1
|
Banu Shaikh G, Das KK. Cilnidipine: An L- and N-Type Blocker of Calcium Channels Ameliorating Renal Damage in Experimental Hypertensive Rats. Cureus 2025; 17:e81404. [PMID: 40296922 PMCID: PMC12035576 DOI: 10.7759/cureus.81404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 03/28/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND Cilnidipine is both an L-type and N-type calcium channel blocker (CCB). Cilnidipine suppresses hyperactivity in the renin-angiotensin system and sympathetic nervous system by blocking N-type calcium channels. We hypothesized that through its N-type calcium channel blockade, cilnidipine may reduce proteinuria and improve creatinine clearance in hypertensive rats. Aim of the study: The current study was done to show the renal protective effect of cilnidipine on chronic hypertensive rats. Our primary objective is to develop a hypertensive rat model by giving L-NAME (NG-nitro-L-Arginine Methyl Ester Hydrochloride) and 4% sodium chloride (4% NaCl). High salt was given along with L-NAME because the combination of L-NAME and high salt intake leads to a synergistic effect on blood pressure. L-NAME impairs NO production, and high salt intake exacerbates this effect by promoting vasoconstriction and fluid retention. Our secondary objective is to evaluate the kidney damage measures, including recording of proteinuria, creatinine clearance, and urinary angiotensin II (Ang II) levels in hypertensive rats with or without cilnidipine treatment. METHODS Six groups of male Albino Wister rats (with six rats in each group) were created by a simple randomization technique. Rats were obtained from the animal house of our institution. Group 1 (control) received vehicle treatment; cilnidipine (2mg/kg/day by oral gavage) was given to Group 2; L-NAME (40mg/kg/day by oral gavage) was given to Group 3; Group 4 received L-NAME plus cilnidipine; Group 5 received L-NAME plus 4% NaCl (mixed with diet) treatment; Group 6 received L-NAME, 4% NaCl and cilnidipine. Creatinine excretion and urinary protein were assessed in a 24-hour urine sample. Serum urea and creatinine levels were also measured. Relative expression of serum and renal tissue ACE (angiotensin-converting enzyme) protein was done by Western blotting. Quantitative estimation of urinary and serum Ang II levels was done by enzyme-linked immunosorbent assay (ELISA). Kidney histopathological analysis was carried out. RESULTS In the renal tissue homogenate and serum of L-NAME and salt-induced hypertensive rats, we found elevated ACE and Ang II levels. We also observed a significant increase in proteinuria (4.65±0.29) compared to control (1.56±0.044) and a decrease in creatinine clearance (0.06±0.0019) compared to control (0.078±0.0044) in hypertensive rats. We observed that in treatment with cilnidipine (groups 4 and 6 rats), there were significant improvements in creatinine clearance (0.077±0.0027) (p < 0.05) and a significant decrease in proteinuria (3.38±0.24) (p < 0.05). In rats treated with cilnidipine, we also observed significantly decreased levels of Ang II in the urine and serum (p < 0.05) and a significantly decreased expression of ACE in the renal tissue and serum. CONCLUSION These results showed that in hypertensive experimental rats, cilnidipine, apart from its hypotensive actions, decreases proteinuria and urinary creatinine and Ang II levels. These actions of cilnidipine may be because of blocking N-type calcium channels. Therefore, cilnidipine dual L/N type CCB may act as a renoprotective drug and decrease glomerular damage. Further mechanistic studies using selective N-type channel blockers or knockout mice are needed to prove the findings.
Collapse
Affiliation(s)
- Gouher Banu Shaikh
- Department of Physiology, BLDE (Deemed to be University) Shri B. M. Patil Medical College, Hospital and Research Center, Vijayapura, IND
| | - Kusal K Das
- Department of Physiology, BLDE (Deemed to be University) Shri B. M. Patil Medical College, Hospital and Research Center, Vijayapura, IND
| |
Collapse
|
2
|
Leierer J, Salib M, Evgeniou M, Rossignol P, Massy ZA, Kratochwill K, Mayer G, Fellström B, Girerd N, Zannad F, Perco P. Identification of endophenotypes supporting outcome prediction in hemodialysis patients based on mechanistic markers of statin treatment. Heliyon 2024; 10:e30709. [PMID: 38765135 PMCID: PMC11098839 DOI: 10.1016/j.heliyon.2024.e30709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 05/02/2024] [Indexed: 05/21/2024] Open
Abstract
Background Statins are widely used to reduce the risk of cardiovascular disease (CVD). Patients with end-stage renal disease (ESRD) on hemodialysis have significantly increased risk of developing CVD. Statin treatment in these patients however did not show a statistically significant benefit in large trials on a patient cohort level. Methods We generated gene expression profiles for statins to investigate the impact on cellular programs in human renal proximal tubular cells and mesangial cells in-vitro. We subsequently selected biomarkers from key statin-affected molecular pathways and assessed these biomarkers in plasma samples from the AURORA cohort, a double-blind, randomized, multi-center study of patients on hemodialysis or hemofiltration that have been treated with rosuvastatin. Patient clusters (phenotypes) were created based on the identified biomarkers using Latent Class Model clustering and the associations with outcome for the generated phenotypes were assessed using Cox proportional hazards regression models. The multivariable models were adjusted for clinical and biological covariates based on previously published data in AURORA. Results The impact of statin treatment on mesangial cells was larger as compared with tubular cells with a large overlap of differentially expressed genes identified for atorvastatin and rosuvastatin indicating a predominant drug class effect. Affected molecular pathways included TGFB-, TNF-, and MAPK-signaling and focal adhesion among others. Four patient clusters were identified based on the baseline plasma concentrations of the eight biomarkers. Phenotype 1 was characterized by low to medium levels of the hepatocyte growth factor (HGF) and high levels of interleukin 6 (IL6) or matrix metalloproteinase 2 (MMP2) and it was significantly associated with outcome showing increased risk of developing major adverse cardiovascular events (MACE) or cardiovascular death. Phenotype 2 had high HGF but low Fas cell surface death receptor (FAS) levels and it was associated with significantly better outcome at 1 year. Conclusions In this translational study, we identified patient subgroups based on mechanistic markers of statin therapy that are associated with disease outcome in patients on hemodialysis.
Collapse
Affiliation(s)
- Johannes Leierer
- Medical University of Innsbruck, Department of Internal Medicine IV, Innsbruck, Austria
| | - Madonna Salib
- Université de Lorraine, Inserm, Centre d’Investigations Cliniques- 1433, and Inserm U1116, CHRU Nancy, F-CRIN INI-CRCT, Nancy, France
| | - Michail Evgeniou
- Medical University of Vienna, Comprehensive Center for Pediatrics, Department of Pediatrics and Adolescent Medicine, Division of Pediatric Nephrology and Gastroenterology, Vienna, Austria
| | - Patrick Rossignol
- Université de Lorraine, Inserm, Centre d’Investigations Cliniques- 1433, and Inserm U1116, CHRU Nancy, F-CRIN INI-CRCT, Nancy, France
- Medical Specialties and Nephrology departments, Princess Grace Hospital, Monaco, Monaco
| | - Ziad A. Massy
- Association pour l'Utilisation du Rein Artificiel (AURA) Paris and Department of Nephrology, CHU Ambroise Paré, APHP, 92104, Boulogne Billancourt, and Centre for Research in Epidemiology and Population Health (CESP), University Paris-Saclay, University Versailles-Saint Quentin, Inserm UMRS, 1018, Clinical Epidemiology Team, Villejuif, France
| | - Klaus Kratochwill
- Medical University of Vienna, Comprehensive Center for Pediatrics, Department of Pediatrics and Adolescent Medicine, Division of Pediatric Nephrology and Gastroenterology, Vienna, Austria
| | - Gert Mayer
- Medical University of Innsbruck, Department of Internal Medicine IV, Innsbruck, Austria
| | - Bengt Fellström
- Uppsala University, Department of Medical Sciences, Uppsala, Sweden
| | - Nicolas Girerd
- Université de Lorraine, Inserm, Centre d’Investigations Cliniques- 1433, and Inserm U1116, CHRU Nancy, F-CRIN INI-CRCT, Nancy, France
| | - Faiez Zannad
- Université de Lorraine, Inserm, Centre d’Investigations Cliniques- 1433, and Inserm U1116, CHRU Nancy, F-CRIN INI-CRCT, Nancy, France
| | - Paul Perco
- Medical University of Innsbruck, Department of Internal Medicine IV, Innsbruck, Austria
| |
Collapse
|
3
|
Martin WP, Nair M, Chuah YH, Malmodin D, Pedersen A, Abrahamsson S, Hutter M, Abdelaal M, Elliott JA, Fearon N, Eckhardt H, Godson C, Brennan EP, Fändriks L, le Roux CW, Docherty NG. Dietary restriction and medical therapy drives PPARα-regulated improvements in early diabetic kidney disease in male rats. Clin Sci (Lond) 2022; 136:1485-1511. [PMID: 36259366 PMCID: PMC7613831 DOI: 10.1042/cs20220205] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 10/14/2022] [Accepted: 10/18/2022] [Indexed: 11/17/2022]
Abstract
The attenuation of diabetic kidney disease (DKD) by metabolic surgery is enhanced by pharmacotherapy promoting renal fatty acid oxidation (FAO). Using the Zucker Diabetic Fatty and Zucker Diabetic Sprague Dawley rat models of DKD, we conducted studies to determine if these effects could be replicated with a non-invasive bariatric mimetic intervention. Metabolic control and renal injury were compared in rats undergoing a dietary restriction plus medical therapy protocol (DMT; fenofibrate, liraglutide, metformin, ramipril, and rosuvastatin) and ad libitum-fed controls. The global renal cortical transcriptome and urinary 1H-NMR metabolomic profiles were also compared. Kidney cell type-specific and medication-specific transcriptomic responses were explored through in silico deconvolution. Transcriptomic and metabolomic correlates of improvements in kidney structure were defined using a molecular morphometric approach. The DMT protocol led to ∼20% weight loss, normalized metabolic parameters and was associated with reductions in indices of glomerular and proximal tubular injury. The transcriptomic response to DMT was dominated by changes in fenofibrate- and peroxisome proliferator-activated receptor-α (PPARα)-governed peroxisomal and mitochondrial FAO transcripts localizing to the proximal tubule. DMT induced urinary excretion of PPARα-regulated metabolites involved in nicotinamide metabolism and reversed DKD-associated changes in the urinary excretion of tricarboxylic acid (TCA) cycle intermediates. FAO transcripts and urinary nicotinamide and TCA cycle metabolites were moderately to strongly correlated with improvements in glomerular and proximal tubular injury. Weight loss plus pharmacological PPARα agonism is a promising means of attenuating DKD.
Collapse
Affiliation(s)
- William P. Martin
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| | - Meera Nair
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| | - Yeong H.D. Chuah
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| | - Daniel Malmodin
- Swedish NMR Centre, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Anders Pedersen
- Swedish NMR Centre, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Sanna Abrahamsson
- Bioinformatics Core Facility, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Michaela Hutter
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| | - Mahmoud Abdelaal
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| | - Jessie A. Elliott
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| | - Naomi Fearon
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| | - Hans Eckhardt
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| | - Catherine Godson
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| | - Eoin P. Brennan
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| | - Lars Fändriks
- Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Carel W. le Roux
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
- Diabetes Research Group, Ulster University, Coleraine BT52 1SA, UK
| | - Neil G. Docherty
- Diabetes Complications Research Centre, School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| |
Collapse
|
4
|
Martin WP, Chuah YHD, Abdelaal M, Pedersen A, Malmodin D, Abrahamsson S, Hutter M, Godson C, Brennan EP, Fändriks L, le Roux CW, Docherty NG. Medications Activating Tubular Fatty Acid Oxidation Enhance the Protective Effects of Roux-en-Y Gastric Bypass Surgery in a Rat Model of Early Diabetic Kidney Disease. Front Endocrinol (Lausanne) 2022; 12:757228. [PMID: 35222262 PMCID: PMC8867227 DOI: 10.3389/fendo.2021.757228] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 12/23/2021] [Indexed: 01/03/2023] Open
Abstract
Background Roux-en-Y gastric bypass surgery (RYGB) improves biochemical and histological parameters of diabetic kidney disease (DKD). Targeted adjunct medical therapy may enhance renoprotection following RYGB. Methods The effects of RYGB and RYGB plus fenofibrate, metformin, ramipril, and rosuvastatin (RYGB-FMRR) on metabolic control and histological and ultrastructural indices of glomerular and proximal tubular injury were compared in the Zucker Diabetic Sprague Dawley (ZDSD) rat model of DKD. Renal cortical transcriptomic (RNA-sequencing) and urinary metabolomic (1H-NMR spectroscopy) responses were profiled and integrated. Transcripts were assigned to kidney cell types through in silico deconvolution in kidney single-nucleus RNA-sequencing and microdissected tubular epithelial cell proteomics datasets. Medication-specific transcriptomic responses following RYGB-FMRR were explored using a network pharmacology approach. Omic correlates of improvements in structural and ultrastructural indices of renal injury were defined using a molecular morphometric approach. Results RYGB-FMRR was superior to RYGB alone with respect to metabolic control, albuminuria, and histological and ultrastructural indices of glomerular injury. RYGB-FMRR reversed DKD-associated changes in mitochondrial morphology in the proximal tubule to a greater extent than RYGB. Attenuation of transcriptomic pathway level activation of pro-fibrotic responses was greater after RYGB-FMRR than RYGB. Fenofibrate was found to be the principal medication effector of gene expression changes following RYGB-FMRR, which led to the transcriptional induction of PPARα-regulated genes that are predominantly expressed in the proximal tubule and which regulate peroxisomal and mitochondrial fatty acid oxidation (FAO). After omics integration, expression of these FAO transcripts positively correlated with urinary levels of PPARα-regulated nicotinamide metabolites and negatively correlated with urinary tricarboxylic acid (TCA) cycle intermediates. Changes in FAO transcripts and nicotinamide and TCA cycle metabolites following RYGB-FMRR correlated strongly with improvements in glomerular and proximal tubular injury. Conclusions Integrative multi-omic analyses point to PPARα-stimulated FAO in the proximal tubule as a dominant effector of treatment response to combined surgical and medical therapy in experimental DKD. Synergism between RYGB and pharmacological stimulation of FAO represents a promising combinatorial approach to the treatment of DKD in the setting of obesity.
Collapse
Affiliation(s)
- William P. Martin
- Diabetes Complications Research Centre, School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
| | - Yeong H. D. Chuah
- Diabetes Complications Research Centre, School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
| | - Mahmoud Abdelaal
- Diabetes Complications Research Centre, School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
| | - Anders Pedersen
- Swedish NMR Centre, University of Gothenburg, Gothenburg, Sweden
| | - Daniel Malmodin
- Swedish NMR Centre, University of Gothenburg, Gothenburg, Sweden
| | - Sanna Abrahamsson
- Bioinformatics Core Facility, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Michaela Hutter
- Diabetes Complications Research Centre, School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
| | - Catherine Godson
- Diabetes Complications Research Centre, School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
| | - Eoin P. Brennan
- Diabetes Complications Research Centre, School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
| | - Lars Fändriks
- Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Carel W. le Roux
- Diabetes Complications Research Centre, School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
- Diabetes Research Group, Ulster University, Coleraine, United Kingdom
| | - Neil G. Docherty
- Diabetes Complications Research Centre, School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
| |
Collapse
|
5
|
Thangaraj SS, Oxlund CS, Fonseca MPD, Svenningsen P, Stubbe J, Palarasah Y, Ketelhuth DFJ, Jacobsen IA, Jensen BL. The mineralocorticoid receptor blocker spironolactone lowers plasma interferon-γ and interleukin-6 in patients with type 2 diabetes and treatment-resistant hypertension. J Hypertens 2022; 40:153-162. [PMID: 34843183 DOI: 10.1097/hjh.0000000000002990] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
BACKGROUND The mineralocorticoid receptor antagonist spironolactone lowers blood pressure in patients with resistant hypertension despite antihypertensive treatment with angiotensin-converting inhibitors (ACEi) and angiotensin-II receptor blockers (ARB). In preclinical studies, spironolactone suppresses pro-hypertensive interleukin 17A (IL-17A). OBJECTIVES Plasma samples were analysed from a randomized, double-blind placebo-controlled trial with spironolactone given to patients with type 2 diabetes mellitus (T2DM) and resistant hypertension on three antihypertensive drugs. We tested the hypothesis that spironolactone-induced antihypertensive effects are associated with suppression of IL-17A and related cytokines. METHODS Interferon-γ (IFN-γ), IL-17A, tumor necrosis factor-α (TNF-α), IL-6, IL-1β and IL-10 were assessed in plasma with immunoassay in samples before and after 16 weeks of treatment with placebo or spironolactone (12.5-25-50 mg/day). RESULTS Spironolactone significantly reduced plasma IFN-γ and IL-6 while IL-17A, TNF-α, IL-1β and IL-10 were unchanged. IL-6 was more sensitive to higher doses of spironolactone. At baseline, serum aldosterone correlated positively with diastolic night blood pressure. Urine albumin/creatinine-ratios correlated positively with plasma IL-6 at baseline. There were no relations between aldosterone and cytokine concentrations at baseline; between cytokine concentration and blood pressure at baseline; and between cytokine concentration decrease and blood pressure decrease, except for IFN-γ, after treatment. The spironolactone-induced elevation in plasma potassium related inversely to blood pressure but not to changes in cytokines. In macrophages in vitro, spironolactone suppressed lipopolysaccharide (LPS)-induced TNF-α, IL-6, IL-1β and IL-10 levels. CONCLUSION The antihypertensive action of spironolactone in resistant hypertensive patients is associated with suppressed IFN-γ and IL-6 and not IL-17A. Spironolactone exerts anti-inflammatory actions in vivo on macrophages and T-cells.
Collapse
Affiliation(s)
- Sai Sindhu Thangaraj
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense
| | | | - Micaella Pereira Da Fonseca
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense
| | - Per Svenningsen
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense
| | - Jane Stubbe
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense
| | - Yaseelan Palarasah
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark
| | - Daniel F J Ketelhuth
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense
| | - Ib Abildgaard Jacobsen
- Research Unit for Cardiovascular and Metabolic Prevention, Department of Endocrinology, Odense University Hospital, Odense C, Denmark
| | - Boye L Jensen
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense
| |
Collapse
|
6
|
Liu Z, Xiao M, Du Z, Li M, Guo H, Yao M, Wan X, Xie Z. Dietary supplementation of Huangshan Maofeng green tea preventing hypertension of older C57BL/6 mice induced by desoxycorticosterone acetate and salt. J Nutr Biochem 2021; 88:108530. [PMID: 33080347 DOI: 10.1016/j.jnutbio.2020.108530] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 08/26/2020] [Accepted: 10/14/2020] [Indexed: 01/12/2023]
Abstract
Senile hypertension affects the life quality of aged population. Dietary intervention plays a pivotal role in the prevention of hypertension. There are few reports concerning the effects and mechanisms of green tea supplementation preventing age related hypertension. The current study investigated the effect and mechanism of dietary supplement of Huangshan Maofeng green tea (HSMF) on prevention of hypertension induced by deoxycorticosterone acetate (DOCA) and salt in old C57BL/6 mice. Our results showed that HSMF dose-dependently prevented the increase of systolic blood pressure and diastolic blood pressure induced by DOCA plus salt (DS) at 51-week-old mice. And HSMF significantly reduced the agonists' stimulated contraction of mesenteric arteries isolated from the old mice. The expression of vasoconstrictor genes and inflammatory cytokines in aorta were suppressed observably by HSMF supplementation compared with DS group. The protein expression of PKCα in the aorta was dose-dependently decreased by HSMF compared to DS group. The phosphorylation level of MYPT1, CPI-17and MLC20 was also restrained by HSMF in the aorta. Furthermore, HSMF protected kidney by maintaining integrity of glomeruli and tubules and remarkably decreased the NGAL level in plasma. HSMF also suppressed the kidney inflammation by decreasing inflammatory cytokines expression and the macrophage infiltration. Our results proved that dietary supplement of HSMF remarkably improved the vascular functions and protected kidney injury, and thus prevented hypertension induced by DS in older C57BL/6 mice. Our data indicated that the dietary supplement of HSMF may potentially be used as a food additive for preventing hypertension for aged people.
Collapse
Affiliation(s)
- Zenghui Liu
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Sciences and Technology, Anhui Agricultural University, Hefei, China; Anhui Academy of Medical Science, Hefei, China
| | - Mengchao Xiao
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Sciences and Technology, Anhui Agricultural University, Hefei, China
| | - Zhaofeng Du
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Sciences and Technology, Anhui Agricultural University, Hefei, China; School of Biology and Food Engineering, Fuyang Normal University, Fuyang, China
| | - Mengwan Li
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Sciences and Technology, Anhui Agricultural University, Hefei, China
| | - Huimin Guo
- Center for Biotechnology, Anhui Agricultural University, Hefei, China
| | - Min Yao
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Sciences and Technology, Anhui Agricultural University, Hefei, China
| | - Xiaochun Wan
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Sciences and Technology, Anhui Agricultural University, Hefei, China.
| | - Zhongwen Xie
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Sciences and Technology, Anhui Agricultural University, Hefei, China.
| |
Collapse
|
7
|
Lindoso RS, Lopes JA, Binato R, Abdelhay E, Takiya CM, Miranda KRD, Lara LS, Viola A, Bussolati B, Vieyra A, Collino F. Adipose Mesenchymal Cells-Derived EVs Alleviate DOCA-Salt-Induced Hypertension by Promoting Cardio-Renal Protection. Mol Ther Methods Clin Dev 2020; 16:63-77. [PMID: 31871958 PMCID: PMC6909095 DOI: 10.1016/j.omtm.2019.11.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 11/02/2019] [Indexed: 12/20/2022]
Abstract
Hypertension is a long-term condition that can increase organ susceptibility to insults and lead to severe complications such as chronic kidney disease (CKD). Extracellular vesicles (EVs) are cell-derived membrane structures that participate in cell-cell communication by exporting encapsulated molecules to target cells, regulating physiological and pathological processes. We here demonstrate that multiple administration of EVs from adipose-derived mesenchymal stromal cells (ASC-EVs) in deoxycorticosterone acetate (DOCA)-salt hypertensive model can protect renal tissue by maintaining its filtration capacity. Indeed, ASC-EVs downregulated the pro-inflammatory molecules monocyte chemoattracting protein-1 (MCP-1) and plasminogen activating inhibitor-1 (PAI1) and reduced recruitment of macrophages in the kidney. Moreover, ASC-EVs prevented cardiac tissue fibrosis and maintained blood pressure within normal levels, thus demonstrating their multiple favorable effects in different organs. By applying microRNA (miRNA) microarray profile of the kidney of DOCA-salt rats, we identified a selective miRNA signature associated with epithelial-mesenchymal transition (EMT). One of the key pathways found was the axis miR-200-TGF-β, that was significantly altered by EV administration, thereby affecting the EMT signaling and preventing renal inflammatory response and fibrosis development. Our results indicate that EVs can be a potent therapeutic tool for the treatment of hypertension-induced CKD in cardio-renal syndrome.
Collapse
Affiliation(s)
- Rafael Soares Lindoso
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
- National Center for Structural Biology and Bioimaging/CENABIO, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
- National Institute of Science and Technology for Regenerative Medicine-REGENERA, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Jarlene Alécia Lopes
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
- National Center for Structural Biology and Bioimaging/CENABIO, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Renata Binato
- Brazilian National Institute of Cancer, 20230-130 Rio de Janeiro, Brazil
| | - Eliana Abdelhay
- Brazilian National Institute of Cancer, 20230-130 Rio de Janeiro, Brazil
| | - Christina Maeda Takiya
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Kildare Rocha de Miranda
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
- National Center for Structural Biology and Bioimaging/CENABIO, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Lucienne Silva Lara
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, 21941-590 Rio de Janeiro, Brazil
| | - Antonella Viola
- Department of Biomedical Sciences and Pediatric Research Institute “Citta della Speranza,” University of Padova, 35131 Padua, Italy
| | - Benedetta Bussolati
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Turin, Italy
| | - Adalberto Vieyra
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
- National Center for Structural Biology and Bioimaging/CENABIO, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
- National Institute of Science and Technology for Regenerative Medicine-REGENERA, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
- Graduate Program of Translational Biomedicine/BIOTRANS, Grande Rio University, 25071-202 Duque de Caxias, Brazil
| | - Federica Collino
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
- National Institute of Science and Technology for Regenerative Medicine-REGENERA, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
- Department of Biomedical Sciences and Pediatric Research Institute “Citta della Speranza,” University of Padova, 35131 Padua, Italy
| |
Collapse
|
8
|
Kubacka M, Zadrożna M, Nowak B, Kotańska M, Filipek B, Waszkielewicz AM, Marona H, Mogilski S. Reversal of cardiac, vascular, and renal dysfunction by non-quinazoline α1-adrenolytics in DOCA-salt hypertensive rats: a comparison with prazosin, a quinazoline-based α1-adrenoceptor antagonist. Hypertens Res 2019; 42:1125-1141. [PMID: 30872813 DOI: 10.1038/s41440-019-0239-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 12/11/2018] [Accepted: 02/03/2019] [Indexed: 12/13/2022]
Abstract
We investigated the therapeutic effect of MH-76 and MH-79, which are non-quinazoline α1-adrenoceptor antagonists with an additional ability to stimulate the nitric oxide (NO)/cyclic guanosine monophosphate (cGMP)/K + pathway, on deoxycorticosterone acetate (DOCA)-salt induced hypertension in rats. Prazosin was used as a reference compound, as quinazoline-based α1-adrenolytics may potentially exert unfavorable proapoptotic and necrotic effects. DOCA-salt hypertension was induced by DOCA (20 mg/kg s.c., twice weekly) administration plus 1% NaCl and 0.2% KCl solutions in drinking water for 12 weeks. The studied compounds MH-76, MH-79 (10 mg/kg i.p.) or prazosin (0.4 mg/kg i.p.) were administered to the DOCA-salt-treated rats, starting from the 6th week of DOCA-salt treatment and continuing for 6 weeks. This study showed that the administration of MH-79 and, to a lesser extent, MH-76 decreased elevated systolic blood pressure and heart rate, reduced heart and kidney hypertrophy, and reversed the histopathological alterations of the heart, kidney, and vessels in DOCA-salt hypertensive rats. MH-79 reversed endothelial dysfunction, which reduced inflammatory cell infiltration, arteriosclerotic alterations in renal and coronary arteries, and tubulointerstitial fibrosis. Prazosin showed a potent hemodynamic effect and reduced cardiac and renal fibrosis but exerted detrimental effects on blood vessels, potentiating fibroplasia of the media of the intrarenal artery and causing calcification of coronary arteries. Prazosin did not reverse endothelial dysfunction. Our results show the beneficial effect of non-quinazoline α1-adrenolytics on cardiac, vascular, and renal dysfunction in DOCA-salt hypertensive rats. Our findings also support the idea that targeting endothelial protection and endothelial integrity would yield beneficial effects against cardiac, blood vessel and renal injury related to hypertension.
Collapse
Affiliation(s)
- Monika Kubacka
- Faculty of Pharmacy, Department of Pharmacodynamics, Medical College, Jagiellonian University, Medyczna 9, 30-688, Kraków, Poland.
| | - Monika Zadrożna
- Faculty of Pharmacy, Department of Cytobiology, Medical College, Jagiellonian University, Medyczna 9, 30-688, Kraków, Poland
| | - Barbara Nowak
- Faculty of Pharmacy, Department of Cytobiology, Medical College, Jagiellonian University, Medyczna 9, 30-688, Kraków, Poland
| | - Magdalena Kotańska
- Faculty of Pharmacy, Department of Pharmacodynamics, Medical College, Jagiellonian University, Medyczna 9, 30-688, Kraków, Poland
| | - Barbara Filipek
- Faculty of Pharmacy, Department of Pharmacodynamics, Medical College, Jagiellonian University, Medyczna 9, 30-688, Kraków, Poland
| | - Anna Maria Waszkielewicz
- Faculty of Pharmacy, Chair of Organic Chemistry, Department of Bioorganic Chemistry, Medical College, Jagiellonian University, Medyczna 9, 30-688, Kraków, Poland
| | - Henryk Marona
- Faculty of Pharmacy, Chair of Organic Chemistry, Department of Bioorganic Chemistry, Medical College, Jagiellonian University, Medyczna 9, 30-688, Kraków, Poland
| | - Szczepan Mogilski
- Faculty of Pharmacy, Department of Pharmacodynamics, Medical College, Jagiellonian University, Medyczna 9, 30-688, Kraków, Poland
| |
Collapse
|
9
|
Renoprotective Effect of the Histone Deacetylase Inhibitor CG200745 in DOCA-Salt Hypertensive Rats. Int J Mol Sci 2019; 20:ijms20030508. [PMID: 30691015 PMCID: PMC6387176 DOI: 10.3390/ijms20030508] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 01/16/2019] [Accepted: 01/22/2019] [Indexed: 12/14/2022] Open
Abstract
The novel histone deacetylase inhibitor CG200745 was initially developed to treat various hematological and solid cancers. We investigated the molecular mechanisms associated with the renoprotective effects of CG200745 using deoxycorticosterone acetate (DOCA)-salt hypertensive (DSH) rats. DOCA strips (200 mg/kg) were implanted into rats one week after unilateral nephrectomy. Two weeks after DOCA implantation, DSH rats were randomly divided into two groups that received either physiological saline or CG200745 (5 mg/kg/day) for another two weeks. The extent of glomerulosclerosis and tubulointerstitial fibrosis was determined by Masson's trichrome staining. The renal expression of fibrosis and inflammatory markers was detected by semiquantitative immunoblotting, a polymerase chain reaction, and immunohistochemistry. Pathological signs such as glomerulosclerosis, tubulointerstitial fibrosis, increased systolic blood pressure, decreased creatinine clearance, and increased albumin-to-creatinine ratios in DSH rats were alleviated by CG200745 treatment compared to those manifestations in positive control animals. Furthermore, this treatment counteracted the increased expression of αSMA, TGF-β1, and Bax, and the decreased expression of Bcl-2 in the kidneys of DSH rats. It also attenuated the increase in the number of apoptotic cells in DSH rats. Thus, CG200745 can effectively prevent the progression of renal injury in DSH rats by exerting anti-inflammatory, anti-fibrotic, and anti-apoptotic effects.
Collapse
|
10
|
Abdel-Zaher AO, Farghaly HSM, El-Refaiy AEM, Abd-Eldayem AM. Protective effect of the standardized extract of ginkgo biloba (EGb761) against hypertension with hypercholesterolemia-induced renal injury in rats: Insights in the underlying mechanisms. Biomed Pharmacother 2017; 95:944-955. [PMID: 28915536 DOI: 10.1016/j.biopha.2017.08.078] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 08/01/2017] [Accepted: 08/20/2017] [Indexed: 02/01/2023] Open
Abstract
The potential protective role of the standardized leaf extract of ginkgo biloba (EGb761) on hypertension with hypercholesterolemia-induced renal injury was investigated in rats. Hypertension was induced by L-N(G)-nitroarginine methyl ester (L-NAME) and hypercholesterolemia was induced by feeding rats with a diet containing 1% cholesterol. In these animals repeated treatment with EGb761 produced a progressive reduction in the systolic, diastolic and mean arterial blood pressure (BP). EGb761 increased the progressive reduction in the systolic, diastolic and mean arterial BP induced by repeated administration of losartan with simvastatin. EGb761 corrected the compromised serum lipid profile and enhanced the effect of losartan with simvastatin on lipid profile. EGb761 protected against hypertension with hypercholesterolemia-induced renal injury as assessed by measurement of serum renal function markers and by histopathological examination. EGb761 enhanced the renoprotective effect of losartan with simvastatin in these rats. Concomitantly, hypertension with hypercholesterolemia-induced elevation of renal tissue malondialdehyde (MDA) and nitrite levels and reduction of intracellular reduced glutathione (GSH) level were inhibited by repeated treatment with EGb761. In addition, hypertension with hypercholesterolemia-induced increases in tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6) and interleukin-1β (IL-1β) levels in renal tissues were inhibited by treatment with EGb761. Also, EGb761 inhibited hypertension with hypercholesterolemia-induced decrease in endothelial nitric oxide synthase (eNOS) protein expression and increase in the protein expressions of inducible NO synthase (iNOS), TNF-α, IL-6 and IL-1β in the kidney tissues. Losartan with simvastatin produced similar effects on renal tissues oxidative stress, nitrite and inflammatory markers levels and on protein expressions of eNOS, iNOS, TNF-α, IL-6 and IL-1β. EGb761 enhanced losartan with simvastatin effects. These results indicate that EGb761 has the ability to protect against hypertension with hypercholesterolemia-induced renal injury. The ability of EGb761 to provide this renoprotective effect may positively correlate, besides its antihypertensive and antihypercholesterolemic effects, to its ability to suppress renal oxidative stress, nitrosative stress and inflammation.
Collapse
Affiliation(s)
- Ahmed O Abdel-Zaher
- Department of Pharmacology, Faculty of Medicine, Assiut University, Assiut, Egypt.
| | - Hanan S M Farghaly
- Department of Pharmacology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Abeer E M El-Refaiy
- Department of Pathology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Ahmed M Abd-Eldayem
- Department of Pharmacology, Faculty of Medicine, Assiut University, Assiut, Egypt
| |
Collapse
|
11
|
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
|
12
|
Chen S, Agrawal DK. Dysregulation of T cell subsets in the pathogenesis of hypertension. Curr Hypertens Rep 2016; 17:8. [PMID: 25633669 DOI: 10.1007/s11906-014-0521-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Essential hypertension (EH) and its complications have had a severe impact on public health. However, the underlying mechanisms of the pathogenesis of EH remain largely unknown. Recent investigations, predominantly in rats and mice, have provided evidence that dysregulation of distinct functions of T lymphocyte subsets is a potentially important mechanism in the pathogenesis of hypertension. We critically reviewed recent findings and propose an alternative explanation on the understanding of dysfunctional T lymphocyte subsets in the pathogenesis of hypertension. The hypothesis is that hypertensive stimuli, directly and indirectly, increase local IL-6 levels in the cardiovascular system and kidney, which may promote peripheral imbalance in the differentiation and ratio of Th17 and T regulatory cells. This results in increased IL-17 and decreased IL-10 in perivascular adipose tissue and adventitia contributing to the development of hypertension in experimental animal models. Further investigation in the field is warranted to inform new translational advances that will promote to understand the pathogenesis of EH and develop novel approaches to prevent and treat EH.
Collapse
Affiliation(s)
- Songcang Chen
- Department of Biomedical Sciences and Center for Clinical & Translational Science, Creighton University School of Medicine, 2500 California Plaza, Omaha, NE, 68178, USA,
| | | |
Collapse
|
13
|
Lother A, Fürst D, Bergemann S, Gilsbach R, Grahammer F, Huber TB, Hilgendorf I, Bode C, Moser M, Hein L. Deoxycorticosterone Acetate/Salt–Induced Cardiac But Not Renal Injury Is Mediated By Endothelial Mineralocorticoid Receptors Independently From Blood Pressure. Hypertension 2016; 67:130-8. [DOI: 10.1161/hypertensionaha.115.06530] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 10/12/2015] [Indexed: 12/23/2022]
Affiliation(s)
- Achim Lother
- From the Department of Cardiology and Angiology I, Heart Center (A.L., I.H., C.B., M.M.), Institute of Experimental and Clinical Pharmacology and Toxicology (A.L., D.F., S.B., R.G., L.H.), Renal Division, Department of Medicine (F.G., T.B.H.), and BIOSS Centre for Biological Signaling Studies (T.B.H., L.H.), University of Freiburg, Freiburg, Germany
| | - David Fürst
- From the Department of Cardiology and Angiology I, Heart Center (A.L., I.H., C.B., M.M.), Institute of Experimental and Clinical Pharmacology and Toxicology (A.L., D.F., S.B., R.G., L.H.), Renal Division, Department of Medicine (F.G., T.B.H.), and BIOSS Centre for Biological Signaling Studies (T.B.H., L.H.), University of Freiburg, Freiburg, Germany
| | - Stella Bergemann
- From the Department of Cardiology and Angiology I, Heart Center (A.L., I.H., C.B., M.M.), Institute of Experimental and Clinical Pharmacology and Toxicology (A.L., D.F., S.B., R.G., L.H.), Renal Division, Department of Medicine (F.G., T.B.H.), and BIOSS Centre for Biological Signaling Studies (T.B.H., L.H.), University of Freiburg, Freiburg, Germany
| | - Ralf Gilsbach
- From the Department of Cardiology and Angiology I, Heart Center (A.L., I.H., C.B., M.M.), Institute of Experimental and Clinical Pharmacology and Toxicology (A.L., D.F., S.B., R.G., L.H.), Renal Division, Department of Medicine (F.G., T.B.H.), and BIOSS Centre for Biological Signaling Studies (T.B.H., L.H.), University of Freiburg, Freiburg, Germany
| | - Florian Grahammer
- From the Department of Cardiology and Angiology I, Heart Center (A.L., I.H., C.B., M.M.), Institute of Experimental and Clinical Pharmacology and Toxicology (A.L., D.F., S.B., R.G., L.H.), Renal Division, Department of Medicine (F.G., T.B.H.), and BIOSS Centre for Biological Signaling Studies (T.B.H., L.H.), University of Freiburg, Freiburg, Germany
| | - Tobias B. Huber
- From the Department of Cardiology and Angiology I, Heart Center (A.L., I.H., C.B., M.M.), Institute of Experimental and Clinical Pharmacology and Toxicology (A.L., D.F., S.B., R.G., L.H.), Renal Division, Department of Medicine (F.G., T.B.H.), and BIOSS Centre for Biological Signaling Studies (T.B.H., L.H.), University of Freiburg, Freiburg, Germany
| | - Ingo Hilgendorf
- From the Department of Cardiology and Angiology I, Heart Center (A.L., I.H., C.B., M.M.), Institute of Experimental and Clinical Pharmacology and Toxicology (A.L., D.F., S.B., R.G., L.H.), Renal Division, Department of Medicine (F.G., T.B.H.), and BIOSS Centre for Biological Signaling Studies (T.B.H., L.H.), University of Freiburg, Freiburg, Germany
| | - Christoph Bode
- From the Department of Cardiology and Angiology I, Heart Center (A.L., I.H., C.B., M.M.), Institute of Experimental and Clinical Pharmacology and Toxicology (A.L., D.F., S.B., R.G., L.H.), Renal Division, Department of Medicine (F.G., T.B.H.), and BIOSS Centre for Biological Signaling Studies (T.B.H., L.H.), University of Freiburg, Freiburg, Germany
| | - Martin Moser
- From the Department of Cardiology and Angiology I, Heart Center (A.L., I.H., C.B., M.M.), Institute of Experimental and Clinical Pharmacology and Toxicology (A.L., D.F., S.B., R.G., L.H.), Renal Division, Department of Medicine (F.G., T.B.H.), and BIOSS Centre for Biological Signaling Studies (T.B.H., L.H.), University of Freiburg, Freiburg, Germany
| | - Lutz Hein
- From the Department of Cardiology and Angiology I, Heart Center (A.L., I.H., C.B., M.M.), Institute of Experimental and Clinical Pharmacology and Toxicology (A.L., D.F., S.B., R.G., L.H.), Renal Division, Department of Medicine (F.G., T.B.H.), and BIOSS Centre for Biological Signaling Studies (T.B.H., L.H.), University of Freiburg, Freiburg, Germany
| |
Collapse
|
14
|
Deng J, Wu G, Yang C, Li Y, Jing Q, Han Y. Rosuvastatin attenuates contrast-induced nephropathy through modulation of nitric oxide, inflammatory responses, oxidative stress and apoptosis in diabetic male rats. J Transl Med 2015; 13:53. [PMID: 25880311 PMCID: PMC4329210 DOI: 10.1186/s12967-015-0416-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Accepted: 01/22/2015] [Indexed: 12/12/2022] Open
Abstract
Background Contrast-induced nephropathy (CIN) is an important cause of acute renal failure. We observe the effect of rosuvastatin on preventing CIN in diabetic rats in current study. Methods Diabetic rats were then divided into five groups: 1 diabetic rats (D), 2 diabetic rats + contrast media (DCM), 3 diabetic rats + rosuvastatin (DR), 4 diabetic rats + contrast media + rosuvastatin (DRCM), 5 non-diabetic rat control (NDCM). Contrast-induced nephropathy was induced by intravenous injection a single dose of indomethacin (10 mg/kg), double doses of N-nitro-L-arginine methyl ester (10 mg/kg) and a single dose of high-osmolar contrast medium meglumine amidotrizoate (6 ml/kg). DR and DRCM group rats were treated with rosuvastatin (10 mg/kg/day) by gavage for 5 days. At the end of treatment, the experimental groups were sacrificed, and their renal tissues were investigated histopathologically beside assessments of functional activities, nitric oxide metabolites, and oxidative stress and apoptic markers. Results After 6 days, serum creatinine and urine microprotein were increased, and creatinine clearance, kidney nitrite were decreased in DCM rats compared with NDCM, D, DR and DRCM groups. Histopathology scores in group DCM were increased compared with groups NDCM, D and DR, but lower in group DRCM than in group DCM (p < 0.01). Kidney thiobarbituric acid-reacting substances (TBARS), serum malondialdehyde (MDA), and serum protein carbonyl content (PCC) were increased, and serum thiol was decreased in the DCM group compared with groups NDCM, D and DR; however, these results were reversed in group DRCM compared with group DCM. Both expression of IL-6, TNF-α and the percentage of apoptotic cells were increased in group DCM than in groups NDCM, D and DR, but they were decreased in group DRCM than in group DCM. The expression of phospho-p38, cleaved capase-3, and the Bax/Bcl-2 ratio, were increased in group DCM than in groups NDCM, D and DR, but were decreased in group DRCM than in group DCM. Conclusions Our study demonstrated that rosuvastatin treatment attenuated both inflammatory processes and apoptosis and inhibited oxidative stress and the p38 MAPK pathway in a diabetic rat model in the setting of CIN.
Collapse
Affiliation(s)
- Jie Deng
- Department of Cardiology, the General Hospital of Shenyang Military Region, Chinese People's Liberation Army, Shenyang, Liaoing, China.
| | - Guijun Wu
- Department of Cardiology, the General Hospital of Shenyang Military Region, Chinese People's Liberation Army, Shenyang, Liaoing, China.
| | - Chen Yang
- Department of Cardiology, the General Hospital of Shenyang Military Region, Chinese People's Liberation Army, Shenyang, Liaoing, China.
| | - Yi Li
- Department of Cardiology, the General Hospital of Shenyang Military Region, Chinese People's Liberation Army, Shenyang, Liaoing, China.
| | - Quanmin Jing
- Department of Cardiology, the General Hospital of Shenyang Military Region, Chinese People's Liberation Army, Shenyang, Liaoing, China.
| | - Yaling Han
- Department of Cardiology, the General Hospital of Shenyang Military Region, Chinese People's Liberation Army, Shenyang, Liaoing, China.
| |
Collapse
|
15
|
Corsetti G, D’Antona G, Ruocco C, Stacchiotti A, Romano C, Tedesco L, Dioguardi F, Rezzani R, Nisoli E. Dietary supplementation with essential amino acids boosts the beneficial effects of rosuvastatin on mouse kidney. Amino Acids 2014; 46:2189-203. [PMID: 24923264 PMCID: PMC4133027 DOI: 10.1007/s00726-014-1772-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Accepted: 05/26/2014] [Indexed: 12/25/2022]
Abstract
The effects of high-potency statins on renal function are controversial. To address the impact of statins on renal morpho-functional aspects, normotensive young mice were treated with rosuvastatin (Rvs). Moreover, because statins may impair mitochondrial function, mice received either dietary supplementation with an amino acid mixture enriched in essential amino acids (EAAm), which we previously demonstrated to increase mitochondrial biogenesis in muscle or an unsupplemented control diet for 1 month. Mitochondrial biogenesis and function, apoptosis, and insulin signaling pathway events were studied, primarily in cortical proximal tubules. By electron microscopy analysis, mitochondria were more abundant and more heterogeneous in size, with dense granules in the inner matrix, in Rvs- and Rvs plus EAAm-treated animals. Rvs administration increased protein kinase B and endothelial nitric oxide synthase phosphorylation, but the mammalian target of rapamycin signaling pathway was not affected. Rvs increased the expression of sirtuin 1, peroxisome proliferator-activated receptor γ coactivator-1α, cytochrome oxidase type IV, cytochrome c, and mitochondrial biogenesis markers. Levels of glucose-regulated protein 75 (Grp75), B-cell lymphoma 2, and cyclin-dependent kinase inhibitor 1 were increased in cortical proximal tubules, and expression of the endoplasmic reticulum-mitochondrial chaperone Grp78 was decreased. EAAm supplementation maintained or enhanced these changes. Rvs promotes mitochondrial biogenesis, with a probable anti-apoptotic effect. EAAm boosts these processes and may contribute to the efficient control of cellular energetics and survival in the mouse kidney. This suggests that appropriate nutritional interventions may enhance the beneficial actions of Rvs, and could potentially prevent chronic renal side effects.
Collapse
Affiliation(s)
- Giovanni Corsetti
- Division of Human Anatomy, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy
| | - Giuseppe D’Antona
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Chiara Ruocco
- Department of Medical Biotechnology and Translational Medicine, University of Milan, via Vanvitelli 32, 20129 Milan, Italy
| | - Alessandra Stacchiotti
- Division of Human Anatomy, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy
| | - Claudia Romano
- Division of Human Anatomy, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy
| | - Laura Tedesco
- Department of Medical Biotechnology and Translational Medicine, University of Milan, via Vanvitelli 32, 20129 Milan, Italy
| | - Francesco Dioguardi
- Department of Clinical Sciences and Community, University of Milan, 20122 Milan, Italy
| | - Rita Rezzani
- Division of Human Anatomy, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy
| | - Enzo Nisoli
- Department of Medical Biotechnology and Translational Medicine, University of Milan, via Vanvitelli 32, 20129 Milan, Italy
| |
Collapse
|
16
|
Kostapanos MS, Rizos CV, Elisaf MS. Benefit-risk assessment of rosuvastatin in the treatment of atherosclerosis and related diseases. Drug Saf 2014; 37:481-500. [PMID: 24788803 DOI: 10.1007/s40264-014-0169-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Rosuvastatin has been marketed for approximately a decade. In this review we critically discuss available evidence on the benefits and risks from its use. In clinical trials using rosuvastatin, 'lowest is best' was relevant for on-treatment low-density lipoprotein cholesterol levels. Targeting levels <50 mg/dl was associated with the greatest decrease in vascular morbidity/mortality in the primary prevention setting. Also, such reduction can induce atherosclerosis regression without increasing the risk of adverse effects. Pooled data suggest that the safety profile of rosuvastatin is not different from that of other statins. It was estimated that rosuvastatin-associated absolute hazards of muscle-, liver- and renal-related adverse effects are lower than the corresponding vascular benefits in moderate vascular risk individuals. However, these data are subject to biases and need confirmation on a prospective basis. Significant liver enzyme elevations are rare. These often imply underlying non-alcoholic fatty liver disease (NAFLD), which is associated with increased vascular risk. Rosuvastatin can improve biochemical biomarkers and histological score of NAFLD. Whether this benefit is associated with vascular risk reduction should be assessed by prospective studies. Both chronic kidney disease and albuminuria independently predict vascular morbidity and mortality. Rosuvastatin improved the estimated glomerular filtration rate and decreased albuminuria in patients with moderately impaired kidney function. Also, vascular morbidity and mortality might be reduced in these patients. The same was not relevant in end-stage renal disease. Rosuvastatin-induced proteinuria appears to be of tubular origin, not relating to kidney injury. Rosuvastatin increases the risk of new-onset diabetes by dose-dependently impairing insulin sensitivity. Obese individuals with prediabetes appear to be predominantly affected. However, absolute vascular benefits of rosuvastatin may counterbalance this risk. Rosuvastatin is effective for the prevention and management of atherosclerotic vascular disease. Individualization of its use can maximize benefits and reduce the risk of adverse effects.
Collapse
Affiliation(s)
- Michael S Kostapanos
- Department of Internal Medicine, Medical School, University of Ioannina, St. Niarchou Avenue, 45110, Ioannina, Greece
| | | | | |
Collapse
|
17
|
Kee HJ, Bae EH, Park S, Lee KE, Suh SH, Kim SW, Jeong MH. HDAC inhibition suppresses cardiac hypertrophy and fibrosis in DOCA-salt hypertensive rats via regulation of HDAC6/HDAC8 enzyme activity. Kidney Blood Press Res 2013; 37:229-39. [PMID: 23868068 DOI: 10.1159/000350148] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/04/2013] [Indexed: 11/19/2022] Open
Abstract
Background : Inhibition of histone deacetylase (HDAC) was reported to suppress cardiac hypertrophy and fibrosis in various hypertrophic animal models. However, the HDAC expression profile and HDAC enzyme activity have not yet been investigated in DOCA-salt hypertensive rats. Methods : Unilaterally nephrectomized rats were implanted with DOCA strips. DOCA-salt rats then received a control diet with vehicle or valproate. We measured the expression of cardiac hypertrophic markers, class I HDACs, class II HDACs, fibrosis, and HDAC enzyme activity. Results : Here we report that sodium valproate inhibits the cardiac hypertrophy accompanied by fibrosis in the heart of chronic hypertensive rats. We show that expression of GATA6 and HDAC6 is upregulated in DOCA-salt hypertension. In addition, HDAC6 and HDAC8 enzyme activity is attenuated by sodium valproate. Conclusion : These results suggest that a novel HDAC6- and HDAC8-selective inhibitor is needed to treat or prevent pathological cardiac hypertrophy. © 2013 S. Karger AG, Basel.
Collapse
Affiliation(s)
- Hae Jin Kee
- Heart Research Center of Chonnam National University Hospital, Gwangju 501-757, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
18
|
Ma SK, Choi JS, Joo SY, Kim HY, Kim CS, Bae EH, Lee JU, Kim SW. Activation of the Renal PI3K/Akt/mTOR Signaling Pathway in a DOCA-Salt Model of Hypertension. Chonnam Med J 2012; 48:150-4. [PMID: 23323219 PMCID: PMC3539094 DOI: 10.4068/cmj.2012.48.3.150] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 10/28/2012] [Accepted: 10/29/2012] [Indexed: 11/24/2022] Open
Abstract
The present study investigated the changes that occurred in the mammalian target of rapamycin (mTOR) signaling pathway in the kidney as a result of deoxycorticosterone acetate (DOCA)-salt hypertension. Rats were implanted with DOCA strips (200 mg/kg) 1 week after unilateral nephrectomy and were then supplied with 0.9% saline to drink. Four weeks after DOCA implantation, systolic blood pressure (SBP) was measured by use of the tail-cuff method. The expression levels of phosphorylated phosphatidylinositol-3-kinase (PI3K), Akt, and mTOR, as well as the protein expression levels of ED-1 and cyclooxygenase-2 (COX-2), transforming growth factor-β1 (TGF-β1), α-smooth muscle actin (SMA), caspase-3, Bax, and Bcl-2, were then examined in the kidney by semiquantitative immunoblotting. DOCA-salt hypertensive rats were found to have significantly increased SBP as well as an increased kidney weight-to-body weight ratio. Moreover, the phosphorylation of PI3K, Akt, and mTOR was increased in the kidney of DOCA-salt hypertensive rats compared with the control, as was the protein expression of ED-1, COX-2, TGF-β1, and α-SMA. The expression levels of caspase-3 and Bax were increased significantly, whereas Bcl-2 expression was decreased. In conclusion, the phosphorylation of PI3K/Akt/mTOR was increased in the kidney of DOCA-salt hypertensive rats.
Collapse
Affiliation(s)
- Seong Kwon Ma
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Nam HK, Lee SJ, Kim MH, Rho JH, Son YK, Lee SM, Kim SE, Kim KH, An WS. Rosuvastatin attenuates inflammation, apoptosis and fibrosis in a rat model of cyclosporine-induced nephropathy. Am J Nephrol 2012; 37:7-15. [PMID: 23258196 DOI: 10.1159/000345990] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Accepted: 11/21/2012] [Indexed: 12/13/2022]
Abstract
BACKGROUND/AIM Cyclosporine (CsA)-induced kidney injury is characterized by renal dysfunction with inflammatory cell infiltrations, apoptosis and fibrosis. Pleiotropic effects of statins may exert anti-inflammatory, antiapoptotic and antifibrotic actions beyond lipid control. The aim of this study is to investigate whether rosuvastatin (RUS) has anti-inflammatory, antiapoptotic and antifibrotic effects on chronic CsA-induced nephropathy in a rat model. METHODS Male Sprague-Dawley rats fed a low-sodium diet were divided into three treatment groups: control (0.9% saline injection), CsA (15 mg/kg/day by subcutaneous injection), CsA + RUS (10 mg/kg/day by gastric gavage). Renal function, CsA level and lipid levels were measured at the end of 4 weeks. The expression of ED-1, transforming growth factor-β(1) (TGF-β(1)) and α-smooth muscle actin (α-SMA) for inflammation and fibrosis were examined by Western blot analysis. The expression levels of apoptosis-associated factors were examined by Western blot analysis. Apoptosis was evaluated using the terminal deoxynucleotidyl transferase-mediated biotin nick end-labeling (TUNEL) method. RESULTS Kidney function was decreased in CsA-treated rats compared with controls, which was attenuated by RUS. RUS did not affect the lipid level or the blood CsA level. TUNEL staining showed that RUS inhibited CsA-induced tubular apoptosis. RUS decreased CsA-induced increased expression of Bax/Bcl-2 ratio. The expressions of ED-1, α-SMA, TGF-β(1), Smad2/3, Smad4 and p-JNK were increased in CsA-treated rats, which were attenuated by RUS. Tubular atrophy and interstitial fibrosis in CsA-treated rats were attenuated by RUS supplementation. CONCLUSION RUS supplementation attenuates proinflammatory and apoptosis-related factors and inhibits the fibrotic pathways including the smad-dependent and smad-independent pathways in a rat model of CsA-induced nephropathy.
Collapse
Affiliation(s)
- Hyun Kyung Nam
- Department of Internal Medicine, Busan Medical Center, Busan, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Abstract
Hydroxy methyl glutaryl CoA inhibitors (statins) are the agents most frequently used to reduce elevated serum cholesterol. In addition to their cholesterol lowering effects, statins also have nonlipid lowering pleiotropic properties. These include reducing oxidative stress, renin-angiotensin and endothelin synthesis and activity, and improving nitric oxide (NO) synthesis and availability. Thus, one would predict that statins might be able to exert an antihypertensive effect. Experimental models bear out the blood pressure lowering effects but the data from clinical trials have been inconsistent perhaps due to inappropriate experimental designs, sample size, blood pressure measurement techniques etc. Moreover, although experimental models strongly suggest a role for salt intake in the potential antihypertensive responses to statins, available clinical trials fail to report salt intake in the studied populations. The statins' antihypertensive effects remain an unsettled hypothesis and calls for a large clinical trial at a wide range of doses and a controlled salt intake. Statins meanwhile remain as a excellent option to control high cholesterol and in tissue injury prevention.
Collapse
|
21
|
Chu PY, Zatta A, Kiriazis H, Chin-Dusting J, Du XJ, Marshall T, Kaye DM. CXCR4 antagonism attenuates the cardiorenal consequences of mineralocorticoid excess. Circ Heart Fail 2011; 4:651-8. [PMID: 21685249 DOI: 10.1161/circheartfailure.110.960831] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Extensive evidence implicates aldosterone excess in the development and progression of cardiovascular disease states including hypertension, metabolic syndrome, cardiac hypertrophy, heart failure, and cardiorenal fibrosis. Recent studies show that activation of inflammatory cascade may play a specific role in the sequelae of mineralocorticoid activation, although the linking mechanism remains unclear. We tested the possibility that secondary stimulation of the stromal-derived factor 1/CXC chemokine receptor 4 (SDF-1/CXCR4) pathway plays a contributory role. METHODS AND RESULTS We investigated the effect of the highly selective CXCR4 antagonist AMD3465 (6 mg/kg per day for 6 weeks through minipump) in dexoycorticosterone acetate (DOCA)-treated, uninephrectomized mice. CXCR4 antagonism significantly attenuated the induction of cardiac fibrosis, renal fibrosis, hypertension, and left ventricular hypertrophy by DOCA. Mineralocorticoid excess also stimulated the accumulation of T-lymphocytes in the heart and kidney and this was significantly blunted by CXCR4 inhibition. CONCLUSIONS Taken together, these data strongly implicate the SDF-1/CXCR4 axis in the pathogenesis of mineralocorticoid excess induced hypertension, inflammation, and cardiorenal fibrosis. This insight provides a new potential therapeutic approach for the treatment of specific aspects of mineralocorticoid mediated cardiovascular disease.
Collapse
Affiliation(s)
- Po-Yin Chu
- Heart Failure Research Group, Baker IDI Heart and Diabetes Research Institute, Melbourne, Australia
| | | | | | | | | | | | | |
Collapse
|
22
|
Rosuvastatin beneficially alters the glomerular structure of kidneys from spontaneously hypertensive rats (SHRs). J Mol Histol 2011; 42:323-31. [PMID: 21670990 DOI: 10.1007/s10735-011-9336-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Accepted: 05/30/2011] [Indexed: 10/18/2022]
Abstract
The incidence of chronic renal diseases is increasing worldwide, and there is a great need to identify therapies capable of arresting or reducing disease progression. The current treatment of chronic nephropathies is limited to angiotensin-converting enzyme inhibitors and angiotensin receptor blockers, but increasing clinical and experimental evidence suggests that statins could play a therapeutic role. Ultrastructural studies have shown the presence of gap junctions within all the cells of the glomerulus and podocytes have been found to contain primarily connexin-43. The present study aims to observe the beneficial effects of rosuvastatin on structural and ultrastructural renal morphology and on glomerular connexin-43 expression in normotensive rats and spontaneously hypertensive rats (SHR). Rats were randomly allocated into four groups: WKY-C: normotensive animals no receiving rosuvastatin; WKY-ROS: normotensive animals receiving rosuvastatin; SHR-C: hypertensive animals no receiving rosuvastatin; SHR-ROS: hypertensive animals receiving rosuvastatin. Our results show no differences in blood urea, creatinine, uric acid and creatine phosphokinase levels between the groups, however, there was an decreasing of 24-h protein excretion in SHR-ROS. Capsular area in SHR-ROS was decreased, however, there was no alteration in urinary space. By transmission electron microscopy the slit diaphragm and podocyte foot processes were more preserved in SHR-ROS. By scanning electron microscopy the podocyte foot processes were more preserved in SHR-ROS. Increased connexin-43 immunofluorescence was observed in glomeruli of WKY-ROS and SHR-ROS. In conclusion, we hypothesize that renal pleiotropic effect of rosuvastatin can be a therapeutic tool for improving kidney ultrastructure and, consequently, renal function in hypertensive individuals.
Collapse
|
23
|
Girardi JM, Farias RE, Ferreira AP, Raposo NRB. Rosuvastatin prevents proteinuria and renal inflammation in nitric oxide-deficient rats. Clinics (Sao Paulo) 2011; 66:1457-62. [PMID: 21915500 PMCID: PMC3161228 DOI: 10.1590/s1807-59322011000800025] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2010] [Accepted: 05/11/2011] [Indexed: 11/28/2022] Open
Abstract
OBJECTIVE The aim of the present study was to assess the effects of rosuvastatin on renal injury and inflammation in a model of nitric oxide deficiency. METHODS Male Wistar rats were randomly divided into four groups (n = 10/group) and treated for 28 days with saline (CTRL); 30 mg/kg/day L-NAME (L-name); L-NAME and 20 mg/kg/day rosuvastatin (L-name+ROS-20); or L-NAME and 2 mg/kg/day rosuvastatin (L-name+ROS-2). Systolic blood pressure was measured by plethysmography in the central artery of the tail. The serum total cholesterol, triglycerides, alanine aminotransferase, aspartate aminotransferase, alkaline phosphatase, creatinine, nitric oxide, interleukin-6, and tumor necrosis factor alpha levels were analyzed. Urine samples were taken to measure the albumin: urinary creatinine ratio. Kidneys were sectioned and stained with hematoxylin/eosin and Masson's trichrome. Immunohistochemical analysis of the renal tissue was performed to detect macrophage infiltration of the glomeruli. RESULTS The systolic blood pressure was elevated in the L-name but not the L-name+rosuvastatin-20 and L-name+rosuvastatin-2 groups. The L-name group had a significantly reduced nitric oxide level and an increased interleukin-6 and tumor necrosis factor alpha level, albumin: urinary creatinine ratio and number of macrophages in the renal glomeruli. Rosuvastatin increased the nitric oxide level in the L-name+rosuvastatin-2 group and reduced the interleukin-6 and tumor necrosis factor alpha levels, glomerular macrophage number and albumin:urinary creatinine ratio in the L-name+rosuvastatin-20 and L-name+rosuvastatin-2 groups. CONCLUSION Rosuvastatin treatment reduced glomerular damage due to improvement in the inflammatory pattern independent of the systolic blood pressure and serum lipid level. These effects may lead to improvements in the treatment of kidney disease.
Collapse
|
24
|
Gluba A, Rysz J, Banach M. Statins in patients with chronic kidney disease: why, who and when? Expert Opin Pharmacother 2010; 11:2665-74. [DOI: 10.1517/14656566.2010.512419] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|