1
|
Du X, Tao Q, Fan S, Ren J, Dong Y, Li G, He S, Cao X, Zhu Y. Traditional Mongolian medicine Wu-Lan thirteen-flavor decoction protects rat from hypertension-induced renal injury via aryl hydrocarbon receptor-mediated pathway. Drug Dev Ind Pharm 2024; 50:952-967. [PMID: 39565140 DOI: 10.1080/03639045.2024.2432596] [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] [Received: 08/03/2024] [Revised: 11/02/2024] [Accepted: 11/14/2024] [Indexed: 11/21/2024]
Abstract
BACKGROUND Wu-Lan Thirteen-Flavour decoction (WLTd), a traditional Mongolian medicine, has been used for treating hypertension in clinical practice, but the chemical basis and underlying mechanisms remain unknown. METHODS The main components of WLTd were identified and quantified using HPLC and UPLC-MS/MS techniques. A compound-target-disease network was constructed using network pharmacology analysis to forecast the potential anti-hypertension targets. In vivo animal and in vitro cellular experiments were performed to validate the efficacy and molecular mechanisms of renal protection of WLTd and its main active components in spontaneous hypertension. RESULTS A total of 136 active compounds in WLTd were collected through relevant databases, and network pharmacology analysis identified that the aryl hydrocarbon receptor (AhR) signaling pathway may serve as a potential anti-hypertension targets. Eight of the active components, including vitexin, kaempferol, toosendanin, ursolic acid, matrine, oxymatrine, gardenoside and quercetin, were identified and quantified by HPLC and UPLC-MS/MS. WLTd effectively lowered the mean blood pressure (159.16 ± 13.91 vs 135 ± 13.37 mmHg), reduced the BUN (391.55 ± 59.96 vs 240.88 ± 51.15 mmol/L) and creatinine (1.78 ± 0.41 vs 0.67 ± 0.34 nmol/L) levels, and reduced hypertension-induced renal damage in SHR. AhR and related key gene expression changes predicted by network pharmacology analysis were validated by immunohistochemistry, RT-qPCR, and Western blot analyses. In vitro, studies also showed that WLTd up-regulated AhR expression in angiotensin II-induced HEK293 cell injury. CONCLUSIONS Wu-Lan Thirteen-Flavour decoction effectively protects hypertension-induced renal injury by regulating the Aryl Hydrocarbon Receptor signaling pathway.
Collapse
Affiliation(s)
- Xiaoli Du
- State Key Laboratory of Component-Based Chinese Medicine and Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Inner Mongolia Key laboratory of Chinese & Mongolian Medicine, Inner Mongolia Medical University, Hohhot, China
| | - Qianqian Tao
- State Key Laboratory of Component-Based Chinese Medicine and Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Siwen Fan
- State Key Laboratory of Component-Based Chinese Medicine and Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jun Ren
- Wuhai Inspection and Testing Center, Wuhai, China
| | - Yu Dong
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, China
| | - Gang Li
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, China
| | - Shuang He
- State Key Laboratory of Component-Based Chinese Medicine and Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaodong Cao
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, China
| | - Yan Zhu
- State Key Laboratory of Component-Based Chinese Medicine and Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
2
|
Buneeva OA, Fedchenko VI, Kaloshina SA, Zavyalova MG, Zgoda VG, Medvedev AE. Comparative proteomic analysis of renal tissue of normotensive and hypertensive rats. BIOMEDITSINSKAIA KHIMIIA 2024; 70:89-98. [PMID: 38711408 DOI: 10.18097/pbmc20247002089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Comparative proteomic analysis of kidney tissue from normotensive (WKY) and spontaneously hypertensive (SHR) rats revealed quantitative and qualitative changes in renal proteins. The number of renal proteins specific for WKY rats (blood pressure 110-120 mm Hg) was 13-16. There were 20-24 renal proteins specific for SHR (blood pressure 180 mm Hg and more). The total number of identified renal proteins common for both rat strains included 972-975 proteins. A pairwise comparison of all possible (SHR-WKY) variants identified 8 proteins specific only for normotensive (WKY) animals, and 7 proteins specific only for hypertensive ones (SHR). Taking into consideration their biological roles, the lack of some enzyme proteins in hypertensive rats (for example, biliverdin reductase A) reduces the production of molecules exhibiting antihypertensive properties, while the appearance of others (e.g. betaine-homocysteine S-methyltransferase 2, septin 2, etc.) can be interpreted as a compensatory reaction. Renal proteins with altered relative content (with more than 2.5-fold change) accounted for no more than 5% of all identified proteins. Among the proteins with an increased relative content in hypertensive animals, the largest group consisted of proteins involved in the processes of energy generation and carbohydrate metabolism, as well as antioxidant and protective proteins. In the context of the development of hypertension, the identified relative changes can apparently be considered compensatory. Among the proteins with the most pronounced decrease in the relative content in hypertensive rats, the dramatic reduction in acyl-CoA medium-chain synthetase-3 (ACSM3) appears to make an important contribution to the development of renal pathology in these animals.
Collapse
Affiliation(s)
- O A Buneeva
- Institute of Biomedical Chemistry, Moscow, Russia
| | | | | | | | - V G Zgoda
- Institute of Biomedical Chemistry, Moscow, Russia
| | - A E Medvedev
- Institute of Biomedical Chemistry, Moscow, Russia
| |
Collapse
|
3
|
Dhande IS, Zhu Y, Joshi AS, Hicks MJ, Braun MC, Doris PA. Polygenic genetic variation affecting antibody formation underlies hypertensive renal injury in the stroke-prone spontaneously hypertensive rat. Am J Physiol Renal Physiol 2023; 325:F317-F327. [PMID: 37439198 PMCID: PMC10511163 DOI: 10.1152/ajprenal.00058.2023] [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: 03/14/2023] [Revised: 07/07/2023] [Accepted: 07/07/2023] [Indexed: 07/14/2023] Open
Abstract
During development of the spontaneously hypertensive rat (SHR), several distinct but closely related lines were generated. Most lines are resistant to hypertensive renal disease. However, the SHR-A3 line (stroke-prone SHR) experiences end-organ injury (EOI) and provides a model of injury susceptibility that can be used to uncover genetic causation. In the present study, we generated a congenic line in which three distinct disease loci in SHR-A3 are concurrently replaced with homologous loci from an injury-resistant SHR line (SHR-B2). Verification that all three loci were homozygously replaced in this triple congenic line [SHR-A3(Trip B2)] while the genetic background of SHR-A3 was fully retained was obtained by whole genome sequencing. Congenic genome substitution was without effect on systolic blood pressure [198.9 ± 3.34 mmHg, mean ± SE, SHR-A3(Trip B2) = 194.7 ± 2.55 mmHg]. Measures of renal injury (albuminuria, histological injury scores, and urinary biomarker levels) were reduced in SHR-A3(Trip B2) animals, even though only 4.5 Mbases of the 2.8 Gbases of the SHR-B2 genome (0.16% of the genome) was transferred into the congenic line. The gene content of the three congenic loci and the functional effects of gene polymorphism within suggest a role of immunoglobulin in EOI pathogenesis. To prove the role of antibodies in EOI in SHR-A3, we generated an SHR-A3 line in which expression from the immunoglobulin heavy chain gene was knocked out (SHR-A3-IGHKO). Animals in the SHR-A3-IGHKO line lack B cells and immunoglobulin, but the hypertensive phenotype is not affected. Renal injury, however, was reduced in this line, confirming a pathogenic role for immunoglobulin in hypertensive EOI in this model of heritable risk.NEW & NOTEWORTHY Here, we used a polygenic animal model of hypertensive renal disease to show that genetic variation affecting antibody formation underlies hypertensive renal disease. We proved the genetic thesis by generating an immunoglobulin knockout in the susceptible animal model.
Collapse
Affiliation(s)
- Isha S Dhande
- Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, United States
| | - Yaming Zhu
- Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, United States
| | - Aniket S Joshi
- Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, United States
| | - M John Hicks
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, United States
| | - Michael C Braun
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, Texas, United States
| | - Peter A Doris
- Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, United States
| |
Collapse
|
4
|
Ansari A, Walton SL, Denton KM. Sex- and age-related differences in renal and cardiac injury and senescence in stroke-prone spontaneously hypertensive rats. Biol Sex Differ 2023; 14:33. [PMID: 37217968 DOI: 10.1186/s13293-023-00519-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 05/05/2023] [Indexed: 05/24/2023] Open
Abstract
BACKGROUND Sex differences play a critical role in the incidence and severity of cardiovascular diseases, whereby men are at a higher risk of developing cardiovascular disease compared to age-matched premenopausal women. Marked sex differences at the cellular and tissue level may contribute to susceptibility to cardiovascular disease and end-organ damage. In this study, we have performed an in-depth histological analysis of sex differences in hypertensive cardiac and renal injury in middle-aged stroke-prone spontaneously hypertensive rats (SHRSPs) to determine the interaction between age, sex and cell senescence. METHODS Kidneys, hearts and urine samples were collected from 6.5- and 8-month-old (Mo) male and female SHRSPs. Urine samples were assayed for albumin and creatinine content. Kidneys and hearts were screened for a suite of cellular senescence markers (senescence-associated β-galactosidase, p16INK4a, p21, γH2AX). Renal and cardiac fibrosis was quantified using Masson's trichrome staining, and glomerular hypertrophy and sclerosis were quantified using Periodic acid-Schiff staining. RESULTS Marked renal and cardiac fibrosis, concomitant with albuminuria, were evident in all SHRSPs. These sequelae were differentially affected by age, sex and organ. That is, the level of fibrosis was greater in the kidney than the heart, males had greater levels of fibrosis than females in both the heart and kidney, and even a 6-week increase in age resulted in greater levels of kidney fibrosis in males. The differences in kidney fibrosis were reflected by elevated levels of cellular senescence in the kidney in males but not females. Senescent cell burden was significantly less in cardiac tissue compared to renal tissue and was not affected by age or sex. CONCLUSIONS Our study demonstrates a clear sex pattern in age-related progression of renal and cardiac fibrosis and cellular senescence in SHRSP rats. A 6-week time frame was associated with increased indices of cardiac and renal fibrosis and cellular senescence in male SHRSPs. Female SHRSP rats were protected from renal and cardiac damage compared to age-matched males. Thus, the SHRSP is an ideal model to investigate the effects of sex and aging on organ injury over a short timeframe.
Collapse
Affiliation(s)
- Aneesa Ansari
- Department of Physiology, Monash University, Melbourne, VIC, Australia
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Sarah L Walton
- Department of Physiology, Monash University, Melbourne, VIC, Australia
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Kate M Denton
- Department of Physiology, Monash University, Melbourne, VIC, Australia.
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia.
| |
Collapse
|
5
|
Kourpa A, Schulz A, Mangelsen E, Kaiser-Graf D, Koppers N, Stoll M, Rothe M, Bader M, Purfürst B, Kunz S, Gladytz T, Niendorf T, Bachmann S, Mutig K, Bolbrinker J, Panáková D, Kreutz R. Studies in Zebrafish and Rat Models Support Dual Blockade of EP2 and EP4 (Prostaglandin E 2 Receptors Type 2 and 4) for Renoprotection in Glomerular Hyperfiltration and Albuminuria. Hypertension 2023; 80:771-782. [PMID: 36715011 DOI: 10.1161/hypertensionaha.122.20392] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Glomerular hyperfiltration (GH) is an important mechanism in the development of albuminuria in hypertension. Upregulation of COX2 (cyclooxygenase 2) and prostaglandin E2 (PGE2) was linked to podocyte damage in GH. We explored the potential renoprotective effects of either separate or combined pharmacological blockade of EP2 (PGE2 receptor type 2) and EP4 (PGE2 receptor type 4) in GH. METHODS We conducted in vivo studies in a transgenic zebrafish model (Tg[fabp10a:gc-EGFP]) suitable for analysis of glomerular filtration barrier function and a genetic rat model with GH, albuminuria, and upregulation of PGE2. Similar pharmacological interventions and primary outcome analysis on albuminuria phenotype development were conducted in both model systems. RESULTS Stimulation of zebrafish embryos with PGE2 induced an albuminuria-like phenotype, thus mimicking the suggested PGE2 effects on glomerular filtration barrier dysfunction. Both separate and combined blockade of EP2 and EP4 reduced albuminuria phenotypes in zebrafish and rat models. A significant correlation between albuminuria and podocyte damage in electron microscopy imaging was identified in the rat model. Dual blockade of both receptors showed a pronounced synergistic suppression of albuminuria. Importantly, this occurred without changes in arterial blood pressure, glomerular filtration rate, or tissue oxygenation in magnetic resonance imaging, while RNA sequencing analysis implicated a potential role of circadian clock genes. CONCLUSIONS Our findings confirm a role of PGE2 in the development of albuminuria in GH and support the renoprotective potential of combined pharmacological blockade of EP2 and EP4 receptors. These data support further translational research to explore this therapeutic option and a possible role of circadian clock genes.
Collapse
Affiliation(s)
- Aikaterini Kourpa
- Institute of Clinical Pharmacology and Toxicology (A.K., A.S., E.M., D.K.-G., J.B., R.K.), Charité-Universitätsmedizin Berlin, Germany.,Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (A.K., M.B., B.P., S.K., T.G., T.N., D.P.)
| | - Angela Schulz
- Institute of Clinical Pharmacology and Toxicology (A.K., A.S., E.M., D.K.-G., J.B., R.K.), Charité-Universitätsmedizin Berlin, Germany
| | - Eva Mangelsen
- Institute of Clinical Pharmacology and Toxicology (A.K., A.S., E.M., D.K.-G., J.B., R.K.), Charité-Universitätsmedizin Berlin, Germany
| | - Debora Kaiser-Graf
- Institute of Clinical Pharmacology and Toxicology (A.K., A.S., E.M., D.K.-G., J.B., R.K.), Charité-Universitätsmedizin Berlin, Germany
| | - Nils Koppers
- Genetic Epidemiology, Institute for Human Genetics, Westfälische Wilhelms University, Münster, Germany (N.K., M.S.)
| | - Monika Stoll
- Genetic Epidemiology, Institute for Human Genetics, Westfälische Wilhelms University, Münster, Germany (N.K., M.S.)
| | | | - Michael Bader
- Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (A.K., M.B., B.P., S.K., T.G., T.N., D.P.).,German Center for Cardiovascular Research, Partner Site Berlin, Germany (M.B.).,Charité-Universitätsmedizin Berlin, Germany (M.B.).,Institute for Biology, University of Lübeck, Germany (M.B.)
| | - Bettina Purfürst
- Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (A.K., M.B., B.P., S.K., T.G., T.N., D.P.)
| | - Severine Kunz
- Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (A.K., M.B., B.P., S.K., T.G., T.N., D.P.)
| | - Thomas Gladytz
- Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (A.K., M.B., B.P., S.K., T.G., T.N., D.P.)
| | - Thoralf Niendorf
- Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (A.K., M.B., B.P., S.K., T.G., T.N., D.P.)
| | - Sebastian Bachmann
- Institute of Vegetative Anatomy (S.B.), Charité-Universitätsmedizin Berlin, Germany
| | - Kerim Mutig
- Institute of Translational Physiology (K.M.), Charité-Universitätsmedizin Berlin, Germany
| | - Juliane Bolbrinker
- Institute of Clinical Pharmacology and Toxicology (A.K., A.S., E.M., D.K.-G., J.B., R.K.), Charité-Universitätsmedizin Berlin, Germany
| | - Daniela Panáková
- Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (A.K., M.B., B.P., S.K., T.G., T.N., D.P.)
| | - Reinhold Kreutz
- Institute of Clinical Pharmacology and Toxicology (A.K., A.S., E.M., D.K.-G., J.B., R.K.), Charité-Universitätsmedizin Berlin, Germany
| |
Collapse
|
6
|
Liu J, Sato Y, Falcone-Juengert J, Kurisu K, Shi J, Yenari MA. Sexual dimorphism in immune cell responses following stroke. Neurobiol Dis 2022; 172:105836. [PMID: 35932990 DOI: 10.1016/j.nbd.2022.105836] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 07/11/2022] [Accepted: 07/31/2022] [Indexed: 11/22/2022] Open
Abstract
Recent bodies of work in regard to stroke have revealed significant sex differences in terms of risk and outcome. While differences in sex hormones have been the focus of earlier research, the reasons for these differences are much more complex and require further identification. This review covers differences in sex related immune responses with a focus on differences in immune cell composition and function. While females are more susceptible to immune related diseases, they seem to have better outcomes from stroke at the experimental level with reduced pro-inflammatory responses. However, at the clinical level, the picture is much more complex with worse neurological outcomes from stroke. While the use of exogenous sex steroids can replicate some of these findings, it is apparent that many other factors are involved in the modulation of immune responses. As a result, more research is needed to better understand these differences and identify appropriate interventions and risk modification.
Collapse
Affiliation(s)
- Jialing Liu
- Dept Neurosurgery, UCSF and SF VAMC, San Francisco, CA, USA
| | - Yoshimichi Sato
- Dept Neurosurgery, UCSF and SF VAMC, San Francisco, CA, USA; Dept Neurosurgery, Tohoku University, Sendai, Japan
| | | | - Kota Kurisu
- Dept Neurosurgery, Hokkaido University, Sapporo, Japan
| | - Jian Shi
- Dept Neurology, UCSF and SF VAMC, San Francisco, CA, USA
| | | |
Collapse
|
7
|
de Jong TV, Chen H, Brashear WA, Kochan KJ, Hillhouse AE, Zhu Y, Dhande IS, Hudson EA, Sumlut MH, Smith ML, Kalbfleisch TS, Doris PA. mRatBN7.2: familiar and unfamiliar features of a new rat genome reference assembly. Physiol Genomics 2022; 54:251-260. [PMID: 35543507 PMCID: PMC9236863 DOI: 10.1152/physiolgenomics.00017.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Rat genomic tools have been slower to emerge than for those of humans and mice and have remained less thorough and comprehensive. The arrival of a new and improved rat reference genome, mRatBN7.2, in late 2020 is a welcome event. This assembly, like predecessor rat reference assemblies, is derived from an inbred Brown Norway rat. In this "user" survey we hope to provide other users of this assembly some insight into its characteristics and some assessment of its improvements as well as a few caveats that arise from the unique aspects of this assembly. mRatBN7.2 was generated by the Wellcome Sanger Institute as part of the large Vertebrate Genomes Project. This rat assembly has now joined human, mouse, chicken, and zebrafish in the National Center for Biotechnology Information (NCBI)'s Genome Reference Consortium, which provides ongoing curation of the assembly. Here we examine the technical procedures by which the assembly was created and assess how this assembly constitutes an improvement over its predecessor. We also indicate the technical limitations affecting the assembly, providing illustrations of how these limitations arise and the impact that results for this reference assembly.
Collapse
Affiliation(s)
- Tristan V. de Jong
- 1Department of Pharmacology, Addiction Science and Toxicology, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Hao Chen
- 1Department of Pharmacology, Addiction Science and Toxicology, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Wesley A. Brashear
- 2Texas A&M Institute for Genome Sciences and Society, Texas A&M University, College Station, Texas
| | - Kelli J. Kochan
- 2Texas A&M Institute for Genome Sciences and Society, Texas A&M University, College Station, Texas
| | - Andrew E. Hillhouse
- 2Texas A&M Institute for Genome Sciences and Society, Texas A&M University, College Station, Texas
| | - Yaming Zhu
- 3Center for Human Genetics, Brown Foundation Institute of Molecular Medicine, University of Texas McGovern School of Medicine, Houston, Texas
| | - Isha S. Dhande
- 3Center for Human Genetics, Brown Foundation Institute of Molecular Medicine, University of Texas McGovern School of Medicine, Houston, Texas
| | - Elizabeth A. Hudson
- 4Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, Kentucky
| | - Mary H. Sumlut
- 4Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, Kentucky
| | - Melissa L. Smith
- 4Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, Kentucky
| | - Theodore S. Kalbfleisch
- 5Department of Veterinary Science, College of Agriculture, Food, and Environment, University of Kentucky, Lexington, Kentucky
| | - Peter A. Doris
- 3Center for Human Genetics, Brown Foundation Institute of Molecular Medicine, University of Texas McGovern School of Medicine, Houston, Texas
| |
Collapse
|
8
|
Dhande IS, Braun MC, Doris PA. Emerging Insights Into Chronic Renal Disease Pathogenesis in Hypertension From Human and Animal Genomic Studies. Hypertension 2021; 78:1689-1700. [PMID: 34757770 PMCID: PMC8577298 DOI: 10.1161/hypertensionaha.121.18112] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The pathogenic links between elevated blood pressure and chronic kidney disease remain obscure. This article examines progress in population genetics and in animal models of hypertension and chronic kidney disease. It also provides a critique of the application of genome-wide association studies to understanding the heritability of renal function. Emerging themes identified indicate that heritable risk of chronic kidney disease in hypertension can arise from genetic variation in (1) glomerular and tubular protein handling mechanisms; (2) autoregulatory capacity of the renal vasculature; and (3) innate and adaptive immune mechanisms. Increased prevalence of hypertension-associated chronic kidney disease that occurs with aging may reflect amplification of heritable risks by normal aging processes affecting immunity and autoregulation.
Collapse
Affiliation(s)
- Isha S. Dhande
- Center for Human Genetics, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas HSC, Houston (I.S.D., P.A.D.)
| | - Michael C. Braun
- Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston (M.C.B.)
| | - Peter A. Doris
- Center for Human Genetics, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas HSC, Houston (I.S.D., P.A.D.)
| |
Collapse
|
9
|
Zhang C, Fang X, Zhang H, Gao W, Hsu HJ, Roman RJ, Fan F. Genetic susceptibility of hypertension-induced kidney disease. Physiol Rep 2021; 9:e14688. [PMID: 33377622 PMCID: PMC7772938 DOI: 10.14814/phy2.14688] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 11/22/2020] [Accepted: 11/27/2020] [Indexed: 02/06/2023] Open
Abstract
Hypertension is the second leading cause of end-stage renal disease (ESRD) after diabetes mellitus. The significant differences in the incidence of hypertensive ESRD between different patient populations worldwide and patients with and without family history indicate that genetic determinants play an important role in the onset and progression of this disease. Recent studies have identified genetic variants and pathways that may contribute to the alteration of renal function. Mechanisms involved include affecting renal hemodynamics (the myogenic and tubuloglomerular feedback responses); increasing the production of reactive oxygen species in the tubules; altering immune cell function; changing the number, structure, and function of podocytes that directly cause glomerular damage. Studies with hypertensive animal models using substitution mapping and gene knockout strategies have identified multiple candidate genes associated with the development of hypertension and subsequent renal injury. Genome-wide association studies have implicated genetic variants in UMOD, MYH9, APOL-1, SHROOM3, RAB38, and DAB2 have a higher risk for ESRD in hypertensive patients. These findings provide genetic evidence of potential novel targets for drug development and gene therapy to design individualized treatment of hypertension and related renal injury.
Collapse
Affiliation(s)
- Chao Zhang
- Department of Pharmacology and ToxicologyUniversity of Mississippi Medical CenterJacksonMississippiUSA
- Department of UrologyZhongshan HospitalFudan UniversityShanghaiChina
| | - Xing Fang
- Department of Pharmacology and ToxicologyUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - Huawei Zhang
- Department of Pharmacology and ToxicologyUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - Wenjun Gao
- Department of Pharmacology and ToxicologyUniversity of Mississippi Medical CenterJacksonMississippiUSA
- Department of UrologyZhongshan HospitalFudan UniversityShanghaiChina
| | - Han Jen Hsu
- Department of UrologyZhongshan HospitalFudan UniversityShanghaiChina
| | - Richard J. Roman
- Department of Pharmacology and ToxicologyUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - Fan Fan
- Department of Pharmacology and ToxicologyUniversity of Mississippi Medical CenterJacksonMississippiUSA
| |
Collapse
|
10
|
Dhande IS, Kneedler SC, Zhu Y, Joshi AS, Hicks MJ, Wenderfer SE, Braun MC, Doris PA. Natural genetic variation in Stim1 creates stroke in the spontaneously hypertensive rat. Genes Immun 2020; 21:182-192. [PMID: 32300198 PMCID: PMC7274944 DOI: 10.1038/s41435-020-0097-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 02/28/2020] [Accepted: 03/20/2020] [Indexed: 12/28/2022]
Abstract
Similar to humans, the risk of cerebrovascular disease in stroke-prone spontaneously hypertensive rats (SHR-A3/SHRSP) arises from naturally occurring genetic variation. In the present study, we show the involvement of genetic variation affecting the store-operated calcium signaling gene, Stim1, in the pathogenesis of stroke in SHR. Stim1 is a key lymphocyte activation signaling molecule and contains functional variation in SHR-A3 that diverges from stroke-resistant SHR-B2. We created a SHR-A3 congenic line in which Stim1 was substituted with the corresponding genomic segment from SHR-B2. Compared with SHR-A3 rats, Stim1 congenic SHR-A3 (SHR-A3(Stim1-B2)) have reduced cerebrovascular disease in response to salt loading including lower neurological deficit scores and cerebral edema. Microbleeds and major hemorrhages occurred in over half of SHR-A3 rats. These lesions were absent in SHR-A3(Stim1-B2) rats. Loss of Stim1 function in mice and humans is associated with antibody-mediated autoimmunity due to defects in T lymphocyte helper function to B cells. We investigated autoantibody formation using a high-density protein array to detect the presence of IgG and IgM autoantibodies in SHR-A3. Autoantibodies to key cerebrovascular stress proteins were detected that were reduced in the congenic line.
Collapse
Affiliation(s)
- Isha S Dhande
- Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Sterling C Kneedler
- Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Yaming Zhu
- Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Aniket S Joshi
- Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - M John Hicks
- Department of Pathology and Immunology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, 77030, USA
| | - Scott E Wenderfer
- Department of Pediatrics, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, 77030, USA
| | - Michael C Braun
- Department of Pediatrics, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, 77030, USA
| | - Peter A Doris
- Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
| |
Collapse
|
11
|
Li D, Cui Z, Xu S, Xu T, Wu S, Bouakaz A, Wan M, Zhang S. Low-Intensity Focused Ultrasound Stimulation Treatment Decreases Blood Pressure in Spontaneously Hypertensive Rats. IEEE Trans Biomed Eng 2020; 67:3048-3056. [PMID: 32086192 DOI: 10.1109/tbme.2020.2975279] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
OBJECTIVE We applied low-intensity focused ultrasound (LIFU) stimulation of the ventrolateral periaqueductal gray (vlPAG) in spontaneously hypertensive rats (SHRs) model to demonstrate the feasibility of LIFU stimulation to decrease blood pressure (BP). METHODS The rats were treated with LIFU stimulation for 20 min every day for one week. The change of BP and heart rate (HR) were recorded to evaluate the antihypertensive effect. Then the plasma levels of epinephrine (EPI), norepinephrine (NE), and angiotensin II (ANGII) were measured to evaluate the activity of the sympathetic nervous system (SNS) and the renin-angiotensin system (RAS). The c-fos immunofluorescence assay was performed to investigate the antihypertensive nerve pathway. Moreover, the biological safety of ultrasound sonication was examined. RESULTS The LIFU stimulation induced a significant reduction of BP in 8 SHRs. The mean systolic blood pressure (SBP) was reduced from 170 ± 4 mmHg to 128 ± 4.5 mmHg after a one-week treatment, p < 0.01. The activity of SNS and RAS were also inhibited. The results of the c-fos immunofluorescence assay showed that US stimulation of the vlPAG significantly enhanced the neuronal activity both in vlPAG and caudal ventrolateral medulla (CVLM) regions. And the US stimulation used in this study did not cause significant tissue damage, hemorrhage and cell apoptosis in the sonication region. CONCLUSION The results support that LIFU stimulation of the vlPAG could relieve hypertension in SHRs. SIGNIFICANCE The LIFU stimulation of the vlPAG could potentially be a new alternative non-invasive device therapy for hypertension.
Collapse
|
12
|
Dhande IS, Zhu Y, Kneedler SC, Joshi AS, Hicks MJ, Wenderfer SE, Braun MC, Doris PA. Stim1 Polymorphism Disrupts Immune Signaling and Creates Renal Injury in Hypertension. J Am Heart Assoc 2020; 9:e014142. [PMID: 32075490 PMCID: PMC7335582 DOI: 10.1161/jaha.119.014142] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background Spontaneously hypertensive rats of the stroke‐prone line (SHR‐A3) develop hypertensive renal disease as a result of naturally occurring genetic variation. Our prior work identified a single‐nucleotide polymorphism unique to SHR‐A3 that results in truncation of the carboxy terminus of STIM1. The SHR‐B2 line, which is also hypertensive but resists hypertensive renal injury, expresses the wild‐type STIM1. STIM1 plays a central role in lymphocyte calcium signaling that directs immune effector responses. Here we show that major defects in lymphocyte function affecting calcium signaling, nuclear factor of activated T cells activation, cytokine production, proliferation, apoptosis, and regulatory T‐cell development are present in SHR‐A3 and attributable to STIM1. Methods and Results To assess the role of Stim1 variation in susceptibility to hypertensive renal injury, we created a Stim1 congenic line, SHR‐A3(Stim1‐B2), and STIM1 function was rescued in SHR‐A3. We found that Stim1 gene rescue restores disturbed lymphocyte function in SHR‐A3. Hypertensive renal injury was compared in SHR‐A3 and the SHR‐A3(Stim1‐B2) congenic line. Histologically assessed renal injury was markedly reduced in SHR‐A3(Stim1‐B2), as were renal injury biomarker levels measured in urine. Stim1 deficiency has been linked to the emergence of antibody‐mediated autoimmunity. Renal glomerular immunoglobulin deposition was greater in SHR‐A3 than SHR‐B2 and was reduced by Stim1 congenic substitution. Serum anti–double‐stranded DNA antibody titers in SHR‐A3 were elevated compared with SHR‐B2 and were reduced in SHR‐A3(Stim1‐B2). Conclusions Stim1 deficiency in lymphocyte function originating from Stim1 truncation in SHR‐A3 combines with hypertension to create end organ disease and may do so as a result of antibody formation.
Collapse
Affiliation(s)
- Isha S Dhande
- Institute of Molecular Medicine University of Texas Health Science Center at Houston Houston TX
| | - Yaming Zhu
- Institute of Molecular Medicine University of Texas Health Science Center at Houston Houston TX
| | - Sterling C Kneedler
- Institute of Molecular Medicine University of Texas Health Science Center at Houston Houston TX
| | - Aniket S Joshi
- Institute of Molecular Medicine University of Texas Health Science Center at Houston Houston TX
| | - M John Hicks
- Department of Pathology and Immunology Baylor College of Medicine and Texas Children's Hospital Houston TX
| | - Scott E Wenderfer
- Department of Pediatrics Baylor College of Medicine and Texas Children's Hospital Houston TX
| | - Michael C Braun
- Department of Pediatrics Baylor College of Medicine and Texas Children's Hospital Houston TX
| | - Peter A Doris
- Institute of Molecular Medicine University of Texas Health Science Center at Houston Houston TX
| |
Collapse
|
13
|
Dhande IS, Kneedler SC, Joshi AS, Zhu Y, Hicks MJ, Wenderfer SE, Braun MC, Doris PA. Germ-line genetic variation in the immunoglobulin heavy chain creates stroke susceptibility in the spontaneously hypertensive rat. Physiol Genomics 2019; 51:578-585. [PMID: 31608789 DOI: 10.1152/physiolgenomics.00054.2019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The risk of cerebrovascular disease in stroke-prone spontaneously hypertensive rats (SHR-A3/SHRSP) arises from naturally occurring genetic variation. In the present study we show the involvement of SHR genetic variation that affects antibody formation and function in the pathogenesis of stroke. We have tested the involvement in susceptibility to stroke of genetic variation in IgH, the gene encoding the immunoglobulin heavy chain by congenic substitution. This gene contains functional natural variation in SHR-A3 that diverges from stroke-resistant SHR-B2. We created a SHR-A3 congenic line in which the IgH gene was substituted with the corresponding haplotype from SHR-B2. Compared with SHR-A3 rats, congenic substitution of the IgH locus [SHR-A3(IgH-B2)] markedly reduced cerebrovascular disease. Given the role in antibody formation of the IgH gene, we investigated the presence of IgG and IgM autoantibodies and their targets using a high-density protein array containing ~20,000 recombinant proteins. High titers of autoantibodies to key cerebrovascular stress proteins were detected, including FABP4, HSP70, and Wnt signaling proteins. Serum levels of these autoantibodies were reduced in the SHR-A3(IgH-B2) congenic line.
Collapse
Affiliation(s)
- Isha S Dhande
- Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas
| | - Sterling C Kneedler
- Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas
| | - Aniket S Joshi
- Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas
| | - Yaming Zhu
- Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas
| | - M John Hicks
- Department of Pathology and Immunology, Baylor College of Medicine and Texas Children's Hospital, Houston
| | - Scott E Wenderfer
- Department of Pediatrics, Baylor College of Medicine and Texas Children's Hospital, Houston, Texas
| | - Michael C Braun
- Department of Pediatrics, Baylor College of Medicine and Texas Children's Hospital, Houston, Texas
| | - Peter A Doris
- Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas
| |
Collapse
|
14
|
Watson CT, Kos JT, Gibson WS, Newman L, Deikus G, Busse CE, Smith ML, Jackson KJ, Collins AM. A comparison of immunoglobulin IGHV, IGHD and IGHJ genes in wild-derived and classical inbred mouse strains. Immunol Cell Biol 2019; 97:888-901. [PMID: 31441114 DOI: 10.1111/imcb.12288] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 08/05/2019] [Accepted: 08/20/2019] [Indexed: 01/20/2023]
Abstract
The genomes of classical inbred mouse strains include genes derived from all three major subspecies of the house mouse, Mus musculus. We recently posited that genetic diversity in the immunoglobulin heavy chain (IGH) gene loci of C57BL/6 and BALB/c mice reflects differences in subspecies origin. To investigate this hypothesis, we conducted high-throughput sequencing of IGH gene rearrangements to document IGH variable (IGHV), joining (IGHJ) and diversity (IGHD) genes in four inbred wild-derived mouse strains (CAST/EiJ, LEWES/EiJ, MSM/MsJ and PWD/PhJ) and a single disease model strain (NOD/ShiLtJ), collectively representing genetic backgrounds of several major mouse subspecies. A total of 341 germline IGHV sequences were inferred in the wild-derived strains, including 247 not curated in the international ImMunoGeneTics information system. By contrast, 83/84 inferred NOD IGHV genes had previously been observed in C57BL/6 mice. Variability among the strains examined was observed for only a single IGHJ gene, involving a description of a novel allele. By contrast, unexpected variation was found in the IGHD gene loci, with four previously unreported IGHD gene sequences being documented. Very few IGHV sequences of C57BL/6 and BALB/c mice were shared with strains representing major subspecies, suggesting that their IGH loci may be complex mosaics of genes of disparate origins. This suggests a similar level of diversity is likely present in the IGH loci of other classical inbred strains. This must now be documented if we are to properly understand interstrain variation in models of antibody-mediated disease.
Collapse
Affiliation(s)
- Corey T Watson
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Justin T Kos
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - William S Gibson
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Leah Newman
- Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Gintaras Deikus
- Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Christian E Busse
- Division of B Cell Immunology, German Cancer Research Center, 69120, Heidelberg, Germany
| | - Melissa L Smith
- Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Katherine Jl Jackson
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia
| | - Andrew M Collins
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| |
Collapse
|
15
|
Vaněčková I, Hojná S, Kadlecová M, Vernerová Z, Kopkan L, Červenka L, Zicha J. Renoprotective effects of ET(A) receptor antagonists therapy in experimental non-diabetic chronic kidney disease: Is there still hope for the future? Physiol Res 2018; 67:S55-S67. [PMID: 29947528 DOI: 10.33549/physiolres.933898] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Chronic kidney disease (CKD) is a life-threatening disease arising as a frequent complication of diabetes, obesity and hypertension. Since it is typically undetected for long periods, it often progresses to end-stage renal disease. CKD is characterized by the development of progressive glomerulosclerosis, interstitial fibrosis and tubular atrophy along with a decreased glomerular filtration rate. This is associated with podocyte injury and a progressive rise in proteinuria. As endothelin-1 (ET-1) through the activation of endothelin receptor type A (ET(A)) promotes renal cell injury, inflammation, and fibrosis which finally lead to proteinuria, it is not surprising that ET(A) receptors antagonists have been proven to have beneficial renoprotective effects in both experimental and clinical studies in diabetic and non-diabetic CKD. Unfortunately, fluid retention encountered in large clinical trials in diabetic CKD led to the termination of these studies. Therefore, several advances, including the synthesis of new antagonists with enhanced pharmacological activity, the use of lower doses of ET antagonists, the addition of diuretics, plus simply searching for distinct pathological states to be treated, are promising targets for future experimental studies. In support of these approaches, our group demonstrated in adult subtotally nephrectomized Ren-2 transgenic rats that the addition of a diuretic on top of renin-angiotensin and ET(A) blockade led to a further decrease of proteinuria. This effect was independent of blood pressure which was normalized in all treated groups. Recent data in non-diabetic CKD, therefore, indicate a new potential for ET(A) antagonists, at least under certain pathological conditions.
Collapse
Affiliation(s)
- I Vaněčková
- Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic.
| | | | | | | | | | | | | |
Collapse
|
16
|
Affiliation(s)
- Richard J Roman
- From the Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson.
| | - Fan Fan
- From the Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson
| |
Collapse
|