ace/ace2 in diabetic nephropathy - CiteSeerX

10 downloads 0 Views 193KB Size Report
Doobay MF, Talman LS, Obr TD, Tian X, Davisson RL, Lazartigues E. ... Ye M, Wysocki J, William J, Soler MJ, Cokic I, Batlle D. Glomerular localization.
ACE/ACE2 IN DIABETIC MICE: IMPLICATIONS FOR NEPHROPATHY María José Soler*, Jan Wysocki, Minghao Ye, Eva Rodriguez and Daniel Batlle. Division of Nephrology & Hypertension, Department of Medicine. The Feinberg School of Medicine, Northwestern University. Chicago, USA. * Nephrology Department Hospital del Mar Universitat Autònoma de Barcelona. Barcelona, Spain.

Introduction Diabetic nephropathy (DN) is a microvascular complication of type 1 and type 2 diabetes, which is associated with end-stage renal disease (ESRD) and premature death from cardiovascular disease. The development of clinical nephropathy is insidious, and macroalbuminuria [urinary albumin excretion rate > 300 mg/24 hours], of the condition, is preceded by a phase of microalbuminuria (UAE 30–300 mg/24 hours), which usually lasts 5 to 10 years. As albuminuria worsens and blood pressure increases, there is a relentless decline in GFR and progression to ESRD. DN is now the most common single cause of end-stage renal disease (ESRD) in the US and Europe. Up to 40% of patients with type 2 diabetes will eventually develop diabetic nephropathy, and the number of patients progressing to ESRD has increased over the past 50 years. Although there is no cure for diabetic nephropathy, the rate of decline in renal function, and therefore progression to ESRD, can be slowed if it is detected and treated at an early stage. The renin-angiotensin system (RAS) plays an important role in the pathophysiology of many progressive renal diseases including diabetic nephropathy, and blockade of the RAS system with either angiotensin converting enzyme (ACE) or Angiotensin (ang) II receptor blockade attenuates progression of glomerular disease including DN . The recent discovery of Angiotensin converting enzyme 2, an active homologue of ACE, has expanded the knowledge of the RAS system. Whereas ACE promotes ang II formation from ang I, ACE2 promotes ang II degradation to the vasodilator peptide ang 1-7 (Figure 1). Moreover, ACE2 also catalyzes ang I to the inactive peptide ang 1-9 (Figure 1) .

1

Figure 1: Renin-angiotensin system pathways for formation and degradation of angiotensin II.

ACE/ACE2 in the kidney Initially, ACE2 was thought to be restricted to the kidney, heart and testes. Subsequently, ACE2 was found in other organs such as lungs, central nervous system and placenta. Using a dual fluorometric method for the concurrent determination of ACE and ACE2 activity, our laboratory found that the activity of both enzymes was higher in kidney than in heart tissue samples . This finding suggests that the high ACE2 expression in the kidney tissue may play a crucial role in the regulation of the intrarenal RAS. We also found that in the kidney, ACE and ACE2 are co-localized in the brush border of proximal tubules, but within the glomerulus both enzymes are localized in distinct structures. Glomerular ACE2 is mainly present in podocytes and, to a lesser extent, in glomerular mesangial cells, whereas glomerular ACE, by contrast, is only in endothelial cells. In kidneys from healthy control subjects, Lely et al. have found ACE2 expression in tubular and glomerular epithelium and in vascular muscular smooth muscle cells and the endothelium of interlobular arteries. In agreement with our findings in mice they did not find ACE2 expression in glomerular endothelium.

2

ACE/ACE2 in diabetic mice We examined the ACE2 and ACE glomerular expression in kidney samples from genetically obese-diabetic mouse (db/db). For this study, we used young female db/db mice to study an early phase of diabetes (3 to 4 wk of onset) without renal pathology by light microscopy. By immunohistochemistry, glomeruli from 8-wk-old db/db mice, showed increased ACE staining as compared with db/m. ACE2, by contrast, was decreased in db/db as compared with db/m. As mentioned before, glomerular ACE is mainly located in the endothelial cells, it seems that the increased ACE staining observed in the glomeruli from db/db mice points to an increase at the level of glomerular endothelial cells. On the other hand, the decreased ACE2 glomerular expression in db/db mice maybe reflects a decrease in protein expression at the level of the podocyte and possibly mesangial cells (or both). Taken these results together, we propose that in the diabetic mice there is an imbalance in the RAS with an excess of glomerular angiotensin II accumulation from increased ACE and decreased ACE2 in the glomeruli. The increased intraglomerular angiotensin II peptide in diabetic nephropathy results in an increase in albumin excretion rate, plasma creatinine, glomerular basement membrane width and a reduction in glomerular filtration rate. We also studied the effect of MLN-4760, a specific ACE2 inhibitor, on diabetic nephropathy. For this purpose we used two experimental models of diabetes: db/db mice (type 2 diabetes) and streptozotocin (STZ)-diabetic mice (type 1 diabetes). After chronic ACE2 inhibition, there was a significant increase in albumin excretion in the db/db mice as compared with vehicle treated db/db mice (figure 2). At the end of the study, 16 weeks of treatment, the albumin excretion was approximately three-fold higher in ML-4760treated db/db mice as compared with vehicle-treated db/db mice (figure 2). Moreover, glomerular staining for fibronectin, an extracellular matrix protein, was increased in db/db treated with MLN-4760. In addition, the specific AT1 blocker, telmisartan, prevented the increase in urinary albumin excretion that was associated with MLN-4760.

3

Figure 2: Monitoring urinary albumin excretion (UAE) in db/db and in STZ mice receiving vehicle or a specific ACE2 inhibitor, MLN-4760. Left panel: diabetic db/db mice receiving MLN-4760 (db/db + MLN-4760) had significantly increased UAR after 12 wk of treatment as compared to vehicle treated db/db mice (db/db + vehicle). The increase in albuminuria remained until the mice were sacrificed. Right panel: diabetic STZ mice receiving MLN-4760 (STZ + MLN-4760) had significantly increased UAE after 4 wk of treatment as compared to vehicle treated STZ mice (STZ).

In concordance with the study mentioned above in db/db mice, chronic ACE2 inhibition, in another model of diabetes produced by STZ-mice increased albumin expression as compared

to

vehicle-treated

STZ-mice

(Figure

2).

Furthermore,

MLN-4760

administration worsened kidney histological lesions such as matrix mesangial expansion (Figure 3) and tubular hypertrophy in STZ-diabetic mice. It should be noted that ACE2 inhibition was associated with enhanced expression of ACE in both glomeruli and vasculature, suggesting that augmentation of this enzyme may play a role in the observed glomerular lesions following ACE2 inhibition. A combination of high ACE and low ACE2 is apt to increase Ang II formation, while decreasing Ang II degradation. Interestingly, renal medulla and papilla were clearly atrophic in STZ-mice treated with MLN-4760. It should be mentioned that in an ACE knockout, marked medullary and papillary atrophy are prominent findings.

4

Figure 3: Ultrastructural analysis of glomeruli from non-diabetic control mice (left panel), STZ-diabetic mice treated with vehicle (middle panel) and STZ-diabetic mice treated with MLN-4760 (right panel): STZ-diabetic mice treated with vehicle shows mild mesangial expansion (M, middle panel), whereas STZdiabetic mice treated with MLN-4760 shows severe mesangial expansion (M, right panel). (magnification X 9500). M:mesangium

It has also been shown that the loss of ACE2 gene in male mice leads to the agedependent development of glomerular mesangial expansion with increased deposition of fibrillar collagens I/III and the extracellular matrix protein fibronectin. Wong et al. studied the effect of deletion of the angiotensin-converting enzyme 2 gene on diabetic kidney injury. For this purpose, ACE2-/- mice were crossed with Akita mice (Ins2WT/C96Y), a model of type 1 diabetes mellitus, and their respective wild type. At three months, ACE2-/y Ins2WT/C96Y mice showed increased urinary albumin excretion rate, in association with increased glomerular volume, increased mesangial matrix expansion, increased fibronectin, and α-SMA expression, and increased glomerular basement membrane thickening as compared with the ACE2+/yIns2WT/C96Y mice. Moreover, angiotensin II type 1 receptor blockade administration in the ACE2-/y Ins2WT/C96Y mice prevented the increase in albumin associated with ACE2 gene deletion. In conclusion, both the deletion of the ACE2 gene and chronic pharmacologic ACE2 inhibition are associated with accelerated kidney injury in diabetic mice. This suggests that ACE2 plays a protective role in diabetic kidney, and its lack leads to increased albumin excretion and mesangial matrix expansion. The fact that the administration of angiotensin II receptor blocker prevents the increase in albumin excretion, suggests that the deleterious effect of ACE2 gene deletion or pharmacological inhibition is mediated by angiotensin II via stimulation of the angiotensin II type 1 receptor. Based on their findings we have surmised that the ACE2 amplification either by gene therapy or with the

5

use of an agonist may be one of the future therapeutic strategies that target the RAS in patients with diabetic nephropathy23. Bibliography 1. Batlle D. Clinical and cellular markers of diabetic nephropathy. Kidney Int 2003;63(6): 2319-30. 2. Incidence of end-stage renal disease among persons with diabetes--United States, 1990-2002. MMWR Morb Mortal Wkly Rep 2005;54(43):1097-100. 3. Berger M, Monks D, Wanner C, Lindner TH. Diabetic nephropathy: an inherited disease or just a diabetic complication? Kidney Blood Press Res 2003;26(3):143-54. 4. Molitch ME. Management of early diabetic nephropathy. Am J Med 1997;102(4): 392-8. 5. Parving HH, Rossing P. The use of antihypertensive agents in prevention and treatment of diabetic nephropathy. Curr Opin Nephrol Hypertens 1994;3(3):292-300. 6. Rossing P, Hougaard P, Parving HH. Risk factors for development of incipient and overt diabetic nephropathy in type 1 diabetic patients: a 10- year prospective observational study. Diabetes Care 2002;25(5):859-64. 7. Taal MW, Brenner BM. Renoprotective benefits of RAS inhibition: from ACEI to angiotensin II antagonists. Kidney Int 2000;57(5):1803-17. 8. Bakris GL. Clinical importance of microalbuminuria in diabetes and hypertension. Curr Hypertens Rep 2004;6(5):352-6. 9. Raij L. Recommendations for the management of special populations: renal disease in diabetes. Am J Hypertens 2003;16(11 Pt 2):46S-9S. 10. Brenner BM, Cooper ME, de Zeeuw D, Keane WF, Mitch WE, Parving HH, Remuzzi G, Snapinn SM, Zhang Z, Shahinfar S. Effects of losartan on renal and cardiovascular outcomes in patients with type 2 diabetes and nephropathy. N Engl J Med 2001;345(12): 861-9. 11. Burnier M, Zanchi A. Blockade of the renin-angiotensin-aldosterone system: a key therapeutic strategy to reduce renal and cardiovascular events in patients with diabetes. J Hypertens 2006;24(1):11-25. 12. Giacchetti G, Sechi LA, Rilli S, Carey RM. The renin-angiotensin-aldosterone system, glucose metabolism and diabetes. Trends Endocrinol Metab 2005;16(3):120-6. 13. Crackower MA, Sarao R, Oudit GY, Yagil C, Kozieradzki I, Scanga SE, Oliveirados-Santos AJ, da Costa J, Zhang L, Pei Y, Scholey J, Ferrario CM, Manoukian AS, Chappell MC, Backx PH, Yagil Y, Penninger JM. Angiotensin-converting enzyme 2 is an essential regulator of heart function. Nature 2002;417(6891):822-8. 14. Donoghue M, Hsieh F, Baronas E, Godbout K, Gosselin M, Stagliano N, Donovan M, Woolf B, Robison K, Jeyaseelan R, Breitbart RE, Acton S. A novel angiotensinconverting enzyme-related carboxypeptidase (ACE2) converts angiotensin I to angiotensin 1-9. Circ Res 2000;87(5):E1-9. 15. Ferrario CM, Trask AJ, Jessup JA. Advances in biochemical and functional roles of angiotensin-converting enzyme 2 and angiotensin-(1-7) in regulation of cardiovascular

6

function. Am J Physiol Heart Circ Physiol 2005;289(6):H2281-90. 16. Doobay MF, Talman LS, Obr TD, Tian X, Davisson RL, Lazartigues E. Differential expression of neuronal ACE2 in transgenic mice with overexpression of the brain reninangiotensin system. Am J Physiol Regul Integr Comp Physiol 2006. 17. Imai Y, Kuba K, Rao S, Huan Y, Guo F, Guan B, Yang P, Sarao R, Wada T, LeongPoi H, Crackower MA, Fukamizu A, Hui CC, Hein L, Uhlig S, Slutsky AS, Jiang C, Penninger JM. Angiotensin-converting enzyme 2 protects from severe acute lung failure. Nature 2005;436(7047):112-6. 18. Valdes G, Neves LA, Anton L, Corthorn J, Chacon C, Germain AM, Merrill DC, Ferrario CM, Sarao R, Penninger J, Brosnihan KB. Distribution of angiotensin-(1-7) and ACE2 in human placentas of normal and pathological pregnancies. Placenta 2006;27(2-3):200-7. 19. Hamming I, Timens W, Bulthuis ML, Lely AT, Navis GJ, van Goor H. Tissue distribution of ACE2 protein, the functional receptor for SARS coronavirus. A first step in understanding SARS pathogenesis. J Pathol 2004;203(2):631-7. 20. Xie X, Chen J, Wang X, Zhang F, Liu Y. Age- and gender-related difference of ACE2 expression in rat lung. Life Sci 2006;78(19):2166-71. 21. Wysocki J, Ye M, Soler MJ, Gurley SB, Xiao HD, Bernstein KE, Coffman TM, Chen S, Batlle D. ACE and ACE2 activity in diabetic mice. Diabetes 2006;55(7):2132-9. 22. Ye M, Wysocki J, William J, Soler MJ, Cokic I, Batlle D. Glomerular localization and expression of Angiotensin-converting enzyme 2 and Angiotensin-converting enzyme: implications for albuminuria in diabetes. J Am Soc Nephrol 2006;17(11):3067-75. 23. Lely AT, Hamming I, van Goor H, Navis GJ. Renal ACE2 expression in human kidney disease. J Pathol 2004;204(5):587-93. 24. Sharma K, McCue P, Dunn SR. Diabetic kidney disease in the db/db mouse. Am J Physiol Renal Physiol 2003;284(6):F1138-44. 25. Breyer MD, Bottinger E, Brosius FC, 3rd, Coffman TM, Harris RC, Heilig CW, Sharma K. Mouse models of diabetic nephropathy. J Am Soc Nephrol 2005;16(1):27-45. 26. Nicholas SB, Mauer M, Basgen JM, Aguiniga E, Chon Y. Effect of angiotensin II on glomerular structure in streptozotocin-induced diabetic rats. Am J Nephrol 2004;24(5): 549-56. 27. Towler P, Staker B, Prasad SG, Menon S, Tang J, Parsons T, Ryan D, Fisher M, Williams D, Dales NA, Patane MA, Pantoliano MW. ACE2 X-ray structures reveal a large hinge-bending motion important for inhibitor binding and catalysis. J Biol Chem 2004;279(17):17996-8007. 28. Soler MJ, Wysocki J, Ye M, Lloveras J, Kanwar Y, Batlle D. ACE2 inhibition worsens glomerular injury in association with increased ACE expression in streptozotocin-induced diabetic mice. Kidney Int 2007;72(5):614-23. 29. Esther CR, Jr., Howard TE, Marino EM, Goddard JM, Capecchi MR, Bernstein KE. Mice lacking angiotensin-converting enzyme have low blood pressure, renal pathology, and reduced male fertility. Lab Invest 1996;74(5):953-65. 30. Oudit GY, Herzenberg AM, Kassiri Z, Wong D, Reich H, Khokha R, Crackower MA, Backx PH, Penninger JM, Scholey JW. Loss of angiotensin-converting enzyme-2 leads to the late development of angiotensin II-dependent glomerulosclerosis. Am J Pathol 2006;168(6):1808-20.

7

31. Wong DW, Oudit GY, Reich H, Kassiri Z, Zhou J, Liu QC, Backx PH, Penninger JM, Herzenberg AM, Scholey JW. Loss of angiotensin-converting enzyme-2 (Ace2) accelerates diabetic kidney injury. Am J Pathol 2007;171(2):438-51.

8