diabetic change, before changes in GFR. It is also interesting that there is a similar disturbance in the ratio of (PGE 2 and PGI 2 ) to thromboxane A 2 in diabetes that has been implicated in the progressive loss of renal function and development of diabetic nephropathy ( 9, 11 - 13 ). Together, a defect in PGE 2 /EP (2 +/or 4) responses relative to PGE 2 /EP (1 +/or 3) responses will surely serve to perpetuate the diabetic complications in the kidney, but further staging of the changes in EP receptor expression as diabetic characteristics arise will be needed to confirm the role of each subtype in the progression of diabetic kidney disease.


In summary, our work shows that renal COX levels and urinary PGE 2 excretion are increased in both STZ-diabetic and B6-Ins2 Akita mice, two models of type I diabetes, before major diabetic changes in renal function. There is an altered expression profile of EP receptors throughout the diabetic kidney, mainly favoring EP 1/3 receptor-mediated responses, suggesting a role in the onset of diabetic change. Further studies will clarify the significance of these findings to disturbances in glomerular and collecting duct function and progression of diabetic nephropathy by staging the disturbances in EP receptor expression as diabetic features occur in B6-Ins2 Akita mouse kidneys. The use of EP receptor knockout technology as well as specific agonists and antagonists should facilitate these future endeavors. Once clarified, this could lead to the advent of better combination therapy to prevent the progression of the disease or reverse diabetic complications ( 16 - 19 ).


GRANTS


This research was supported by the Canadian Institute for Health Research and the Kidney Foundation of Canada.

【参考文献】
  Bakris Gl, Starke U, Heifets M, Polack D, Smith M, Leurgans S. Renal effects of oral prostaglandin supplementation after ibuprofen in diabetic subjects: a double-blind, placebo-controlled, multicenter trial. J Am Soc Nephrol 5: 1684-1688, 1995.

Bankir L, Bardoux P, Ahloulay M. Vasopressin and diabetes mellitus. Nephron 87: 8-18, 2001.

Barber AJ, Antonetti DA, Kern TS, Reiter CE, Soans RS, Krady JK, Levison SW, Gardner TW, Bronson SK. The Ins2Akita mouse as a model of early retinal complications in diabetes. Invest Ophthalmol Vis Sci 46: 2210-2218, 2005.

Barnett AH. Preventing renal complications in diabetic patients: the Diabetics Exposed to Telmisartan And enalaprIL (DETAIL) study. Acta Diabetol 42, Suppl 1: S42-S49, 2005.

Bolzan AD, Bianchi MS. Genotoxicity of streptozotocin. Mutat Res 512: 121-134, 2002.

Bonvalet JP, Pradelles P, Farman N. Segmental synthesis and actions of prostaglandins along the nephron. Am J Physiol Renal Fluid Electrolyte Physiol 253: F377-F387, 1987.

Breyer MD, Bottinger E, Brosius FC III, Coffman TM, Harris RC, Heilig CW, Sharma K, AMDCC. Mouse models of diabetic nephropathy. J Am Soc Nephrol 16: 27-45, 2005.

Breyer M, Breyer R. Prostaglandin E receptors and the kidney. Am J Physiol Renal Physiol 279: F12-F23, 2000.

Burns KD. Angiotensin II and its receptors in the diabetic kidney. Am J Kidney Dis 36: 449-467, 2000.

Cheng HF, Wang CJ, Moeckel GW, Zhang MZ, McKanna JA, Harris RC. Cyclooxygenase-2 inhibitor blocks expression of mediators of renal injury in a model of diabetes and hypertension. Kidney Int 62: 929-939, 2002.

Craven PA, Caines MA, DeRubertis FR. Sequential alterations in glomerular prostaglandin and thromboxane synthesis in diabetic rats: relationship to the hyperfiltration of early diabetes. Metabolism 36: 95-103, 1987.

Craven PA, Melhem MF, DeRubertis FR. Thromboxane in the pathogenesis of glomerular injury in diabetes. Kidney Int 42: 937-946, 1992.

DeRubertis FR, Craven PA. Eicosanoids in the pathogenesis of the functional and structural alterations of the kidney in diabetes. Am J Kidney Dis 22: 727-735, 1993.

Fleming EF, Athirakul K, Oliverio MI, Key M, Goulet J, Koller BH, Coffman TM. Urinary concentrating function in mice lacking EP 3 receptors for prostaglandin E 2. Am J Physiol Renal Physiol 275: F955-F961, 1998.

Gurley SB, Clare SE, Snow KP, Hu A, Meyer TW, Coffman TM. Impact of genetic background on nephropathy in diabetic mice. Am J Physiol Renal Physiol 290: F214-F222, 2006.

Haseyama T, Fujita T, Hirasawa F, Tsukada M. Complications of IgA nephropathy in a non-insulin-dependent diabetes model, the Akita mouse. Tohoku J Exp Med 198: 233-244, 2002.

Hébert RL, Breyer RM, Jacobson HR, Breyer MD. Functional and molecular aspects of prostaglandin E receptors in the cortical collecting duct. Can J Physiol Pharmacol 73: 172-179, 1995.

Hébert RL, Jacobson HR, Fredin D, Breyer MD. Evidence that separate PGE 2 receptors modulate water and sodium transport in rabbit cortical collecting duct. Am J Physiol Renal Fluid Electrolyte Physiol 265: F643-F650, 1993.

Hébert RL, Jacobson HR, Breyer MD. PGE 2 inhibits AVP-induced water flow in cortical collecting ducts by protein kinase C activation. Am J Physiol Renal Fluid Electrolyte Physiol 259: F318-F325, 1990.

Ishibashi R, Tanaka I, Kotani M, Muro S, Goto M, Sugawara A, Mukoyama M, Sugimoto Y, Ichikawa A, Narumiya S, Nakao K. Roles of prostaglandin E receptors in mesangial cells under high-glucose conditions. Kidney Int 56: 589-600, 1999.

Jensen B, Stubbe J, Hansen P, Andreasen D, Skøtt O. Localization of prostaglandin E 2 EP 2 and EP 4 receptors in the rat kidney. Am J Physiol Renal Physiol 280: F1001-F1009, 2001.

Kakoki M, Takahashi N, Jennette JC, Smithies O. Diabetic nephropathy is markedly enhanced in mice lacking the bradykinin B2 receptor. Proc Natl Acad Sci USA 101: 13302-13305, 2004.

Kennedy C, Zhang Y, Brandon S, Guan Y, Coffee K, Funk CD, Magnuson MA, Oates JA, Breyer MD, Breyer RM. Salt-sensitive hypertension and reduced fertility in mice lacking the EP 2 receptor. Nat Med 5: 217-220, 1999.

Knepper MA, Wade JB, Terris J, Ecelbarger CA, Marples D, Mandon B, Chou C, Kishore BK, Nielsen S. Renal aquaporins. Kidney Int 49: 1712-1717, 1996.

Koh E, Morimoto S, Jiang B, Inoue T, Nabata T, Kitano S, Yasuda O, Fukuo K, Ogihara T. Effects of beraprost sodium, a stable analogue of prostacyclin, on hyperplasia, hypertrophy and glycosaminoglycan synthesis of rat aortic smooth muscle cells. Artery 20: 242-252, 1999.

Komers R, Zdychova J, Cahova M, Kazdova L, Lindsley JN, Anderson S. Renal cyclooxygenase-2 in obese Zucker (fatty) rats. Kidney Int 67: 2151-2158, 2005.

Komers R, Lindsley JN, Oyama TT, Schutzer WE, Reed JF, Mader SL, Anderson S. Immunohistochemical and functional correlations of renal cyclooxygenase-2 in experimental diabetes. J Clin Invest 107: 889-898, 2001.

Leung JCK, Chan LYY, Tsang AWL, Tang SCW, Lai KN. Differential expression of aquaporins in the kidneys of streptozotocin-induced diabetic mice. Nephrology 10: 63-72, 2005.

Makino H, Tanaka I, Mukoyama M, Sugawara A, Mori K, Muro S, Suganami T, Yahata K, Ishibashi R, Ohuchida S, Maruyama T, Narumiya S, Nakao K. Prevention of diabetic nephropathy in rats by PGE receptor EP 1 -selective antagonist. J Am Soc Nephrol 13: 1757-1765, 2002.

Martineau LC, McVeigh LI, Jasmin BJ, Kennedy CR. p38 MAP kinase mediates mechanically induced COX-2 and PG EP 4 receptor expression in podocytes: implications for the actin cytoskeleton. Am J Physiol Renal Physiol 286: F693-F701, 2004.

Mathiesen ER, Hommel E, Olsen UB, Parving HH. Elevated urinary prostaglandin excretion and the effect of indomethacin on renal function in incipient diabetic nephropathy. Diabet Med 5: 145-149, 1988.

Narumiya S, Sugimoto Y, Ushikubi F. Prostanoid receptors: structures, properties and functions. Physiol Rev 79: 1193-1226, 1999.

Nasrallah R, Hébert RL. Reduced IP receptors in STZ-induced diabetic rat kidneys and high-glucose-treated mesangial cells. Am J Physiol Renal Physiol 287: F673-F681, 2004.

Nasrallah R, Landry A, Scholey JW, Hébert RL. Characterization of the PGI 2 /IP system in cultured rat mesangial cells. Prostaglandins Leukot Essent Fatty Acids 70: 455-464, 2004.

Nasrallah R, Landry A, Singh S, Sklepowicz M, Hébert RL. Increased expression of cyclooxygenase-1 and -2 in the diabetic rat renal medulla. Am J Physiol Renal Physiol 285: F1068-F1077, 2003.

Nguyen M, Camenisch T, Snouwaert JN, Hicks E, Coffman TM, Anderson PAW, Malouf NN, Koller BH. The prostaglandin receptor EP 4 triggers remodelling of the cardiovascular system at birth. Nature 390: 78-81, 1997.

Owada A, Suda S, Hata T. Effect of long-term administration of prostaglandin I 2 in incipient diabetic nephropathy. Nephron 92: 788-796, 2002.

Robles RG, Villa E, Rabano A, Alvarez E, Martinez J, Ruilope LM, Sancho JM. Effect of cicaprost on the progression of diabetic nephropathy in uninephrectomized streptozotocin-induced diabetic rats. J Hypertens 11, Suppl 5: S208-S209, 1993.

Rossat J, Maillard M, Nussberger J, Brunner HR, Burnier M. Renal effect of selective cyclooxygenase-2 inhibition in normotensive salt-depleted subjects. Clin Pharmacol Ther 66: 76-84, 1999.

Schlein PS, O?Connell MJ, Blom J, Hubbard S, Magrath IT, Bergevin P, Wiernik PH, Ziegler JL, DeVita VT. Clinical antitumor activity and toxicity of streptozotocin. Cancer 34: 993-1000, 1974. <a href="/cgi/external_ref?access_num=10.1002/1097-0142(197410)34:4

Stock JL, Shinjo K, Burkhardt J, Roach M, Taniguchi K, Ishikawa T, Kim HS, Flannery PJ, Coffman TM, McNeish JD, Audoly LP. The prostaglandin E 2 EP 1 receptor mediates pain perception and regulates blood pressure. J Clin Invest 107: 325-331, 2001.

Sugimoto Y, Namba T, Shigemoto R, Negishi M, Ichikawa A, Narumiya S. Distinct cellular localization of mRNAs for three subtypes of prostaglandin E receptor in kidney. Am J Physiol Renal Fluid Electrolyte Physiol 266: F823-F828, 1994.

Tajiri Y, Umeda F, Inoguchi T, Nawata H. Effects of thromboxane synthase inhibitor (OKY-046) on urinary prostaglandin excretion and renal function in streptozotocin-induced diabetic rat. J Diabetes Complications 8: 126-132, 1994.

Tamma G, Wiesner B, Furkert J, Hahm D, Oksche A, Schaefer M, Valenti G, Rosenthal W, Klussmann E. The prostaglandin E 2 analogue sulprostone antagonizes vasopressin-induced antidiuresis through activation of Rho. J Cell Sci 116: 3285-3294, 2003.

Waldner C, Heise G, Schror K, Heering P. COX-2 inhibition and prostaglandin receptors in experimental nephritis. Eur J Clin Invest 33: 969-975, 2003.

Ziyadeh FN. Mediators of diabetic renal disease: the case for TGF- as the major mediator. J Am Soc Nephrol 15, Suppl 1: S55-S57, 2004.


作者单位:Department of Cellular and Molecular Medicine, Kidney Research Centre, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada

作者: Rania Nasrallah, Huaqi Xiong, and Richard L. H&eac 2008-7-4
医学百科App—中西医基础知识学习工具
  • 相关内容
  • 近期更新
  • 热文榜
  • 医学百科App—健康测试工具