1
|
Rouaud F, Meens MJ, Yvon R, Hautefort A, Legouis D, Mean I, Jond L, Maillard M, Kwak BR, Moll S, Seigneux SD, Feraille E, Citi S. The knock-out of paracingulin attenuates hypertension through modulation of kidney ion transport. Am J Physiol Renal Physiol 2025; 328:F737-F751. [PMID: 40204428 DOI: 10.1152/ajprenal.00271.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/21/2024] [Accepted: 03/26/2025] [Indexed: 04/11/2025] Open
Abstract
Hypertension is a major risk factor for human morbidity and mortality, and the junctional protein paracingulin (CGNL1, JACOP) is required for the development of hypertension in a Dahl salt-sensitive rat model and is linked to human hypertension in genome wide association studies. However, the mechanism through which CGNL1 may regulate hypertension is unknown. Here, we address this question using a mouse model, where hypertension is induced by unilateral nephrectomy and angiotensin II infusion (N+A protocol). Although untreated WT and CGNL1-KO mice showed similar blood pressure, the N+A protocol induced hypertension in WT mice but not in CGNL1-KO mice. We show by immunolocalization and transcriptomic analysis that CGNL1 is expressed throughout the kidney tubules and in the endothelium of blood vessels, but not in smooth muscle. The N+A protocol induced decreased potassium urinary excretion in wild-type (WT), but not in CGNL1-KO mice. Immunoblot analysis shows that the KO of CGNL1 blunted the N+A-induced changes in the expression levels and activation of tubular ion transporters, including the Na/H exchanger 3 (NHE3) and the thiazide-sensitive Na-Cl cotransporter (NCC), and blunted the angiotensin II-dependent changes in the levels and/or activation of AMP-activated protein kinase (AMPK), ERK and myosin light chain. In contrast, myography showed comparable vascular reactivity in thoracic aortas and mesenteric arteries isolated from WT or CGNL1-KO mice. Together, these results suggest the KO of CGNL1 attenuates hypertension by uncoupling angiotensin II signaling in kidney tubule cells, indicating a novel pathway of regulation of signaling by a junctional protein.NEW & NOTEWORTHY The knock-out of paracingulin (CGNL1) prevents the development of hypertension in a unilateral nephrectomy/angiotensin II infusion model (N+A) in mice and this antihypertensive effect likely depends on uncoupling of angiotensin II from stimulation of sodium transporter activity in kidney tubules rather than on alteration of resistance blood vessel contractility.
Collapse
Affiliation(s)
- Florian Rouaud
- Department of Molecular and Cellular Biology, Faculty of Sciences, University of Geneva, Geneva, Switzerland
| | - Merlijn J Meens
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Research Office, Faculty of Health, Medicine and Life Sciences, Maastricht, The Netherlands
| | - Raphaël Yvon
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Aurélie Hautefort
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - David Legouis
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Division of Intensive Care, Department of Anesthesiology, Pharmacology, Intensive Care and Emergency Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Isabelle Mean
- Department of Molecular and Cellular Biology, Faculty of Sciences, University of Geneva, Geneva, Switzerland
| | - Lionel Jond
- Department of Molecular and Cellular Biology, Faculty of Sciences, University of Geneva, Geneva, Switzerland
| | - Marc Maillard
- Department of Nephrology and Hypertension, Faculty of Medicine, University of Lausanne, Lausanne, Switzerland
| | - Brenda R Kwak
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Solange Moll
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Sophie De Seigneux
- Department of Internal Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Eric Feraille
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Sandra Citi
- Department of Molecular and Cellular Biology, Faculty of Sciences, University of Geneva, Geneva, Switzerland
| |
Collapse
|
2
|
Yang J, Xiao S, Li L, Zhu A, Xiao W, Wang Q. Actin Dysregulation Mediates Nephrotoxicity of Cassiae Semen Aqueous Extracts. TOXICS 2024; 12:556. [PMID: 39195658 PMCID: PMC11360101 DOI: 10.3390/toxics12080556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/05/2024] [Accepted: 07/26/2024] [Indexed: 08/29/2024]
Abstract
Cassiae semen, commonly consumed as roasted tea, has been widely used for both medicinal purposes and dietary supplements. In this study, we investigated the nephrotoxic effects and underlying mechanisms of Cassiae semen aqueous extracts (CSAEs) using computational and animal models. Both male and female Sprague Dawley rats were treated with 4.73-47.30 g/kg (body weight) of CSAEs by oral gavage twice a day for 7-28 days. We found that serum and urinary biomarkers of kidney injury and kidney coefficients were increased in a dose-dependent manner, and were accompanied by morphological alterations in the kidneys of CSAEs-treated rats. Computational and molecular docking approaches predicted that the three most abundant components of CSAEs-obtusifolin, aurantio-obtusin, and obtusin-exhibited strong affinity for the binding of F-actin, ROCK1, and Rac1, and the RhoA-ROCK pathway was identified as the most likely regulatory mechanism mediating the nephrotoxicity of CSAEs. Consistently, immunofluorescence staining revealed F-actin and cytoskeleton were frequently disturbed in renal cells and brush borders at high doses of CSAEs. Results from gene expression analyses confirmed that CSAEs suppressed the key proteins in the RhoA-ROCK signaling pathway and consequently the expression of F-actin and its stabilization genes. In summary, our findings suggest that Cassiae semen can depolymerize and destabilize actin cytoskeleton by inhibition of the RhoA-ROCK pathway and/or direct binding to F-actin, leading to nephrotoxicity. The consumption of Cassiae semen as a supplement and medicine warrants attention.
Collapse
Affiliation(s)
- Jinlan Yang
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, China; (J.Y.); (S.X.); (L.L.); (A.Z.); (W.X.)
| | - Sheng Xiao
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, China; (J.Y.); (S.X.); (L.L.); (A.Z.); (W.X.)
| | - Ludi Li
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, China; (J.Y.); (S.X.); (L.L.); (A.Z.); (W.X.)
| | - An Zhu
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, China; (J.Y.); (S.X.); (L.L.); (A.Z.); (W.X.)
| | - Wusheng Xiao
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, China; (J.Y.); (S.X.); (L.L.); (A.Z.); (W.X.)
| | - Qi Wang
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, China; (J.Y.); (S.X.); (L.L.); (A.Z.); (W.X.)
- Key Laboratory of State Administration of Traditional Chinese Medicine (TCM) for Compatibility Toxicology, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, China
| |
Collapse
|
3
|
Chivers JM, Whiles SA, Miles CB, Biederman BE, Ellison MF, Lovingood CW, Wright MH, Hoover DB, Raafey MA, Youngberg GA, Venkatachalam MA, Zheleznova NN, Yang C, Liu P, Kriegel AJ, Cowley AW, O'Connor PM, Picken MM, Polichnowski AJ. Brown-Norway chromosome 1 mitigates the upregulation of proinflammatory pathways in mTAL cells and subsequent age-related CKD in Dahl SS/JrHsdMcwi rats. Am J Physiol Renal Physiol 2023; 324:F193-F210. [PMID: 36475869 PMCID: PMC9886360 DOI: 10.1152/ajprenal.00145.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 11/23/2022] [Accepted: 11/25/2022] [Indexed: 12/13/2022] Open
Abstract
Chronic kidney disease (CKD) has a strong genetic component; however, the underlying pathways are not well understood. Dahl salt-sensitive (SS)/Jr rats spontaneously develop CKD with age and are used to investigate the genetic determinants of CKD. However, there are currently several genetically diverse Dahl SS rats maintained at various institutions and the extent to which some exhibit age-related CKD is unclear. We assessed glomerulosclerosis (GS) and tubulointerstitial fibrosis (TIF) in 3- and 6-mo-old male and female SS/JrHsdMcwi, BN/NHsd/Mcwi [Brown-Norway (BN)], and consomic SS-Chr 1BN/Mcwi (SS.BN1) rats, in which chromosome 1 from the BN rat was introgressed into the genome of the SS/JrHsdMcwi rat. Rats were fed a 0.4% NaCl diet. GS (31 ± 3% vs. 7 ± 1%) and TIF (2.3 ± 0.2 vs. 0.5 ± 0.1) were significantly greater in 6-mo-old compared with 3-mo-old SS/JrHsdMcwi rats, and CKD was exacerbated in males. GS was minimal in 6- and 3-mo-old BN (3.9 ± 0.6% vs. 1.2 ± 0.4%) and SS.BN1 (2.4 ± 0.5% vs. 1.0 ± 0.3%) rats, and neither exhibited TIF. In SS/JrHsdMcwi and SS.BN1 rats, mean arterial blood pressure was significantly greater in 6-mo-old compared with 3-mo-old SS/JrHsdMcwi (162 ± 4 vs. 131 ± 2 mmHg) but not SS.BN1 (115 ± 2 vs. 116 ± 1 mmHg) rats. In 6-mo-old SS/JrHsdMcwi rats, blood pressure was significantly greater in females. RNA-sequencing analysis revealed that inflammatory pathways were upregulated in isolated medullary thick ascending tubules in 7-wk-old SS/JrHsdMcwi rats, before the development of tubule pathology, compared with SS.BN1 rats. In summary, SS/JrHsdMcwi rats exhibit robust age-related progression of medullary thick ascending limb abnormalities, CKD, and hypertension, and gene(s) on chromosome 1 have a major pathogenic role in such changes.NEW & NOTEWORTHY This study shows that the robust age-related progression of kidney disease in Dahl SS/JrHsdMcw rats maintained on a normal-salt diet is abolished in consomic SS.BN1 rats. Evidence that medullary thick ascending limb segments of SS/JrHsdMcw rats are structurally abnormal and enriched in proinflammatory pathways before the development of protein casts provides new insights into the pathogenesis of kidney disease in this model.
Collapse
Affiliation(s)
- Jacqueline M Chivers
- Department of Biomedical Sciences, East Tennessee State University, Johnson City, Tennessee
| | - Shannon A Whiles
- Department of Biomedical Sciences, East Tennessee State University, Johnson City, Tennessee
| | - Conor B Miles
- Department of Biomedical Sciences, East Tennessee State University, Johnson City, Tennessee
| | - Brianna E Biederman
- Department of Biomedical Sciences, East Tennessee State University, Johnson City, Tennessee
| | - Megan F Ellison
- Department of Biomedical Sciences, East Tennessee State University, Johnson City, Tennessee
| | - Connor W Lovingood
- Department of Biomedical Sciences, East Tennessee State University, Johnson City, Tennessee
| | - Marie H Wright
- Department of Biomedical Sciences, East Tennessee State University, Johnson City, Tennessee
| | - Donald B Hoover
- Department of Biomedical Sciences, East Tennessee State University, Johnson City, Tennessee
- Center of Excellence in Inflammation, Infectious Disease and Immunity, East Tennessee State University, Johnson City, Tennessee
| | - Muhammad A Raafey
- Department of Pathology, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| | - George A Youngberg
- Department of Pathology, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| | | | | | - Chun Yang
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Pengyuan Liu
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Alison J Kriegel
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Allen W Cowley
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Paul M O'Connor
- Department of Physiology, Augusta University, Augusta, Georgia
| | - Maria M Picken
- Department of Pathology, Loyola University Medical Center, Maywood, Illinois
| | - Aaron J Polichnowski
- Department of Biomedical Sciences, East Tennessee State University, Johnson City, Tennessee
- Center of Excellence in Inflammation, Infectious Disease and Immunity, East Tennessee State University, Johnson City, Tennessee
| |
Collapse
|
4
|
Gokula V, Terrero D, Joe B. Six Decades of History of Hypertension Research at the University of Toledo: Highlighting Pioneering Contributions in Biochemistry, Genetics, and Host-Microbiota Interactions. Curr Hypertens Rep 2022; 24:669-685. [PMID: 36301488 PMCID: PMC9708772 DOI: 10.1007/s11906-022-01226-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/21/2022] [Indexed: 01/31/2023]
Abstract
PURPOSE OF REVIEW The study aims to capture the history and lineage of hypertension researchers from the University of Toledo in Ohio and showcase their collective scientific contributions dating from their initial discoveries of the physiology of adrenal and renal systems and genetics regulating blood pressure (BP) to its more contemporary contributions including microbiota and metabolomic links to BP regulation. RECENT FINDINGS The University of Toledo College of Medicine and Life Sciences (UTCOMLS), previously known as the Medical College of Ohio, has contributed significantly to our understanding of the etiology of hypertension. Two of the scientists, Patrick Mulrow and John Rapp from UTCOMLS, have been recognized with the highest honor, the Excellence in Hypertension award from the American Heart Association for their pioneering work on the physiology and genetics of hypertension, respectively. More recently, Bina Joe has continued their legacy in the basic sciences by uncovering previously unknown novel links between microbiota and metabolites to the etiology of hypertension, work that has been recognized by the American Heart Association with multiple awards. On the clinical research front, Christopher Cooper and colleagues lead the CORAL trials and contributed importantly to the investigations on renal artery stenosis treatment paradigms. Hypertension research at this institution has not only provided these pioneering insights, but also grown careers of scientists as leaders in academia as University Presidents and Deans of Medical Schools. Through the last decade, the university has expanded its commitment to Hypertension research as evident through the development of the Center for Hypertension and Precision Medicine led by Bina Joe as its founding Director. Hypertension being the top risk factor for cardiovascular diseases, which is the leading cause of human mortality, is an important area of research in multiple international universities. The UTCOMLS is one such university which, for the last 6 decades, has made significant contributions to our current understanding of hypertension. This review is a synthesis of this rich history. Additionally, it also serves as a collection of audio archives by more recent faculty who are also prominent leaders in the field of hypertension research, including John Rapp, Bina Joe, and Christopher Cooper, which are cataloged at Interviews .
Collapse
Affiliation(s)
- Veda Gokula
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, College of Medicine and Life Sciences, University of Toledo College of Medicine and Life Sciences, Block Health Science Building, 3000 Arlington Ave, Toledo, OH, 43614-2598, USA
| | - David Terrero
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy, University of Toledo, Toledo, OH, USA
| | - Bina Joe
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, College of Medicine and Life Sciences, University of Toledo College of Medicine and Life Sciences, Block Health Science Building, 3000 Arlington Ave, Toledo, OH, 43614-2598, USA.
| |
Collapse
|
5
|
Wang W, Li C, Zhuang C, Zhang H, Wang Q, Fan X, Qi M, Sun R, Yu J. Research on the Mechanism and Prevention of Hypertension Caused by Apatinib Through the RhoA/ROCK Signaling Pathway in a Mouse Model of Gastric Cancer. Front Cardiovasc Med 2022; 9:873829. [PMID: 35811723 PMCID: PMC9262125 DOI: 10.3389/fcvm.2022.873829] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 06/08/2022] [Indexed: 11/13/2022] Open
Abstract
Hypertension is one of the main adverse effects of antiangiogenic tumor drugs and thus limits their application. The mechanism of hypertension caused by tyrosine kinase inhibitors (TKIs) targeting vascular endothelial growth factors is mainly related to inhibition of the nitric oxide (NO) pathway and activation of the endothelin pathway, as well as vascular rarefaction and increased salt sensitivity; consequently, prevention and treatment differ for this type of hypertension compared with primary hypertension. Apatinib is a highly selective TKI approved in China for the treatment of advanced or metastatic gastric cancer. The RhoA/ROCK pathway is involved in the pathogenesis of hypertension and mediates smooth muscle contraction, eNOS inhibition, endothelial dysfunction and vascular remodeling. In this study, in vivo experiments were performed to explore whether the RhoA/ROCK signaling pathway is part of a possible mechanism of apatinib in the treatment of gastric cancer-induced hypertension and the impairment of vascular remodeling and left ventricular function. Y27632, a selective small inhibitor of both ROCK1 and ROCK2, was combined with apatinib, and its efficacy was evaluated, wherein it can reduce hypertension induced by apatinib treatment in gastric cancer mice and weaken the activation of the RhoA/ROCK pathway by apatinib and a high-salt diet (HSD). Furthermore, Y-27632 improved aortic remodeling, fibrosis, endothelial dysfunction, superior mesenteric artery endothelial injury, left ventricular dysfunction and cardiac fibrosis in mice by weakening the activation of the RhoA/ROCK pathway. The expression of RhoA/ROCK pathway-related proteins and relative mRNA levels in mice after apatinib intervention were analyzed by various methods, and blood pressure and cardiac function indexes were compared. Endothelial and cardiac function and collagen levels in the aorta were also measured to assess vascular and cardiac fibrosis and to provide a basis for the prevention and treatment of this type of hypertension.
Collapse
|
6
|
Cobb MB, Wu W, Attipoe EM, Johnson AC, Garrett MR. Nephron-deficient HSRA rats exhibit renal injury with age but have limited renal damage from streptozotocin-induced hyperglycemia. Am J Physiol Renal Physiol 2021; 320:F1093-F1105. [PMID: 33843272 PMCID: PMC8285653 DOI: 10.1152/ajprenal.00487.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 04/05/2021] [Accepted: 04/05/2021] [Indexed: 01/13/2023] Open
Abstract
Hypertension and diabetes are the greatest factors influencing the progression of chronic kidney disease (CKD). Investigation into the role of nephron number in CKD alone or with hypertension has revealed a strong inverse relationship between the two; however, not much is known about the connection between nephron number and diabetic kidney disease. The heterogeneous stock-derived model of unilateral renal agenesis (HSRA) rat, a novel model of nephron deficiency, provides a unique opportunity to study the association between nephron number and hypertension and diabetes on CKD. HSRA rats exhibit failure of one kidney to develop in 50-75% of offspring, whereas the remaining offspring are born with two kidneys. Rats born with one kidney (HSRA-S) develop significant renal injury with age compared with two-kidney littermates (HSRA-C). The induction of hypertension as a secondary stressor leads to significantly more renal injury in HSRA-S compared with HSRA-C rats and nephrectomized HSRA-C (HSRA-UNX) rats. The present study sought to address the hypothesis that nephron deficiency in the HSRA rat would hasten renal injury in the presence of a secondary stressor of hyperglycemia. HSRA animals did not exhibit diabetes-related traits at any age; thus, streptozotocin (STZ) was used to induce hyperglycemia in HSRA-S, HSRA-C, and HSRA-UNX rats. STZ- and vehicle-treated animals were followed for 15 wk. STZ-treated animals developed robust hyperglycemia, but in contrast to the response to hypertension, neither HSRA-S nor HSRA-UNX animals developed proteinuria compared with vehicle treatment. In total, our data indicate that hyperglycemia from STZ alone does not have a significant impact on the onset or progression of injury in young one-kidney HSRA animals.NEW & NOTEWORTHY The HSRA rat, a novel model of nephron deficiency, provides a unique opportunity to study the association between nephron number and confounding cardiovascular complications that impact kidney health. Although hypertension was previously shown to exacerbate renal injury in young HSRA animals, diabetic hyperglycemia did not lead to worse renal injury, suggesting that nephron number has limited impact on kidney injury, at least in this model.
Collapse
Affiliation(s)
- Meredith B Cobb
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Wenjie Wu
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Esinam M Attipoe
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Ashley C Johnson
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Michael R Garrett
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
- Department of Medicine (Nephrology), University of Mississippi Medical Center, Jackson, Mississippi
- Department of Pediatrics (Genetics), University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
7
|
Cooperation of augmented calcium sensitization and increased calcium entry contributes to high blood pressure in salt-sensitive Dahl rats. Hypertens Res 2021; 44:1067-1078. [PMID: 33875859 DOI: 10.1038/s41440-021-00659-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 02/11/2021] [Accepted: 03/14/2021] [Indexed: 11/09/2022]
Abstract
Salt hypertensive Dahl rats are characterized by sympathoexcitation and relative NO deficiency. We tested the hypothesis that the increased blood pressure (BP) response to fasudil in salt hypertensive Dahl rats is due to augmented calcium sensitization in the salt-sensitive strain and/or due to their decreased baroreflex efficiency. BP reduction after acute administration of nifedipine (an L-type voltage-dependent calcium channel blocker) or fasudil (a Rho kinase inhibitor) was studied in conscious intact rats and in rats subjected to acute NO synthase inhibition or combined blockade of the renin-angiotensin system (captopril), sympathetic nervous system (pentolinium), and NO synthase (L-NAME). Intact salt-sensitive (SS) Dahl rats fed a low-salt diet had greater BP responses to nifedipine (-31 ± 6 mmHg) or fasudil (-34 ± 7 mmHg) than salt-resistant (SR) Dahl rats (-16 ± 4 and -17 ± 2 mmHg, respectively), and a high-salt intake augmented the BP response only in SS rats. These BP responses were doubled after acute NO synthase inhibition, indicating that endogenous NO attenuates both calcium entry and calcium sensitization. Additional pentolinium administration, which minimized sympathetic compensation for the drug-induced BP reduction, magnified the BP responses to nifedipine or fasudil in all groups except for salt hypertensive SS rats due to their lower baroreflex efficiency. The BP response to the calcium channel blocker nifedipine can distinguish SS and SR rats even after calcium sensitization inhibition by fasudil, which was not seen when fasudil was administered to nifedipine-pretreated rats. Thus, enhanced calcium entry (potentiated by sympathoexcitation) in salt hypertensive Dahl rats is the abnormality that is essential for their BP increase, which was further augmented by increased calcium sensitization in salt-sensitive Dahl rats.
Collapse
|
8
|
Kawarazaki W, Fujita T. Role of Rho in Salt-Sensitive Hypertension. Int J Mol Sci 2021; 22:ijms22062958. [PMID: 33803946 PMCID: PMC8001214 DOI: 10.3390/ijms22062958] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/09/2021] [Accepted: 03/12/2021] [Indexed: 12/21/2022] Open
Abstract
A high amount of salt in the diet increases blood pressure (BP) and leads to salt-sensitive hypertension in individuals with impaired renal sodium excretion. Small guanosine triphosphatase (GTP)ase Rho and Rac, activated by salt intake, play important roles in the pathogenesis of salt-sensitive hypertension as key switches of intracellular signaling. Focusing on Rho, high salt intake in the central nervous system increases sodium concentrations of cerebrospinal fluid in salt-sensitive subjects via Rho/Rho kinase and renin-angiotensin system activation and causes increased brain salt sensitivity and sympathetic nerve outflow in BP control centers. In vascular smooth muscle cells, Rho-guanine nucleotide exchange factors and Rho determine sensitivity to vasoconstrictors such as angiotensin II (Ang II), and facilitate vasoconstriction via G-protein and Wnt pathways, leading to increased vascular resistance, including in the renal arteries, in salt-sensitive subjects with high salt intake. In the vascular endothelium, Rho/Rho kinase inhibits nitric oxide (NO) production and function, and high salt amounts further augment Rho activity via asymmetric dimethylarginine, an endogenous inhibitor of NO synthetase, causing aberrant relaxation and increased vascular tone. Rho-associated mechanisms are deeply involved in the development of salt-sensitive hypertension, and their further elucidation can help in developing effective protection and new therapies.
Collapse
|
9
|
Keele GR, Prokop JW, He H, Holl K, Littrell J, Deal AW, Kim Y, Kyle PB, Attipoe E, Johnson AC, Uhl KL, Sirpilla OL, Jahanbakhsh S, Robinson M, Levy S, Valdar W, Garrett MR, Solberg Woods LC. Sept8/SEPTIN8 involvement in cellular structure and kidney damage is identified by genetic mapping and a novel human tubule hypoxic model. Sci Rep 2021; 11:2071. [PMID: 33483609 PMCID: PMC7822875 DOI: 10.1038/s41598-021-81550-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 01/05/2021] [Indexed: 01/29/2023] Open
Abstract
Chronic kidney disease (CKD), which can ultimately progress to kidney failure, is influenced by genetics and the environment. Genes identified in human genome wide association studies (GWAS) explain only a small proportion of the heritable variation and lack functional validation, indicating the need for additional model systems. Outbred heterogeneous stock (HS) rats have been used for genetic fine-mapping of complex traits, but have not previously been used for CKD traits. We performed GWAS for urinary protein excretion (UPE) and CKD related serum biochemistries in 245 male HS rats. Quantitative trait loci (QTL) were identified using a linear mixed effect model that tested for association with imputed genotypes. Candidate genes were identified using bioinformatics tools and targeted RNAseq followed by testing in a novel in vitro model of human tubule, hypoxia-induced damage. We identified two QTL for UPE and five for serum biochemistries. Protein modeling identified a missense variant within Septin 8 (Sept8) as a candidate for UPE. Sept8/SEPTIN8 expression increased in HS rats with elevated UPE and tubulointerstitial injury and in the in vitro hypoxia model. SEPTIN8 is detected within proximal tubule cells in human kidney samples and localizes with acetyl-alpha tubulin in the culture system. After hypoxia, SEPTIN8 staining becomes diffuse and appears to relocalize with actin. These data suggest a role of SEPTIN8 in cellular organization and structure in response to environmental stress. This study demonstrates that integration of a rat genetic model with an environmentally induced tubule damage system identifies Sept8/SEPTIN8 and informs novel aspects of the complex gene by environmental interactions contributing to CKD risk.
Collapse
Affiliation(s)
| | - Jeremy W Prokop
- HudsonAlpha Institute, Huntsville, AL, USA
- Department of Pediatrics and Human Development, Department of Pharmacology, Michigan State University, Grand Rapids, MI, USA
| | - Hong He
- Departments of Pediatrics and Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Katie Holl
- Departments of Pediatrics and Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - John Littrell
- Departments of Pediatrics and Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Aaron W Deal
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Yunjung Kim
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Patrick B Kyle
- Department of Pharmacology, Medicine (Nephrology), Pediatrics (Genetics), University of Mississippi Medical Center, Jackson, MS, USA
- Department of Pathology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Esinam Attipoe
- Department of Pharmacology, Medicine (Nephrology), Pediatrics (Genetics), University of Mississippi Medical Center, Jackson, MS, USA
| | - Ashley C Johnson
- Department of Pharmacology, Medicine (Nephrology), Pediatrics (Genetics), University of Mississippi Medical Center, Jackson, MS, USA
| | - Katie L Uhl
- Department of Pediatrics and Human Development, Department of Pharmacology, Michigan State University, Grand Rapids, MI, USA
| | - Olivia L Sirpilla
- Department of Pediatrics and Human Development, Department of Pharmacology, Michigan State University, Grand Rapids, MI, USA
| | - Seyedehameneh Jahanbakhsh
- Department of Pediatrics and Human Development, Department of Pharmacology, Michigan State University, Grand Rapids, MI, USA
| | | | - Shawn Levy
- HudsonAlpha Institute, Huntsville, AL, USA
| | - William Valdar
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Michael R Garrett
- Department of Pharmacology, Medicine (Nephrology), Pediatrics (Genetics), University of Mississippi Medical Center, Jackson, MS, USA
| | - Leah C Solberg Woods
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
10
|
Szpirer C. Rat models of human diseases and related phenotypes: a systematic inventory of the causative genes. J Biomed Sci 2020; 27:84. [PMID: 32741357 PMCID: PMC7395987 DOI: 10.1186/s12929-020-00673-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 07/09/2020] [Indexed: 12/13/2022] Open
Abstract
The laboratory rat has been used for a long time as the model of choice in several biomedical disciplines. Numerous inbred strains have been isolated, displaying a wide range of phenotypes and providing many models of human traits and diseases. Rat genome mapping and genomics was considerably developed in the last decades. The availability of these resources has stimulated numerous studies aimed at discovering causal disease genes by positional identification. Numerous rat genes have now been identified that underlie monogenic or complex diseases and remarkably, these results have been translated to the human in a significant proportion of cases, leading to the identification of novel human disease susceptibility genes, helping in studying the mechanisms underlying the pathological abnormalities and also suggesting new therapeutic approaches. In addition, reverse genetic tools have been developed. Several genome-editing methods were introduced to generate targeted mutations in genes the function of which could be clarified in this manner [generally these are knockout mutations]. Furthermore, even when the human gene causing a disease had been identified without resorting to a rat model, mutated rat strains (in particular KO strains) were created to analyze the gene function and the disease pathogenesis. Today, over 350 rat genes have been identified as underlying diseases or playing a key role in critical biological processes that are altered in diseases, thereby providing a rich resource of disease models. This article is an update of the progress made in this research and provides the reader with an inventory of these disease genes, a significant number of which have similar effects in rat and humans.
Collapse
Affiliation(s)
- Claude Szpirer
- Université Libre de Bruxelles, B-6041, Gosselies, Belgium.
- , Waterloo, Belgium.
| |
Collapse
|
11
|
Garrett MR, Korstanje R. Using Genetic and Species Diversity to Tackle Kidney Disease. Trends Genet 2020; 36:499-509. [PMID: 32362446 DOI: 10.1016/j.tig.2020.04.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 03/26/2020] [Accepted: 04/03/2020] [Indexed: 12/18/2022]
Abstract
Progress in the identification of causal genes and understanding of the mechanism underlying kidney disease is hindered by the almost exclusive use of a few animal models with restrictive monogenic backgrounds that may be more resistant to kidney disease compared with humans and, therefore, poor models. Exploring the large genetic diversity in classical animal models, such as mice and rats, and leveraging species diversity will allow us to use the genetic advantages of zebrafish, Drosophila, and other species, to develop both new animal models that are more relevant to the study of human kidney disease and potential therapies.
Collapse
Affiliation(s)
- Michael R Garrett
- Department of Pharmacology, University of Mississippi Medical Center, Jackson, MS, USA; Department of Medicine (Nephrology), University of Mississippi Medical Center, Jackson, MS, USA; Department of Pediatrics (Genetics), University of Mississippi Medical Center, Jackson, MS, USA
| | - Ron Korstanje
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, Maine, ME, USA; Mount Desert Island Biological Laboratory, Bar Harbor, Maine, ME, USA.
| |
Collapse
|
12
|
Johnson AC, Wu W, Attipoe EM, Sasser JM, Taylor EB, Showmaker KC, Kyle PB, Lindsey ML, Garrett MR. Loss of Arhgef11 in the Dahl Salt-Sensitive Rat Protects Against Hypertension-Induced Renal Injury. Hypertension 2020; 75:1012-1024. [PMID: 32148127 DOI: 10.1161/hypertensionaha.119.14338] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Arhgef11 is a Rho-guanine nucleotide exchange factor that was previously implicated in kidney injury in the Dahl salt-sensitive (SS) rat, a model of hypertension-related chronic kidney disease. Reduced Arhgef11 expression in an SS-Arhgef11SHR-minimal congenic strain (spontaneously hypertensive rat allele substituted for S allele) significantly decreased proteinuria, fibrosis, and improved renal hemodynamics, without impacting blood pressure compared with the control SS (SS-wild type). Here, SS-Arhgef11-/- and SS-wild type rats were placed on either low or elevated salt (0.3% or 2% NaCl) from 4 to 12 weeks of age. On low salt, starting at week 6 and through week 12, SS-Arhgef11-/- animals demonstrated a 3-fold decrease in proteinuria compared with SS-wild type. On high salt, beginning at week 6, SS-Arhgef11-/- animals demonstrated >2-fold lower proteinuria from weeks 8 to 12 and 30 mm Hg lower BP compared with SS-wild type. To better understand the molecular mechanisms of the renal protection from loss of Arhgef11, both RNA sequencing and discovery proteomics were performed on kidneys from week 4 (before onset of renal injury/proteinuria between groups) and at week 12 (low salt). The omics data sets revealed loss of Arhgef11 (SS-Arhgef11-/-) initiates early transcriptome/protein changes in the cytoskeleton starting as early as week 4 that impact a number of cellular functions, including actin cytoskeletal regulation, mitochondrial metabolism, and solute carrier transporters. In summary, in vivo phenotyping coupled with a multi-omics approach provides strong evidence that increased Arhgef11 expression in the Dahl SS rat leads to actin cytoskeleton-mediated changes in cell morphology and cell function that promote kidney injury, hypertension, and decline in kidney function.
Collapse
Affiliation(s)
- Ashley C Johnson
- From the Department of Pharmacology and Toxicology (A.C.J., W.W., E.M.A., J.M.S., M.R.G., K.C.S.), University of Mississippi Medical Center
| | - Wenjie Wu
- From the Department of Pharmacology and Toxicology (A.C.J., W.W., E.M.A., J.M.S., M.R.G., K.C.S.), University of Mississippi Medical Center
| | - Esinam M Attipoe
- From the Department of Pharmacology and Toxicology (A.C.J., W.W., E.M.A., J.M.S., M.R.G., K.C.S.), University of Mississippi Medical Center
| | - Jennifer M Sasser
- From the Department of Pharmacology and Toxicology (A.C.J., W.W., E.M.A., J.M.S., M.R.G., K.C.S.), University of Mississippi Medical Center
| | - Erin B Taylor
- Department of Physiology (E.B.T., M.L.L.), University of Mississippi Medical Center
| | - Kurt C Showmaker
- From the Department of Pharmacology and Toxicology (A.C.J., W.W., E.M.A., J.M.S., M.R.G., K.C.S.), University of Mississippi Medical Center
| | - Patrick B Kyle
- Department of Pathology (P.B.K.), University of Mississippi Medical Center
| | - Merry L Lindsey
- Department of Physiology (E.B.T., M.L.L.), University of Mississippi Medical Center
| | - Michael R Garrett
- From the Department of Pharmacology and Toxicology (A.C.J., W.W., E.M.A., J.M.S., M.R.G., K.C.S.), University of Mississippi Medical Center.,Department of Medicine (Nephrology) (M.R.G.), University of Mississippi Medical Center
| |
Collapse
|
13
|
Kumar V, Yang C, Cowley AW. Temporal Expression and Cellular Localization of PAPPA2 in the Developing Kidney of Rat. J Histochem Cytochem 2020; 68:209-222. [PMID: 31989854 DOI: 10.1369/0022155420904478] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
PAPPA2 is a metalloproteinase which cleaves insulin-like growth factor binding protein (IGFBP)-3 and IGFBP-5, and its role in pregnancy and postnatal growth is primarily studied. Using exclusion mapping, we reported a subcongenic (26-P) rat where a 0.71-Mbp region containing the pregnancy-associated plasma protein a2 (Pappa2) allele of salt-insensitive Brown Norway (BN) was introgressed into Dahl saltsensitive (SS) genetic background, resulting in the reduction of salt sensitivity. Pappa2 was differentially expressed in the adult kidney of 26-P and SS rats. Here, the expression and cellular localization of Pappa2 in embryonic and postnatal kidneys of 26-P and SS rats were examined. Pappa2 mRNA expression was 5-fold higher in the embryonic kidney (day 20.5) of the 26-P rat compared with the SS rat. Pappa2 mRNA expression progressively increased with the development of kidney, reaching a peak at postnatal day 5 before trending downward in subsequent stages of development in both strains. At all tested time points, Pappa2 remained higher in the 26-P compared with the SS rat kidney. Immunohistochemistry studies localized PAPPA2 in the ureteric bud (UB) and distal part of S-shaped body. PAPPA2 was colocalized with IGFBP-5 in the UB and Na+/K+/2Cl- cotransporter-stained tubules, respectively. Future studies are needed to determine the role of Pappa2 in kidney development and mechanistic pathways involved in this process.
Collapse
Affiliation(s)
- Vikash Kumar
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Chun Yang
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Allen W Cowley
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
14
|
Rapp JP, Garrett MR. Will the real Dahl S rat please stand up? Am J Physiol Renal Physiol 2019; 317:F1231-F1240. [PMID: 31545925 PMCID: PMC6879929 DOI: 10.1152/ajprenal.00359.2019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 09/12/2019] [Accepted: 09/16/2019] [Indexed: 11/22/2022] Open
Affiliation(s)
- John P Rapp
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio
| | - Michael R Garrett
- Department of Pharmacology, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
15
|
Spires D, Poudel B, Shields CA, Pennington A, Fizer B, Taylor L, McPherson KC, Cornelius DC, Williams JM. Prevention of the progression of renal injury in diabetic rodent models with preexisting renal disease with chronic endothelin A receptor blockade. Am J Physiol Renal Physiol 2018; 315:F977-F985. [PMID: 29846112 DOI: 10.1152/ajprenal.00182.2018] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The endothelin (ET) system has emerged as a therapeutic target for the treatment of diabetic nephropathy (DN). The present study examined whether chronic endothelin A (ETA) receptor blockade with atrasentan prevents the progression of renal injury in two models of DN with preexisting renal disease that exhibit an increased renal ET-1 system compared with nondiabetic rats: streptozotocin-treated Dahl salt-sensitive (STZ-SS) and type 2 diabetic nephropathy (T2DN) rats. Nine week-old SS rats were treated with (STZ; 50 mg/kg ip) to induce diabetes. After 3 wk of diabetes, proteinuria increased to 353 ± 34 mg/day. The rats were then separated into two groups: 1) vehicle and 2) atrasentan (5 mg·kg-1·day-1) via drinking water. After 6 wk of treatment with atrasentan, mean arterial pressure (MAP) and proteinuria decreased by 12 and 40%, respectively, in STZ-SS rats. The degree of glomerulosclerosis and renal fibrosis was significantly reduced in the kidneys of atrasentan-treated STZ-SS rats compared with vehicle STZ-SS rats. Interestingly, treatment with atrasentan did not affect GFR but significantly increased renal blood flow by 33% and prevented the elevations in filtration fraction and renal vascular resistance by 23 and 20%, respectively, in STZ-SS rats. In contrast to the STZ-SS study, atrasentan had no effect on MAP or proteinuria in T2DN rats. However, treatment with atrasentan significantly decreased glomerular injury and renal fibrosis and prevented the decline in renal function in T2DN rats. These data indicate that chronic ETA blockade produces advantageous changes in renal hemodynamics that slow the progression of renal disease and also reduces renal histopathology in the absence of reducing arterial pressure and proteinuria.
Collapse
Affiliation(s)
- Denisha Spires
- Departments of Pharmacology and Emergency Medicine, University of Mississippi Medical Center , Jackson, Mississippi
| | - Bibek Poudel
- Departments of Pharmacology and Emergency Medicine, University of Mississippi Medical Center , Jackson, Mississippi
| | - Corbin A Shields
- Departments of Pharmacology and Emergency Medicine, University of Mississippi Medical Center , Jackson, Mississippi
| | - Alyssa Pennington
- Departments of Pharmacology and Emergency Medicine, University of Mississippi Medical Center , Jackson, Mississippi
| | - Brianca Fizer
- Departments of Pharmacology and Emergency Medicine, University of Mississippi Medical Center , Jackson, Mississippi
| | - Lateia Taylor
- Departments of Pharmacology and Emergency Medicine, University of Mississippi Medical Center , Jackson, Mississippi
| | - Kasi C McPherson
- Departments of Pharmacology and Emergency Medicine, University of Mississippi Medical Center , Jackson, Mississippi
| | - Denise C Cornelius
- Departments of Pharmacology and Emergency Medicine, University of Mississippi Medical Center , Jackson, Mississippi
| | - Jan M Williams
- Departments of Pharmacology and Emergency Medicine, University of Mississippi Medical Center , Jackson, Mississippi
| |
Collapse
|
16
|
Velma V, Dasari SR, Tchounwou PB. Low Doses of Cisplatin Induce Gene Alterations, Cell Cycle Arrest, and Apoptosis in Human Promyelocytic Leukemia Cells. Biomark Insights 2016; 11:113-21. [PMID: 27594783 PMCID: PMC4998075 DOI: 10.4137/bmi.s39445] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 04/17/2016] [Accepted: 04/26/2016] [Indexed: 12/20/2022] Open
Abstract
Cisplatin is a known antitumor drug, but its mechanisms of action are not fully elucidated. In this research, we studied the anticancer potential of cisplatin at doses of 1, 2, or 3 µM using HL-60 cells as a test model. We investigated cisplatin effects at the molecular level using RNA sequencing, cell cycle analysis, and apoptotic assay after 24, 48, 72, and 96 hours of treatment. The results show that many genes responsible for molecular and cellular functions were significantly altered. Cisplatin treatment also caused the cells to be arrested at the DNA synthesis phase, and as the time increases, the cells gradually accumulated at the sub-G1 phase. Also, as the dose increases, a significant number of cells entered into the apoptotic and necrotic stages. Altogether, the data show that low doses of cisplatin significantly impact the viability of HL-60 cells, through modulation of gene expression, cell cycle, and apoptosis.
Collapse
Affiliation(s)
- Venkatramreddy Velma
- Cellomics and Toxicogenomics Research Laboratory, NIH/NIMHD RCMI-Center for Environmental Health, College of Science, Engineering and Technology, Jackson State University, Jackson, MS, USA
| | - Shaloam R Dasari
- Cellomics and Toxicogenomics Research Laboratory, NIH/NIMHD RCMI-Center for Environmental Health, College of Science, Engineering and Technology, Jackson State University, Jackson, MS, USA
| | - Paul B Tchounwou
- Cellomics and Toxicogenomics Research Laboratory, NIH/NIMHD RCMI-Center for Environmental Health, College of Science, Engineering and Technology, Jackson State University, Jackson, MS, USA
| |
Collapse
|
17
|
Elijovich F, Weinberger MH, Anderson CAM, Appel LJ, Bursztyn M, Cook NR, Dart RA, Newton-Cheh CH, Sacks FM, Laffer CL. Salt Sensitivity of Blood Pressure: A Scientific Statement From the American Heart Association. Hypertension 2016; 68:e7-e46. [PMID: 27443572 DOI: 10.1161/hyp.0000000000000047] [Citation(s) in RCA: 360] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
18
|
Wang X, Johnson AC, Sasser JM, Williams JM, Solberg Woods LC, Garrett MR. Spontaneous one-kidney rats are more susceptible to develop hypertension by DOCA-NaCl and subsequent kidney injury compared with uninephrectomized rats. Am J Physiol Renal Physiol 2016; 310:F1054-64. [PMID: 26936874 PMCID: PMC5002061 DOI: 10.1152/ajprenal.00555.2015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 02/25/2016] [Indexed: 12/12/2022] Open
Abstract
There is little clinical data of how hypertension may influence individuals with nephron deficiency in the context of being born with a single kidney. We recently developed a new rat model (the heterogeneous stock-derived model of unilateral renal agenesis rat) that is born with a single kidney and exhibits progressive kidney injury and decline in kidney function with age. We hypothesized that DOCA-salt would induce a greater increase in blood pressure and therefore accelerate the progression of kidney injury in rats born with a solitary kidney compared with rats that have undergone unilateral nephrectomy. Time course evaluation of blood pressure, kidney injury, and renal hemodynamics was performed in the following six groups of animals from weeks 13 to 18: 1) DOCA-treated rats with a solitary kidney (DOCA+S group), 2) placebo-treated rats with a solitary kidney, 3) DOCA-treated control rats with two kidneys (DOCA+C group), 4) placebo-treated control rats with two kidneys, 5) DOCA-treated rats with two kidneys that underwent uninephrectomy (DOCA+UNX8 group), and 6) placebo-treated rats with two kidneys that underwent uninephrectomy. DOCA+S rats demonstrated a significant rise (P < 0.05) in blood pressure (192 ± 4 mmHg), proteinuria (205 ± 31 mg/24 h), and a decline in glomerular filtration rate (600 ± 42 μl·min(-1)·g kidney weight(-1)) relative to the DOCA+UNX8 (173 ± 3 mmHg, 76 ± 26 mg/24 h, and 963 ± 36 μl·min(-1)·g kidney weight(-1)) and DOCA+C (154 ± 2 mmHg, 7 ± 1 mg/24 h, and 1,484 ± 121 μl·min(-1)·g kidney weight(-1)) groups. Placebo-treated groups showed no significant change among the three groups. An assessment of renal injury markers via real-time PCR/Western blot analysis and histological analysis was concordant with the measured physiological parameters. In summary, congenital solitary kidney rats are highly susceptible to the induction of hypertension compared with uninephrectomized rats, suggesting that low nephron endowment is an important driver of elevated blood pressure, hastening nephron injury through the transmission of elevated systemic blood pressure and thereby accelerating decline in kidney function.
Collapse
Affiliation(s)
- Xuexiang Wang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Ashley C Johnson
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Jennifer M Sasser
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Jan M Williams
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | | | - Michael R Garrett
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi; Department of Medicine (Nephrology), University of Mississippi Medical Center, Jackson, Mississippi; and
| |
Collapse
|
19
|
Abstract
The establishment and maintenance of epithelial cell-cell junctions is crucially important to regulate adhesion, apico-basal polarity and motility of epithelial cells, and ultimately controls the architecture and physiology of epithelial organs. Junctions are supported, shaped and regulated by cytoskeletal filaments, whose dynamic organization and contractility are finely tuned by GTPases of the Rho family, primarily RhoA, Rac1 and Cdc42. Recent research has identified new molecular mechanisms underlying the cross-talk between these GTPases and epithelial junctions. Here we briefly summarize the current knowledge about the organization, molecular evolution and cytoskeletal anchoring of cell-cell junctions, and we comment on the most recent advances in the characterization of the interactions between Rho GTPases and junctional proteins, and their consequences with regards to junction assembly and regulation of cell behavior in vertebrate model systems. The concept of “zonular signalosome” is proposed, which highlights the close functional relationship between proteins of zonular junctions (zonulae occludentes and adhaerentes) and the control of cytoskeletal organization and signaling through Rho GTPases, transcription factors, and their effectors.
Collapse
Key Words
- AJ, adherens junction
- AMOT, angiomotin
- AMPK, Adenosine Monophosphate-Activated Protein Kinase
- APC, adenomatous poliposis coli
- CD2AP, CD2-associated protein
- CGN, cingulin
- CGNL1, paracingulin
- Cdc42
- Cdc42, cell division cycle 42
- DLC, deleted in liver cancer
- Dbl, diffuse B-cell lymphoma
- EPLIN, epithelial protein lost in neoplasm
- ERK, extracellular regulated kinase
- FERM, four.point.one, ezrin, radixin, moesin
- FGD5, FYVE, RhoGEF and PH domain containing 5
- GAP, GTPase activating protein
- GEF, guanine nucleotide exchange factor
- GST, glutathione -S- transferase; JAM = junctional adhesion molecule
- MCF-7, Michigan Cancer Foundation - 7
- MDCK, Madin Darby Canine Kidney
- MKLP1, mitotic kinesin-like protein-1
- MRCK, myotonic dystrophy-related Cdc42-binding kinase
- MgcRacGAP, male germ cell racGAP
- PA, puncta adhaerentia
- PAK, p21-activated kinase; PATJ, Pals1 associated tight junction protein
- PCNA, proliferating cell nuclear antigen
- PDZ, Post synaptic density protein (PSD95), Drosophila, disc large tumour suppressor (DlgA), and zonula occludens-1
- PLEKHA7, pleckstrin homology domain containing, family A member 7
- RICH-1, RhoGAP interacting with CIP4 homologues
- ROCK, Rho-associated protein kinase
- Rac
- Rho
- SH3BP1, (SH3 domain 490 binding protein-1)
- TJ, tight junction
- Tbx-3, T-box-3
- Tiam, Tumor invasion and metastasis
- WASP, Wiskott-Aldrich Syndrome Protein
- WAVE, WASP family Verprolin-homologous protein
- ZA, zonula adhaerens
- ZO, zonula occludens
- ZONAB, (ZO-1)–associated nucleic acid binding protein.
- cytoseleton
- epithelium
- junctions
Collapse
Affiliation(s)
- Sandra Citi
- a Department of Cell Biology ; University of Geneva ; Geneva , Switzerland
| | | | | | | |
Collapse
|
20
|
Abstract
Emerging integrative analysis of genomic and anatomical imaging data which has not been well developed, provides invaluable information for the holistic discovery of the genomic structure of disease and has the potential to open a new avenue for discovering novel disease susceptibility genes which cannot be identified if they are analyzed separately. A key issue to the success of imaging and genomic data analysis is how to reduce their dimensions. Most previous methods for imaging information extraction and RNA-seq data reduction do not explore imaging spatial information and often ignore gene expression variation at the genomic positional level. To overcome these limitations, we extend functional principle component analysis from one dimension to two dimensions (2DFPCA) for representing imaging data and develop a multiple functional linear model (MFLM) in which functional principal scores of images are taken as multiple quantitative traits and RNA-seq profile across a gene is taken as a function predictor for assessing the association of gene expression with images. The developed method has been applied to image and RNA-seq data of ovarian cancer and kidney renal clear cell carcinoma (KIRC) studies. We identified 24 and 84 genes whose expressions were associated with imaging variations in ovarian cancer and KIRC studies, respectively. Our results showed that many significantly associated genes with images were not differentially expressed, but revealed their morphological and metabolic functions. The results also demonstrated that the peaks of the estimated regression coefficient function in the MFLM often allowed the discovery of splicing sites and multiple isoforms of gene expressions.
Collapse
|
21
|
Jia Z, Johnson AC, Wang X, Guo Z, Dreisbach AW, Lewin JR, Kyle PB, Garrett MR. Allelic Variants in Arhgef11 via the Rho-Rock Pathway Are Linked to Epithelial-Mesenchymal Transition and Contributes to Kidney Injury in the Dahl Salt-Sensitive Rat. PLoS One 2015; 10:e0132553. [PMID: 26172442 PMCID: PMC4501567 DOI: 10.1371/journal.pone.0132553] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 06/17/2015] [Indexed: 12/21/2022] Open
Abstract
Previously, genetic analyses identified that variants in Arhgef11 may influence kidney injury in the Dahl salt-sensitive (S) rat, a model of hypertensive chronic kidney disease. To understand the potential mechanism by which altered expression and/or protein differences in Arhgef11 could play a role in kidney injury, stably transduced Arhgef11 knockdown cell lines as well as primary cultures of proximal tubule cells were studied. Genetic knockdown of Arhgef11 in HEK293 and NRK resulted in reduced RhoA activity, decreased activation of Rho-ROCK pathway, and less stress fiber formation versus control, similar to what was observed by pharmacological inhibition (fasudil). Primary proximal tubule cells (PTC) cultured from the S exhibited increased expression of Arhgef11, increased RhoA activity, and up regulation of Rho-ROCK signaling compared to control (small congenic). The cells were also more prone (versus control) to TGFβ-1 induced epithelial-mesenchymal transition (EMT), a hallmark feature of the development of renal interstitial fibrosis, and characterized by development of spindle shape morphology, gene expression changes in EMT markers (Col1a3, Mmp9, Bmp7, and Ocln) and increased expression of N-Cadherin and Vimentin. S derived PTC demonstrated a decreased ability to uptake FITC-albumin compared to the small congenic in vitro, which was confirmed by assessment of albumin re-uptake in vivo by infusion of FITC-albumin and immunofluorescence imaging. In summary, these studies suggest that genetic variants in the S form of Arhgef11 via increased expression and/or protein activity play a role in promoting kidney injury in the S rat through changes in cell morphology (Rho-Rock and/or EMT) that impact the function of tubule cells.
Collapse
Affiliation(s)
- Zhen Jia
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, United States of America
| | - Ashley C. Johnson
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, United States of America
| | - Xuexiang Wang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, United States of America
| | - Zibiao Guo
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, United States of America
- Molecular and Genomics Core Facility, University of Mississippi Medical Center, Jackson, MS, United States of America
| | - Albert W. Dreisbach
- Department of Medicine (Nephrology), University of Mississippi Medical Center, Jackson, MS, United States of America
| | - Jack R. Lewin
- Department of Pathology, University of Mississippi Medical Center, Jackson, MS, United States of America
| | - Patrick B. Kyle
- Department of Pathology, University of Mississippi Medical Center, Jackson, MS, United States of America
| | - Michael R. Garrett
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, United States of America
- Department of Medicine (Nephrology), University of Mississippi Medical Center, Jackson, MS, United States of America
- * E-mail:
| |
Collapse
|
22
|
Samad MA, Kim UK, Kang JJ, Ke Q, Kang PM. Endothelin A receptor antagonist, atrasentan, attenuates renal and cardiac dysfunction in Dahl salt-hypertensive rats in a blood pressure independent manner. PLoS One 2015; 10:e0121664. [PMID: 25775254 PMCID: PMC4361570 DOI: 10.1371/journal.pone.0121664] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 02/02/2015] [Indexed: 02/02/2023] Open
Abstract
Proteinuria is a hallmark of chronic kidney disease (CKD) and cardiovascular disease (CVD), and a good predictor of clinical outcome. Selective endothelin A (ETA) receptor antagonist used with renin-angiotensin system (RAS) inhibitors prevents development of proteinuria in CKD. However, whether the improvement in proteinuria would have beneficial effects on CVD, independent of RAS inhibition, is not well understood. In this study, we investigated whether atrasentan, an ETA receptor antagonist, has renal and cardiovascular effects independent of RAS inhibition. Male Dahl salt sensitive (DSS) rats, at six weeks of age, received water with or without different doses of atrasentan and/or enalapril under high salt (HS) diet or normal diet (ND) for 6 weeks. At the end of 12th week, atrasentan at a moderate dose significantly attenuated proteinuria and serum creatinine without reducing mean arterial pressure (MAP), thereby preventing cardiac hypertrophy and improving cardiac function. ACE inhibitor enalapril at a dose that did not significantly lowered BP, attenuated cardiac hypertrophy while moderately improving cardiac function without reducing proteinuria and serum creatinine level. Nonetheless, combined therapy of atrasentan and enalapril that does not altering BP exerted additional cardioprotective effect. Based on these findings, we conclude that BP independent monotherapy of ETA receptor antagonist attenuates the progression of CKD and significantly mitigates CVD independent of RAS inhibition.
Collapse
Affiliation(s)
- Mohammed A. Samad
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of BIN Fusion Technology, Chonbuk National University, Jeonju, South Korea
| | - Ui Kyoung Kim
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Joshua J. Kang
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Qingen Ke
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Peter M. Kang
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of BIN Fusion Technology, Chonbuk National University, Jeonju, South Korea
- * E-mail:
| |
Collapse
|
23
|
Wang X, Johnson AC, Williams JM, White T, Chade AR, Zhang J, Liu R, Roman RJ, Lee JW, Kyle PB, Solberg-Woods L, Garrett MR. Nephron Deficiency and Predisposition to Renal Injury in a Novel One-Kidney Genetic Model. J Am Soc Nephrol 2014; 26:1634-46. [PMID: 25349207 DOI: 10.1681/asn.2014040328] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 09/08/2014] [Indexed: 11/03/2022] Open
Abstract
Some studies have reported up to 40% of patients born with a single kidney develop hypertension, proteinuria, and in some cases renal failure. The increased susceptibility to renal injury may be due, in part, to reduced nephron numbers. Notably, children who undergo nephrectomy or adults who serve as kidney donors exhibit little difference in renal function compared with persons who have two kidneys. However, the difference in risk between being born with a single kidney versus being born with two kidneys and then undergoing nephrectomy are unclear. Animal models used previously to investigate this question are not ideal because they require invasive methods to model congenital solitary kidney. In this study, we describe a new genetic animal model, the heterogeneous stock-derived model of unilateral renal agenesis (HSRA) rat, which demonstrates 50%-75% spontaneous incidence of a single kidney. The HSRA model is characterized by reduced nephron number (more than would be expected by loss of one kidney), early kidney/glomerular hypertrophy, and progressive renal injury, which culminates in reduced renal function. Long-term studies of temporal relationships among BP, renal hemodynamics, and renal function demonstrate that spontaneous single-kidney HSRA rats are more likely than uninephrectomized normal littermates to exhibit renal impairment because of the combination of reduced nephron numbers and prolonged exposure to renal compensatory mechanisms (i.e., hyperfiltration). Future studies with this novel animal model may provide additional insight into the genetic contributions to kidney development and agenesis and the factors influencing susceptibility to renal injury in individuals with congenital solitary kidney.
Collapse
Affiliation(s)
| | | | - Jan M Williams
- Departments of *Pharmacology and Toxicology, Medicine (Nephrology)
| | | | - Alejandro R Chade
- Physiology and Biophysics, Radiology, University of Mississippi Medical Center, Jackson, Mississippi; and
| | | | | | - Richard J Roman
- Departments of *Pharmacology and Toxicology, Medicine (Nephrology)
| | | | | | - Leah Solberg-Woods
- Department of Pediatrics, Medical College of Wisconsin, Madison, Wisconsin
| | | |
Collapse
|
24
|
Abstract
Rho kinase (ROCK) is a major downstream effector of the small GTPase RhoA. ROCK family, consisting of ROCK1 and ROCK2, plays central roles in the organization of actin cytoskeleton and is involved in a wide range of fundamental cellular functions, such as contraction, adhesion, migration, proliferation, and apoptosis. Due to the discovery of effective inhibitors, such as fasudil and Y27632, the biological roles of ROCK have been extensively explored with particular attention on the cardiovascular system. In many preclinical models of cardiovascular diseases, including vasospasm, arteriosclerosis, hypertension, pulmonary hypertension, stroke, ischemia-reperfusion injury, and heart failure, ROCK inhibitors have shown a remarkable efficacy in reducing vascular smooth muscle cell hypercontraction, endothelial dysfunction, inflammatory cell recruitment, vascular remodeling, and cardiac remodeling. Moreover, fasudil has been used in the clinical trials of several cardiovascular diseases. The continuing utilization of available pharmacological inhibitors and the development of more potent or isoform-selective inhibitors in ROCK signaling research and in treating human diseases are escalating. In this review, we discuss the recent molecular, cellular, animal, and clinical studies with a focus on the current understanding of ROCK signaling in cardiovascular physiology and diseases. We particularly note that emerging evidence suggests that selective targeting ROCK isoform based on the disease pathophysiology may represent a novel therapeutic approach for the disease treatment including cardiovascular diseases.
Collapse
|
25
|
Abstract
Arterial hypertension is a major health problem, accounting for 12 % of the global death rate. A large proportion of patients treated for high blood pressure do not reach target blood pressure values. The question arises if new antihypertensive drugs could improve present hypertension treatment. Rho-kinases (ROCKs) are ubiquitously expressed serine/threonine kinases and involved in a variety of cell functions. They contribute to the pathogenesis of human and experimental hypertension. Pharmacological ROCK inhibition has been shown to effectively lower blood pressure in patients and experimental animals. Progress has been made towards the understanding on how non-selective ROCK inhibitors lower arterial pressure and efforts are currently undertaken to develop ROCK inhibitors to improve their specificity and isoenzyme selectivity. If introduction of ROCK inhibitors for the treatment of high blood pressure can significantly advance currently available options of antihypertensive pharmacotherapy awaits further experimental and clinical research.
Collapse
Affiliation(s)
- Olaf Grisk
- Department of Physiology, University of Greifswald, Greifswalder Str. 11c, 17495, Greifswald, Karlsburg, Germany,
| |
Collapse
|
26
|
Westbrook L, Johnson AC, Regner KR, Williams JM, Mattson DL, Kyle PB, Henegar JR, Garrett MR. Genetic susceptibility and loss of Nr4a1 enhances macrophage-mediated renal injury in CKD. J Am Soc Nephrol 2014; 25:2499-510. [PMID: 24722447 DOI: 10.1681/asn.2013070786] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Nuclear hormone receptors of the NR4A subgroup have been implicated in cancer, atherosclerosis, and metabolic disease. However, little is known about the role of these receptors in kidney health or disease. Nr4a1-deficient rats (Nr4a1(-/-)) developed on a genetic background susceptible to kidney injury (fawn-hooded hypertensive rat [FHH]) were evaluated for BP, proteinuria, renal function, and metabolic parameters from 4 to 24 weeks-of-age. By week 24, Nr4a1(-/-) rats exhibited significantly higher proteinuria (approximately 4-fold) and decreased GFR compared with FHH controls. The severity of tubular atrophy, tubular casts, and interstitial fibrosis increased significantly in Nr4a1(-/-) rats and was accompanied by a large increase in immune cell infiltration, predominantly macrophages and to a lesser extent T cells and B cells. Global transcriptome and network analyses at weeks 8, 16, and 24 identified several proinflammatory genes and pathways differentially regulated between strains. Bone marrow crosstransplantation studies demonstrated that kidney injury in Nr4a1(-/-) rats was almost completely rescued by bone marrow transplanted from FHH controls. In vitro, macrophages isolated from Nr4a1(-/-) rats demonstrated increased immune activation compared with FHH-derived macrophages. In summary, the loss of Nr4a1 in immune cells appears to cause the increased kidney injury and reduced renal function observed in the Nr4a1(-/-) model.
Collapse
Affiliation(s)
| | | | | | - Jan M Williams
- Departments of Pharmacology and Toxicology, Medicine, and
| | - David L Mattson
- Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Patrick B Kyle
- Pathology, University of Mississippi Medical Center, Jackson, Mississippi; and
| | - Jeffery R Henegar
- Pathology, University of Mississippi Medical Center, Jackson, Mississippi; and
| | | |
Collapse
|
27
|
Brown Norway chromosome 1 congenic reduces symptoms of renal disease in fatty Zucker rats. PLoS One 2014; 9:e87770. [PMID: 24498189 PMCID: PMC3909223 DOI: 10.1371/journal.pone.0087770] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 12/30/2013] [Indexed: 11/19/2022] Open
Abstract
We previously reported that a congenic rat with Brown Norway (BN) alleles on chromosome 1 reduces renal disease of 15-week old fatty Zucker rats (ZUC). Development of renal disease in fatty BN congenic and fatty ZUC rats from 9 through 28 weeks is now examined. Analysis of urine metabolites by 1H nuclear magnetic resonance (NMR) spectroscopy revealed a significantly increased urinary loss of glucose, myo-inositol, urea, creatine, and valine in ZUC. Food intake was lower in the BN congenic rats at weeks 9–24, but they weighed significantly more at 28 weeks compared with the ZUC group. Fasting glucose was significantly higher in ZUC than congenic and adiponectin levels were significantly lower in ZUC, but there was no significant genotype effect on Insulin levels. Glucose tolerance tests exhibited no significant differences between ZUC and congenic when values were normalized to basal glucose levels. Quantitative PCR on livers revealed evidence for higher gluconeogenesis in congenics than ZUC at 9 weeks. Plasma urea nitrogen and creatinine were more than 2-fold higher in 28-week ZUC. Twelve urine protein markers of glomerular, proximal and distal tubule disease were assayed at three ages. Several proteins that indicate glomerular and proximal tubular disease increased with age in both congenic and ZUC. Epidermal growth factor (EGF) level, a marker whose levels decrease with distal tubule disease, was significantly higher in congenics. Quantitative histology of 28 week old animals revealed the most significant genotype effect was for tubular dilation and intratubular protein. The congenic donor region is protective of kidney disease, and effects on Type 2 diabetes are likely limited to fasting glucose and adiponectin. The loss of urea together with a small increase of food intake in ZUC support the hypothesis that nitrogen balance is altered in ZUC from an early age.
Collapse
|
28
|
Mouawad F, Tsui H, Takano T. Role of Rho-GTPases and their regulatory proteins in glomerular podocyte function. Can J Physiol Pharmacol 2013; 91:773-82. [PMID: 24144047 DOI: 10.1139/cjpp-2013-0135] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Podocytes play a critical role in maintaining glomerular permselectivity. It has been long recognized that their intricate actin-based structures are tightly associated with their normal function; however, the precise mechanisms by which podocytes form and maintain their complex structure had been poorly understood until the intensive investigations on podocyte biology began in 1998, triggered by the breakthrough discovery of nephrin. This review summarizes the recent discoveries of the molecular mechanisms by which the actin cytoskeleton is regulated in podocytes. A particular focus will be on the role of the Rho-family of small GTPases, represented by RhoA, Rac1, and Cdc42. Rho-GTPases are known for their versatile cellular functions, most importantly for the actin regulatory roles. We will also discuss the potential roles of the 3 groups of proteins known to regulate Rho-GTPases, namely GTPase-activating proteins, guanine nucleotide exchange factors, and guanine nucleotide dissociation inhibitors.
Collapse
Affiliation(s)
- Flaviana Mouawad
- Department of Medicine, McGill University Health Centre, Montreal, QC H3A 2B4, Canada
| | | | | |
Collapse
|
29
|
Johnson AC, Lee JW, Harmon AC, Morris Z, Wang X, Fratkin J, Rapp JP, Gomez-Sanchez E, Garrett MR. A mutation in the start codon of γ-crystallin D leads to nuclear cataracts in the Dahl SS/Jr-Ctr strain. Mamm Genome 2013; 24:95-104. [PMID: 23404175 PMCID: PMC3628938 DOI: 10.1007/s00335-013-9447-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Accepted: 12/17/2012] [Indexed: 10/27/2022]
Abstract
Cataracts are a major cause of blindness. The most common forms of cataracts are age- and UV-related and develop mostly in the elderly, while congenital cataracts appear at birth or in early childhood. The Dahl salt-sensitive (SS/Jr) rat is an extensively used model of salt-sensitive hypertension that exhibits concomitant renal disease. In the mid-1980s, cataracts appeared in a few animals in the Dahl S colony, presumably the result of a spontaneous mutation. The mutation was fixed and bred to establish the SS/Jr-Ctr substrain. The SS/Jr-Ctr substrain has been used exclusively by a single investigator to study the role of steroids and hypertension. Using a classical positional cloning approach, we localized the cataract gene with high resolution to a less than 1-Mbp region on chromosome 9 using an F1(SS/Jr-Ctr × SHR) × SHR backcross population. The 1-Mbp region contained only 13 genes, including 4 genes from the γ-crystallins (Cryg) gene family, which are known to play a role in cataract formation. All of the γ-crystallins were sequenced and a novel point mutation in the start codon (ATG → GTG) of the Crygd gene was identified. This led to the complete absence of the CRYGD protein in the eyes of the SS/Jr-Ctr strain. In summary, the identification of the genetic cause in this novel cataract model may provide an opportunity to better understand the development of cataracts, particularly in the context of hypertension.
Collapse
Affiliation(s)
- Ashley C. Johnson
- Department of Pharmacology, University of Mississippi Medical Center, Jackson, MS 39216
| | - Jonathan W. Lee
- Department of Pharmacology, University of Mississippi Medical Center, Jackson, MS 39216
| | - Ashlyn C. Harmon
- Department of Pharmacology, University of Mississippi Medical Center, Jackson, MS 39216
| | - Zaliya Morris
- Department of Pharmacology, University of Mississippi Medical Center, Jackson, MS 39216
| | - Xuexiang Wang
- Department of Pharmacology, University of Mississippi Medical Center, Jackson, MS 39216
| | - Jonathan Fratkin
- Department of Pathology, University of Mississippi Medical Center, Jackson, MS 39216
| | | | - Elise Gomez-Sanchez
- Department of Pharmacology, University of Mississippi Medical Center, Jackson, MS 39216
- GV(Sonny) Montgomery VAMC
| | - Michael R. Garrett
- Department of Pharmacology, University of Mississippi Medical Center, Jackson, MS 39216
- Department of Medicine (Nephrology), University of Mississippi Medical Center, Jackson, MS 39216
| |
Collapse
|