1
|
Nath KA, Juncos LA, Singh RD, Grande JP, Croatt AJ, Ackerman AW, Kanamori KS, Adams CM, Tchkonia T, Kirkland JL, Katusic ZS. The Occurrence of Senescence in the Arteriovenous Fistula in the Rat. KIDNEY360 2025; 6:27-37. [PMID: 39418108 PMCID: PMC11793188 DOI: 10.34067/kid.0000000605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 09/30/2024] [Indexed: 10/19/2024]
Abstract
Key Points The rat arteriovenous fistula (AVF) model exhibits marked upregulation of p16Ink4a and p21Cip1 and multiple markers of senescence. Fisetin, an established vasoprotective senolytic agent, when administered for 3 weeks, increases AVF blood flow and AVF outward remodeling. Heme is shown to be a novel prosenescence metabolite, and when chronically administered, it decreases AVF blood flow. Background Maturational failure of dialysis arteriovenous fistulas (AVFs) not uncommonly occurs and is of considerable and timely importance. Our prior studies demonstrate that senescence, a phenotypic process that promotes vascular and other diseases, occurs in the murine AVF. In this study, we examined whether senescence also occurs in the rat AVF model and the effect of compounds that inhibit or accelerate senescence. Methods The rat AVF was created in the femoral vessels by an end vein-side artery anastomosis. In the AVF, we assessed the expression of critical drivers of senescence, specifically, the cell cycle inhibitors p16Ink4a and p21Cip1, and such indices of a senescence phenotype as senescence-associated β -galactosidase (SA-β -gal) activity, SA-β -gal staining, and a senescence-associated secretory phenotype. We examined the effects of compounds that retard or accelerate senescence on AVF blood flow. Results The AVF evinced upregulation of p16Ink4a and p21Cip1 when assessed 3 days after AVF creation. The AVF also demonstrated increased SA-β -gal activity in the artery and vein; staining for SA-β -gal in the AVF artery, anastomosis, and vein; and a prominent senescence-associated secretory phenotype. Fisetin, an established senolytic that is protective in other models of vascular injury, when administered for 3 weeks, increased AVF blood flow and outward remodeling. Hemin, when administered for 3 weeks, decreased AVF blood flow. We demonstrate that hemin is a novel inducer of a senescence phenotype in endothelial cells, as reflected by several senescence indices. However, when administered relatively acutely (for 5 days), hemin increased AVF blood flow by heme oxygenase–dependent mechanisms because the latter was entirely prevented by a competitive inhibitor of heme oxygenase activity. Conclusions The rat AVF exhibits senescence within 3 days of its creation. Chronic administration of a senolytic compound (fisetin) increases AVF blood flow, whereas chronic administration of a prosenescence compound (hemin) decreases AVF blood flow.
Collapse
Affiliation(s)
- Karl A. Nath
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Luis A. Juncos
- University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Raman Deep Singh
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Joseph P. Grande
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Anthony J. Croatt
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Allan W. Ackerman
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Karina S. Kanamori
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Christopher M. Adams
- Division of Endocrinology, Diabetes, Metabolism and Nutrition, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Tamara Tchkonia
- Division of Endocrinology, Department of Medicine, Center for Advanced Gerotherapeutics, Cedars-Sinai Health Sciences Center, Los Angeles, California
| | - James L. Kirkland
- Division of Endocrinology, Department of Medicine, Center for Advanced Gerotherapeutics, Cedars-Sinai Health Sciences Center, Los Angeles, California
| | - Zvonimir S. Katusic
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
2
|
Saleem M, Aden LA, Mutchler AL, Basu C, Ertuglu LA, Sheng Q, Penner N, Hemnes AR, Park JH, Ishimwe JA, Laffer CL, Elijovich F, Wanjalla CN, de la Visitacion N, Kastner PD, Albritton CF, Ahmad T, Haynes AP, Yu J, Graber MK, Yasmin S, Wagner KU, Sayeski PP, Hatzopoulos AK, Gamazon ER, Bick AG, Kleyman TR, Kirabo A. Myeloid-Specific JAK2 Contributes to Inflammation and Salt Sensitivity of Blood Pressure. Circ Res 2024; 135:890-909. [PMID: 39263750 PMCID: PMC11466692 DOI: 10.1161/circresaha.124.323595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 08/23/2024] [Accepted: 08/28/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND Salt sensitivity of blood pressure (SSBP), characterized by acute changes in blood pressure with changes in dietary sodium intake, is an independent risk factor for cardiovascular disease and mortality in people with and without hypertension. We previously found that elevated sodium concentration activates antigen-presenting cells (APCs), resulting in high blood pressure, but the mechanisms are unknown. Here, we hypothesized that APC-specific JAK2 (Janus kinase 2) through STAT3 (signal transducer and activator of transcription 3) and SMAD3 (small mothers against decapentaplegic homolog 3) contributes to SSBP. METHODS We performed bulk or single-cell transcriptomic analyses following in vitro monocytes exposed to high salt and in vivo high sodium treatment in humans using a rigorous salt-loading/depletion protocol to phenotype SSBP. We also used a myeloid cell-specific CD11c+ JAK2 knockout mouse model and measured blood pressure with radiotelemetry after N-omega-nitro-L-arginine-methyl ester and a high salt diet treatment. We used flow cytometry for immunophenotyping and measuring cytokine levels. Fluorescence in situ hybridization and immunohistochemistry were performed to spatially visualize the kidney's immune cells and cytokine levels. Echocardiography was performed to assess cardiac function. RESULTS We found that high salt treatment upregulates gene expression of the JAK/STAT/SMAD pathway while downregulating inhibitors of this pathway, such as suppression of cytokine signaling and cytokine-inducible SH2, in human monocytes. Expression of the JAK2 pathway genes mirrored changes in blood pressure after salt loading and depletion in salt-sensitive but not salt-resistant humans. Ablation of JAK2, specifically in CD11c+ APCs, attenuated salt-induced hypertension in mice with SSBP. Mechanistically, we found that SMAD3 acted downstream of JAK2 and STAT3, leading to increased production of highly reactive isolevuglandins and proinflammatory cytokine IL (interleukin)-6 in renal APCs, which activate T cells and increase production of IL-17A, IL-6, and TNF-α (tumor necrosis factor-alpha). CONCLUSIONS Our findings reveal the APC JAK2 signaling pathway as a potential target for the diagnosis and treatment of SSBP in humans.
Collapse
Affiliation(s)
- Mohammad Saleem
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Luul A Aden
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Ashley L Mutchler
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Chitra Basu
- Department of Medicine, Division of Genetic Medicine (C.B., E.R.G.), Vanderbilt University Medical Center, Nashville, TN
- Department of Medicine, Division of Cardiovascular Medicine (C.B., A.K.H.), Vanderbilt University Medical Center, Nashville, TN
| | - Lale A Ertuglu
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Quanhu Sheng
- Department of Biostatistics (Q.S.), Vanderbilt University Medical Center, Nashville, TN
| | - Niki Penner
- Division of Allergy, Pulmonary, and Critical Care Medicine (N.P., A.R.H.)
| | - Anna R Hemnes
- Division of Allergy, Pulmonary, and Critical Care Medicine (N.P., A.R.H.)
| | - Jennifer H Park
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Jeanne A Ishimwe
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Cheryl L Laffer
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
| | | | - Celestine N Wanjalla
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Nestor de la Visitacion
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Paul D Kastner
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Claude F Albritton
- School of Graduate Studies, Meharry Medical College, Nashville, TN (C.F.A.)
| | - Taseer Ahmad
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
- Department of Pharmacology, College of Pharmacy, University of Sargodha, Punjab, Pakistan (T.A.)
| | - Alexandria P Haynes
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Justin Yu
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Meghan K Graber
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Sharia Yasmin
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Kay-Uwe Wagner
- Wayne State University, Department of Oncology and Tumor Biology Program, Barbara Ann Karmanos Cancer Institute, Detroit, MI (K.-U.W.)
| | - Peter P Sayeski
- Center for Integrative Cardiovascular and Metabolic Disease, University of Florida, Gainesville (P.P.S.)
| | - Antonis K Hatzopoulos
- Department of Medicine, Division of Cardiovascular Medicine (C.B., A.K.H.), Vanderbilt University Medical Center, Nashville, TN
| | - Eric R Gamazon
- Department of Medicine, Division of Genetic Medicine (C.B., E.R.G.), Vanderbilt University Medical Center, Nashville, TN
| | - Alexander G Bick
- Division of Genetic Medicine (A.G.B.), Vanderbilt University Medical Center, Nashville, TN
| | - Thomas R Kleyman
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, PA (T.R.K.)
| | - Annet Kirabo
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Center for Immunobiology (A.K.)
- Vanderbilt Institute for Infection, Immunology and Inflammation (A.K.)
- Vanderbilt Institute for Global Health, Vanderbilt University Medical Center, Nashville, TN (A.K.)
| |
Collapse
|
3
|
Abstract
Almost a hundred years have passed since obstruction of the renal artery has been recognized to raise blood pressure. By now chronic renovascular disease (RVD) due to renal artery stenosis is recognized as a major source of renovascular hypertension and renal disease. In some patients, RVD unaccompanied by noteworthy renal dysfunction or blood pressure elevation may be incidentally identified during peripheral angiography. Nevertheless, in others, RVD might present as a progressive disease associated with diffuse atherosclerosis, leading to loss of renal function, renovascular hypertension, hemodynamic compromise, and a magnified risk for cardiovascular morbidity and mortality. Atherosclerotic RVD leads to renal atrophy, inflammation, and hypoxia but represents a potentially treatable cause of chronic renal failure because until severe fibrosis sets in the ischemic kidney, it retains a robust potential for vascular and tubular regeneration. This remarkable recovery capacity of the kidney begs for early diagnosis and treatment. However, accumulating evidence from both animal studies and randomized clinical trials has convincingly established the inadequate efficacy of renal artery revascularization to fully restore renal function or blood pressure control and has illuminated the potential of therapies targeted to the ischemic renal parenchyma to instigate renal regeneration. Some of the injurious mechanisms identified as potential therapeutic targets included oxidative stress, microvascular disease, inflammation, mitochondrial injury, and cellular senescence. This review recapitulates the intrinsic mechanisms that orchestrate renal damage and recovery in RVD and can be harnessed to introduce remedial opportunities.
Collapse
Affiliation(s)
- Alfonso Eirin
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Alejandro R. Chade
- Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, MO
| | - Lilach O. Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
4
|
Pizzo TRF, Valverde AP, Orzari LE, Terciotti LG, de Lima RD, Costa do Bomfim FR, Esquisatto MAM, de Andrade TAM, Corezola do Amaral ME, de Oliveira CA, Felonato M. Caloric restriction improves inflammation in different tissues of the Wistar rats with obesity and 2K1C renovascular hypertension. Can J Physiol Pharmacol 2023; 101:661-671. [PMID: 37746936 DOI: 10.1139/cjpp-2022-0452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Renovascular hypertension (RHV) is the cause of high blood pressure due to left renal ischemia, and obesity and hypertension cause an inflammatory response. This work analyzed the inflammatory and tissue repair profile in renal, hepatic, and cardiac tissues in an animal model of RVH associated with a high-fat diet and caloric restriction. The expressions of RORγ-t, IL-17, T-bet, and TNF-α decreased and IFN-γ increased in the right kidney. In relation to the left kidney, caloric restriction decreased the expression of IFN-γ. In the liver, caloric restriction decreased RORγ-t, IL-17, and T-bet. Hypertension associated with obesity decreased the expression of IFN-γ, while caloric restriction increased. In the right kidney, hypertension and obesity, associated or not with caloric restriction, increased the area of collagen fibers. In the heart and liver, caloric restriction reduced the area of collagen fibers. Caloric restriction increased vascular endothelial growth factor, reduced levels of growth transformation factor-β1 (TGF-β), and increased collagen I in the left kidney. Hypertension/obesity, submitted or not having caloric restriction, increased TGF-β in liver. The results suggest that caloric restriction has beneficial effects in lowering blood pressure and regulating tissue proinflammatory cytokines. However, there was no change in the structure and composition of tissue repair markers.
Collapse
Affiliation(s)
- Thayane Rafaela Feola Pizzo
- Graduate Program of Biomedical Sciences, University Center of Herminio Ometto Foundation-FHO, Av. Dr. Maximiliano Baruto, 500-Jd. Universitário, 13607-339, Araras, São Paulo, Brasil
| | - Ana Paula Valverde
- Graduate Program of Biomedical Sciences, University Center of Herminio Ometto Foundation-FHO, Av. Dr. Maximiliano Baruto, 500-Jd. Universitário, 13607-339, Araras, São Paulo, Brasil
| | - Lucas Eduardo Orzari
- Graduate Program of Biomedical Sciences, University Center of Herminio Ometto Foundation-FHO, Av. Dr. Maximiliano Baruto, 500-Jd. Universitário, 13607-339, Araras, São Paulo, Brasil
| | - Luiz Gustavo Terciotti
- Graduate Program of Biomedical Sciences, University Center of Herminio Ometto Foundation-FHO, Av. Dr. Maximiliano Baruto, 500-Jd. Universitário, 13607-339, Araras, São Paulo, Brasil
| | - Robson Damasceno de Lima
- Graduate Program of Biomedical Sciences, University Center of Herminio Ometto Foundation-FHO, Av. Dr. Maximiliano Baruto, 500-Jd. Universitário, 13607-339, Araras, São Paulo, Brasil
| | - Fernando Russo Costa do Bomfim
- Graduate Program of Biomedical Sciences, University Center of Herminio Ometto Foundation-FHO, Av. Dr. Maximiliano Baruto, 500-Jd. Universitário, 13607-339, Araras, São Paulo, Brasil
| | - Marcelo Augusto Marreto Esquisatto
- Graduate Program of Biomedical Sciences, University Center of Herminio Ometto Foundation-FHO, Av. Dr. Maximiliano Baruto, 500-Jd. Universitário, 13607-339, Araras, São Paulo, Brasil
| | - Thiago Antônio Moretti de Andrade
- Graduate Program of Biomedical Sciences, University Center of Herminio Ometto Foundation-FHO, Av. Dr. Maximiliano Baruto, 500-Jd. Universitário, 13607-339, Araras, São Paulo, Brasil
| | - Maria Esméria Corezola do Amaral
- Graduate Program of Biomedical Sciences, University Center of Herminio Ometto Foundation-FHO, Av. Dr. Maximiliano Baruto, 500-Jd. Universitário, 13607-339, Araras, São Paulo, Brasil
| | - Camila Andrea de Oliveira
- Graduate Program of Biomedical Sciences, University Center of Herminio Ometto Foundation-FHO, Av. Dr. Maximiliano Baruto, 500-Jd. Universitário, 13607-339, Araras, São Paulo, Brasil
| | - Maíra Felonato
- Graduate Program of Biomedical Sciences, University Center of Herminio Ometto Foundation-FHO, Av. Dr. Maximiliano Baruto, 500-Jd. Universitário, 13607-339, Araras, São Paulo, Brasil
| |
Collapse
|
5
|
Kazemi F, Mohebbati R, Shafei MN. Antihypertensive and Antioxidant Effects of an Aqueous Extract of Asafetida in Renovascular Hypertensive Rats. SAUDI JOURNAL OF KIDNEY DISEASES AND TRANSPLANTATION 2023; 34:S86-S95. [PMID: 38995276 DOI: 10.4103/sjkdt.sjkdt_9_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2024] Open
Abstract
Recently, the effect of an aqueous extract of asafetida on acute angiotensin II hypertensive rats was evaluated. The present study evaluated the antihypertensive and antioxidant effects of asafetida on a rat model of renovascular hypertension (RVH) using four groups. RVH was induced by clipping the renal artery; the sham group underwent surgery but without clipping. The RVH rats received losartan (Los, an AT1 receptor antagonist) or asafetida by gavage for 4 weeks. On the 28th day, the femoral artery was cannulated, and the systolic blood pressure (SBP), mean arterial pressure (MAP), and heart rate (HR) were recorded. Finally, the levels of superoxide dismutase (SOD) activity, malondialdehyde (MDA), and total thiol content in the kidney and heart tissues were measured. In RVH rats, SBP and MAP significantly increased compared with the control. Los and the extract significantly reduced the changes in SBP, MAP, and HR that were induced in the RVH rats (P <0.05-0.001). In RVH rats, levels of MDA significantly increased and the content of total thiol and SOD decreased in both the heart and kidney tissues. Los plus the extract significantly decreased MDA and increased total thiol and SOD in the heart and kidney tissues. We concluded that an aqueous extract of asafetida gum has antihypertensive and antioxidant effects in the RVH rat model. The effect of the extract is similar to that of Los, which suggests that this effect of asafetida is mediated via an effect on the angiotensin Type I receptor.
Collapse
Affiliation(s)
- Farzaneh Kazemi
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reza Mohebbati
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Naser Shafei
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Division of Neurocognitive Sciences, Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
6
|
Cobb MS, Tao S, Shortt K, Girgis M, Hauptman J, Schriewer J, Chin Z, Dorfman E, Campbell K, Heruth DP, Shohet RV, Dawn B, Konorev EA. Smad3 promotes adverse cardiovascular remodeling and dysfunction in doxorubicin-treated hearts. Am J Physiol Heart Circ Physiol 2022; 323:H1091-H1107. [PMID: 36269647 PMCID: PMC9678413 DOI: 10.1152/ajpheart.00312.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 09/26/2022] [Accepted: 10/14/2022] [Indexed: 01/21/2023]
Abstract
Many anticancer therapies cause serious cardiovascular complications that degrade quality of life and cause early mortality in treated patients. Specifically, doxorubicin is known as an effective anticancer agent that causes cardiomyopathy in treated patients. There has been growing interest in defining the role of endothelial cells in cardiac damage by doxorubicin. We have shown in the present study that endothelial nuclei accumulate more intravenously administered doxorubicin than other cardiac cell types. Doxorubicin enhanced cardiac production of the transforming growth factor-β (TGF-β) ligands and nuclear translocation of phospho-Smad3 in both cultured and in vivo cardiac endothelial cells. To examine the role of the TGF-β/mothers against decapentaplegic homolog 3 (Smad3) pathway in cardiac damage by doxorubicin, we used both Smad3 shRNA stable endothelial cell lines and Smad3-knockout mice. We demonstrated using endothelial transcriptome analysis that upregulation of the TGF-β and inflammatory cytokine/cytokine receptor pathways, as well as suppression of cell cycle and angiogenesis by doxorubicin, were alleviated in Smad3-deficient endothelial cells. The results of transcriptomic analysis were validated using qPCR, immunoblotting, and ex vivo aortic ring sprouting assays. Similarly, increased cardiac expression of cytokines and chemokines observed in treated wild-type mice was diminished in treated Smad3-knockout animals. We also detected increased end-diastolic diameter and depressed systolic function in doxorubicin-treated wild-type but not Smad3-knockout mice. This work provides evidence for the critical role of the canonical TGF-β/Smad3 pathway in cardiac damage by doxorubicin.NEW & NOTEWORTHY Microvascular endothelial cells in the heart accumulate more intravenously administered doxorubicin than nonendothelial cardiac cell types. The treatment enhanced the TGF-β/Smad3 pathway and elicited endothelial cell senescence and inflammatory responses followed by adverse cardiac remodeling and dysfunction in wild-type but not Smad3-deficient animals. Our study suggests that the TGF-β/Smad3 pathway contributes to the development of doxorubicin cardiomyopathy and the potential value of novel approaches to ameliorate cardiotoxicity by targeting the Smad3 transcription factor.
Collapse
Affiliation(s)
- Melissa S Cobb
- Department of Basic Sciences, Kansas City University, Kansas City, Missouri
| | - Shixin Tao
- Department of Basic Sciences, Kansas City University, Kansas City, Missouri
| | - Katherine Shortt
- Ambry Genetics, Department of Advanced Analytics, Aliso Viejo, California
| | - Magdy Girgis
- Department of Internal Medicine, Kirk Kerkorian School of Medicine at UNLV, Las Vegas, Nevada
| | - Jeryl Hauptman
- Department of Internal Medicine, Kirk Kerkorian School of Medicine at UNLV, Las Vegas, Nevada
| | - Jill Schriewer
- Department of Basic Sciences, Kansas City University, Kansas City, Missouri
| | - Zaphrirah Chin
- Department of Basic Sciences, Kansas City University, Kansas City, Missouri
| | - Edward Dorfman
- Department of Basic Sciences, Kansas City University, Kansas City, Missouri
| | - Kyle Campbell
- Department of Basic Sciences, Kansas City University, Kansas City, Missouri
| | - Daniel P Heruth
- The Children's Mercy Research Institute, Kansas City, Missouri
- Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, Missouri
| | - Ralph V Shohet
- Department of Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| | - Buddhadeb Dawn
- Department of Internal Medicine, Kirk Kerkorian School of Medicine at UNLV, Las Vegas, Nevada
| | - Eugene A Konorev
- Department of Basic Sciences, Kansas City University, Kansas City, Missouri
| |
Collapse
|
7
|
Kim SR, Puranik AS, Jiang K, Chen X, Zhu XY, Taylor I, Khodadadi-Jamayran A, Lerman A, Hickson LJ, Childs BG, Textor SC, Tchkonia T, Niewold TB, Kirkland JL, Lerman LO. Progressive Cellular Senescence Mediates Renal Dysfunction in Ischemic Nephropathy. J Am Soc Nephrol 2021; 32:1987-2004. [PMID: 34135081 PMCID: PMC8455278 DOI: 10.1681/asn.2020091373] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 03/29/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Peripheral vascular diseases may induce chronic ischemia and cellular injury distal to the arterial obstruction. Cellular senescence involves proliferation arrest in response to stress, which can damage neighboring cells. Renal artery stenosis (RAS) induces stenotic-kidney dysfunction and injury, but whether these arise from cellular senescenceand their temporal pattern remain unknown. METHODS Chronic renal ischemia was induced in transgenic INK-ATTAC and wild type C57BL/6 mice by unilateral RAS, and kidney function (in vivo micro-MRI) and tissue damage were assessed. Mouse healthy and stenotic kidneys were analyzed using unbiased single-cell RNA-sequencing. To demonstrate translational relevance, cellular senescence was studied in human stenotic kidneys. RESULTS Using intraperitoneal AP20187 injections starting 1, 2, or 4 weeks after RAS, selective clearance of cells highly expressing p16Ink4a attenuated cellular senescence and improved stenotic-kidney function; however, starting treatment immediately after RAS induction was unsuccessful. Broader clearance of senescent cells, using the oral senolytic combination dasatinib and quercetin, in C57BL/6 RAS mice was more effective in clearing cells positive for p21 (Cdkn1a) and alleviating renal dysfunction and damage. Unbiased, single-cell RNA sequencing in freshly dissociated cells from healthy and stenotic mouse kidneys identified stenotic-kidney epithelial cells undergoing both mesenchymal transition and senescence. As in mice, injured human stenotic kidneys exhibited cellular senescence, suggesting this process is conserved. CONCLUSIONS Maladaptive tubular cell senescence, involving upregulated p16 (Cdkn2a), p19 (Cdkn2d), and p21 (Cdkn1a) expression, is associated with renal dysfunction and injury in chronic ischemia. These findings support development of senolytic strategies to delay chronic ischemic renal injury.
Collapse
Affiliation(s)
- Seo Rin Kim
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota,Department of Nephrology and Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Korea
| | - Amrutesh S. Puranik
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota,Colton Center for Autoimmunity, Division of Rheumatology, New York University Langone Medical Center, New York, New York
| | - Kai Jiang
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Xiaojun Chen
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota,Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xiang-Yang Zhu
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Ian Taylor
- FlowJo, BD Life Sciences, Ashland, Oregon
| | | | - Amir Lerman
- Department of Cardiology, Mayo Clinic, Rochester, Minnesota
| | - LaTonya J. Hickson
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Bennett G. Childs
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Stephen C. Textor
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Tamara Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota
| | - Timothy B. Niewold
- Colton Center for Autoimmunity, Division of Rheumatology, New York University Langone Medical Center, New York, New York
| | - James L. Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota
| | - Lilach O. Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
8
|
Almeida A, Lira R, Oliveira M, Martins M, Azevedo Y, Silva K, Carvalho S, Cortez E, Stumbo AC, Carvalho L, Thole A. Bone marrow-derived mesenchymal stem cells transplantation ameliorates renal injury through anti-fibrotic and anti-inflammatory effects in chronic experimental renovascular disease. Biomed J 2021; 45:629-641. [PMID: 34333108 PMCID: PMC9486239 DOI: 10.1016/j.bj.2021.07.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 05/11/2021] [Accepted: 07/23/2021] [Indexed: 12/11/2022] Open
Abstract
Background Progressive renal fibrosis is an underlying pathological process of chronic kidney disease (CKD) evolution. This study aimed to evaluate the roles of bone-marrow-derived mesenchymal stem cells (MSC) in the remodeling of fibrotic kidney parenchyma in the two kidneys-one clip (2K1C) CKD animal model. Methods Wistar rats were allocated into three groups: Sham, 2K1C, and 2K1C þ MSC. MSCs (106) were transplanted into the renal subcapsular region two weeks after clipping the left renal artery. Six weeks after clipping, left kidney samples were analyzed using histological and western blotting techniques. ANOVA tests were performed and differences between groups were considered statistically significant if p < 0.05. Results Clipped kidneys of 2K1C rats displayed renal fibrosis, with excessive collagen deposition, glomerulosclerosis and renal basement membrane disruption. Clipped kidneys of 2K1C þ MSC rats showed preserved Bowman's capsule and tubular basement membranes, medullary tubules morphological reconstitution and reduced collagen deposits. Expression levels of matrix metalloproteinase (MMP)-2 and MMP-9 were elevated, whereas tissue inhibitor of MMPs (TIMP)-1 and TIMP-2 levels were decreased in clipped kidneys of 2K1C rats. MSCs transplantation restored these expression levels. Moreover, MSCs suppressed macrophages and myofibroblasts accumulation, as well as TNF-a expression in clipped kidneys of 2K1C animals. MSCs transplantation significantly increased IL-10 expression. Conclusions Transplanted MSCs orchestrate anti-fibrotic and anti-inflammatory events, which reverse renal fibrosis and promote renal morphological restoration. This study supports the notion that only one MSCs delivery into the renal subcapsular region represents a possible therapeutic strategy against renal fibrosis for CKD treatment.
Collapse
Affiliation(s)
- Aline Almeida
- Laboratory of Stem Cell Research, Histology and Embryology Department, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, 20550-170, Brazil.
| | - Rafaelle Lira
- Laboratory of Stem Cell Research, Histology and Embryology Department, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, 20550-170, Brazil
| | - Mariana Oliveira
- Laboratory of Stem Cell Research, Histology and Embryology Department, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, 20550-170, Brazil
| | - Marcela Martins
- Laboratory of Stem Cell Research, Histology and Embryology Department, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, 20550-170, Brazil
| | - Yanca Azevedo
- Laboratory of Stem Cell Research, Histology and Embryology Department, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, 20550-170, Brazil
| | - Karina Silva
- Laboratory of Stem Cell Research, Histology and Embryology Department, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, 20550-170, Brazil
| | - Simone Carvalho
- Laboratory of Stem Cell Research, Histology and Embryology Department, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, 20550-170, Brazil
| | - Erika Cortez
- Laboratory of Stem Cell Research, Histology and Embryology Department, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, 20550-170, Brazil
| | - Ana Carolina Stumbo
- Laboratory of Stem Cell Research, Histology and Embryology Department, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, 20550-170, Brazil
| | - Lais Carvalho
- Laboratory of Stem Cell Research, Histology and Embryology Department, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, 20550-170, Brazil
| | - Alessandra Thole
- Laboratory of Stem Cell Research, Histology and Embryology Department, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, 20550-170, Brazil
| |
Collapse
|
9
|
Farahani RA, Afarideh M, Zhu XY, Tang H, Jordan KL, Saadiq IM, Ferguson CM, Lerman A, Textor SC, Lerman LO, Eirin A. Percutaneous transluminal renal angioplasty attenuates poststenotic kidney mitochondrial damage in pigs with renal artery stenosis and metabolic syndrome. J Cell Physiol 2021; 236:4036-4049. [PMID: 33151557 PMCID: PMC7920930 DOI: 10.1002/jcp.30146] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 10/09/2020] [Accepted: 10/26/2020] [Indexed: 02/06/2023]
Abstract
Percutaneous transluminal renal angioplasty (PTRA) has been used to treat renovascular disease (RVD), a chronic condition characterized by renal ischemia and metabolic abnormalities. Mitochondrial injury has been implicated as a central pathogenic mechanism in RVD, but whether it can be reversed by PTRA remains uncertain. We hypothesized that PTRA attenuates mitochondrial damage, renal injury, and dysfunction in pigs with coexisting renal artery stenosis (RAS) and metabolic syndrome (MetS). Four groups of pigs (n = 6 each) were studied after 16 weeks of diet-induced MetS and RAS (MetS + RAS), MetS + RAS treated 4 weeks earlier with PTRA, and Lean and MetS Sham controls. Single-kidney renal blood flow (RBF) and glomerular filtration rate (GFR) were assessed in vivo with multidetector computed tomography, and renal tubular mitochondrial structure and function and renal injury ex vivo. PTRA successfully restored renal artery patency, but mean arterial pressure remained unchanged. Stenotic kidney RBF and GFR, which fell in MetS + RAS compared to MetS, rose after PTRA. PTRA attenuated MetS + RAS-induced mitochondrial structural abnormalities in tubular cells and peritubular capillary endothelial cells, decreased mitochondrial H2 02 production, and increased renal cytochrome-c oxidase-IV activity and ATP production. PTRA also improved cortical microvascular and peritubular capillary density and ameliorated tubular injury and tubulointerstitial fibrosis in the poststenotic kidney. Importantly, renal mitochondrial damage correlated with poststenotic injury and dysfunction. Renal revascularization attenuated mitochondrial injury and improved renal hemodynamics and function in swine poststenotic kidneys. This study suggests a novel mechanism by which PTRA might be relatively effective in ameliorating mitochondrial damage and improving renal function in coexisting MetS and RAS.
Collapse
Affiliation(s)
- Rahele A. Farahani
- Department of Internal Medicine, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN
| | - Mohsen Afarideh
- Department of Internal Medicine, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN
| | - Xiang-Yang Zhu
- Department of Internal Medicine, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN
| | - Hui Tang
- Department of Internal Medicine, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN
| | - Kyra L. Jordan
- Department of Internal Medicine, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN
| | - Ishran M. Saadiq
- Department of Internal Medicine, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN
| | - Christopher M. Ferguson
- Department of Internal Medicine, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN
| | - Amir Lerman
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN
| | - Stephen C. Textor
- Department of Internal Medicine, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN
| | - Lilach O. Lerman
- Department of Internal Medicine, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN
| | - Alfonso Eirin
- Department of Internal Medicine, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN
| |
Collapse
|
10
|
Chen XJ, Kim SR, Jiang K, Ferguson CM, Tang H, Zhu XY, Lerman A, Eirin A, Lerman LO. Renovascular Disease Induces Senescence in Renal Scattered Tubular-Like Cells and Impairs Their Reparative Potency. Hypertension 2021; 77:507-518. [PMID: 33390051 DOI: 10.1161/hypertensionaha.120.16218] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Scattered tubular-like cells (STCs), dedifferentiated renal tubular epithelial cells, contribute to renal self-healing, but severe injury might blunt their effectiveness. We hypothesized that ischemic renovascular disease (RVD) induces senescence in STC and impairs their reparative potency. CD24+/CD133+ STCs were isolated from swine kidneys after 16 weeks of RVD or healthy controls. To test their reparative capabilities in injured kidneys, control or RVD-STC (5×105) were prelabeled and injected into the aorta of 2 kidneys, 1-clip (2k,1c) mice 2 weeks after surgery. Murine renal function and oxygenation were studied in vivo 2 weeks after injection using micro-magnetic resonance imaging, and fibrosis, tubulointerstitial injury, capillary density, and expression of profibrotic and inflammatory genes ex vivo. STC isolated from swine RVD kidneys showed increased gene expression of senescence and senescence-associated secretory phenotype markers and positive SA-β-gal staining. Delivery of normal pig STCs in 2k,1c mice improved murine renal perfusion, blood flow, and glomerular filtration rate, and downregulated profibrotic and inflammatory gene expression. These renoprotective effects were blunted using STC harvested from RVD kidneys, which also failed to attenuate hypoxia, fibrosis, tubular injury, and capillary loss in injured mouse 2k,1c kidneys. Hence, RVD may induce senescence in endogenous STC and impair their reparative capacity. These observations implicate cellular senescence in the pathophysiology of ischemic kidney disease and support senolytic therapy to permit self-healing of senescent kidneys.
Collapse
Affiliation(s)
- Xiao-Jun Chen
- From the Division of Nephrology and Hypertension (X.-J.C., S.R.K., K.J., C.M.F., H.T., X.-Y.Z., A.E., L.O.L.), Mayo Clinic, Rochester, MN.,Department of Nephrology, The Second Xiangya Hospital of Central-South University, Changsha, Hunan, China (X.-J.C.)
| | - Seo Rin Kim
- From the Division of Nephrology and Hypertension (X.-J.C., S.R.K., K.J., C.M.F., H.T., X.-Y.Z., A.E., L.O.L.), Mayo Clinic, Rochester, MN.,Division of Nephrology, Pusan National University Yangsan Hospital, Korea (S.R.K.)
| | - Kai Jiang
- From the Division of Nephrology and Hypertension (X.-J.C., S.R.K., K.J., C.M.F., H.T., X.-Y.Z., A.E., L.O.L.), Mayo Clinic, Rochester, MN
| | - Christopher M Ferguson
- From the Division of Nephrology and Hypertension (X.-J.C., S.R.K., K.J., C.M.F., H.T., X.-Y.Z., A.E., L.O.L.), Mayo Clinic, Rochester, MN
| | - Hui Tang
- From the Division of Nephrology and Hypertension (X.-J.C., S.R.K., K.J., C.M.F., H.T., X.-Y.Z., A.E., L.O.L.), Mayo Clinic, Rochester, MN
| | - Xiang-Yang Zhu
- From the Division of Nephrology and Hypertension (X.-J.C., S.R.K., K.J., C.M.F., H.T., X.-Y.Z., A.E., L.O.L.), Mayo Clinic, Rochester, MN
| | - Amir Lerman
- Department of Cardiovascular Diseases (A.L.), Mayo Clinic, Rochester, MN
| | - Alfonso Eirin
- From the Division of Nephrology and Hypertension (X.-J.C., S.R.K., K.J., C.M.F., H.T., X.-Y.Z., A.E., L.O.L.), Mayo Clinic, Rochester, MN
| | - Lilach O Lerman
- From the Division of Nephrology and Hypertension (X.-J.C., S.R.K., K.J., C.M.F., H.T., X.-Y.Z., A.E., L.O.L.), Mayo Clinic, Rochester, MN
| |
Collapse
|
11
|
Chen XJ, Zhang X, Jiang K, Krier JD, Zhu X, Conley S, Lerman A, Lerman LO. Adjunctive mesenchymal stem/stromal cells augment microvascular function in poststenotic kidneys treated with low-energy shockwave therapy. J Cell Physiol 2020; 235:9806-9818. [PMID: 32430932 DOI: 10.1002/jcp.29794] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 05/05/2020] [Accepted: 05/05/2020] [Indexed: 12/21/2022]
Abstract
Effective therapeutic strategies are needed to preserve renal function in patients with atherosclerotic renal artery stenosis (ARAS). Low-energy shockwave therapy (SW) and adipose tissue-derived mesenchymal stem/stromal cells (MSCs) both stimulate angiogenesis repair of stenotic kidney injury. This study tested the hypothesis that intrarenal delivery of adipose tissue-derived MSCs would enhance the capability of SW to preserve stenotic kidney function and structure. Twenty-two pigs were studied after 16 weeks of ARAS, ARAS treated with a SW regimen (bi-weekly for 3 weeks) with or without subsequent intrarenal delivery of adipose tissue-derived MSCs and controls. Four weeks after treatment, single-kidney renal blood flow (RBF) before and after infusion of acetylcholine, glomerular filtration rate (GFR), and oxygenation were assessed in vivo and the renal microcirculation, fibrosis, and oxidative stress ex vivo. Mean arterial pressure remained higher in ARAS, ARAS + SW, and ARAS + SW + MSC compared with normal. Both SW and SW + MSC similarly elevated the decreased stenotic kidney GFR and RBF observed in ARAS to normal levels. Yet, SW + MSC significantly improved RBF response to acetylcholine in ARAS, and attenuated capillary loss and oxidative stress more than SW alone. Density of larger microvessels was similarly increased by both interventions. Therefore, although significant changes in functional outcomes were not observed in a short period of time, adjunct MSCs enhanced pro-angiogenic effect of SW to improve renal microvascular outcomes, suggesting this as an effective stratege for long-term management of renovascular disease.
Collapse
Affiliation(s)
- Xiao-Jun Chen
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota.,Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xin Zhang
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Kai Jiang
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - James D Krier
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Xiangyang Zhu
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Sabena Conley
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Amir Lerman
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota
| | - Lilach O Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota.,Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
12
|
DeLalio LJ, Hahn S, Katayama PL, Wenner MM, Farquhar WB, Straub AC, Stocker SD. Excessive dietary salt promotes aortic stiffness in murine renovascular hypertension. Am J Physiol Heart Circ Physiol 2020; 318:H1346-H1355. [PMID: 32302491 PMCID: PMC7346535 DOI: 10.1152/ajpheart.00601.2019] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 03/18/2020] [Accepted: 04/12/2020] [Indexed: 12/22/2022]
Abstract
Renovascular hypertension is characterized by activation of the renin-angiotensin-aldosterone system, blunted natriuretic responses, and elevated sympathetic nerve activity. Excess dietary salt intake exaggerates arterial blood pressure (ABP) in multiple models of experimental hypertension. The present study tested whether a high-salt diet exaggerated ABP and vascular dysfunction in a 2-kidney, 1-clip (2K1C) murine model. Male C57BL/6J mice (8-12 wk) were randomly assigned, and fed a 0.1% or 4.0% NaCl diet, and instrumented with telemetry units to measure ABP. Then, the 2K1C model was produced by placing a cuff around the right renal artery. Systolic, diastolic, and mean ABP were significantly higher in mice fed 4.0% vs. 0.1% NaCl at 1 wk but not after 3 wk. Interestingly, 2K1C hypertension progressively increased arterial pulse pressure in both groups; however, the magnitude was significantly greater in mice fed 4.0% vs. 0.1% NaCl at 3 wk. Moreover, pulse wave velocity was significantly greater in 2K1C mice fed 4.0% vs. 0.1% NaCl diet or sham-operated mice fed either diet. Histological assessment of aortas indicated no structural differences among groups. Finally, endothelium-dependent vasodilation was significantly and selectively attenuated in the aorta but not mesenteric arteries of 2K1C mice fed 4.0% NaCl vs. 0.1% NaCl or sham-operated control mice. The findings suggest that dietary salt loading transiently exaggerates 2K1C renovascular hypertension but promotes chronic aortic stiffness and selective aortic vascular dysfunction.NEW & NOTEWORTHY High dietary salt exaggerates hypertension in multiple experimental models. Here we demonstrate that a high-salt diet produces a greater increase in arterial blood pressure at 1 wk after induction of 2-kidney, 1-clip (2K1C) hypertension but not at 3 wk. Interestingly, 2K1C mice fed a high-salt diet displayed an exaggerated pulse pressure, elevated pulse wave velocity, and reduced endothelium-dependent vasodilation of the aorta but not mesenteric arteries. These findings suggest that dietary salt may interact with underlying cardiovascular disease to promote selective vascular dysfunction and aortic stiffness.
Collapse
Affiliation(s)
- Leon J DeLalio
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Scott Hahn
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Pedro L Katayama
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Megan M Wenner
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, Delaware
| | - William B Farquhar
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, Delaware
| | - Adam C Straub
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Pittsburgh, Pennsylvania
| | - Sean D Stocker
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
13
|
Abstract
PURPOSE OF REVIEW Renovascular disease (RVD) remains an important cause of hypertension and renal dysfunction. Given the failure of renal revascularization to provide consistent clinical benefit in the Cardiovascular Outcomes for Renal Artery Lesions trial among others, further research has underscored the need for mechanistically targeted interventions to improve renal outcomes in patients in RVD. This review discusses novel therapeutic approaches for RVD in the post-Cardiovascular Outcomes for Renal Artery Lesions era. RECENT FINDINGS Emerging evidence indicates that renal inflammation, microvascular remodeling, and mitochondrial damage accelerate progression of renal injury and are important determinants of the response to revascularization. Experimental studies have identified interventions capable of ameliorating renal inflammation (e.g., cytokine inhibitors, mesenchymal stem cells), microvascular remodeling (proangiogenic interventions), and mitochondrial injury (mito-protective drugs), alone or combined with renal revascularization, to preserve the structure and function of the poststenotic kidney. Recent prospective pilot studies in patients with atherosclerotic RVD demonstrate the safety and feasibility of some of such interventions to protect the kidney. SUMMARY Experimental studies and pilot clinical trials suggest that therapies targeting renal inflammation, microvascular remodeling, and mitochondrial damage have the potential to preserve the structure and function of the stenotic kidney. Further studies in larger cohorts are needed to confirm their renoprotective effects and clinical role in human RVD.
Collapse
|
14
|
Ghayur A, Padwal MK, Liu L, Zhang J, Margetts PJ. SMAD3-dependent and -independent pathways in glomerular injury associated with experimental glomerulonephritis. Am J Physiol Renal Physiol 2019; 317:F152-F162. [PMID: 31141397 DOI: 10.1152/ajprenal.00406.2018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Glomerulonephritis (GN) is a common cause of end-stage kidney disease and is characterized by glomerular inflammation, hematuria, proteinuria, and progressive renal dysfunction. Transforming growth factor (TGF)-β is involved in glomerulosclerosis and interstitial fibrosis. TGF-β activates multiple signaling pathways, including the canonical SMAD pathway. We evaluated the role of SMAD signaling in renal injury and proteinuria in a murine model of GN. SMAD3+/+ or SMAD3-/- mice received anti-glomerular basement membrane antibodies to induce GN. We confirmed previous reports that demonstrated that SMAD3 is an important mediator of glomerulosclerosis and renal interstitial fibrosis. Proteinuria was highly SMAD3 dependent. We found differential effects of SMAD3 deletion on podocytes and glomerular endothelial cells. GN led to podocyte injury, including foot process effacement and loss of podocyte-specific markers. Interestingly, these changes were not SMAD3 dependent. Furthermore, there were significant changes to glomerular endothelial cells, including loss of fenestrations, swelling, and basement membrane reduplication, which were SMAD3 dependent. Despite ongoing markers of podocyte injury in SMAD3-/- mice, proteinuria was transient. Renal injury in the setting of GN involves TGF-β and SMAD3 signaling. Cell populations within the glomerulus respond differently to SMAD3 deletion. Proteinuria correlated more with endothelial cell changes as opposed to podocyte injury in this model.
Collapse
Affiliation(s)
- Ayesha Ghayur
- Department of Medicine, McMaster University , Hamilton, Ontario , Canada
| | | | - Limin Liu
- Department of Medicine, McMaster University , Hamilton, Ontario , Canada
| | - Jing Zhang
- Department of Medicine, McMaster University , Hamilton, Ontario , Canada
| | - Peter J Margetts
- Department of Medicine, McMaster University , Hamilton, Ontario , Canada
| |
Collapse
|
15
|
Ong J, Kinsman BJ, Sved AF, Rush BM, Tan RJ, Carattino MD, Stocker SD. Renal sensory nerves increase sympathetic nerve activity and blood pressure in 2-kidney 1-clip hypertensive mice. J Neurophysiol 2019; 122:358-367. [PMID: 31091159 DOI: 10.1152/jn.00173.2019] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Renal denervation lowers arterial blood pressure (ABP) in multiple clinical trials and some experimental models of hypertension. These antihypertensive effects have been attributed to the removal of renal afferent nerves. The purpose of the present study was to define the function, anatomy, and contribution of mouse renal sensory neurons to a renal nerve-dependent model of hypertension. First, electrical stimulation of mouse renal afferent nerves produced frequency-dependent increases in ABP that were eliminated by ganglionic blockade. Stimulus-triggered averaging revealed renal afferent stimulation significantly increased splanchnic, renal, and lumbar sympathetic nerve activity (SNA). Second, kidney injection of wheat germ agglutinin into male C57Bl6 mice (12-14 wk; Jackson Laboratories) produced ipsilateral labeling in the T11-L2 dorsal root ganglia. Next, 2-kidney 1-clip (2K1C) hypertension was produced in male C57Bl6 mice (12-14 wk; Jackson Laboratories) by placement of a 0.5-mm length of polytetrafluoroethylene tubing around the left renal artery. 2K1C mice displayed an elevated ABP measured via telemetry and a greater fall in mean ABP to ganglionic blockade at day 14 or 21 vs. day 0. Renal afferent discharge was significantly higher in 2K1C-clipped vs. 2K1C-unclipped or sham kidneys. In addition, 2K1C-clipped vs. 2K1C-unclipped or sham kidneys had lower renal mass and higher mRNA levels of several proinflammatory cytokines. Finally, both ipsilateral renal denervation (10% phenol) or selective denervation of renal afferent nerves (periaxonal application of 33 mM capsaicin) at time of clipping resulted in lower ABP of 2K1C mice at day 14 or 21. These findings suggest mouse renal sensory neurons are activated to increase SNA and ABP in 2K1C hypertension. NEW & NOTEWORTHY This study documents the function, anatomy, and contribution of mouse renal sensory nerves to neurogenic hypertension produced by renal stenosis. Activation of renal afferents increased sympathetic nerve activity and blood pressure. Renal afferent activity was elevated in hypertensive mice, and renal afferent denervation lowered blood pressure. Clinically, patients with renal stenosis have been excluded from clinical trials for renal denervation, but this study highlights the potential therapeutic efficacy to target renal nerves in these patients.
Collapse
Affiliation(s)
- Jason Ong
- Division of Renal-Electrolyte, Department of Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania.,Department of Neuroscience, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Brian J Kinsman
- Division of Renal-Electrolyte, Department of Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Alan F Sved
- Department of Neuroscience, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Brittney M Rush
- Division of Renal-Electrolyte, Department of Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Roderick J Tan
- Division of Renal-Electrolyte, Department of Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Marcelo D Carattino
- Division of Renal-Electrolyte, Department of Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Sean D Stocker
- Division of Renal-Electrolyte, Department of Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| |
Collapse
|
16
|
Affiliation(s)
- Alfonso Eirin
- From the Divisions of Nephrology and Hypertension (A.E., S.C.T., L.O.L.), Mayo Clinic, Rochester, MN
| | - Stephen C Textor
- From the Divisions of Nephrology and Hypertension (A.E., S.C.T., L.O.L.), Mayo Clinic, Rochester, MN
| | - Lilach O Lerman
- From the Divisions of Nephrology and Hypertension (A.E., S.C.T., L.O.L.), Mayo Clinic, Rochester, MN
- Department of Cardiovascular Diseases (L.O.L.), Mayo Clinic, Rochester, MN
| |
Collapse
|
17
|
Kashyap S, Osman M, Ferguson CM, Nath MC, Roy B, Lien KR, Nath KA, Garovic VD, Lerman LO, Grande JP. Ccl2 deficiency protects against chronic renal injury in murine renovascular hypertension. Sci Rep 2018; 8:8598. [PMID: 29872089 PMCID: PMC5988825 DOI: 10.1038/s41598-018-26870-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 05/22/2018] [Indexed: 01/11/2023] Open
Abstract
Inflammation plays an important role in the pathogenesis of renal and cardiovascular disease in renovascular hypertension (RVH). Ccl2 is an important mediator of inflammation, and is induced within 24 hours following surgery to establish RVH in the murine 2 kidney 1 clip model, a time prior to onset of interstitial inflammation, fibrosis, or tubular atrophy. We tested the hypothesis that Ccl2 deficiency protects the stenotic kidney (STK) from development of chronic renal damage in mice with renovascular hypertension due to renal artery stenosis (RAS). RAS surgery was performed on wild type (WT) and Ccl2 knock out (KO) mice; animals were studied for four weeks. Renal blood flow was reduced to similar extent in both WT and Ccl2 KO mice with RVH. Perfusion of the stenotic kidney was significantly reduced in Ccl2 KO mice as assessed by magnetic resonance imaging (MRI). Stenotic kidney volume in WT, but not in Ccl2 KO mice, was significantly reduced following surgery. Cortical hypoxia was observed in the stenotic kidney of Ccl2 KO mice, as assessed by blood oxygen level-dependent MRI (BOLD-MRI). Ccl2 KO mice showed less cortical atrophy than WT RAS mice. Ccl2 deficiency reduced the number of infiltrating mononuclear cells and expression of Ccl5, Ccl7, Ccl8, Ccr2 and Cd206. We conclude that Ccl2 is a critical mediator of chronic renal injury in RVH.
Collapse
Affiliation(s)
- Sonu Kashyap
- Department of Laboratory Medicine & Pathology, Mayo Clinic, Rochester, MN, USA
| | - Mazen Osman
- Department of Laboratory Medicine & Pathology, Mayo Clinic, Rochester, MN, USA
| | | | - Meryl C Nath
- Department of Laboratory Medicine & Pathology, Mayo Clinic, Rochester, MN, USA
| | - Bhaskar Roy
- Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, USA
| | - Karen R Lien
- Department of Laboratory Medicine & Pathology, Mayo Clinic, Rochester, MN, USA
| | - Karl A Nath
- Division of Nephrology & Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Vesna D Garovic
- Division of Nephrology & Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Lilach O Lerman
- Division of Nephrology & Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Joseph P Grande
- Department of Laboratory Medicine & Pathology, Mayo Clinic, Rochester, MN, USA. .,Division of Nephrology & Hypertension, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
18
|
Zhang X, Zhu X, Ferguson CM, Jiang K, Burningham T, Lerman A, Lerman LO. Magnetic resonance elastography can monitor changes in medullary stiffness in response to treatment in the swine ischemic kidney. MAGMA (NEW YORK, N.Y.) 2018; 31:375-382. [PMID: 29289980 PMCID: PMC5976551 DOI: 10.1007/s10334-017-0671-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 12/14/2017] [Accepted: 12/20/2017] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Low-energy shockwave (SW) therapy attenuates damage in the stenotic kidney (STK) caused by atherosclerotic renal artery stenosis (ARAS). We hypothesized that magnetic resonance elastography (MRE) would detect attenuation of fibrosis following SW in unilateral ARAS kidneys. MATERIALS AND METHODS Domestic pigs were randomized to control, unilateral ARAS, and ARAS treated with 6 sessions of SW over 3 consecutive weeks (n = 7 each) starting after 3 weeks of ARAS or sham. Four weeks after SW treatment, renal fibrosis was evaluated with MRE in vivo or trichrome staining ex vivo. Blood pressure, single-kidney renal-blood-flow (RBF) and glomerular-filtration-rate (GFR) were assessed. RESULTS MRE detected increased stiffness in the STK medulla (15.3 ± 2.1 vs. 10.1 ± 0.8 kPa, p < 0.05) that moderately correlated with severity of fibrosis (R2 = 0.501, p < 0.01), but did not identify mild STK cortical or contralateral kidney fibrosis. Trichrome staining showed that medullary fibrosis was increased in ARAS and alleviated by SW (10.4 ± 1.8% vs. 2.9 ± 0.2%, p < 0.01). SW slightly decreased blood pressure and normalized STK RBF and GFR in ARAS. In the contralateral kidney, SW reversed the increase in RBF and GFR. CONCLUSION MRE might be a tool for noninvasive monitoring of medullary fibrosis in response to treatment in kidney disease.
Collapse
Affiliation(s)
- Xin Zhang
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Xiangyang Zhu
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | | | - Kai Jiang
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Tyson Burningham
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Amir Lerman
- Cardiovascular Diseases, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Lilach Orly Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
- Cardiovascular Diseases, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
19
|
Kashyap S, Warner G, Hu Z, Gao F, Osman M, Al Saiegh Y, Lien KR, Nath K, Grande JP. Cardiovascular phenotype in Smad3 deficient mice with renovascular hypertension. PLoS One 2017; 12:e0187062. [PMID: 29073282 PMCID: PMC5658153 DOI: 10.1371/journal.pone.0187062] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 10/12/2017] [Indexed: 11/18/2022] Open
Abstract
Renovascular hypertension (RVH) has deleterious effects on both the kidney and the heart. TGF-β signaling through Smad3 directs tissue fibrosis in chronic injury models. In the 2-kidney 1-clip (2K1C) model of RVH, employing mice on the 129 genetic background, Smad3 deficiency (KO) protects the stenotic kidney (STK) from development of interstitial fibrosis. However, these mice have an increased incidence of sudden cardiac death following 2K1C surgery. The purpose of this study was to characterize the cardiovascular phenotype of these mice. Renal artery stenosis (RAS) was established in Wild-type (WT) and Smad3 KO mice (129 genetic background) by placement of a polytetrafluoroethylene cuff on the right renal artery. Mortality was 25.5% for KO mice with RAS, 4.1% for KO sham mice, 1.2% for WT with RAS, and 1.8% for WT sham mice. Myocardial tissue of mice studied at 3 days following surgery showed extensive myocyte necrosis in KO but not WT mice. Myocyte necrosis was associated with a rapid induction of Ccl2 expression, macrophage influx, and increased MMP-9 activity. At later time points, both KO and WT mice developed myocardial fibrosis. No aortic aneurysms or dissections were observed at any time point. Smad3 KO mice were backcrossed to the C57BL/6J strain and subjected to RAS. Sudden death was observed at 10-14 days following surgery in 62.5% of mice; necropsy revealed aortic dissections as the cause of death. As observed in the 129 mice, the STK of Smad3 KO mice on the C57BL/6J background did not develop significant chronic renal damage. We conclude that the cardiovascular manifestations of Smad3 deficient mice are strain-specific, with myocyte necrosis in 129 mice and aortic rupture in C57BL/6J mice. Future studies will define mechanisms underlying this strain-specific effect on the cardiovascular system.
Collapse
Affiliation(s)
- Sonu Kashyap
- Department of Laboratory Medicine & Pathology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Gina Warner
- Kogod Aging Center, Department of Anesthesiology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Zeng Hu
- Department of Laboratory Medicine & Pathology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Feng Gao
- UT Southwestern Medical School, Dallas, Texas, United States of America
| | - Mazen Osman
- Department of Laboratory Medicine & Pathology, Mayo Clinic, Rochester, Minnesota, United States of America
| | | | - Karen R. Lien
- Department of Laboratory Medicine & Pathology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Karl Nath
- Division of Nephrology & Hypertension, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Joseph P. Grande
- Department of Laboratory Medicine & Pathology, Mayo Clinic, Rochester, Minnesota, United States of America
- Division of Nephrology & Hypertension, Mayo Clinic, Rochester, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
20
|
Ma Z, Jin X, He L, Wang Y. CXCL16 regulates renal injury and fibrosis in experimental renal artery stenosis. Am J Physiol Heart Circ Physiol 2016; 311:H815-21. [PMID: 27496882 PMCID: PMC5142186 DOI: 10.1152/ajpheart.00948.2015] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 07/27/2016] [Indexed: 12/27/2022]
Abstract
Recent studies have shown that inflammation plays a critical role in the initiation and progression of hypertensive kidney disease, including renal artery stenosis. However, the signaling mechanisms underlying the induction of inflammation are poorly understood. We found that CXCL16 was induced in the kidney in a murine model of renal artery stenosis. To determine whether CXCL16 is involved in renal injury and fibrosis, wild-type and CXCL16 knockout mice were subjected to renal artery stenosis induced by placing a cuff on the left renal artery. Wild-type and CXCL16 knockout mice had comparable blood pressure at baseline. Renal artery stenosis caused an increase in blood pressure that was similar between wild-type and CXCL16 knockout mice. CXCL16 knockout mice were protected from RAS-induced renal injury and fibrosis. CXCL16 deficiency suppressed bone marrow-derived fibroblast accumulation and myofibroblast formation in the stenotic kidneys, which was associated with less expression of extracellular matrix proteins. Furthermore, CXCL16 deficiency inhibited infiltration of F4/80(+) macrophages and CD3(+) T cells in the stenotic kidneys compared with those of wild-type mice. Taken together, our results indicate that CXCL16 plays a pivotal role in the pathogenesis of renal artery stenosis-induced renal injury and fibrosis through regulation of bone marrow-derived fibroblast accumulation and macrophage and T-cell infiltration.
Collapse
Affiliation(s)
- Zhiheng Ma
- Selzman Institute for Kidney Health and Section of Nephrology, Department of Medicine, Baylor College of Medicine, Houston, Texas; Section of Nephrology, Department of Medicine, Shuguang Hospital, Shanghai, China; and
| | - Xiaogao Jin
- Selzman Institute for Kidney Health and Section of Nephrology, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Liqun He
- Section of Nephrology, Department of Medicine, Shuguang Hospital, Shanghai, China; and
| | - Yanlin Wang
- Selzman Institute for Kidney Health and Section of Nephrology, Department of Medicine, Baylor College of Medicine, Houston, Texas; Center for Translational Research on Inflammatory Diseases and Renal Section, Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas
| |
Collapse
|
21
|
Kashyap S, Boyilla R, Zaia PJ, Ghossan R, Nath KA, Textor SC, Lerman LO, Grande JP. Development of renal atrophy in murine 2 kidney 1 clip hypertension is strain independent. Res Vet Sci 2016; 107:171-177. [PMID: 27473991 DOI: 10.1016/j.rvsc.2016.06.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 05/20/2016] [Accepted: 06/06/2016] [Indexed: 01/02/2023]
Abstract
The murine 2-kidney 1-clip (2K1C) model has been used to identify mechanisms underlying chronic renal disease in human renovascular hypertension. Although this model recapitulates many of the features of human renovascular disease, strain specific variability in renal outcomes and animal-to-animal variation in the degree of arterial stenosis are well recognized limitations. In particular, the C57BL/6J strain is considered to be resistant to chronic renal damage in other models. Our objectives were to determine strain dependent variations in renal disease progression and to identify parameters that predict renal atrophy in murine 2K1C hypertension. We used a 0.20mm polytetrafluoroethylene cuff to establish RAS in 3 strains of mice C57BL/6J (N=321), C57BLKS/J (N=177) and129Sv (N=156). The kidneys and hearts were harvested for histopathologic analysis after 3days or after 1, 2, 4, 6, 7, 11 or 17weeks. We performed multivariate analysis to define associations between blood pressure, heart and kidney weights, ratio of stenotic kidney/contralateral kidney (STK/CLK) weight, percent atrophy (% atrophy) and plasma renin content. The STK of all 3 strains showed minimal histopathologic alterations after 3days, but later developed progressive interstitial fibrosis, tubular atrophy, and inflammation. The STK weight negatively correlated with maximum blood pressure and % atrophy, and positively correlated with STK/CLK ratio. RAS produces severe chronic renal injury in the STK of all murine strains studied, including C57BL/6J. Systolic blood pressure is negatively associated with STK weight, STK/CLK ratio and positively with atrophy and may be used to assess adequacy of vascular stenosis in this model.
Collapse
Affiliation(s)
- Sonu Kashyap
- Department of Laboratory Medicine & Pathology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Rajendra Boyilla
- Department of Laboratory Medicine & Pathology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Paula J Zaia
- Fundacao Lusiada-UNILUS Rua Oswaldo Cruz, 179 11045-101 Boqueirao-Santos, SP, Brazil
| | - Roba Ghossan
- Saint Joseph University, Rue de Damas, Beirut, Lebanon
| | - Karl A Nath
- Division of Nephrology & Hypertension Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Stephen C Textor
- Division of Nephrology & Hypertension Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Lilach O Lerman
- Division of Nephrology & Hypertension Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Joseph P Grande
- Department of Laboratory Medicine & Pathology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA; Division of Nephrology & Hypertension Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.
| |
Collapse
|
22
|
Kashyap S, Engel S, Osman M, Al-Saiegh Y, Wongjarupong A, Grande JP. Cardiovascular manifestations of renovascular hypertension in diabetic mice. PeerJ 2016; 4:e1736. [PMID: 26925344 PMCID: PMC4768709 DOI: 10.7717/peerj.1736] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2015] [Accepted: 02/03/2016] [Indexed: 01/19/2023] Open
Abstract
Purpose. Type 2 diabetes is the leading cause of end stage renal disease in the United States. Atherosclerotic renal artery stenosis is commonly observed in diabetic patients and impacts the rate of renal and cardiovascular disease progression. We sought to test the hypothesis that renovascular hypertension, induced by unilateral renal artery stenosis, exacerbates cardiac remodeling in leptin-deficient (db/db) mice, which serves as a model of human type II diabetes. Methods. We employed a murine model of renovascular hypertension through placement of a polytetrafluoroethylene cuff on the right renal artery in db/db mice. We studied 109 wild-type (non-diabetic, WT) and 95 db/db mice subjected to renal artery stenosis (RAS) or sham surgery studied at 1, 2, 4, and 6+ weeks following surgery. Cardiac remodeling was assessed by quantitative analysis of the percent of myocardial surface area occupied by interstitial fibrosis tissue, as delineated by trichrome stained slides. Aortic pathology was assessed by histologic sampling of grossly apparent structural abnormalities or by section of ascending aorta of vessels without apparent abnormalities. Results. We noted an increased mortality in db/db mice subjected to RAS. The mortality rate of db/db RAS mice was about 23.5%, whereas the mortality rate of WT RAS mice was only 1.5%. Over 60% of mortality in the db/db mice occurred in the first two weeks following RAS surgery. Necropsy showed massive intrathoracic hemorrhage associated with aortic dissection, predominantly in the ascending aorta and proximal descending aorta. Aortas from db/db RAS mice showed more smooth muscle dropout, loss of alpha smooth muscle actin expression, medial disruption, and hemorrhage than aortas from WT mice with RAS. Cardiac tissue from db/db RAS mice had more fibrosis than did cardiac tissue from WT RAS mice. Conclusions. db/db mice subjected to RAS are prone to develop fatal aortic dissection, which is not observed in WT mice with RAS. The db/db RAS model provides the basis for future studies directed towards defining basic mechanisms underlying the interaction of hypertension and diabetes on the development of aortic lesions.
Collapse
Affiliation(s)
- Sonu Kashyap
- Department of Laboratory Medicine and Pathology, Mayo Clinic , Rochester, MN , USA
| | - Sean Engel
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA; Wartburg College, Waverly, IA, United States
| | - Mazen Osman
- Department of Laboratory Medicine and Pathology, Mayo Clinic , Rochester, MN , USA
| | | | | | - Joseph P Grande
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA; Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
23
|
Kashyap S, Warner GM, Hartono SP, Boyilla R, Knudsen BE, Zubair AS, Lien K, Nath KA, Textor SC, Lerman LO, Grande JP. Blockade of CCR2 reduces macrophage influx and development of chronic renal damage in murine renovascular hypertension. Am J Physiol Renal Physiol 2015; 310:F372-84. [PMID: 26661648 DOI: 10.1152/ajprenal.00131.2015] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 12/01/2015] [Indexed: 01/01/2023] Open
Abstract
Renovascular hypertension (RVH) is a common cause of both cardiovascular and renal morbidity and mortality. In renal artery stenosis (RAS), atrophy in the stenotic kidney is associated with an influx of macrophages and other mononuclear cells. We tested the hypothesis that chemokine receptor 2 (CCR2) inhibition would reduce chronic renal injury by reducing macrophage influx in the stenotic kidney of mice with RAS. We employed a well-established murine model of RVH to define the relationship between macrophage infiltration and development of renal atrophy in the stenotic kidney. To determine the role of chemokine ligand 2 (CCL2)/CCR2 signaling in the development of renal atrophy, mice were treated with the CCR2 inhibitor RS-102895 at the time of RAS surgery and followed for 4 wk. Renal tubular epithelial cells expressed CCL2 by 3 days following surgery, a time at which no significant light microscopic alterations, including interstitial inflammation, were identified. Macrophage influx increased with time following surgery. At 4 wk, the development of severe renal atrophy was accompanied by an influx of inducible nitric oxide synthase (iNOS)+ and CD206+ macrophages that coexpressed F4/80, with a modest increase in macrophages coexpressing arginase 1 and F4/80. The CCR2 inhibitor RS-102895 attenuated renal atrophy and significantly reduced the number of dual-stained F4/80+ iNOS+ and F4/80+ CD206+ but not F4/80+ arginase 1+ macrophages. CCR2 inhibition reduces iNOS+ and CD206+ macrophage accumulation that coexpress F4/80 and renal atrophy in experimental renal artery stenosis. CCR2 blockade may provide a novel therapeutic approach to humans with RVH.
Collapse
Affiliation(s)
- Sonu Kashyap
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Gina M Warner
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Stella P Hartono
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota; Mayo Medical School, Mayo Clinic, Rochester, Minnesota; and
| | - Rajendra Boyilla
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Bruce E Knudsen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Adeel S Zubair
- Mayo Medical School, Mayo Clinic, Rochester, Minnesota; and
| | - Karen Lien
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Karl A Nath
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Stephen C Textor
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Lilach O Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Joseph P Grande
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota; Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
24
|
Kwon SH, Lerman LO. Atherosclerotic renal artery stenosis: current status. Adv Chronic Kidney Dis 2015; 22:224-31. [PMID: 25908472 DOI: 10.1053/j.ackd.2014.10.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 10/14/2014] [Accepted: 10/15/2014] [Indexed: 12/29/2022]
Abstract
Atherosclerotic renal artery stenosis (ARAS) remains a major cause of secondary hypertension and kidney failure. Randomized prospective trials show that medical treatment should constitute the main therapeutic approach in ARAS. Regardless of intensive treatment and adequate blood pressure control, however, renal and extrarenal complications are not uncommon. Yet, the precise mechanisms, accurate detection, and optimal treatment in ARAS remain elusive. Strategies oriented to early detection and targeting these pathogenic pathways might prevent development of clinical end points. Here, we review the results of recent clinical trials, current understanding of the pathogenic mechanisms, novel imaging techniques to assess kidney damage in ARAS, and treatment options.
Collapse
|
25
|
Textor SC, Lerman LO. Paradigm Shifts in Atherosclerotic Renovascular Disease: Where Are We Now? J Am Soc Nephrol 2015; 26:2074-80. [PMID: 25868641 DOI: 10.1681/asn.2014121274] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Results of recent clinical trials and experimental studies indicate that whereas atherosclerotic renovascular disease can accelerate both systemic hypertension and tissue injury in the poststenotic kidney, restoring vessel patency alone is insufficient to recover kidney function for most subjects. Kidney injury in atherosclerotic renovascular disease reflects complex interactions among vascular rarefication, oxidative stress injury, and recruitment of inflammatory cellular elements that ultimately produce fibrosis. Classic paradigms for simply restoring blood flow are shifting to implementation of therapy targeting mitochondria and cell-based functions to allow regeneration of vascular, glomerular, and tubular structures sufficient to recover, or at least stabilize, renal function. These developments offer exciting possibilities of repair and regeneration of kidney tissue that may limit progressive CKD in atherosclerotic renovascular disease and may apply to other conditions in which inflammatory injury is a major common pathway.
Collapse
Affiliation(s)
- Stephen C Textor
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Lilach O Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
26
|
Franchi F, Peterson KM, Xu R, Miller B, Psaltis PJ, Harris PC, Lerman LO, Rodriguez-Porcel M. Mesenchymal Stromal Cells Improve Renovascular Function in Polycystic Kidney Disease. Cell Transplant 2014; 24:1687-98. [PMID: 25290249 DOI: 10.3727/096368914x684619] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Polycystic kidney disease (PKD) is a common cause of end-stage renal failure, for which there is no accepted treatment. Progenitor and stem cells have been shown to restore renal function in a model of renovascular disease, a disease that shares many features with PKD. The objective of this study was to examine the potential of adult stem cells to restore renal structure and function in PKD. Bone marrow-derived mesenchymal stromal cells (MSCs, 2.5 × 10(5)) were intrarenally infused in 6-week-old PCK rats. At 10 weeks of age, PCK rats had an increase in systolic blood pressure (SBP) versus controls (126.22 ± 2.74 vs. 116.45 ± 3.53 mmHg, p < 0.05) and decreased creatinine clearance (3.76 ± 0.31 vs. 6.10 ± 0.48 µl/min/g, p < 0.01), which were improved in PKD animals that received MSCs (SBP: 114.67 ± 1.34 mmHg, and creatinine clearance: 4.82 ± 0.24 µl/min/g, p = 0.001 and p = 0.003 vs. PKD, respectively). MSCs preserved vascular density and glomeruli diameter, measured using microcomputed tomography. PCK animals had increased urine osmolality (843.9 ± 54.95 vs. 605.6 ± 45.34 mOsm, p < 0.01 vs. control), which was improved after MSC infusion and not different from control (723.75 ± 56.6 mOsm, p = 0.13 vs. control). Furthermore, MSCs reduced fibrosis and preserved the expression of proangiogenic molecules, while cyst size and number were unaltered by MSCs. Delivery of exogenous MSCs improved vascular density and renal function in PCK animals, and the benefit was observed up to 4 weeks after a single infusion. Cell-based therapy constitutes a novel approach in PKD.
Collapse
Affiliation(s)
- Federico Franchi
- Division of Cardiovascular Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Al-Suraih M, Grande JP. Management of renal artery stenosis: What does the experimental evidence tell us? World J Cardiol 2014; 6:855-860. [PMID: 25228964 PMCID: PMC4163714 DOI: 10.4330/wjc.v6.i8.855] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Revised: 03/04/2014] [Accepted: 06/11/2014] [Indexed: 02/06/2023] Open
Abstract
Optimal management of patients with renal artery stenosis (RAS) is a subject of considerable controversy. There is incontrovertible evidence that renal artery stenosis has profound effects on the heart and cardiovascular system in addition to the kidney. Recent evidence indicates that restoration of blood flow alone does not improve renal or cardiovascular outcomes in patients with renal artery stenosis. A number of human and experimental studies have documented the clinical, hemodynamic, and histopathologic features in renal artery stenosis. New approaches to the treatment of renovascular hypertension due to RAS depend on better understanding of basic mechanisms underlying the development of chronic renal disease in these patients. Several groups have employed the two kidney one clip model of renovascular hypertension to define basic signaling mechanisms responsible for the development of chronic renal disease. Recent studies have underscored the importance of inflammation in the development and progression of renal damage in renal artery stenosis. In particular, interactions between the renin-angiotensin system, oxidative stress, and inflammation appear to play a critical role in this process. In this overview, results of recent studies to define basic pathways responsible for renal disease progression will be highlighted. These studies may provide the rationale for novel therapeutic approaches to treat patients with renovascular hypertension.
Collapse
|
28
|
Hartono SP, Knudsen BE, Lerman LO, Textor SC, Grande JP. Combined effect of hyperfiltration and renin angiotensin system activation on development of chronic kidney disease in diabetic db/db mice. BMC Nephrol 2014; 15:58. [PMID: 24708836 PMCID: PMC3984262 DOI: 10.1186/1471-2369-15-58] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Accepted: 03/31/2014] [Indexed: 12/17/2022] Open
Abstract
Background Hypertension is a major risk factor for renal disease progression. However, the mechanisms by which hypertension aggravates the effects of diabetes on the kidney are incompletely understood. We tested the hypothesis that renovascular hypertension accelerates angiotensin-II-dependent kidney damage and inflammation in the db/db mouse, a model of type II diabetes. Methods Renovascular hypertension was established in db/db and wild-type control mice through unilateral renal artery stenosis (RAS); the non-stenotic contralateral kidneys evaluated 2, 4 and 6 weeks later. Angiotensin-II infusion (1000 ng/kg/min), unilateral nephrectomy, or both were also performed in db/db mice to discern the contributions of hypertension versus hyperfiltration to development of chronic renal injury in db/db mice with RAS. The effect of blood pressure reduction in db/db mice with RAS was assessed using angiotensin-receptor-blocker (ARB) or hydralazine treatment. Results Db/db mice with renovascular hypertension developed greater and more prolonged elevation of renin activity than all other groups studied. Stenotic kidneys of db/db mice developed progressive interstitial fibrosis, tubular atrophy, and interstitial inflammation. Contralateral kidneys of wild type mice with RAS showed minimal histopathologic abnormalities, whereas db/db mice with RAS developed severe diffuse mesangial sclerosis, interstitial fibrosis, tubular atrophy, and interstitial inflammation. Db/db mice with Angiotensin II-induced hypertension developed interstitial lesions and albuminuria but not mesangial matrix expansion, while nephrectomized db/db mice exhibited modest mesangial expansion and interstitial fibrosis, but not significant albuminuria. The combination of unilateral nephrectomy and angiotensin II infusion reproduced all the features of the injury albeit in a less severe manner. ARB and hydralazine were equally effective in attenuating the development of mesangial expansion in the contralateral kidneys of db/db mice with RAS. However, only ARB prevented elevation of urinary albumin/creatinine in db/db mice with RAS. Conclusion Renovascular hypertension superimposed on diabetes exacerbates development of chronic renal disease in db/db mice at least in part through interaction with the renin-angiotensin system. Both ARB and hydralazine were equally effective in reducing systolic blood pressure and in preventing renal injury in the contralateral kidney of db/db mice with renal artery stenosis. ARB but not hydralazine prevented elevation of urinary albumin/creatinine in the db/db RAS model.
Collapse
Affiliation(s)
| | | | | | | | - Joseph P Grande
- Department of Laboratory Medicine & Pathology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.
| |
Collapse
|
29
|
Renovascular hypertension: is there still a role for stent revascularization? Curr Opin Nephrol Hypertens 2014; 22:525-30. [PMID: 23917028 DOI: 10.1097/mnh.0b013e328363ffe0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Management of renovascular hypertension remains controversial and problematic, in part, due to failure of prospective trials to demonstrate added benefit to revascularization. RECENT FINDINGS Effective drug therapy often can achieve satisfactory blood pressure control, although concerns persist of the potential for progressive, delayed loss of kidney function beyond a stenotic lesion. Recent studies highlight benefits of renal artery stenting in subsets of patients including those with recurrent pulmonary edema and those intolerant to blockade of the renin-angiotensin system. Occasional patients with recent deterioration in renal function recover sufficient glomerular filtration rate after stenting to avoid requirements for renal replacement therapy. Emerging paradigms from both clinical and experimental studies suggest that hypoxic injury within the kidney activates inflammatory injury pathways and microvascular rarification that may not recover after technically successful revascularization alone. Initial data suggest that additional measures to repair the kidney, including the use of cell-based therapy, may offer the potential to recover kidney function in advanced renovascular disease. SUMMARY Specific patient groups benefit from renal revascularization. Nephrologists will increasingly be asked to manage complex renovascular patients, different from those in randomized trials, that require intensely individualized management.
Collapse
|
30
|
Kellenberger T, Krag S, Danielsen CC, Wang XF, Nyengaard JR, Pedersen L, Yang C, Gao S, Wogensen L. Differential effects of Smad3 targeting in a murine model of chronic kidney disease. Physiol Rep 2013; 1:e00181. [PMID: 24744860 PMCID: PMC3970747 DOI: 10.1002/phy2.181] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Revised: 11/01/2013] [Accepted: 11/03/2013] [Indexed: 01/06/2023] Open
Abstract
Transforming growth factor (TGF)‐β1 has a pivotal role in the pathogenesis of progressive kidney diseases that are characterized by fibrosis. The main intracellular signaling pathway of TGF‐β1 is the Smad system, where Smad2 and Smad3 play a central role in transcriptional regulation of target genes involved in extracellular matrix (ECM) metabolism. This study analyzes the hypothesis that blockade of Smad3 attenuates the development of TGF‐β1‐driven renal fibrosis. This was examined in vivo in a transgenic model of TGF‐β1‐induced chronic kidney disease with Smad3 or without Smad3 expression and in vitro in mesangial cells and glomerular endothelial cells with Smad2/3 inhibitors or Smad3‐knockdown. Electron microscopy was used for evaluation of morphological changes, real‐time polymerase chain reaction for detection of RNA expression, and immunohistochemistry for localization of ECM components. Matrix metalloproteinase (MMP) level was assessed by gelatin zymography electrophoresis and located by in situ zymography. The results show TGF‐β1‐induced mesangial matrix expansion, tubulointerstitial fibrosis, and tubular basement membrane thickening that are attenuated by Smad3 deletion, whereas TGF‐β1‐induced glomerular basement membrane thickening is not shown. The amount and distribution profile of MMP‐2 may suggest a role of the enzyme herein. We conclude that Smad3 targeting is not exclusively beneficial as Smad3 has diverse transcriptional regulatory effects in different cell types in the kidney. Deletion of Smad3 protects the kidney from developing transforming growth factor (TGF)‐β1‐induced tubulointerstitial fibrosis, mesangial matrix expansion, and tubular basement membrane thickening, but not glomerular basement membrane thickening. The favorable effects of Smad3 deficiency can be explained by reduced deposition of collagen subtypes. The cell‐specific changes of matrix metalloproteinase expression can be a result of altered TGF‐β1 signaling.
Collapse
Affiliation(s)
- Terese Kellenberger
- Research Laboratory for Biochemical Pathology, Aarhus University Hospital, Institute of Clinical Medicine, University of Aarhus, Aarhus, Denmark
| | - Søren Krag
- Research Laboratory for Biochemical Pathology, Aarhus University Hospital, Institute of Clinical Medicine, University of Aarhus, Aarhus, Denmark
| | - Carl Christian Danielsen
- Department of Connective Tissue Biology, Institute of Biomedicine, University of Aarhus, Aarhus, Denmark
| | - Xiao-Fan Wang
- Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC
| | - Jens Randel Nyengaard
- Stereology and Electron Microscopy Laboratory, Centre for Stochastic Geometry and Advanced Bioimaging, Institute of Clinical Medicine, University of Aarhus, Aarhus, Denmark
| | - Lea Pedersen
- Research Laboratory for Biochemical Pathology, Aarhus University Hospital, Institute of Clinical Medicine, University of Aarhus, Aarhus, Denmark
| | - Chuanxu Yang
- Department of Molecular Biology, University of Aarhus, Aarhus, Denmark
| | - Shan Gao
- Department of Molecular Biology, University of Aarhus, Aarhus, Denmark
| | - Lise Wogensen
- Research Laboratory for Biochemical Pathology, Aarhus University Hospital, Institute of Clinical Medicine, University of Aarhus, Aarhus, Denmark
| |
Collapse
|
31
|
Eirin A, Lerman LO. Darkness at the end of the tunnel: poststenotic kidney injury. Physiology (Bethesda) 2013; 28:245-53. [PMID: 23817799 DOI: 10.1152/physiol.00010.2013] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Renal artery stenosis remains an important contributor to renal failure in the elderly population, but uncertainty continues to surround the mechanisms underlying progressive renal dysfunction. Here, we present the current understanding of the pathogenic mechanisms responsible for renal injury in these patients, with emphasis on those involved in disease progression.
Collapse
Affiliation(s)
- Alfonso Eirin
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | | |
Collapse
|
32
|
Ebrahimi B, Crane JA, Knudsen BE, Macura SI, Grande JP, Lerman LO. Evolution of cardiac and renal impairment detected by high-field cardiovascular magnetic resonance in mice with renal artery stenosis. J Cardiovasc Magn Reson 2013; 15:98. [PMID: 24160179 PMCID: PMC3874758 DOI: 10.1186/1532-429x-15-98] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 10/16/2013] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Renal artery stenosis (RAS) promotes hypertension and cardiac dysfunction. The 2-kidney, 1-clip mouse model in many ways resembles RAS in humans and is amenable for genetic manipulation, but difficult to evaluate noninvasively. We hypothesized that cardiovascular magnetic resonance (CMR) is capable of detecting progressive cardiac and renal dysfunction in mice with RAS and monitoring the progression of the disease longitudinally. METHODS RAS was induced at baseline in eighteen mice by constricting the renal artery. Nine additional animals served as normal controls. CMR scans (16.4 T) were performed in all mice one week before and 2 and 4 weeks after baseline. Renal volumes and hemodynamics were assessed using 3D fast imaging with steady-state precession and arterial spin labelling, and cardiac function using CMR cine. Renal hypoxia was investigated using blood oxygen-level dependent (BOLD) MR. RESULTS Two weeks after surgery, mean arterial pressure was elevated in RAS mice. The stenotic kidney (STK) showed atrophy, while the contra-lateral kidney (CLK) showed hypertrophy. Renal blood flow (RBF) and cortical oxygenation level declined in the STK but remained unchanged in CLK. Moreover, cardiac end-diastolic and stroke volumes decreased and myocardial mass increased. At 4 weeks, STK RBF remained declined and the STK cortex and medulla showed development of hypoxia. Additionally, BOLD detected a mild hypoxia in CLK cortex. Cardiac end-diastolic and stroke volumes remained reduced and left ventricular hypertrophy worsened. Left ventricular filling velocities (E/A) indicated progression of cardiac dysfunction towards restrictive filling. CONCLUSIONS CMR detected longitudinal progression of cardiac and renal dysfunction in 2K, 1C mice. These observations support the use of high-field CMR to obtain useful information regarding chronic cardiac and renal dysfunction in small animals.
Collapse
MESH Headings
- Animals
- Arterial Pressure
- Atrophy
- Cardio-Renal Syndrome/diagnosis
- Cardio-Renal Syndrome/etiology
- Cardio-Renal Syndrome/physiopathology
- Disease Models, Animal
- Disease Progression
- Heart Rate
- Hypertension, Renovascular/diagnosis
- Hypertension, Renovascular/etiology
- Hypertension, Renovascular/physiopathology
- Hypertrophy
- Hypertrophy, Left Ventricular/diagnosis
- Hypertrophy, Left Ventricular/etiology
- Hypertrophy, Left Ventricular/physiopathology
- Kidney/blood supply
- Kidney/pathology
- Magnetic Resonance Imaging, Cine
- Male
- Mice
- Mice, 129 Strain
- Predictive Value of Tests
- Renal Artery Obstruction/complications
- Renal Artery Obstruction/diagnosis
- Renal Artery Obstruction/physiopathology
- Renal Circulation
- Time Factors
- Ventricular Dysfunction, Left/diagnosis
- Ventricular Dysfunction, Left/etiology
- Ventricular Dysfunction, Left/physiopathology
- Ventricular Function, Left
Collapse
Affiliation(s)
- Behzad Ebrahimi
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - John A Crane
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Bruce E Knudsen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Slobodan I Macura
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Joseph P Grande
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Lilach O Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
33
|
Redox signaling is an early event in the pathogenesis of renovascular hypertension. Int J Mol Sci 2013; 14:18640-56. [PMID: 24025423 PMCID: PMC3794800 DOI: 10.3390/ijms140918640] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 08/30/2013] [Accepted: 09/02/2013] [Indexed: 01/11/2023] Open
Abstract
Activation of the renin-angiotensin-aldosterone system plays a critical role in the development of chronic renal damage in patients with renovascular hypertension. Although angiotensin II (Ang II) promotes oxidative stress, inflammation, and fibrosis, it is not known how these pathways intersect to produce chronic renal damage. We tested the hypothesis that renal parenchymal cells are subjected to oxidant stress early in the development of RVH and produce signals that promote influx of inflammatory cells, which may then propagate chronic renal injury. We established a reproducible murine model of RVH by placing a tetrafluoroethylene cuff on the right renal artery. Three days after cuff placement, renal tissue demonstrates no histologic abnormalities despite up regulation of both pro- and anti-oxidant genes. Mild renal atrophy was observed after seven days and was associated with induction of Tnfα and influx of CD3⁺ T cells and F4/80⁺ macrophages. By 28 days, kidneys developed severe renal atrophy with interstitial inflammation and fibrosis, despite normalization of plasma renin activity. Based on these considerations, we propose that renal parenchymal cells initiate a progressive cascade of events leading to oxidative stress, interstitial inflammation, renal fibrosis, and atrophy.
Collapse
|
34
|
|
35
|
Xu R, Franchi F, Miller B, Crane JA, Peterson KM, Psaltis PJ, Harris PC, Lerman LO, Rodriguez-Porcel M. Polycystic kidneys have decreased vascular density: a micro-CT study. Microcirculation 2013; 20:183-9. [PMID: 23167921 DOI: 10.1111/micc.12022] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2012] [Accepted: 10/26/2012] [Indexed: 12/25/2022]
Abstract
OBJECTIVE Polycystic kidney disease (PKD) is a common cause of end-stage renal failure and many of these patients suffer vascular dysfunction and hypertension. It remains unclear whether PKD is associated with abnormal microvascular structure. Thus, this study examined the renovascular structure in PKD. METHODS PKD rats (PCK model) and controls were studied at 10 weeks of age, and mean arterial pressure (MAP), renal blood flow, and creatinine clearance were measured. Microvascular architecture and cyst number and volume were assessed using micro-computed tomography, and angiogenic pathways evaluated. RESULTS Compared with controls, PKD animals had an increase in MAP (126.4 ± 4.0 vs. 126.2 ± 2.7 mmHg) and decreased clearance of creatinine (0.39 ± 0.09 vs. 0.30 ± 0.05 mL/min), associated with a decrease in microvascular density, both in the cortex (256 ± 22 vs. 136 ± 20 vessels per cm2) and medullar (114 ± 14 vs. 50 ± 9 vessels/cm2) and an increase in the average diameter of glomeruli (104.14 ± 2.94 vs. 125.76 ± 9.06 mm). PKD animals had increased fibrosis (2.2 ± 0.2 fold vs. control) and a decrease in the cortical expression in hypoxia inducible factor 1-α and vascular endothelial growth factor. CONCLUSIONS PKD animals have impaired renal vascular architecture, which can have significant functional consequences. The PKD microvasculature could represent a therapeutic target to decrease the impact of this disease.
Collapse
Affiliation(s)
- Rende Xu
- Division of Cardiovascular Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota 55905, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Saad A, Herrmann SMS, Crane J, Glockner JF, McKusick MA, Misra S, Eirin A, Ebrahimi B, Lerman LO, Textor SC. Stent revascularization restores cortical blood flow and reverses tissue hypoxia in atherosclerotic renal artery stenosis but fails to reverse inflammatory pathways or glomerular filtration rate. Circ Cardiovasc Interv 2013; 6:428-35. [PMID: 23899868 DOI: 10.1161/circinterventions.113.000219] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND Atherosclerotic renal artery stenosis (ARAS) is known to reduce renal blood flow, glomerular filtration rate (GFR) and amplify kidney hypoxia, but the relationships between these factors and tubulointerstitial injury in the poststenotic kidney are poorly understood. The purpose of this study was to examine the effect of renal revascularization in ARAS on renal tissue hypoxia and renal injury. METHODS AND RESULTS Inpatient studies were performed in patients with ARAS (n=17; >60% occlusion) before and 3 months after stent revascularization, or in patients with essential hypertension (n=32), during fixed Na(+) intake and angiotensin converting enzyme/angiotensin receptors blockers Rx. Single kidney cortical, medullary perfusion, and renal blood flow were measured using multidetector computed tomography, and GFR by iothalamate clearance. Tissue deoxyhemoglobin levels (R(2)*) were measured by blood oxygen level-dependent MRI at 3T, as was fractional kidney hypoxia (percentage of axial area with R(2)*>30/s). In addition, we measured renal vein levels of neutrophil gelatinase-associated lipocalin, monocyte chemoattractant protein-1, and tumor necrosis factor-α. Pre-stent single kidney renal blood flow, perfusion, and GFR were reduced in the poststenotic kidney. Renal vein neutrophil gelatinase-associated lipocalin, tumor necrosis factor-α, monocyte chemoattractant protein-1, and fractional hypoxia were higher in untreated ARAS than in essential hypertension. After stent revascularization, fractional hypoxia fell (P<0.002) with increased cortical perfusion and blood flow, whereas GFR and neutrophil gelatinase-associated lipocalin, monocyte chemoattractant protein-1, and tumor necrosis factor-α remained unchanged. CONCLUSIONS These data demonstrate that despite reversal of renal hypoxia and partial restoration of renal blood flow after revascularization, inflammatory cytokines and injury biomarkers remained elevated and GFR failed to recover in ARAS. Restoration of vessel patency alone failed to reverse tubulointerstitial damage and partly explains the limited clinical benefit of renal stenting. These results identify potential therapeutic targets for recovery of kidney function in renovascular disease.
Collapse
Affiliation(s)
- Ahmed Saad
- Division of Nephrology and Hypertension, and Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Franchi F, Knudsen BE, Oehler E, Textor SC, Lerman LO, Grande JP, Rodriguez-Porcel M. Non-invasive assessment of cardiac function in a mouse model of renovascular hypertension. Hypertens Res 2013; 36:770-5. [PMID: 23676847 DOI: 10.1038/hr.2013.43] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Revised: 01/22/2013] [Accepted: 02/14/2013] [Indexed: 01/19/2023]
Abstract
Hypertension continues to be a significant cause of morbidity and mortality, underscoring the need to better understand its early effects on the myocardium. The aim of this study is to determine the feasibility of in vivo longitudinal assessment of cardiac function, particularly diastolic function, in a mouse model of renovascular hypertension. Renovascular hypertension (RVH) was induced in 129S1/SvImJ male mice (n=9). To assess left ventricular (LV) systolic and diastolic function, M-mode echocardiography, pulsed-wave Doppler echocardiography and tissue Doppler imaging were performed at baseline, 2 and 4 weeks after the induction of renal artery stenosis. Myocardial tissue was collected to assess cellular morphology, fibrosis, extracellular matrix remodeling and inflammation ex vivo. RVH led to a significant increase in systolic blood pressure after 2 and 4 weeks (baseline: 99.26±1.09 mm Hg; 2 weeks: 140.90±7.64 mm Hg; 4 weeks: 147.52±5.91 mm Hg, P<0.05), resulting in a significant decrease in LV end-diastolic volume, associated with a significant elevation in ejection fraction and preserved cardiac output. Furthermore, the animals developed an abnormal diastolic function profile, with a shortening in the E velocity deceleration time as well as increases in the E/e' and the E/A ratio. The ex vivo analysis revealed a significant increase in myocyte size and deposition of extracellular matrix. Non-invasive high-resolution ultrasonography allowed assessment of the diastolic function profile in a small animal model of renovascular hypertension.
Collapse
Affiliation(s)
- Federico Franchi
- Department of Internal Medicine, Divisions of Cardiovascular Diseases, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | | | | | | | | |
Collapse
|
38
|
Bai S, Huang ZG, Chen L, Wang JT, Ding BP. Effects of felodipine combined with puerarin on ACE2-Ang (1-7)-Mas axis in renovascular hypertensive rat. ACTA ACUST UNITED AC 2013; 184:54-61. [PMID: 23523569 DOI: 10.1016/j.regpep.2013.03.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Revised: 02/25/2013] [Accepted: 03/03/2013] [Indexed: 11/26/2022]
Abstract
This study aimed to investigate the effect of combination of felodipine+puerarin on ACE2-Ang (1-7)-Mas axis, and to explore the protective effect of the combination against kidney in renovascular hypertensive rats. Goldblatt rats were randomly divided into 5 groups as follows: 4 groups which were treated with felodipine (Felo), puerarin (Pue), Felo+Pue, and Felo+captopril (Cap), respectively, and a control group of animals that were administrated with distilled water. Contents of Ang II and Ang (1-7) in renal tissues were determined by ELISA kit. The mRNA expression of ACE2/Mas and ACE/AT1 in kidneys was analyzed by RT-PCR. After 8weeks of treatment, compared with Goldblatt group, Felo+Pue reduced SBP, DBP and HR (p<0.01 or p<0.05), ameliorated renal interstitial fibrosis, decreased the level of Ang II and increased that of Ang (1-7), upregulated mRNA expression of ACE2 and Mas, decreased that of ACE and AT1, and downregulated protein expression of TGF-β1 in kidneys (p<0.01). Compared with Felo group, Felo+Pue decreased DBP and HR more markedly, attenuated fibrosis, decreased Ang II levels and increased those of Ang (1-7), upregulated mRNA expression of ACE2 in bilateral kidneys and that of Mas in ischemic kidney, downregulated that of ACE in bilateral kidneys and that of AT1 in ischemic kidney, and decreased expression of TGF-β1 protein significantly. In a word, a combination of Felo+Pue has a more efficient therapeutic effect on DBP and HR, and contributes to a better protection against renal interstitial fibrosis.
Collapse
Affiliation(s)
- Song Bai
- Department of Pharmacology & Pharmacology of TCM Grade Three Laboratory, State Administration of Traditional Chinese Medicine of PR China, Wannan Medical College, Wuhu City, Anhui Province 241002, China
| | | | | | | | | |
Collapse
|
39
|
Wang D, Warner GM, Yin P, Knudsen BE, Cheng J, Butters KA, Lien KR, Gray CE, Garovic VD, Lerman LO, Textor SC, Nath KA, Simari RD, Grande JP. Inhibition of p38 MAPK attenuates renal atrophy and fibrosis in a murine renal artery stenosis model. Am J Physiol Renal Physiol 2013; 304:F938-47. [PMID: 23364805 DOI: 10.1152/ajprenal.00706.2012] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Renal artery stenosis (RAS) is an important cause of chronic renal dysfunction. Recent studies have underscored a critical role for CCL2 (MCP-1)-mediated inflammation in the progression of chronic renal damage in RAS and other chronic renal diseases. In vitro studies have implicated p38 MAPK as a critical intermediate for the production of CCL2. However, a potential role of p38 signaling in the development and progression of chronic renal disease in RAS has not been previously defined. We sought to test the hypothesis that inhibition of p38 MAPK ameliorates chronic renal injury in mice with RAS. We established a murine RAS model by placing a cuff on the right renal artery and treated mice with the p38 inhibitor SB203580 or vehicle for 2 wk. In mice treated with vehicle, the cuffed kidney developed interstitial fibrosis, tubular atrophy, and interstitial inflammation. In mice treated with SB203580, the RAS-induced renal atrophy was reduced (70% vs. 39%, P < 0.05). SB203580 also reduced interstitial inflammation and extracellular matrix deposition but had no effect on the development of hypertension. SB203580 partially blocked the induction of CCL2, CCL7 (MCP-3), CC chemokine receptor 2 (CCR2), and collagen 4 mRNA expression in the cuffed kidneys. In vitro, blockade of p38 hindered both TNF-α and TGF-β-induced CCL2 upregulation. Based on these observations, we conclude that p38 MAPK plays a critical role in the induction of CCL2/CCL7/CCR2 system and the development of interstitial inflammation in RAS.
Collapse
Affiliation(s)
- Diping Wang
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Gloviczki ML, Keddis MT, Garovic VD, Friedman H, Herrmann S, McKusick MA, Misra S, Grande JP, Lerman LO, Textor SC. TGF expression and macrophage accumulation in atherosclerotic renal artery stenosis. Clin J Am Soc Nephrol 2012; 8:546-53. [PMID: 23258796 DOI: 10.2215/cjn.06460612] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND OBJECTIVES Atherosclerotic renal artery stenosis (ARAS) reduces renal blood flow and is a potential cause of chronic kidney injury, yet little is known regarding inflammatory pathways in this disorder in human participants. This study aimed to examine the hypothesis that reduced renal blood flow (RBF) in ARAS would be associated with tissue TGF-β activation and inflammatory cell accumulation. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS This cross-sectional study of ARAS of varying severity compared transjugular biopsy specimens in patients with ARAS (n=12, recruited between 2008 and 2012) with tissue from healthy kidney donors (n=15) and nephrectomy specimens from individuals with total vascular occlusion (n=65). ARAS patients were studied under controlled conditions to measure RBF by multidetector computed tomography and tissue oxygenation by blood oxygen level-dependent magnetic resonance imaging. RESULTS Compared with the nonstenotic contralateral kidneys, RBF was reduced in poststenotic kidneys (242±149 versus 365+174 ml/min; P<0.01) as was single-kidney GFR (28±17 versus 41±19 ml/min; P<0.01), whereas cortical and medullary oxygenation were relatively preserved. Tissue TGF-β immunoreactivity was higher in ARAS patients compared with those with both normal kidneys and those with total occlusion (mean score 2.4±0.7 versus 1.5+1.1 in the nephrectomy group and versus 0±0 in donors; P<0.01). By contrast, the number of CD68+ macrophages was higher with greater disease severity (from 2.2±2.7 in normal to 22.4±18 cells/high-power field in nephrectomy samples; P<0.001). CONCLUSIONS The results of this study indicate robust stimulation of TGF-β associated with macrophage infiltration within the human kidney with vascular occlusive disease.
Collapse
Affiliation(s)
- Monika L Gloviczki
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|