1
|
Zhao XP, Chang SY, Pang Y, Liao MC, Peng J, Ingelfinger JR, Chan JSD, Zhang SL. Hedgehog interacting protein activates sodium-glucose cotransporter 2 expression and promotes renal tubular epithelial cell senescence in a mouse model of type 1 diabetes. Diabetologia 2023; 66:223-240. [PMID: 36260124 DOI: 10.1007/s00125-022-05810-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 08/17/2022] [Indexed: 12/13/2022]
Abstract
AIMS/HYPOTHESIS Senescent renal tubular cells may be linked to diabetic kidney disease (DKD)-related tubulopathy. We studied mice with or without diabetes in which hedgehog interacting protein (HHIP) was present or specifically knocked out in renal tubules (HhipRT-KO), hypothesising that local deficiency of HHIP in the renal tubules would attenuate tubular cell senescence, thereby preventing DKD tubulopathy. METHODS Low-dose streptozotocin was employed to induce diabetes in both HhipRT-KO and control (Hhipfl/fl) mice. Transgenic mice overexpressing Hhip in renal proximal tubular cells (RPTC) (HhipRPTC-Tg) were used for validation, and primary RPTCs and human RPTCs (HK2) were used for in vitro studies. Kidney morphology/function, tubular senescence and the relevant molecular measurements were assessed. RESULTS Compared with Hhipfl/fl mice with diabetes, HhipRT-KO mice with diabetes displayed lower blood glucose levels, normalised GFR, ameliorated urinary albumin/creatinine ratio and less severe DKD, including tubulopathy. Sodium-glucose cotransporter 2 (SGLT2) expression was attenuated in RPTCs of HhipRT-KO mice with diabetes compared with Hhipfl/fl mice with diabetes. In parallel, an increased tubular senescence-associated secretory phenotype involving release of inflammatory cytokines (IL-1β, IL-6 and monocyte chemoattractant protein-1) and activation of senescence markers (p16, p21, p53) in Hhipfl/fl mice with diabetes was attenuated in HhipRT-KO mice with diabetes. In contrast, HhipRPTC-Tg mice had increased tubular senescence, which was inhibited by canagliflozin in primary RPTCs. In HK2 cells, HHIP overexpression or recombinant HHIP increased SGLT2 protein expression and promoted cellular senescence by targeting both ataxia-telangiectasia mutated and ataxia-telangiectasia and Rad3-related-mediated cell arrest. CONCLUSIONS/INTERPRETATION Tubular HHIP deficiency prevented DKD-related tubulopathy, possibly via the inhibition of SGLT2 expression and cellular senescence.
Collapse
Affiliation(s)
- Xin-Ping Zhao
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Department of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Shiao-Ying Chang
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Department of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Yuchao Pang
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Department of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Min-Chun Liao
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Department of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Junzheng Peng
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Department of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Julie R Ingelfinger
- Harvard Medical School, Pediatric Nephrology Unit, Massachusetts General Hospital, Boston, MA, USA
| | - John S D Chan
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Department of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Shao-Ling Zhang
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Department of Medicine, Université de Montréal, Montréal, QC, Canada.
| |
Collapse
|
2
|
Overstreet JM, Gifford CC, Tang J, Higgins PJ, Samarakoon R. Emerging role of tumor suppressor p53 in acute and chronic kidney diseases. Cell Mol Life Sci 2022; 79:474. [PMID: 35941392 PMCID: PMC11072039 DOI: 10.1007/s00018-022-04505-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/20/2022] [Accepted: 07/21/2022] [Indexed: 02/06/2023]
Abstract
p53 is a major regulator of cell cycle arrest, apoptosis, and senescence. While involvement of p53 in tumorigenesis is well established, recent studies implicate p53 in the initiation and progression of several renal diseases, which is the focus of this review. Ischemic-, aristolochic acid (AA) -, diabetic-, HIV-associated-, obstructive- and podocyte-induced nephropathies are accompanied by activation and/or elevated expression of p53. Studies utilizing chemical or renal-specific inhibition of p53 in mice confirm the pathogenic role of this transcription factor in acute kidney injury and chronic kidney disease. TGF-β1, NOX, ATM/ATR kinases, Cyclin G, HIPK, MDM2 and certain micro-RNAs are important determinants of renal p53 function in response to trauma. AA, cisplatin or TGF-β1-mediated ROS generation via NOXs promotes p53 phosphorylation and subsequent tubular dysfunction. p53-SMAD3 transcriptional cooperation downstream of TGF-β1 orchestrates induction of fibrotic factors, extracellular matrix accumulation and pathogenic renal cell communication. TGF-β1-induced micro-RNAs (such as mir-192) could facilitate p53 activation, leading to renal hypertrophy and matrix expansion in response to diabetic insults while AA-mediated mir-192 induction regulates p53 dependent epithelial G2/M arrest. The widespread involvement of p53 in tubular maladaptive repair, interstitial fibrosis, and podocyte injury indicate that p53 clinical targeting may hold promise as a novel therapeutic strategy for halting progression of certain acute and chronic renal diseases, which affect hundreds of million people worldwide.
Collapse
Affiliation(s)
| | - Cody C Gifford
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, 12208, USA
| | - Jiaqi Tang
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Paul J Higgins
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, 12208, USA.
- Center for Cell Biology and Cancer Research, Albany Medical College, 47 New Scotland Avenue, Albany, NY, 12208, USA.
| | - Rohan Samarakoon
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, 12208, USA.
- Center for Cell Biology and Cancer Research, Albany Medical College, 47 New Scotland Avenue, Albany, NY, 12208, USA.
| |
Collapse
|
3
|
Re RN. A Pathogenic Mechanism Potentially Operative in Multiple Progressive Diseases and Its Therapeutic Implications. J Clin Pharmacol 2017; 57:1507-1518. [DOI: 10.1002/jcph.997] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 07/17/2017] [Indexed: 01/07/2023]
Affiliation(s)
- Richard N. Re
- Division of Academics-Research; Ochsner Clinic Foundation; New Orleans LA USA
| |
Collapse
|
4
|
An Expanded View of Progressive Cardiorenal Disorders. Am J Med Sci 2016; 351:626-33. [DOI: 10.1016/j.amjms.2016.03.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 03/17/2016] [Indexed: 11/23/2022]
|
5
|
Casare FAM, Thieme K, Costa-Pessoa JM, Rossoni LV, Couto GK, Fernandes FB, Casarini DE, Oliveira-Souza M. Renovascular remodeling and renal injury after extended angiotensin II infusion. Am J Physiol Renal Physiol 2016; 310:F1295-307. [PMID: 26962104 DOI: 10.1152/ajprenal.00471.2015] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 03/05/2016] [Indexed: 11/22/2022] Open
Abstract
Chronic angiotensin II (ANG II) infusion for 1 or 2 wk leads to progressive hypertension and induces inward hypertrophic remodeling in preglomerular vessels, which is associated with increased renal vascular resistance (RVR) and decreased glomerular perfusion. Considering the ability of preglomerular vessels to exhibit adaptive responses, the present study was performed to evaluate glomerular perfusion and renal function after 6 wk of ANG II infusion. To address this study, male Wistar rats were submitted to sham surgery (control) or osmotic minipump insertion (ANG II 200 ng·kg(-1)·min(-1), 42 days). A group of animals was treated or cotreated with losartan (10 mg·kg(-1)·day(-1)), an AT1 receptor antagonist, between days 28 and 42 Chronic ANG II infusion increased systolic blood pressure to 185 ± 4 compared with 108 ± 2 mmHg in control rats. Concomitantly, ANG II-induced hypertension increased intrarenal ANG II level and consequently, preglomerular and glomerular injury. Under this condition, ANG II enhanced the total renal plasma flow (RPF), glomerular filtration rate (GFR), urine flow and induced pressure natriuresis. These changes were accompanied by lower RVR and enlargement of the lumen of interlobular arteries and afferent arterioles, consistent with impairment of renal autoregulatory capability and outward preglomerular remodeling. The glomerular injury culminated with podocyte effacement, albuminuria, tubulointerstitial macrophage infiltration and intrarenal extracellular matrix accumulation. Losartan attenuated most of the effects of ANG II. Our findings provide new information regarding the contribution of ANG II infusion over 2 wk to renal hemodynamics and function via the AT1 receptor.
Collapse
Affiliation(s)
| | - Karina Thieme
- Laboratory of Cellular and Molecular Endocrinology, Medical School, University of Sao Paulo, Sao Paulo, Brazil; and
| | - Juliana Martins Costa-Pessoa
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Luciana Venturini Rossoni
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Gisele Kruger Couto
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | | | - Dulce Elena Casarini
- Division of Nephrology, Department of Medicine, Federal University of Sao Paulo, Sao Paulo, Brazil
| | - Maria Oliveira-Souza
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil;
| |
Collapse
|
6
|
Lo CS, Shi Y, Chang SY, Abdo S, Chenier I, Filep JG, Ingelfinger JR, Zhang SL, Chan JSD. Overexpression of heterogeneous nuclear ribonucleoprotein F stimulates renal Ace-2 gene expression and prevents TGF-β1-induced kidney injury in a mouse model of diabetes. Diabetologia 2015; 58:2443-54. [PMID: 26232095 PMCID: PMC4572079 DOI: 10.1007/s00125-015-3700-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 06/26/2015] [Indexed: 01/09/2023]
Abstract
AIMS/HYPOTHESIS We investigated whether heterogeneous nuclear ribonucleoprotein F (hnRNP F) stimulates renal ACE-2 expression and prevents TGF-β1 signalling, TGF-β1 inhibition of Ace-2 gene expression and induction of tubulo-fibrosis in an Akita mouse model of type 1 diabetes. METHODS Adult male Akita transgenic (Tg) mice overexpressing specifically hnRNP F in their renal proximal tubular cells (RPTCs) were studied. Non-Akita littermates and Akita mice served as controls. Immortalised rat RPTCs stably transfected with plasmid containing either rat Hnrnpf cDNA or rat Ace-2 gene promoter were also studied. RESULTS Overexpression of hnRNP F attenuated systemic hypertension, glomerular filtration rate, albumin/creatinine ratio, urinary angiotensinogen (AGT) and angiotensin (Ang) II levels, renal fibrosis and profibrotic gene (Agt, Tgf-β1, TGF-β receptor II [Tgf-βrII]) expression, stimulated anti-profibrotic gene (Ace-2 and Ang 1-7 receptor [MasR]) expression, and normalised urinary Ang 1-7 level in Akita Hnrnpf-Tg mice as compared with Akita mice. In vitro, hnRNP F overexpression stimulated Ace-2 gene promoter activity, mRNA and protein expression, and attenuated Agt, Tgf-β1 and Tgf-βrII gene expression. Furthermore, hnRNP F overexpression prevented TGF-β1 signalling and TGF-β1 inhibition of Ace-2 gene expression. CONCLUSIONS/INTERPRETATION These data demonstrate that hnRNP F stimulates Ace-2 gene transcription, prevents TGF-β1 inhibition of Ace-2 gene transcription and induction of kidney injury in diabetes. HnRNP F may be a potential target for treating hypertension and renal fibrosis in diabetes.
Collapse
Affiliation(s)
- Chao-Sheng Lo
- Centre de recherche, Centre hospitalier de l'Université de Montréal (CRCHUM) - Tour Viger Pavillon R, Université de Montréal, 900 Saint-Denis Street, Montreal, QC, H2X 0A9, Canada
| | - Yixuan Shi
- Centre de recherche, Centre hospitalier de l'Université de Montréal (CRCHUM) - Tour Viger Pavillon R, Université de Montréal, 900 Saint-Denis Street, Montreal, QC, H2X 0A9, Canada
| | - Shiao-Ying Chang
- Centre de recherche, Centre hospitalier de l'Université de Montréal (CRCHUM) - Tour Viger Pavillon R, Université de Montréal, 900 Saint-Denis Street, Montreal, QC, H2X 0A9, Canada
| | - Shaaban Abdo
- Centre de recherche, Centre hospitalier de l'Université de Montréal (CRCHUM) - Tour Viger Pavillon R, Université de Montréal, 900 Saint-Denis Street, Montreal, QC, H2X 0A9, Canada
| | - Isabelle Chenier
- Centre de recherche, Centre hospitalier de l'Université de Montréal (CRCHUM) - Tour Viger Pavillon R, Université de Montréal, 900 Saint-Denis Street, Montreal, QC, H2X 0A9, Canada
| | - Janos G Filep
- Research Centre, Maisonneuve-Rosemont Hospital, Université de Montréal, Montreal, QC, Canada
| | - Julie R Ingelfinger
- Pediatric Nephrology Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Shao-Ling Zhang
- Centre de recherche, Centre hospitalier de l'Université de Montréal (CRCHUM) - Tour Viger Pavillon R, Université de Montréal, 900 Saint-Denis Street, Montreal, QC, H2X 0A9, Canada.
| | - John S D Chan
- Centre de recherche, Centre hospitalier de l'Université de Montréal (CRCHUM) - Tour Viger Pavillon R, Université de Montréal, 900 Saint-Denis Street, Montreal, QC, H2X 0A9, Canada.
| |
Collapse
|
7
|
Padda RS, Shi Y, Lo CS, Zhang SL, Chan JSD. Angiotensin-(1-7): A Novel Peptide to Treat Hypertension and Nephropathy in Diabetes? ACTA ACUST UNITED AC 2015; 6. [PMID: 26793405 DOI: 10.4172/2155-6156.1000615] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The renin-angiotensin system (RAS) plays a pivotal role in mammalian homeostasis physiology. The RAS can be delineated into a classical RAS (the pressor arm) including angiotensinogen (Agt), renin, angiotensin-converting enzyme (ACE), angiotensin II (Ang II) and angiotensin type 1 receptor (AT1R), and a counterbalancing novel RAS (the depressor arm) including Agt, renin, angiotensin-converting enzyme-2 (ACE-2), angiotensin-(1-7) (Ang 1-7) and Ang 1-7 receptor (or Mas receptor (MasR)). Hyperglycemia (diabetes) induces severe tissue oxidative stress, which stimulates the pressor arm of the renal RAS axis and leads to an increase in ACE/ACE-2 ratio, with excessive formation of Ang II. There is a growing body of evidence for beneficial effects of the depressor arm of RAS (ACE-2/Ang 1-7/MasR) axis in diabetes, hypertension and several other diseased conditions. Evidence from in vitro, in vivo and clinical studies reflects anti-oxidant, anti-fibrotic, and anti-inflammatory properties of Ang 1-7. Most of the currently available therapies only target suppression of the pressor arm of RAS with angiotensin receptor blockers (ARBs) and ACE inhibitors (ACEi). However, it is time to consider simultaneous activation of the depressor arm for more effective outcomes. This review summarizes the recent updates on the protective role of Ang 1-7 in hypertension and kidney injury in diabetes, as well as the possible underlying mechanism(s) of Ang 1-7 action, suggesting that the ACE-2/Ang 1-7/MasR axis can be developed as a therapeutic target for the treatment of diabetes-induced hypertension and renal damage.
Collapse
Affiliation(s)
- Ranjit Singh Padda
- Université de Montréal, Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Tour Viger, 900 Saint Denis Street, Montreal, Quebec, Canada H2X 0A9
| | - Yixuan Shi
- Université de Montréal, Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Tour Viger, 900 Saint Denis Street, Montreal, Quebec, Canada H2X 0A9
| | - Chao-Sheng Lo
- Université de Montréal, Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Tour Viger, 900 Saint Denis Street, Montreal, Quebec, Canada H2X 0A9
| | - Shao-Ling Zhang
- Université de Montréal, Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Tour Viger, 900 Saint Denis Street, Montreal, Quebec, Canada H2X 0A9
| | - John S D Chan
- Université de Montréal, Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Tour Viger, 900 Saint Denis Street, Montreal, Quebec, Canada H2X 0A9
| |
Collapse
|
8
|
Rukavina Mikusic NL, Kravetz MC, Kouyoumdzian NM, Della Penna SL, Rosón MI, Fernández BE, Choi MR. Signaling pathways involved in renal oxidative injury: role of the vasoactive peptides and the renal dopaminergic system. JOURNAL OF SIGNAL TRANSDUCTION 2014; 2014:731350. [PMID: 25436148 PMCID: PMC4243602 DOI: 10.1155/2014/731350] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 10/16/2014] [Indexed: 12/24/2022]
Abstract
The physiological hydroelectrolytic balance and the redox steady state in the kidney are accomplished by an intricate interaction between signals from extrarenal and intrarenal sources and between antinatriuretic and natriuretic factors. Angiotensin II, atrial natriuretic peptide and intrarenal dopamine play a pivotal role in this interactive network. The balance between endogenous antioxidant agents like the renal dopaminergic system and atrial natriuretic peptide, by one side, and the prooxidant effect of the renin angiotensin system, by the other side, contributes to ensuring the normal function of the kidney. Different pathological scenarios, as nephrotic syndrome and hypertension, where renal sodium excretion is altered, are associated with an impaired interaction between two natriuretic systems as the renal dopaminergic system and atrial natriuretic peptide that may be involved in the pathogenesis of renal diseases. The aim of this review is to update and comment the most recent evidences about the intracellular pathways involved in the relationship between endogenous antioxidant agents like the renal dopaminergic system and atrial natriuretic peptide and the prooxidant effect of the renin angiotensin system in the pathogenesis of renal inflammation.
Collapse
Affiliation(s)
- N. L. Rukavina Mikusic
- Department of Pathophysiology, Faculty of Pharmacy and Biochemistry, University of Buenos Aires, CONICET, INFIBIOC, 1113 Buenos Aires, Argentina
| | - M. C. Kravetz
- Department of Pathophysiology, Faculty of Pharmacy and Biochemistry, University of Buenos Aires, CONICET, INFIBIOC, 1113 Buenos Aires, Argentina
| | - N. M. Kouyoumdzian
- Department of Pathophysiology, Faculty of Pharmacy and Biochemistry, University of Buenos Aires, CONICET, INFIBIOC, 1113 Buenos Aires, Argentina
| | - S. L. Della Penna
- Department of Pathophysiology, Faculty of Pharmacy and Biochemistry, University of Buenos Aires, CONICET, INFIBIOC, 1113 Buenos Aires, Argentina
| | - M. I. Rosón
- Department of Pathophysiology, Faculty of Pharmacy and Biochemistry, University of Buenos Aires, CONICET, INFIBIOC, 1113 Buenos Aires, Argentina
| | - B. E. Fernández
- Department of Pathophysiology, Faculty of Pharmacy and Biochemistry, University of Buenos Aires, CONICET, INFIBIOC, 1113 Buenos Aires, Argentina
| | - M. R. Choi
- Department of Pathophysiology, Faculty of Pharmacy and Biochemistry, University of Buenos Aires, CONICET, INFIBIOC, 1113 Buenos Aires, Argentina
| |
Collapse
|
9
|
Matsuki K, Hathaway CK, Lawrence MG, Smithies O, Kakoki M. The role of transforming growth factor β1 in the regulation of blood pressure. Curr Hypertens Rev 2014; 10:223-38. [PMID: 25801626 PMCID: PMC4842018 DOI: 10.2174/157340211004150319123313] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 02/19/2015] [Accepted: 02/23/2015] [Indexed: 01/21/2023]
Abstract
Although human association studies suggest a link between polymorphisms in the gene encoding transforming growth factor (TGF) β1 and differing blood pressure levels, a causative mechanism for this correlation remains elusive. Recently we have generated a series of mice with graded expression of TGFβ1, ranging from approximately 10% to 300% compared to normal. We have found that blood pressure and plasma volume are negatively regulated by TGFβ1. Of note, the 10% hypomorph exhibits primary aldosteronism and markedly impaired urinary excretion of water and electrolytes. We here review previous literature highlighting the importance of TGFβ signaling as a natriuretic system, which we postulate is a causative mechanism explaining how polymorphisms in TGFβ1 could influence blood pressure levels.
Collapse
Affiliation(s)
| | | | | | | | - Masao Kakoki
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, CB #7525, 701 Brinkhous-Bullitt Building, Chapel Hill, NC 27599-7525, USA.
| |
Collapse
|
10
|
Fibulin-2 deficiency attenuates angiotensin II-induced cardiac hypertrophy by reducing transforming growth factor-β signalling. Clin Sci (Lond) 2013; 126:275-88. [PMID: 23841699 DOI: 10.1042/cs20120636] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
AngII (angiotensin II) is a potent neurohormone responsible for cardiac hypertrophy, in which TGF (transforming growth factor)-β serves as a principal downstream mediator. We recently found that ablation of fibulin-2 in mice attenuated TGF-β signalling, protected mice against progressive ventricular dysfunction, and significantly reduced the mortality after experimental MI (myocardial infarction). In the present study, we investigated the role of fibulin-2 in AngII-induced TGF-β signalling and subsequent cardiac hypertrophy. We performed chronic subcutaneous infusion of AngII in fibulin-2 null (Fbln2-/-), heterozygous (Fbln2+/-) and WT (wild-type) mice by a mini-osmotic pump. After 4 weeks of subpressor dosage of AngII infusion (0.2 μg/kg of body weight per min), WT mice developed significant hypertrophy, whereas the Fbln2-/- showed no response. In WT, AngII treatment significantly up-regulated mRNAs for fibulin-2, ANP (atrial natriuretic peptide), TGF-β1, Col I (collagen type I), Col III (collagen type III), MMP (matrix metalloproteinase)-2 and MMP-9, and increased the phosphorylation of TGF-β-downstream signalling markers, Smad2, TAK1 (TGF-β-activated kinase 1) and p38 MAPK (mitogen-activated protein kinase), which were all unchanged in AngII-treated Fbln2-/- mice. The Fbln2+/- mice consistently displayed AngII-induced effects intermediate between WT and Fbln2-/-. Pressor dosage of AngII (2 mg/kg of body weight per min) induced significant fibrosis in WT but not in Fbln2-/- mice with comparable hypertension and hypertrophy in both groups. Isolated CFs (cardiac fibroblasts) were treated with AngII, in which direct AngII effects and TGF-β-mediated autocrine effects was observed in WT. The latter effects were totally abolished in Fbln2-/- cells, suggesting that fibulin-2 is essential for AngII-induced TGF-β activation. In conclusion our data indicate that fibulin-2 is essential for AngII-induced TGF-β-mediated cardiac hypertrophy via enhanced TGF-β activation and suggest that fibulin-2 is a potential therapeutic target to inhibit AngII-induced cardiac remodelling.
Collapse
|
11
|
Guo W, Wong S, Bhasin S. AAV-mediated administration of myostatin pro-peptide mutant in adult Ldlr null mice reduces diet-induced hepatosteatosis and arteriosclerosis. PLoS One 2013; 8:e71017. [PMID: 23936482 PMCID: PMC3731267 DOI: 10.1371/journal.pone.0071017] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Accepted: 07/01/2013] [Indexed: 12/25/2022] Open
Abstract
UNLABELLED Genetic disruption of myostatin or its related signaling is known to cause strong protection against diet-induced metabolic disorders. The translational value of these prior findings, however, is dependent on whether such metabolically favorable phenotype can be reproduced when myostatin blockade begins at an adult age. Here, we reported that AAV-mediated delivery of a myostatin pro-peptide D76A mutant in adult mice attenuates the development of hepatic steatosis and arteriosclerosis, two common diet-induced metabolic diseases. A single dose of AAV-D76A in adult Ldlr null mice resulted in sustained expression of myostatin pro-peptide in the liver. Compared to vehicle-treated mice, D76A-treated mice gained similar amount of lean and fat mass when fed a high fat diet. However, D76A-treated mice displayed significantly reduced aortic lesions and liver fat, in association with a reduction in hepatic expression of lipogenic genes and improvement in liver insulin sensitivity. This suggests that muscle and fat may not be the primary targets of treatment under our experimental condition. In support to this argument, we show that myostatin directly up-regulated lipogenic genes and increased fat accumulation in cultured liver cells. We also show that both myostatin and its receptor were abundantly expressed in mouse aorta. Cultured aortic endothelial cells responded to myostatin with a reduction in eNOS phosphorylation and an increase in ICAM-1 and VCAM-1 expression. CONCLUSIONS AAV-mediated expression of myostatin pro-peptide D76A mutant in adult Ldlr null mice sustained metabolic protection without remarkable impacts on body lean and fat mass. Further investigations are needed to determine whether direct impact of myostatin on liver and aortic endothelium may contribute to the related metabolic phenotypes.
Collapse
Affiliation(s)
- Wen Guo
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA.
| | | | | |
Collapse
|
12
|
Samarakoon R, Dobberfuhl AD, Cooley C, Overstreet JM, Patel S, Goldschmeding R, Meldrum KK, Higgins PJ. Induction of renal fibrotic genes by TGF-β1 requires EGFR activation, p53 and reactive oxygen species. Cell Signal 2013; 25:2198-209. [PMID: 23872073 DOI: 10.1016/j.cellsig.2013.07.007] [Citation(s) in RCA: 124] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Accepted: 07/10/2013] [Indexed: 12/14/2022]
Abstract
While transforming growth factor-β (TGF-β1)-induced SMAD2/3 signaling is a critical event in the progression of chronic kidney disease, the role of non-SMAD mechanisms in the orchestration of fibrotic gene changes remains largely unexplored. TGF-β1/SMAD3 pathway activation in renal fibrosis (induced by ureteral ligation) correlated with epidermal growth factor receptor(Y845) (EGFR(Y845)) and p53(Ser15) phosphorylation and induction of disease causative target genes plasminogen activator inhibitor-1 (PAI-1) and connective tissue growth factor (CTGF) prompting an investigation of the mechanistic involvement of EGFR and tumor suppressor p53 in profibrotic signaling. TGF-β1, PAI-1, CTGF, p53 and EGFR were co-expressed in the obstructed kidney localizing predominantly to the tubular and interstitial compartments. Indeed, TGF-β1 activated EGFR and p53 as well as SMAD2/3. Genetic deficiency of either EGFR or p53 or functional blockade with AG1478 or Pifithrin-α, respectively, effectively inhibited PAI-1and CTGF induction and morphological transformation of renal fibroblasts as did SMAD3 knockdown or pretreatment with the SMAD3 inhibitor SIS3. Reactive oxygen species (ROS)-dependent mechanisms initiated by TGF-β1 were critical for EGFR(Y845) and p53(Ser15) phosphorylation and target gene expression. The p22(Phox) subunit of NADPH oxidase was also elevated in the fibrotic kidney with an expression pattern similar to p53 and EGFR. EGF stimulation alone initiated, albeit delayed, c-terminal SMAD3 phosphorylation (that required the TGF-β1 receptor) and rapid ERK2 activation both of which are necessary for PAI-1 and CTGF induction in renal fibroblasts. These data highlight the extensive cross-talk among SMAD2/3, EGFR and p53 pathways essential for expression of TGF-β1-induced fibrotic target genes.
Collapse
Affiliation(s)
- Rohan Samarakoon
- Center for Cell Biology & Cancer Research, Albany Medical College, Albany, NY 12208, USA
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Samarakoon R, Overstreet JM, Higgins PJ. TGF-β signaling in tissue fibrosis: redox controls, target genes and therapeutic opportunities. Cell Signal 2012; 25:264-8. [PMID: 23063463 DOI: 10.1016/j.cellsig.2012.10.003] [Citation(s) in RCA: 269] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Accepted: 10/07/2012] [Indexed: 10/27/2022]
Abstract
During development of TGF-β1-initiated fibroproliferative disorders, NADPH oxidases (NOX family members) generate reactive oxygen species (ROS) resulting in downstream transcription of a subset genes encoding matrix structural elements and profibrotic factors. Prominent among the repertoire of disease-implicated genes is the TGF-β1 target gene encoding the potent profibrotic matricellular protein plasminogen activator inhibitor-1 (PAI-1 or SERPINE1). PAI-1 is the major physiologic inhibitor of the plasmin-based pericellular cascade and a causative factor in the development of vascular thrombotic and fibroproliferative disorders. ROS generation in response to TGF-β1 stimulation is rapid and precedes PAI-1 induction; engagement of non-SMAD (e.g., EGFR, Src kinase, MAP kinases, p53) and SMAD2/3 pathways are both required for PAI-1 expression and are ROS-dependent. Recent findings suggest a novel role for p53 in TGF-β1-induced PAI-1 transcription that involves ROS generation and p53/SMAD interactions. Targeting ROS and ROS-activated cellular events is likely to have therapeutic implications in the management of fibrotic disorders, particularly in the context of prolonged TGF-β1 signaling.
Collapse
Affiliation(s)
- Rohan Samarakoon
- Center for Cell Biology and Cancer Research, Albany Medical College, 47 New Scotland Avenue, Albany, NY 12208, USA
| | | | | |
Collapse
|
14
|
|
15
|
Chang SY, Chen YW, Zhao XP, Chenier I, Tran S, Sauvé A, Ingelfinger JR, Zhang SL. Catalase prevents maternal diabetes-induced perinatal programming via the Nrf2-HO-1 defense system. Diabetes 2012; 61:2565-74. [PMID: 22733796 PMCID: PMC3447903 DOI: 10.2337/db12-0248] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
We investigated whether overexpression of catalase (CAT) in renal proximal tubular cells (RPTCs) could prevent the programming of hypertension and kidney disease in the offspring of dams with maternal diabetes. Male offspring of nondiabetic and diabetic dams from two transgenic (Tg) lines (Hoxb7-green fluorescent protein [GFP]-Tg [controls] and Hoxb7/CAT-GFP-Tg, which overexpress CAT in RPTCs) were studied from the prenatal period into adulthood. Nephrogenesis, systolic blood pressure, renal hyperfiltration, kidney injury, and reactive oxygen species (ROS) generation were assessed. Gene expression of transforming growth factor-β1 (TGF-β1), nuclear factor erythroid 2p45-related factor-2 (Nrf2), and heme oxygenase-1 (HO-1) was tested in both in vitro and in vivo studies. Renal dysmorphogenesis was observed in offspring of Hoxb7-GFP-Tg dams with severe maternal diabetes; the affected male offspring displayed higher renal ROS generation and developed hypertension and renal hyperfiltration as well as renal injury with heightened TGF-β1 expression in adulthood. These changes were ameliorated in male offspring of diabetic Hoxb7/CAT-GFP-Tg dams via the Nrf2-HO-1 defense system. CAT promoted Nrf2 nuclear translocation and HO-1 gene expression, seen in both in vitro and in vivo studies. In conclusion, CAT overexpression in the RPTCs ameliorated maternal diabetes-induced perinatal programming, mediated, at least in part, by triggering the Nrf2-HO-1 defense system.
Collapse
Affiliation(s)
- Shiao-Ying Chang
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Hôpital Hôtel-Dieu, Université de Montréal, Montréal, Québec, Canada
| | - Yun-Wen Chen
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Hôpital Hôtel-Dieu, Université de Montréal, Montréal, Québec, Canada
| | - Xin-Ping Zhao
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Hôpital Hôtel-Dieu, Université de Montréal, Montréal, Québec, Canada
| | - Isabelle Chenier
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Hôpital Hôtel-Dieu, Université de Montréal, Montréal, Québec, Canada
| | - Stella Tran
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Hôpital Hôtel-Dieu, Université de Montréal, Montréal, Québec, Canada
| | - Alexandre Sauvé
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Hôpital Hôtel-Dieu, Université de Montréal, Montréal, Québec, Canada
| | - Julie R. Ingelfinger
- Pediatric Nephrology Unit, Massachusetts General Hospital, and Harvard Medical School, Boston, Massachusetts
| | - Shao-Ling Zhang
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Hôpital Hôtel-Dieu, Université de Montréal, Montréal, Québec, Canada
- Corresponding author: Shao-Ling Zhang,
| |
Collapse
|
16
|
|
17
|
Lau GJ, Godin N, Maachi H, Lo CS, Wu SJ, Zhu JX, Brezniceanu ML, Chénier I, Fragasso-Marquis J, Lattouf JB, Ethier J, Filep JG, Ingelfinger JR, Nair V, Kretzler M, Cohen CD, Zhang SL, Chan JS. Bcl-2-modifying factor induces renal proximal tubular cell apoptosis in diabetic mice. Diabetes 2012; 61:474-84. [PMID: 22210314 PMCID: PMC3266424 DOI: 10.2337/db11-0141] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
This study investigated the mechanisms underlying tubular apoptosis in diabetes by identifying proapoptotic genes that are differentially upregulated by reactive oxygen species in renal proximal tubular cells (RPTCs) in models of diabetes. Total RNAs isolated from renal proximal tubules (RPTs) of 20-week-old heterozygous db/m+, db/db, and db/db catalase (CAT)-transgenic (Tg) mice were used for DNA chip microarray analysis. Real-time quantitative PCR assays, immunohistochemistry, and mice rendered diabetic with streptozotocin were used to validate the proapoptotic gene expression in RPTs. Cultured rat RPTCs were used to confirm the apoptotic activity and regulation of proapoptotic gene expression. Additionally, studies in kidney tissues from patients with and without diabetes were used to confirm enhanced proapoptotic gene expression in RPTs. Bcl-2-modifying factor (Bmf) was differentially upregulated (P<0.01) in RPTs of db/db mice compared with db/m+ and db/db CAT-Tg mice and in RPTs of streptozotocin-induced diabetic mice in which insulin reversed this finding. In vitro, Bmf cDNA overexpression in rat RPTCs coimmunoprecipated with Bcl-2, enhanced caspase-3 activity, and promoted apoptosis. High glucose (25 mmol/L) induced Bmf mRNA expression in RPTCs, whereas rotenone, catalase, diphenylene iodinium, and apocynin decreased it. Knockdown of Bmf with small interfering RNA reduced high glucose-induced apoptosis in RPTCs. More important, enhanced Bmf expression was detected in RPTs of kidneys from patients with diabetes. These data demonstrate differential upregulation of Bmf in diabetic RPTs and suggest a potential role for Bmf in regulating RPTC apoptosis and tubular atrophy in diabetes.
Collapse
Affiliation(s)
- Garnet J. Lau
- Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Hôtel-Dieu Hospital, Université de Montréal, Montreal, Quebec, Canada
| | - Nicolas Godin
- Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Hôtel-Dieu Hospital, Université de Montréal, Montreal, Quebec, Canada
| | - Hasna Maachi
- Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Hôtel-Dieu Hospital, Université de Montréal, Montreal, Quebec, Canada
| | - Chao-Sheng Lo
- Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Hôtel-Dieu Hospital, Université de Montréal, Montreal, Quebec, Canada
| | - Shyh-Jong Wu
- Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Hôtel-Dieu Hospital, Université de Montréal, Montreal, Quebec, Canada
| | - Jian-Xin Zhu
- Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Hôtel-Dieu Hospital, Université de Montréal, Montreal, Quebec, Canada
| | - Marie-Luise Brezniceanu
- Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Hôtel-Dieu Hospital, Université de Montréal, Montreal, Quebec, Canada
| | - Isabelle Chénier
- Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Hôtel-Dieu Hospital, Université de Montréal, Montreal, Quebec, Canada
| | - Joelle Fragasso-Marquis
- Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Hôtel-Dieu Hospital, Université de Montréal, Montreal, Quebec, Canada
| | - Jean-Baptiste Lattouf
- Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Hôtel-Dieu Hospital, Université de Montréal, Montreal, Quebec, Canada
| | - Jean Ethier
- Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Hôtel-Dieu Hospital, Université de Montréal, Montreal, Quebec, Canada
| | - Janos G. Filep
- Research Centre, Maisonneuve-Rosemont Hospital, Montreal, Quebec, Canada
| | - Julie R. Ingelfinger
- Pediatric Nephrology Unit, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Viji Nair
- Nephrology/Internal Medicine, Center for Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | - Matthias Kretzler
- Nephrology/Internal Medicine, Center for Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | - Clemens D. Cohen
- Division of Nephrology, Institute of Physiology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Shao-Ling Zhang
- Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Hôtel-Dieu Hospital, Université de Montréal, Montreal, Quebec, Canada
| | - John S.D. Chan
- Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Hôtel-Dieu Hospital, Université de Montréal, Montreal, Quebec, Canada
- Corresponding author: John S.D. Chan,
| |
Collapse
|
18
|
Grenz A, Bauerle JD, Dalton JH, Ridyard D, Badulak A, Tak E, McNamee EN, Clambey E, Moldovan R, Reyes G, Klawitter J, Ambler K, Magee K, Christians U, Brodsky KS, Ravid K, Choi DS, Wen J, Lukashev D, Blackburn MR, Osswald H, Coe IR, Nürnberg B, Haase VH, Xia Y, Sitkovsky M, Eltzschig HK. Equilibrative nucleoside transporter 1 (ENT1) regulates postischemic blood flow during acute kidney injury in mice. J Clin Invest 2012; 122:693-710. [PMID: 22269324 DOI: 10.1172/jci60214] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Accepted: 12/07/2011] [Indexed: 01/09/2023] Open
Abstract
A complex biologic network regulates kidney perfusion under physiologic conditions. This system is profoundly perturbed following renal ischemia, a leading cause of acute kidney injury (AKI) - a life-threatening condition that frequently complicates the care of hospitalized patients. Therapeutic approaches to prevent and treat AKI are extremely limited. Better understanding of the molecular pathways promoting postischemic reflow could provide new candidate targets for AKI therapeutics. Due to its role in adapting tissues to hypoxia, we hypothesized that extracellular adenosine has a regulatory function in the postischemic control of renal perfusion. Consistent with the notion that equilibrative nucleoside transporters (ENTs) terminate adenosine signaling, we observed that pharmacologic ENT inhibition in mice elevated renal adenosine levels and dampened AKI. Deletion of the ENTs resulted in selective protection in Ent1-/- mice. Comprehensive examination of adenosine receptor-knockout mice exposed to AKI demonstrated that renal protection by ENT inhibitors involves the A2B adenosine receptor. Indeed, crosstalk between renal Ent1 and Adora2b expressed on vascular endothelia effectively prevented a postischemic no-reflow phenomenon. These studies identify ENT1 and adenosine receptors as key to the process of reestablishing renal perfusion following ischemic AKI. If translatable from mice to humans, these data have important therapeutic implications.
Collapse
Affiliation(s)
- Almut Grenz
- Mucosal Inflammation Program, Department of Anesthesiology, School of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Angiotensin II type II receptor deficiency accelerates the development of nephropathy in type I diabetes via oxidative stress and ACE2. EXPERIMENTAL DIABETES RESEARCH 2011; 2011:521076. [PMID: 22110472 PMCID: PMC3205615 DOI: 10.1155/2011/521076] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Revised: 07/11/2011] [Accepted: 07/20/2011] [Indexed: 01/06/2023]
Abstract
Since the functional role(s) of angiotensin II (Ang II) type II receptor (AT2R) in type I diabetes is unknown, we hypothesized that AT2R is involved in decreasing the effects of type I diabetes on the kidneys. We induced diabetes with low-dose streptozotocin (STZ) in both AT2R knockout (AT2RKO) and wild-type (WT) male mice aged 12 weeks and followed them for 4 weeks. Three subgroups nondiabetic, diabetic, and insulin-treated diabetic (Rx insulin implant) were studied. Systolic blood pressure (SBP), physiological parameters, glomerular filtration rate (GFR), renal morphology, gene expression, and apoptosis were assessed. After 4 weeks of diabetes, compared to WT controls, AT2RKO mice clearly developed features of early diabetic nephropathy (DN), such as renal hypertrophy, tubular apoptosis, and progressive extracellular matrix (ECM) protein accumulation as well as increased GFR. AT2RKO mice presented hypertension unaffected by diabetes. Renal oxidative stress (measured as heme oxygenase 1 (HO-1) gene expression and reactive oxygen species (ROS) generation) and intrarenal renin angiotensin system components, such as angiotensinogen (Agt), AT1R, and angiotensin-converting enzyme (ACE) gene expression, were augmented whereas angiotensin-converting enzyme2 (ACE2) gene expression was decreased in renal proximal tubules (RPTs) of AT2RKO mice. The renal changes noted above were significantly enhanced in diabetic AT2RKO mice but partially attenuated in insulin-treated diabetic WT and AT2RKO mice. In conclusion, AT2R deficiency accelerates the development of DN, which appears to be mediated, at least in part, via heightened oxidative stress and ACE/ACE2 ratio in RPTs.
Collapse
|
20
|
|
21
|
|
22
|
Chen YW, Chenier I, Tran S, Scotcher M, Chang SY, Zhang SL. Maternal diabetes programs hypertension and kidney injury in offspring. Pediatr Nephrol 2010; 25:1319-29. [PMID: 20422227 DOI: 10.1007/s00467-010-1506-1] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2009] [Revised: 03/01/2010] [Accepted: 03/10/2010] [Indexed: 01/13/2023]
Abstract
We investigated whether maternal diabetes programs the offspring to develop hypertension and kidney injury in adulthood and examined potential underlying mechanisms. In a murine model we studied the offspring of three groups of dams (non-diabetic, diabetic, and diabetic treated with insulin). Mean systolic blood pressure in the offspring was monitored from 8 to 20 weeks. Body and kidney weights in the offspring of diabetic mothers were significantly lower than in offspring of non-diabetic mothers. Offspring of diabetic mothers developed hypertension, microalbuminuria, and glucose intolerance. Increased accumulation of extracellular matrix proteins in the glomeruli and marked upregulation of angiotensinogen, angiotensin II type 1 receptor, angiotensin-converting enzyme, transforming growth factor beta-1 (TGF-beta1), and plasminogen activator inhibitor-1 (PAI-1) gene expression were evident in the renal cortex of hypertensive offspring of diabetic mothers. By contrast, angiotensin-converting enzyme-2 (ACE2) gene expression was lower in the hypertensive offspring of diabetic mothers than in that of non-diabetic mothers. These changes were prevented in the offspring of insulin-treated diabetic mothers. These data indicate that maternal diabetes induces perinatal programming of hypertension, renal injury, and glucose intolerance in the offspring and suggest a central role for the activation of the intrarenal renin-angiotensin system and TGF-beta1 gene expression in this process.
Collapse
Affiliation(s)
- Yun-Wen Chen
- Department of Medicine, Université de Montréal and Research Centre, Centre hospitalier de l'Université de Montréal (CRCHUM)-Hôtel-Dieu, 8-227, Pavillon Masson, 3850 Saint Urbain Street, Montreal, QC, H2W 1T7, Canada
| | | | | | | | | | | |
Collapse
|
23
|
Brezniceanu ML, Lau CJ, Godin N, Chénier I, Duclos A, Ethier J, Filep JG, Ingelfinger JR, Zhang SL, Chan JSD. Reactive oxygen species promote caspase-12 expression and tubular apoptosis in diabetic nephropathy. J Am Soc Nephrol 2010; 21:943-54. [PMID: 20299359 DOI: 10.1681/asn.2009030242] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Apoptosis of tubular epithelial cells contributes to the tubular atrophy that accompanies diabetic nephropathy. Reactive oxygen species (ROS) promote tubular apoptosis, but the mechanisms by which this occurs are incompletely understood. Here, we sought proapoptotic genes that ROS differentially upregulate in renal proximal tubular cells of diabetic (db/db) mice. We performed microarray analysis using total RNA from freshly isolated renal proximal tubules of nondiabetic, diabetic, and diabetic transgenic mice overexpressing catalase in the proximal tubule (thereby attenuating ROS). We observed greater expression of caspase-12 in the proximal tubules of the diabetic mice compared with the nondiabetic and diabetic transgenic mice. Quantitative PCR and immunohistochemistry confirmed the enhanced expression of caspase-12, as well as members of the endoplasmic reticulum stress-induced apoptotic pathway. Ex vivo, albumin induced caspase-12 activity and expression (protein and mRNA) and mRNA expression of the CCAT/enhancer-binding protein homologous protein in freshly isolated wild-type proximal tubules but not in catalase-overexpressing proximal tubules. In vitro, albumin stimulated activity of both caspase-12 and caspase-3 as well as expression of caspase-12 and CCAT/enhancer-binding protein homologous protein in a human proximal tubule cell line (HK-2). The free radical scavenger tiron inhibited these effects. Furthermore, knockdown of caspase-12 with small interfering RNA reduced albumin-induced apoptosis in HK-2 cells. Taken together, these studies demonstrate that albuminuria may induce tubular apoptosis through generation of ROS and the subsequent expression and activation of endoplasmic reticulum stress genes in the diabetic kidney.
Collapse
Affiliation(s)
- Marie-Luise Brezniceanu
- Université de Montréal, Centre de recherche du Centre hospitalier de l'Université de Montréal, Hôtel-Dieu Hospital, Pavillon Masson, 3850 Saint Urbain Street, Montréal, Québec, Canada H2W 1T8
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Abstract
Exposing rodents to brief episodes of hypoxia mimics the hypoxemia and the cardiovascular and metabolic effects observed in patients with obstructive sleep apnoea (OSA), a condition that affects between 5% and 20% of the population. Apart from daytime sleepiness, OSA is associated with a high incidence of systemic and pulmonary hypertension, peripheral vascular disease, stroke and sudden cardiac death. The development of animal models to study sleep apnoea has provided convincing evidence that recurrent exposure to intermittent hypoxia (IH) has significant vascular and haemodynamic impact that explain much of the cardiovascular morbidity and mortality observed in patients with sleep apnoea. However, the molecular and cellular mechanisms of how IH causes these changes is unclear and under investigation. This review focuses on the most recent findings addressing these mechanisms. It includes a discussion of the contribution of the nervous system, circulating and vascular factors, inflammatory mediators and transcription factors to IH-induced cardiovascular disease. It also highlights the importance of reactive oxygen species as a primary mediator of the systemic and pulmonary hypertension that develops in response to exposure to IH.
Collapse
Affiliation(s)
- Laura V González Bosc
- Vascular Physiology Group, Department of Cell Biology and Physiology, School of Medicine, University of New Mexico, Albuquerque, NM, USA.
| | | | | | | |
Collapse
|
25
|
Uchida Y, Miyajima A, Kikuchi E, Kozakai N, Kosaka T, Ieda M, Fukuda K, Ohigashi T, Oya M. Renal Damage Inhibited in Mice Lacking Angiotensinogen Gene Subjected to Unilateral Ureteral Obstruction. Urology 2009; 74:938-43. [DOI: 10.1016/j.urology.2009.02.059] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2008] [Revised: 01/24/2009] [Accepted: 02/26/2009] [Indexed: 11/28/2022]
|
26
|
Suthanthiran M, Gerber LM, Schwartz JE, Sharma VK, Medeiros M, Marion R, Pickering TG, August P. Circulating transforming growth factor-beta1 levels and the risk for kidney disease in African Americans. Kidney Int 2009; 76:72-80. [PMID: 19279557 PMCID: PMC3883576 DOI: 10.1038/ki.2009.66] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Transforming growth factor-β1 (TGF-β1) is well known to induce progression of experimental renal disease. Here we determined whether there is an association between serum levels of TGF-β1 and the risk factors for progression of clinically relevant renal disorders in 186 black and 147 white adults none of whom had kidney disease or diabetes. Serum TGF-β1 protein levels were positively and significantly associated with plasma renin activity along with the systolic and diastolic blood pressure in blacks but not whites after controlling for age, gender and body mass index. These TGF-β1 protein levels were also significantly associated with body mass index and metabolic syndrome and more predictive of microalbuminuria in blacks than in whites. The differential association between TGF-β1 and renal disease risk factors in blacks and whites suggests an explanation for the excess burden of end-stage renal disease in the black population but this requires validation in an independent cohort. Whether these findings show that it is the circulating levels of TGF-β1 that contributes to renal disease progression or reflects other unmeasured factors will need to be tested in longitudinal studies.
Collapse
Affiliation(s)
- Manikkam Suthanthiran
- Department of Medicine, Weill Cornell Medical College, New York-Presbyterian Hospital, New York, NY 10065, USA
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Rosón MI, Cao G, Della Penna S, Gorzalczany S, Pandolfo M, Toblli JE, Fernández BE. Angiotensin II increases intrarenal transforming growth factor-beta1 in rats submitted to sodium overload independently of blood pressure. Hypertens Res 2008; 31:707-15. [PMID: 18633183 DOI: 10.1291/hypres.31.707] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Angiotensin II (Ang II) promotes sodium-retention, cell growth and fibrosis in addition to its classical effects on blood pressure and fluid homeostasis. In this study we examined whether low and non-hypertensive doses of exogenous Ang II could enhance the intrarenal expression of transforming growth factor-beta1 (TGF-beta1) observed in rats submitted to sodium overload. Sprague-Dawley-rats were infused for 2 h with 0.1 and 5 microg kg(-1) h(-1) Ang II (Ang 0.1 and Ang 5, respectively) together with saline solution at four different concentrations (isotonic and Na 0.5 mol L(-1), Na 1.0 mol L(-1) and Na 1.5 mol L(-1)). Renal function and mean arterial blood pressure (BP) were measured. The renal distributions of TGF-beta1, alpha-smooth-muscle-actin (alpha-SMA) and nuclear factor-kappaB (NF-kappaB) were evaluated by immunohistochemistry. While the Ang 0.1 groups were normotensive, the Ang 5 groups developed arterial hypertension progressively, and the highest blood pressure values were observed when rats were simultaneously infused with Na 1.5 mol L(-1). Glomerular function was not altered in any group. In cortical tubules, all groups infused with Ang II (0.1 and 5) and hypertonic saline solution (HSS) showed an increase in TGF-beta1 immunostaining compared to those infused with HSS alone. In medullary tubules, only the Ang 5-Na 0.5 group showed a significant increase in TGF-beta 1 immunostaining compared to the Na 0.5 group. Peritubular positive staining for alpha-SMA was present in groups receiving Ang alone or Ang-Na, in a sodium concentration-dependent manner. In cortical-tubules, NF-kappaB immunostaining was significantly increased in the Ang groups in comparison with the control and in Ang-Na 0.5 and Ang-Na 1.0 groups in comparison with the Na 0.5 mol L(-1) and Na 1.5 mol L(-1) groups, respectively, except in the case of the Ang 0.1-Na 1.5 mol L(-1) and Ang 5-Na 1.5 mol L(-1) groups. Moreover, Ang II and sodium overload induced additional changes in TGF-beta1, alpha-SMA and NF-kappaB immunostanding in glomeruli, medullary tubules and renal vessels. In conclusion, the interaction of Ang II with acute-sodium overload exacerbated intrarenal TGF-beta1, alpha-SMA and NF-kappaB expression, independently from changes in blood pressure levels, in normal rats.
Collapse
Affiliation(s)
- María I Rosón
- Department of Pathophysiology, School of Pharmacy and Biochemistry, University of Buenos Aires, Buenos Aires, Argentina.
| | | | | | | | | | | | | |
Collapse
|
28
|
Walsh MF, Ampasala DR, Hatfield J, Vander Heide R, Suer S, Rishi AK, Basson MD. Transforming growth factor-beta stimulates intestinal epithelial focal adhesion kinase synthesis via Smad- and p38-dependent mechanisms. THE AMERICAN JOURNAL OF PATHOLOGY 2008; 173:385-399. [PMID: 18583311 PMCID: PMC2475776 DOI: 10.2353/ajpath.2008.070729] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/24/2008] [Indexed: 01/18/2023]
Abstract
Focal adhesion kinase (FAK) regulates cell migration, proliferation, and apoptosis. FAK protein is reduced at the edge of migrating gut epithelial sheets in vitro, but it has not been characterized in restitutive gut mucosa in vivo. Here we show that FAK and activated phospho-FAK (FAK(397)) immunoreactivity was lower in epithelial cells immediately adjacent to human gastric and colonic ulcers in vivo, but dramatically increased in epithelia near the ulcers, possibly reflecting stimulation by growth factors absent in vitro. Transforming growth factor (TGF)-beta, but not fibroblast growth factor, platelet-derived growth factor, or vascular endothelial growth factor, increased FAK levels in Caco-2 and IEC-6 cells. Epithelial immunoreactivity to TGF-beta and phospho-Smad3 was also higher near the ulcers, varying in parallel with FAK. The TGF-beta receptor antagonist SB431542 completely blocked TGF-beta-induced Smad2/3 and p38 activation in IEC-6 cells. SB431542, the p38 antagonist SB203580, and siRNA-mediated reduction of Smad2 and p38alpha prevented TGF-beta stimulation of both FAK transcription and translation (as measured via a FAK promoter-luciferase construct). FAK(397) levels were directly related to total FAK protein expression. Although gut epithelial motility is associated with direct inhibition of FAK protein adjacent to mucosal wounds, TGF-beta may increase FAK protein near but not bordering mucosal ulcers via Smad2/3 and p38 signals. Our results show that regulation of FAK expression may be as important as FAK phosphorylation in critically influencing gut epithelial cell migration after mucosal injury.
Collapse
Affiliation(s)
- Mary F Walsh
- Departments of Surgery and Pathology, John D. Dingell VA Medical Center, Wayne State University, 4646 John R Detroit, MI 48201-1932, USA
| | | | | | | | | | | | | |
Collapse
|
29
|
Shin GT, Kim DR, Lim JE, Yim H, Kim H. Upregulation and function of GADD45gamma in unilateral ureteral obstruction. Kidney Int 2008; 73:1251-65. [PMID: 18354378 DOI: 10.1038/ki.2008.93] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
We performed differential display analysis to determine transcriptional activity in the rat kidney, following unilateral ureteral obstruction and found a 12-fold increase in the expression of Growth Arrest and DNA Damage-45gamma (GADD45gamma), a stress-responsive molecule that interacts with cell-cycle proteins. GADD45gamma was strongly expressed in as little as 6 h following ureteric obstruction in the renal tubules, and was also found in kidney tissue of patients with chronic glomerulonephritis. Adenovirus-mediated expression of GADD45gamma in cultured renal tubular cells activated p38 along with a significant upregulation of C-C and C-X3-C chemokine ligands and fibrosis-related factors such as several matrix metalloproteinases, transforming growth factor-beta1, decorin, and bone morphogenetic protein 2. Silencing of GADD45gamma expression significantly blunted the upregulation of these inflammatory and fibrogenic mediators and monocyte infiltration in the ureteral obstructed rat kidney. Our study shows that GADD45gamma is quickly upregulated in the kidney with an obstructed ureter, enhancing the production of factors regulating the pathogenesis of kidney disease.
Collapse
Affiliation(s)
- G-T Shin
- Department of Nephrology, Ajou University School of Medicine, Suwon, Republic of Korea.
| | | | | | | | | |
Collapse
|
30
|
Do vasculature reactive oxygen species play a role in the mobilization of bone marrow endothelial progenitor cells? J Hypertens 2008; 26:188-90. [PMID: 18192830 DOI: 10.1097/hjh.0b013e3282f2851a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
31
|
Tran S, Chen YW, Chenier I, Chan JSD, Quaggin S, Hébert MJ, Ingelfinger JR, Zhang SL. Maternal diabetes modulates renal morphogenesis in offspring. J Am Soc Nephrol 2008; 19:943-52. [PMID: 18305124 DOI: 10.1681/asn.2007080864] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Maternal diabetes leads to an adverse in utero environment, but whether maternal diabetes impairs nephrogenesis is unknown. Diabetes was induced with streptozotocin in pregnant Hoxb7-green fluorescence protein mice at embryonic day 13, and the offspring were examined at several time points after birth. Compared with offspring of nondiabetic controls, offspring of diabetic mice had lower body weight, body size, kidney weight, and nephron number. The observed renal dysmorphogenesis may be the result of increased apoptosis, because immunohistochemical analysis revealed significantly more apoptotic podocytes as well as increased active caspase-3 immunostaining in the renal tubules compared with control mice. Regarding potential mediators of these differences, offspring of diabetic mice had increased expression of intrarenal angiotensinogen and renin mRNA, upregulation of NF-kappaB isoforms p50 and p65, and activation of the NF-kappaB pathway. In conclusion, maternal diabetes impairs nephrogenesis, possibly via enhanced intrarenal activation of the renin-angiotensin system and NF-kappaB signaling.
Collapse
Affiliation(s)
- Stella Tran
- University of Montreal, Centre Hospitalier de l'Université de Montréal-Hôtel-Dieu, Research Centre, Montreal, Quebec, Canada
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Brezniceanu ML, Liu F, Wei CC, Chénier I, Godin N, Zhang SL, Filep JG, Ingelfinger JR, Chan JSD. Attenuation of interstitial fibrosis and tubular apoptosis in db/db transgenic mice overexpressing catalase in renal proximal tubular cells. Diabetes 2008; 57:451-9. [PMID: 17977949 DOI: 10.2337/db07-0013] [Citation(s) in RCA: 138] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
OBJECTIVE The present study investigated the relationships between reactive oxygen species (ROS), interstitial fibrosis, and renal proximal tubular cell (RPTC) apoptosis in type 2 diabetic db/db mice and in db/db transgenic (Tg) mice overexpressing rat catalase (rCAT) in their RPTCs (db/db rCAT-Tg). RESEARCH DESIGN AND METHODS Blood pressure, blood glucose, and albuminuria were monitored for up to 5 months. Kidneys were processed for histology and apoptosis studies (terminal transferase-mediated dUTP nick-end labeling or immunostaining for active caspase-3 and Bax). Real-time quantitative PCR assays were used to quantify angiotensinogen (ANG), p53, and Bax mRNA levels. RESULTS db/db mice developed obesity, hyperglycemia, hypertension, and albuminuria. In contrast, db/db rCAT-Tg mice became obese and hyperglycemic but had normal blood pressure and attenuated albuminuria compared with db/db mice. Kidneys from db/db mice displayed progressive glomerular hypertrophy, glomerulosclerosis, interstitial fibrosis, and tubular apoptosis and increased expression of collagen type IV, Bax, and active caspase-3, as well as increased ROS production. These changes, except glomerular hypertrophy, were markedly attenuated in kidneys of db/db rCAT-Tg mice. Furthermore, ANG, p53, and Bax mRNA expression was increased in renal proximal tubules of db/db mice but not of db/db rCAT-Tg mice. CONCLUSIONS Our results indicate a crucial role for intra-renal ROS in the progression of hypertension, albuminuria, interstitial fibrosis, and tubular apoptosis in type 2 diabetes and demonstrate the beneficial effects of suppressing ROS formation.
Collapse
Affiliation(s)
- Marie-Luise Brezniceanu
- Université de Montréal, Centre hospitalier de l'Université de Montréal (CHUM) Hôtel-Dieu, Research Centre, Pavillon Masson, 3850 Saint Urbain St., Montreal, Quebec, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Liu F, Brezniceanu ML, Wei CC, Chénier I, Sachetelli S, Zhang SL, Filep JG, Ingelfinger JR, Chan JSD. Overexpression of angiotensinogen increases tubular apoptosis in diabetes. J Am Soc Nephrol 2007; 19:269-80. [PMID: 18057217 DOI: 10.1681/asn.2007010074] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The intrarenal renin-angiotensin system (RAS) plays an important role in the progression of diabetic nephropathy. We have previously reported that mice overexpressing angiotensinogen in renal proximal tubular cells (RPTC) develop hypertension, albuminuria, and renal injury. Here, we investigated whether activation of the intrarenal RAS contributes to apoptosis of RPTC in diabetes. Induction of diabetes with streptozotocin in these transgenic mice led to significant increases in BP, albuminuria, RPTC apoptosis, and proapoptotic gene expression compared with diabetic nontransgenic littermates. Insulin and/or RAS blockers markedly attenuated these changes. Hydralazine prevented hypertension but not albuminuria, RPTC apoptosis, or proapoptotic gene expression. In vitro, high-glucose medium significantly increased apoptosis and caspase-3 activity in rat immortalized RPTC overexpressing angiotensinogen compared with control cells, and these changes were prevented by insulin and/or RAS blockers. In conclusion, intrarenal RAS activation and high glucose may act in concert to increase tubular apoptosis in diabetes, independent of systemic hypertension.
Collapse
Affiliation(s)
- Fang Liu
- Université de Montréal Centre hospitalier de l'Université de Montréal-Hôtel-Dieu, Research Centre Pavillon Masson, 3850 Saint Urbain Street, Montreal, Quebec, Canada H2W 1T8
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Lee YJ, Lee YJ, Han HJ. Regulatory mechanisms of Na(+)/glucose cotransporters in renal proximal tubule cells. Kidney Int 2007:S27-35. [PMID: 17653207 DOI: 10.1038/sj.ki.5002383] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Glucose is a key fuel and an important metabolic substrate in mammals. Renal proximal tubular cells (PTCs) not only reabsorb filtered glucose but are also believed to play a role in the glucotoxicity associated with renal pathogenesis, such as in diabetes. The proximal tubule environment is where 90% of the filtered glucose is reabsorbed by the low-affinity/high-capacity Na(+)/glucose cotransporter 2 (SGLT2) and facilitated diffusion glucose transporter 2 (GLUT2). Both active and facilitative glucose transporters have distinct distribution profiles along the proximal tubule related to their particular kinetic characteristics. A number of mechanisms contribute to the changes in the cellular functions, which occur in response to exposure to various endogenous factors. Hyperglycemia was reported to regulate the renal SGLT activities through the reactive oxygen species-nuclear factor-kappaB pathways, which suggests that the transcellular glucose uptake within the PTCs contribute to the development of diabetic-like nephropathy. Angiotensin II (ANG II) plays an important role in its development through epidermal growth factor receptor (EGFR) transactivation. Therefore, a combination of high glucose, ANG II, and EGF are involved in diabetic-like nephropathy by regulating the SGLT activity. In addition, endogenously enhanced SGLTs have a cytoprotective function. The renal proximal tubules play a major role in regulating the plasma glucose levels, and there is increasing interest in the renal glucose transporters on account of their potential implications in the treatment of various conditions including diabetes mellitus.
Collapse
Affiliation(s)
- Y J Lee
- Biotherapy Human Resources Center, College of Veterinary Medicine, Chonnam National University, Gwangju, Republic of Korea
| | | | | |
Collapse
|
35
|
Jiang T, Wang XX, Scherzer P, Wilson P, Tallman J, Takahashi H, Li J, Iwahashi M, Sutherland E, Arend L, Levi M. Farnesoid X receptor modulates renal lipid metabolism, fibrosis, and diabetic nephropathy. Diabetes 2007; 56:2485-93. [PMID: 17660268 DOI: 10.2337/db06-1642] [Citation(s) in RCA: 182] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Recent studies indicate an important role for nuclear receptors in regulating lipid and carbohydrate metabolism, fibrosis, and inflammation. Farnesoid X receptor (FXR) is a member of the nuclear hormone receptor superfamily. FXR is highly expressed in the liver, intestine, adrenal gland, and kidney. The primary bile acids are the highest affinity endogenous ligands for FXR. The effects of FXR agonists in diabetic kidney disease, the main cause of end-stage renal disease, however, have not been determined. RESEARCH DESIGN AND METHODS To identify the effect of FXR activation in modulation of diabetic nephropathy, we treated 1) C57BL/6J mice on low-fat diet or high-fat diet with FXR agonists (GW4064 or cholic acid) for 1 week; 2) C57BLKS/J-db/db mice and their lean mates with GW4064 for 1 week; and 3) C57BL/6J-db/db mice and their lean mates with cholic acid for 12 weeks. RESULTS We found that FXR agonists modulate renal sterol regulatory element-binding protein-1 (SREBP-1) expression and lipid metabolism and renal expression of profibrotic growth factors, proinflammatory cytokines, and oxidative stress enzymes and decrease glomerulosclerosis, tubulointerstitial fibrosis, and proteinuria. In renal mesangial cells, overexpression of FXR or treatment with GW4064 also inhibited SREBP-1c and other lipogenic genes, transforming growth factor-beta, and interleukin-6, suggesting a direct role of FXR in modulating renal lipid metabolism and modulation of fibrosis and inflammation. CONCLUSIONS These results therefore indicate a new and important role for FXR in the kidney and provide new therapeutic avenues for the treatment of diabetic nephropathy.
Collapse
Affiliation(s)
- Tao Jiang
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado Health Sciences Center, 4200 East 9th Ave., Denver, CO 80262, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Martin MM, Buckenberger JA, Jiang J, Malana GE, Knoell DL, Feldman DS, Elton TS. TGF-beta1 stimulates human AT1 receptor expression in lung fibroblasts by cross talk between the Smad, p38 MAPK, JNK, and PI3K signaling pathways. Am J Physiol Lung Cell Mol Physiol 2007; 293:L790-9. [PMID: 17601799 PMCID: PMC2413071 DOI: 10.1152/ajplung.00099.2007] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Both angiotensin II (ANG II) and transforming growth factor-beta1 (TGF-beta1) are thought to be involved in mediating pulmonary fibrosis. Interactions between the renin-angiotensin system (RAS) and TGF-beta1 have been well documented, with most studies describing the effect of ANG II on TGF-beta1 expression. However, recent gene expression profiling experiments demonstrated that the angiotensin II type 1 receptor (AT(1)R) gene was a novel TGF-beta1 target in human adult lung fibroblasts. In this report, we show that TGF-beta1 augments human AT(1)R (hAT(1)R) steady-state mRNA and protein levels in a dose- and time-dependent manner in primary human fetal pulmonary fibroblasts (hPFBs). Nuclear run-on experiments demonstrate that TGF-beta1 transcriptionally activates the hAT(1)R gene and does not influence hAT(1)R mRNA stability. Pharmacological inhibitors and specific siRNA knockdown experiments demonstrate that the TGF-beta1 type 1 receptor (TbetaRI/ALK5), Smad2/3, and Smad4 are essential for TGF-beta1-stimulated hAT(1)R expression. Additional pharmacological inhibitor and small interference RNA experiments also demonstrated that p38 MAPK, JNK, and phosphatidylinositol 3-kinase (PI3K) signaling pathways are also involved in the TGF-beta1-stimulated increase in hAT(1)R density. Together, our results suggest an important role for cross talk among Smad, p38 MAPK, JNK, and PI3K pathways in mediating the augmented expression of hAT(1)R following TGF-beta1 treatment in hPFB. This study supports the hypothesis that a self-potentiating loop exists between the RAS and the TGF-beta1 signaling pathways and suggests that ANG II and TGF-beta1 may cooperate in the pathogenesis of pulmonary fibrosis. The synergy between these systems may require that both pathways be simultaneously inhibited to treat fibrotic lung disease.
Collapse
MESH Headings
- Cells, Cultured
- Dose-Response Relationship, Drug
- Enzyme Activation/drug effects
- Extracellular Signal-Regulated MAP Kinases/metabolism
- Fetus/cytology
- Fetus/drug effects
- Fetus/metabolism
- Fibroblasts/drug effects
- Fibroblasts/enzymology
- Humans
- JNK Mitogen-Activated Protein Kinases/metabolism
- Lung/cytology
- Lung/drug effects
- Lung/enzymology
- MAP Kinase Signaling System/drug effects
- Models, Biological
- Phosphatidylinositol 3-Kinases/metabolism
- Protein Serine-Threonine Kinases/metabolism
- RNA Stability/drug effects
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Small Interfering/metabolism
- Receptor, Angiotensin, Type 1/genetics
- Receptor, Angiotensin, Type 1/metabolism
- Receptor, Transforming Growth Factor-beta Type I
- Receptors, Transforming Growth Factor beta/metabolism
- Smad Proteins/metabolism
- Transcription, Genetic/drug effects
- Transforming Growth Factor beta1/pharmacology
- Up-Regulation/drug effects
- p38 Mitogen-Activated Protein Kinases/metabolism
Collapse
Affiliation(s)
- Mickey M Martin
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio 43210, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Gagliardini E, Benigni A. Therapeutic potential of TGF-beta inhibition in chronic renal failure. Expert Opin Biol Ther 2007; 7:293-304. [PMID: 17309322 DOI: 10.1517/14712598.7.3.293] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Chronic kidney diseases are emerging as a worldwide public health problem. The progression of kidney diseases closely correlates with the accumulation of extracellular matrix leading to glomerulosclerosis and tubulointerstitial injury. Transforming growth factor (TGF)-beta has been identified as a key mediator of kidney matrix accumulation. Overexpression of TGF-beta isoforms and their receptors was observed in a variety of renal diseases in both animals and humans. Given its crucial role in fibrotic kidney disease, TGF-beta has been recently considered as a possible target in the management of chronic renal diseases. This review discusses the role of TGF-beta in renal fibrosis and provides an overview of the strategies that, when interfering with TGF-beta expression and signalling, could be employed as new renoprotective treatments.
Collapse
Affiliation(s)
- Elena Gagliardini
- Mario Negri Institute for Pharmacological Research, Via Gavazzeni 11, 24125 Bergamo, Italy
| | | |
Collapse
|
38
|
Zhang SL, Chen YW, Tran S, Chenier I, Hébert MJ, Ingelfinger JR. Reactive oxygen species in the presence of high glucose alter ureteric bud morphogenesis. J Am Soc Nephrol 2007; 18:2105-15. [PMID: 17538188 DOI: 10.1681/asn.2006101124] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Renal malformations are a major cause of childhood renal failure. During the development of the kidney, ureteric bud (UB) branching morphogenesis is critical for normal nephrogenesis. These studies investigated whether renal UB branching morphogenesis is altered by a high ambient glucose environment and studied underlying mechanism(s). Kidney explants that were isolated from different periods of gestation (embryonic days 12 to 18) from Hoxb7-green fluorescence protein mice were cultured for 24 h in either normal d-glucose (5 mM) or high d-glucose (25 mM) medium with or without various inhibitors. Alterations in renal morphogenesis were assessed by fluorescence microscopy. Paired-homeobox 2 (Pax-2) gene expression was determined by real-time quantitative PCR, Western blotting, and immunohistology. The results revealed that high d-glucose (25 mM) specifically stimulates UB branching morphogenesis via Pax-2 gene expression, whereas other glucose analogs, such as d-mannitol, l-glucose, and 2-deoxy-d-glucose, had no effect. The stimulatory effect of high glucose on UB branching was blocked in the presence of catalase and inhibitors of NADPH oxidase, mitochondrial electron transport chain complex I, and Akt signaling. Moreover, in in vivo studies, it seems that high glucose induces, via Pax-2 (mainly localized in UB), acceleration of UB branching but not nephron formation. Taken together, these data demonstrate that high glucose alters UB branching morphogenesis. This occurs, at least in part, via reactive oxygen species generation, activation of Akt signaling, and upregulation of Pax-2 gene expression.
Collapse
Affiliation(s)
- Shao-Ling Zhang
- University of Montreal, Centre hospitalier de l'Université de Montréal-Hôtel-Dieu, Pavillon Masson, Montreal, Quebec, Canada.
| | | | | | | | | | | |
Collapse
|
39
|
Brezniceanu ML, Liu F, Wei CC, Tran S, Sachetelli S, Zhang SL, Guo DF, Filep JG, Ingelfinger JR, Chan JSD. Catalase overexpression attenuates angiotensinogen expression and apoptosis in diabetic mice. Kidney Int 2007; 71:912-23. [PMID: 17342175 DOI: 10.1038/sj.ki.5002188] [Citation(s) in RCA: 135] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Increased generation of reactive oxygen species (ROS) leads to oxidative stress in diabetes. Catalase is a highly conserved heme-containing protein that reduces hydrogen peroxide to water and oxygen and is an important factor decreasing cellular injury owing to oxidative stress. Hyperglycemic conditions increase oxidative stress and angiotensinogen gene expression. Angiotensinogen conversion to angiotensin II leads to a furtherance in oxidative stress through increased generation of reactive oxygen species. In this study, we utilized mice transgenically overexpressing rat catalase in a kidney-specific manner to determine the impact on ROS, angiotensinogen and apoptotic gene expression in proximal tubule cells of diabetic animals. Proximal tubules isolated from wild-type and transgenic animals without or with streptozotocin-induced diabetes were incubated in low glucose media in the absence or presence of angiotensin II or in a high-glucose media. Our results show that the overexpression of catalase prevents the stimulation of ROS and angiotensinogen mRNA in tubules owing to elevated glucose or angiotensin II in vitro. Additionally, overexpression of catalase attenuated ROS generation, angiotensinogen and proapoptotic gene expression and apoptosis in the kidneys of diabetic mice in vivo. Our studies point to an important role of ROS in the pathophysiology of diabetic nephropathy.
Collapse
Affiliation(s)
- M-L Brezniceanu
- Research Centre, Centre hospitalier de l'Université de Montréal (CHUM)-Hôtel-Dieu, Montreal, Quebec, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Wolf G. Renal injury due to renin-angiotensin-aldosterone system activation of the transforming growth factor-beta pathway. Kidney Int 2006; 70:1914-9. [PMID: 16985515 DOI: 10.1038/sj.ki.5001846] [Citation(s) in RCA: 237] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Glomerulosclerosis, interstitial fibrosis, and tubular atrophy occur with end-stage kidney failure, irrespective of the primary etiology. The transforming growth factor-beta (TGF-beta) is a key factor in these alterations either directly, by stimulating synthesis of extracellular matrix components and reducing collagenase production, or indirectly through other profibrogenic factors such as connective tissue growth factor (CTGF). TGF-beta is important for the proliferation of intrarenal fibroblasts and the epithelial-mesenchymal transition through which tubular cells become fibroblasts. Although several factors induce TGF-beta expression in the kidney, one very interesting aspect is the link between the renin-angiotensin-aldosterone (Aldo) system (RAAS) and TGF-beta. Angiotensin II (ANG II) stimulates TGF-beta expression in the kidney by various mechanisms and upregulates receptors for TGF-beta. ANG II can directly phosphorylate Smads without inducing TGF-beta. Recent data provide compelling evidence that other components of the RAAS including ANG III, renin, and Aldo also activate the TGF-beta system. As direct modulation of the TGF-beta system is not yet feasible in humans, angiotensin-converting enzyme (ACE) inhibitors and angiotensin type 1 (AT1)-receptor blockers are currently the most potential drugs to interfere with this ANG II-mediated TGF-beta expression. This review highlights some current aspects of the interaction between the RAAS and the TGF-beta axis.
Collapse
Affiliation(s)
- G Wolf
- Klinik für Innere Medizin III, Klinikum der Friedrich-Schiller-Universität, Jena, Germany.
| |
Collapse
|