1
|
Noels H, van der Vorst EPC, Rubin S, Emmett A, Marx N, Tomaszewski M, Jankowski J. Renal-Cardiac Crosstalk in the Pathogenesis and Progression of Heart Failure. Circ Res 2025; 136:1306-1334. [PMID: 40403103 DOI: 10.1161/circresaha.124.325488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 02/14/2025] [Accepted: 03/11/2025] [Indexed: 05/24/2025]
Abstract
Chronic kidney disease (CKD) represents a global health issue with a high socioeconomic impact. Beyond a progressive decline of kidney function, patients with CKD are at increased risk of cardiovascular diseases, including heart failure (HF) and sudden cardiac death. HF in CKD can manifest both as HF with reduced ejection fraction and HF with preserved ejection fraction, with the latter further increasing in relative importance in the more advanced stages of CKD. Typical cardiac remodeling characteristics in uremic cardiomyopathy include left ventricular hypertrophy, myocardial fibrosis, cardiac electrical dysregulation, capillary rarefaction, and microvascular dysfunction, which are triggered by increased cardiac preload, cardiac afterload, and preload and afterload-independent factors. The pathophysiological mechanisms underlying cardiac remodeling in CKD are multifactorial and include neurohormonal activation (with increased activation of the renin-angiotensin-aldosterone system, the sympathetic nervous system, and mineralocorticoid receptor signaling), cardiac steroid activation, mitochondrial dysfunction, inflammation, innate immune activation, and oxidative stress. Furthermore, disturbances in cardiac metabolism and calcium homeostasis, macrovascular and microvascular dysfunction, increased cellular profibrotic responses, the accumulation of uremic retention solutes, and mineral and bone disorders also contribute to cardiovascular disease and HF in CKD. Here, we review the current knowledge of HF in CKD, including the clinical characteristics and pathophysiological mechanisms revealed in animal studies. We also elaborate on the detrimental impact of comorbidities of CKD on HF using hypertension as an example and discuss the clinical characteristics of hypertensive heart disease and the genetic predisposition. Overall, this review aims to increase the understanding of HF in CKD to support future research and clinical translational approaches for improved diagnosis and therapy of this vulnerable patient population.
Collapse
Affiliation(s)
- Heidi Noels
- Institute for Molecular Cardiovascular Research (H.N., E.P.C.v.d.V., J.J.), Uniklinik RWTH Aachen, RWTH Aachen University, Germany
- Aachen-Maastricht Institute for Cardiorenal Disease (H.N., E.P.C.v.d.V., J.J.), Uniklinik RWTH Aachen, RWTH Aachen University, Germany
- Biochemistry Department (H.N.), Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands
| | - Emiel P C van der Vorst
- Institute for Molecular Cardiovascular Research (H.N., E.P.C.v.d.V., J.J.), Uniklinik RWTH Aachen, RWTH Aachen University, Germany
- Aachen-Maastricht Institute for Cardiorenal Disease (H.N., E.P.C.v.d.V., J.J.), Uniklinik RWTH Aachen, RWTH Aachen University, Germany
- Interdisciplinary Center for Clinical Research (IZKF) (E.P.C.v.d.V.), RWTH Aachen University, Germany
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Germany (E.P.C.v.d.V.)
| | - Sébastien Rubin
- L'Institut national de la santé et de la recherche médicale (INSERM), BMC, U1034, University of Bordeaux, Pessac, France (S.R.)
- Renal Unit, University Hospital of Bordeaux, France (S.R.)
| | - Amber Emmett
- Faculty of Medicine, Biology and Health, Division of Cardiovascular Sciences, The University of Manchester, United Kingdom (A.E., M.T.)
| | - Nikolaus Marx
- Department of Internal Medicine I-Cardiology, Angiology and Internal Intensive Care Medicine (N.M.), RWTH Aachen University, Germany
| | - Maciej Tomaszewski
- Faculty of Medicine, Biology and Health, Division of Cardiovascular Sciences, The University of Manchester, United Kingdom (A.E., M.T.)
- British Heart Foundation Manchester Centre of Research Excellence, United Kingdom (M.T.)
- Manchester Academic Health Science Centre, Manchester University National Health Service (NHS) Foundation Trust, United Kingdom (M.T.)
- Signature Research Programme in Health Services and Systems Research, Duke-National University of Singapore (M.T.)
| | - Joachim Jankowski
- Institute for Molecular Cardiovascular Research (H.N., E.P.C.v.d.V., J.J.), Uniklinik RWTH Aachen, RWTH Aachen University, Germany
- Biochemistry Department (H.N.), Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands
- Pathology Department (J.J.), Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands
| |
Collapse
|
2
|
Landim-Vieira M, Nieto Morales PF, ElSafty S, Kahmini AR, Ranek MJ, Solís C. The role of mechanosignaling in the control of myocardial mass. Am J Physiol Heart Circ Physiol 2025; 328:H622-H638. [PMID: 39739566 DOI: 10.1152/ajpheart.00277.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 12/03/2024] [Accepted: 12/17/2024] [Indexed: 01/02/2025]
Abstract
Regulation of myocardial mass is key for maintaining cardiovascular health. This review highlights the complex and regulatory relationship between mechanosignaling and myocardial mass, influenced by many internal and external factors including hemodynamic and microgravity, respectively. The heart is a dynamic organ constantly adapting to changes in workload (preload and afterload) and mechanical stress exerted on the myocardium, influencing both physiological adaptations and pathological remodeling. Mechanosignaling pathways, such as the mitogen-activated protein kinases (MAPKs) and the phosphoinositide 3-kinases and serine/threonine kinase (PI3K/Akt) pathways, mediate downstream effects on gene expression and play key roles in transducing mechanical cues into biochemical signals, thereby modulating cellular processes, including control of myocardial mass. Dysregulation of these processes can lead to pathological cardiac remodeling, such as hypertrophic cardiomyopathy. Furthermore, recent studies have highlighted the importance of protein quality control mechanisms, such as the ubiquitin-proteasome system, in settings of extreme physiological conditions that alter the heart workload such as pregnancy and microgravity. Overall, this review provides a thorough insight into how mechanical signals are converted into chemical signals to regulate myocardial mass in both healthy and diseased conditions.
Collapse
Affiliation(s)
- Maicon Landim-Vieira
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, Florida, United States
| | - Paula F Nieto Morales
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, Florida, United States
| | - Summer ElSafty
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, Florida, United States
| | - Aida Rahimi Kahmini
- Department of Health, Nutrition, and Food Science, Florida State University, Tallahassee, Florida, United States
| | - Mark J Ranek
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University, Baltimore, Maryland, United States
| | - Christopher Solís
- Department of Health, Nutrition, and Food Science, Florida State University, Tallahassee, Florida, United States
| |
Collapse
|
3
|
Sun Y, Xu M, Duan Q, Bryant JL, Xu X. The role of autophagy in the progression of HIV infected cardiomyopathy. Front Cell Dev Biol 2024; 12:1372573. [PMID: 39086659 PMCID: PMC11289186 DOI: 10.3389/fcell.2024.1372573] [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: 01/18/2024] [Accepted: 06/05/2024] [Indexed: 08/02/2024] Open
Abstract
Although highly active antiretroviral therapy (HAART) has changed infection with human immunodeficiency virus (HIV) from a diagnosis with imminent mortality to a chronic illness, HIV positive patients who do not develop acquired immunodeficiency syndrome (AIDs) still suffer from a high rate of cardiac dysfunction and fibrosis. Regardless of viral load and CD count, HIV-associated cardiomyopathy (HIVAC) still causes a high rate of mortality and morbidity amongst HIV patients. While this is a well characterized clinical phenomena, the molecular mechanism of HIVAC is not well understood. In this review, we consolidate, analyze, and discuss current research on the intersection between autophagy and HIVAC. Multiple studies have linked dysregulation in various regulators and functional components of autophagy to HIV infection regardless of mode of viral entry, i.e., coronary, cardiac chamber, or pericardial space. HIV proteins, including negative regulatory factor (Nef), glycoprotein 120 (gp120), and transactivator (Tat), have been shown to interact with type II microtubule-associated protein-1 β light chain (LC3-II), Rubiquitin, SQSTM1/p62, Rab7, autophagy-specific gene 7 (ATG7), and lysosomal-associated membrane protein 1 (LAMP1), all molecules critical to normal autophagy. HIV infection can also induce dysregulation of mitochondrial bioenergetics by altering production and equilibrium of adenosine triphosphate (ATP), mitochondrial reactive oxygen species (ROS), and calcium. These changes alter mitochondrial mass and morphology, which normally trigger autophagy to clear away dysfunctional organelles. However, with HIV infection also triggering autophagy dysfunction, these abnormal mitochondria accumulate and contribute to myocardial dysfunction. Likewise, use of HAART, azidothymidine and Abacavir, have been shown to induce cardiac dysfunction and fibrosis by inducing abnormal autophagy during antiretroviral therapy. Conversely, studies have shown that increasing autophagy can reduce the accumulation of dysfunctional mitochondria and restore cardiomyocyte function. Interestingly, Rapamycin, a mammalian target of rapamycin (mTOR) inhibitor, has also been shown to reduce HIV-induced cytotoxicity by regulating autophagy-related proteins, making it a non-antiviral agent with the potential to treat HIVAC. In this review, we synthesize these findings to provide a better understanding of the role autophagy plays in HIVAC and discuss the potential pharmacologic targets unveiled by this research.
Collapse
Affiliation(s)
- Yuting Sun
- Laboratory of Cell Biology, Genetics and Developmental Biology, College of Life Sciences and University Hospital, Shaanxi Normal University, Xi’an, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Mengmeng Xu
- Department of Pediatrics, Morgan Stanley Children’s Hospital, Columbia University, New York, NY, United States
| | - Qinchun Duan
- Laboratory of Cell Biology, Genetics and Developmental Biology, College of Life Sciences and University Hospital, Shaanxi Normal University, Xi’an, China
| | - Joseph L. Bryant
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Xuehong Xu
- Laboratory of Cell Biology, Genetics and Developmental Biology, College of Life Sciences and University Hospital, Shaanxi Normal University, Xi’an, China
| |
Collapse
|
4
|
Opałka B, Żołnierczuk M, Grabowska M. Immunosuppressive Agents-Effects on the Cardiovascular System and Selected Metabolic Aspects: A Review. J Clin Med 2023; 12:6935. [PMID: 37959400 PMCID: PMC10647341 DOI: 10.3390/jcm12216935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 10/28/2023] [Accepted: 11/02/2023] [Indexed: 11/15/2023] Open
Abstract
The widespread use of immunosuppressive drugs makes it possible to reduce inflammation in autoimmune diseases, as well as prevent transplant rejection in organ recipients. Despite their key action in blocking the body's immune response, these drugs have many side effects. These actions primarily affect the cardiovascular system, and the incidence of complications in patients using immunosuppressive drugs is significant, being associated with a higher incidence of cardiovascular incidents such as myocardial infarction and stroke. This paper analyzes the mechanisms of action of commonly used immunosuppressive drugs and their impact on the cardiovascular system. The adverse effect of immunosuppressive drugs is associated with toxicity within the cardiovascular system, which may be a problem in the clinical management of patients after transplantation. Immunosuppressants act on the cardiovascular system in a variety of ways, including fibrosis and myocardial remodeling, endothelium disfunction, hypertension, atherosclerosis, dyslipidemia or hyperglycaemia, metabolic syndrome, and hyperuricemia. The use of multidrug protocols makes it possible to develop regimens that can reduce the incidence of cardiovascular events. A better understanding of their mechanism of action and the range of complications could enable physicians to select the appropriate therapy for a given patient, as well as to reduce complications and prolong life.
Collapse
Affiliation(s)
- Bianka Opałka
- Department of Histology and Developmental Biology, Faculty of Health Sciences, Pomeranian Medical University, 71-210 Szczecin, Poland;
| | - Michał Żołnierczuk
- Department of Plastic, Endocrine and General Surgery, Pomeranian Medical University, 72-010 Szczecin, Poland;
| | - Marta Grabowska
- Department of Histology and Developmental Biology, Faculty of Health Sciences, Pomeranian Medical University, 71-210 Szczecin, Poland;
| |
Collapse
|
5
|
Dube P, Aradhyula V, Lad A, Khalaf FK, Breidenbach JD, Kashaboina E, Gorthi S, Varatharajan S, Stevens TW, Connolly JA, Soehnlen SM, Sood A, Marellapudi A, Ranabothu M, Kleinhenz AL, Domenig O, Dworkin LD, Malhotra D, Haller ST, Kennedy DJ. Novel Model of Oxalate Diet-Induced Chronic Kidney Disease in Dahl-Salt-Sensitive Rats. Int J Mol Sci 2023; 24:10062. [PMID: 37373209 DOI: 10.3390/ijms241210062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 05/12/2023] [Accepted: 05/20/2023] [Indexed: 06/29/2023] Open
Abstract
Diet-induced models of chronic kidney disease (CKD) offer several advantages, including clinical relevance and animal welfare, compared with surgical models. Oxalate is a plant-based, terminal toxic metabolite that is eliminated by the kidneys through glomerular filtration and tubular secretion. An increased load of dietary oxalate leads to supersaturation, calcium oxalate crystal formation, renal tubular obstruction, and eventually CKD. Dahl-Salt-Sensitive (SS) rats are a common strain used to study hypertensive renal disease; however, the characterization of other diet-induced models on this background would allow for comparative studies of CKD within the same strain. In the present study, we hypothesized that SS rats on a low-salt, oxalate rich diet would have increased renal injury and serve as novel, clinically relevant and reproducible CKD rat models. Ten-week-old male SS rats were fed either 0.2% salt normal chow (SS-NC) or a 0.2% salt diet containing 0.67% sodium oxalate (SS-OX) for five weeks.Real-time PCR demonstrated an increased expression of inflammatory marker interleukin-6 (IL-6) (p < 0.0001) and fibrotic marker Timp-1 metalloproteinase (p < 0.0001) in the renal cortex of SS-OX rat kidneys compared with SS-NC. The immunohistochemistry of kidney tissue demonstrated an increase in CD-68 levels, a marker of macrophage infiltration in SS-OX rats (p < 0.001). In addition, SS-OX rats displayed increased 24 h urinary protein excretion (UPE) (p < 0.01) as well as significant elevations in plasma Cystatin C (p < 0.01). Furthermore, the oxalate diet induced hypertension (p < 0.05). A renin-angiotensin-aldosterone system (RAAS) profiling (via liquid chromatography-mass spectrometry; LC-MS) in the SS-OX plasma showed significant (p < 0.05) increases in multiple RAAS metabolites including angiotensin (1-5), angiotensin (1-7), and aldosterone. The oxalate diet induces significant renal inflammation, fibrosis, and renal dysfunction as well as RAAS activation and hypertension in SS rats compared with a normal chow diet. This study introduces a novel diet-induced model to study hypertension and CKD that is more clinically translatable and reproducible than the currently available models.
Collapse
Affiliation(s)
- Prabhatchandra Dube
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43606, USA
| | - Vaishnavi Aradhyula
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43606, USA
| | - Apurva Lad
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43606, USA
| | - Fatimah K Khalaf
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43606, USA
- Department of Medicine, University of Alkafeel College of Medicine, Najaf 54001, Iraq
| | - Joshua D Breidenbach
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43606, USA
| | - Eshita Kashaboina
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43606, USA
| | - Snigdha Gorthi
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43606, USA
| | - Shangari Varatharajan
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43606, USA
| | - Travis W Stevens
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43606, USA
| | - Jacob A Connolly
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43606, USA
| | - Sophia M Soehnlen
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43606, USA
| | - Ambika Sood
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43606, USA
| | - Amulya Marellapudi
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43606, USA
| | - Meghana Ranabothu
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43606, USA
| | - Andrew L Kleinhenz
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43606, USA
| | | | - Lance D Dworkin
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43606, USA
| | - Deepak Malhotra
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43606, USA
| | - Steven T Haller
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43606, USA
| | - David J Kennedy
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43606, USA
| |
Collapse
|
6
|
Gomez-Manjarres DC, Axell-House DB, Patel DC, Odackal J, Yu V, Burdick MD, Mehrad B. Sirolimus suppresses circulating fibrocytes in idiopathic pulmonary fibrosis in a randomized controlled crossover trial. JCI Insight 2023; 8:e166901. [PMID: 36853800 PMCID: PMC10243828 DOI: 10.1172/jci.insight.166901] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 02/23/2023] [Indexed: 03/01/2023] Open
Abstract
BACKGROUNDFibrocytes are BM-derived circulating cells that traffic to the injured lungs and contribute to fibrogenesis. The mTOR inhibitor, sirolimus, inhibits fibrocyte CXCR4 expression, reducing fibrocyte traffic and attenuating lung fibrosis in animal models. We sought to test the hypothesis that short-term treatment with sirolimus reduces the concentration of CXCR4+ circulating fibrocytes in patients with idiopathic pulmonary fibrosis (IPF).METHODSWe conducted a short-term randomized double-blind placebo-controlled crossover pilot trial to assess the safety and tolerability of sirolimus in IPF. Participants were randomly assigned to sirolimus or placebo for approximately 6 weeks, and after a 4-week washout, they were assigned to the alternate treatment. Toxicity, lung function, and the concentration of circulating fibrocytes were measured before and after each treatment.RESULTSIn the 28 study participants, sirolimus resulted in a statistically significant 35% decline in the concentration of total fibrocytes, 34% decline in CXCR4+ fibrocytes, and 42% decline in fibrocytes expressing α-smooth muscle actin, but no significant change in these populations occurred on placebo. Respiratory adverse events occurred more frequently during treatment with placebo than sirolimus; the incidence of adverse events and drug tolerability did not otherwise differ during therapy with drug and placebo. Lung function was unaffected by either treatment, with the exception of a small decline in gas transfer during treatment with placebo.CONCLUSIONAs compared with placebo, short-term treatment with sirolimus resulted in reduction of circulating fibrocyte concentrations in participants with IPF, with an acceptable safety profile.TRIAL REGISTRATIONClinicalTrials.gov, accession no. NCT01462006.FUNDINGNIH R01HL098329 and American Heart Association 18TPA34170486.
Collapse
Affiliation(s)
- Diana C. Gomez-Manjarres
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Florida, Gainesville, Florida, USA
| | - Dierdre B. Axell-House
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Divya C. Patel
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Florida, Gainesville, Florida, USA
| | - John Odackal
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Victor Yu
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Marie D. Burdick
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Florida, Gainesville, Florida, USA
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Borna Mehrad
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Florida, Gainesville, Florida, USA
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| |
Collapse
|
7
|
Elezaby A, Dexheimer R, Sallam K. Cardiovascular effects of immunosuppression agents. Front Cardiovasc Med 2022; 9:981838. [PMID: 36211586 PMCID: PMC9534182 DOI: 10.3389/fcvm.2022.981838] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 08/30/2022] [Indexed: 11/26/2022] Open
Abstract
Immunosuppressive medications are widely used to treat patients with neoplasms, autoimmune conditions and solid organ transplants. Key drug classes, namely calcineurin inhibitors, mammalian target of rapamycin (mTOR) inhibitors, and purine synthesis inhibitors, have direct effects on the structure and function of the heart and vascular system. In the heart, immunosuppressive agents modulate cardiac hypertrophy, mitochondrial function, and arrhythmia risk, while in vasculature, they influence vessel remodeling, circulating lipids, and blood pressure. The aim of this review is to present the preclinical and clinical literature examining the cardiovascular effects of immunosuppressive agents, with a specific focus on cyclosporine, tacrolimus, sirolimus, everolimus, mycophenolate, and azathioprine.
Collapse
Affiliation(s)
- Aly Elezaby
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, United States
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, CA, United States
| | - Ryan Dexheimer
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, United States
| | - Karim Sallam
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, United States
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, CA, United States
- *Correspondence: Karim Sallam
| |
Collapse
|
8
|
Liu J, Tian J, Sodhi K, Shapiro JI. The Na/K-ATPase Signaling and SGLT2 Inhibitor-Mediated Cardiorenal Protection: A Crossed Road? J Membr Biol 2021; 254:513-529. [PMID: 34297135 PMCID: PMC8595165 DOI: 10.1007/s00232-021-00192-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/06/2021] [Indexed: 12/17/2022]
Abstract
In different large-scale clinic outcome trials, sodium (Na+)/glucose co-transporter 2 (SGLT2) inhibitors showed profound cardiac- and renal-protective effects, making them revolutionary treatments for heart failure and kidney disease. Different theories are proposed according to the emerging protective effects other than the original purpose of glucose-lowering in diabetic patients. As the ATP-dependent primary ion transporter providing the Na+ gradient to drive other Na+-dependent transporters, the possible role of the sodium–potassium adenosine triphosphatase (Na/K-ATPase) as the primary ion transporter and its signaling function is not explored.
Collapse
Affiliation(s)
- Jiang Liu
- Department of Biomedical Sciences, JCE School of Medicine, Marshall University, Huntington, WV, USA.
| | - Jiang Tian
- Department of Biomedical Sciences, JCE School of Medicine, Marshall University, Huntington, WV, USA
| | - Komal Sodhi
- Department of Surgery, JCE School of Medicine, Marshall University, Huntington, WV, USA
| | - Joseph I Shapiro
- Departments of Medicine, JCE School of Medicine, Marshall University, Huntington, WV, USA
| |
Collapse
|
9
|
Zhong M, Cui B, Xiang J, Wu X, Wen Q, Li Q, Zhang F. Rapamycin is Effective for Upper but not for Lower Gastrointestinal Crohn's Disease-Related Stricture: A Pilot Study. Front Pharmacol 2021; 11:617535. [PMID: 33628182 PMCID: PMC7898031 DOI: 10.3389/fphar.2020.617535] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 12/23/2020] [Indexed: 11/18/2022] Open
Abstract
Crohn's disease (CD)-related fibrotic stricture remains a clinical challenge because of no effective treatments. This study aimed to evaluate the potential efficacy of rapamycin in patients with CD-related strictures in different locations in gastrointestinal tract. A pilot prospective study on using rapamycin for CD-related stricture was performed from April 2015 to August 2020 in a single center in China. Fifteen patients were enrolled into the study. The clinical efficacy was evaluated by diet score and gastrointestinal obstruction symptoms score. Clinical responses were defined as the ability to tolerate the regular diet with vegetable fiber combined with a reduction of ≥75% in overall target score and a score of less than two points for each item. Three patients discontinued rapamycin for less than 1-month due to intolerance to adverse events, then, 12 patients received ≥1 dose of the rapamycin and provided ≥1 post-baseline target score after baseline were included for intent-to-treat (ITT) analysis. 100% (5/5) of patients with upper gastrointestinal strictures achieved clinical response after using rapamycin. However, no clinical response was observed in those patients with CD lesions in lower gastrointestinal tract. Adverse events occurred in 40% (6/15) of patients. No death or serious opportunistic infections were observed in the present study. This study firstly reported that rapamycin might be effective for CD-related stricture in the upper, but not in lower gastrointestinal tract.
Collapse
Affiliation(s)
- Min Zhong
- Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Key Lab of Holistic Integrative Enterology, Nanjing Medical University, Nanjing, China
| | - Bota Cui
- Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Key Lab of Holistic Integrative Enterology, Nanjing Medical University, Nanjing, China
| | - Jie Xiang
- Department of Gastroenterology, The Central Hospital of Enshi Autonomous Prefecture, Enshi, China
| | - Xia Wu
- Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Key Lab of Holistic Integrative Enterology, Nanjing Medical University, Nanjing, China
| | - Quan Wen
- Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Key Lab of Holistic Integrative Enterology, Nanjing Medical University, Nanjing, China
| | - Qianqian Li
- Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Key Lab of Holistic Integrative Enterology, Nanjing Medical University, Nanjing, China
| | - Faming Zhang
- Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Key Lab of Holistic Integrative Enterology, Nanjing Medical University, Nanjing, China
| |
Collapse
|
10
|
Abdelkarim Maki MA, Kumar PV, Elumalai M, Bayazeid O. In silico-based Approach to Investigate the Ability of PEGylated Rapamycin to Inhibit Galectin-3. Curr Drug Discov Technol 2021; 18:451-456. [PMID: 31969105 DOI: 10.2174/1570163817666200122162042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 12/20/2019] [Accepted: 12/30/2019] [Indexed: 02/08/2023]
Abstract
AIMS To utilize in silico-based approach for investigating the ability of PEGylated rapamycin as a competitive inhibitor to Galectin-3 for curing various diseases or that may provide an attractive strategy for treatment of a wide variety of tumors. BACKGROUND Galectin-3 (Gal-3) signaling protein is a unique member of lectin family present at the cell surface, intracellularly in both the nucleus and cytoplasm and extracellularly in the general circulation. Circulating Gal-3 is present in both normal and cancer cells. High levels of circulating Gal-3 have been proven to be associated with inflammation and fibrosis in several acute and chronic conditions, which include neurological degeneration, inflammatory and immune responses, autoimmune diseases, diabetes, heart failure, atherosclerosis, response to infection, wound healing, liver, lung, and kidney disease. Gal-3 is known to regulate many biological activities including cell adhesion, angiogenesis, growth, apoptosis, migration, and metastasis. Rapamycin has been examined alone or in combination with other drugs for treatment of various cancers in clinical studies. Although it has shown promising therapeutic effects, its clinical development was interrupted by poor aqueous solubility and limited preferential distribution. To overcome these limitations, RA has been chemically modified to hydrophilic analogues, such as everolimus (EV). However, all these approaches can only partially increase the solubility, but has little effect on the blood distribution and pharmacokinetics. Therefore, it is necessary to explore other RA conjugates to improve aqueous solubility and tissue distribution profile. Recently we reported that RP-MPEG inhibits the growth of various cancer cell lines by acting on mammalian target of RP (mTOR) receptor site and it can be used for gastric cancer. OBJECTIVE To construct various molecular weight RP-MPEG by replacing MPEG chain in 40-O-(2- hydroxyethyl) position of the EV and analyze their binding affinity to Gal-3. METHODS The chemical structures of various molecular weight RP-MPEG were built using ChemSketch software. The molecular docking study was performed to find the best probable structure of RP-MPEG for competitive inhibition of the CRD, based on the interaction score. For that purpose, the 3D structures of RP and EV were obtained from NCBI PubChem compound database, where the structural protein-co-crystallized ligand complex of Gal-3 (TD2, as a native ligand) was retrieved from RCSB Protein Data Bank. All structures of the selected compounds, served as molecules for molecular modeling, were optimized through MOE.2014 software before docking. Hydrogen atoms and partial charges were added to the protein. Protein minimization was performed in gas solvation with the side chains, keeping it rigid and the ligand flexible. The selected site was isolated and minimized, followed by protonating the protein. The 3D ligands were minimized using MMFF94x with cutoffs of 10 to 12 Å. The hydrogens and charges were fixed, and the RMS gradient was set to 0.001 kcal/mol. The docking results were analyzed to identify and assess the binding affinity of these compounds to CRD using drug discovery software. RESULTS Our results indicated that RP-MPEG with MW 1178.51 g/mol has a logP value of 3.79 and has possessed the strongest binding affinity toward CRD of Gal-3 with a docking score of -6.87. Compared with TD2, there were additional close contacts for RP-MPEG (MW 1178.51 g/mol), coming from three hydrogen bonds with Asp148, Arg162, and Arg144 which suggest that this ligand is a strong competitive inhibitor among the other molecules for Gal-3. CONCLUSION RP-MPEG with the MW 1178.51 g/mol could be a promising blocker for various biological action of Gal-3 includes profibrotic activity, modulation of immune responses and inflammatory responses to cancer that contributes to neoplastic transformation, angiogenesis and metastasis. Other: The 95% confidence intervals (CIs) of the binding affinity (according to their mean and standard errors) were estimated with 2.5 and 97.5 percentile as the lower and upper bounds.
Collapse
Affiliation(s)
| | - Palanirajan Vijayaraj Kumar
- Faculty of Pharmaceutical Sciences, UCSI University, No. 1, Jalan Menara Gading, Taman Connaught, Cheras 56000, Kuala Lumpur, Malaysia
| | - Manogaran Elumalai
- Faculty of Pharmaceutical Sciences, UCSI University, No. 1, Jalan Menara Gading, Taman Connaught, Cheras 56000, Kuala Lumpur, Malaysia
| | - Omer Bayazeid
- Department of Pharmacognosy, Faculty of Pharmacy, Hacettepe University, 06100 Ankara, Turkey
| |
Collapse
|
11
|
Elucidating Potential Profibrotic Mechanisms of Emerging Biomarkers for Early Prognosis of Hepatic Fibrosis. Int J Mol Sci 2020; 21:ijms21134737. [PMID: 32635162 PMCID: PMC7369895 DOI: 10.3390/ijms21134737] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 06/29/2020] [Accepted: 07/01/2020] [Indexed: 02/06/2023] Open
Abstract
Hepatic fibrosis has been associated with a series of pathophysiological processes causing excessive accumulation of extracellular matrix proteins. Several cellular processes and molecular mechanisms have been implicated in the diseased liver that augments fibrogenesis, fibrogenic cytokines and associated liver complications. Liver biopsy remains an essential diagnostic tool for histological evaluation of hepatic fibrosis to establish a prognosis. In addition to being invasive, this methodology presents with several limitations including poor cost-effectiveness, prolonged hospitalizations, and risks of peritoneal bleeding, while the clinical use of this method does not reveal underlying pathogenic mechanisms. Several alternate noninvasive diagnostic strategies have been developed, to determine the extent of hepatic fibrosis, including the use of direct and indirect biomarkers. Immediate diagnosis of hepatic fibrosis by noninvasive means would be more palatable than a biopsy and could assist clinicians in taking early interventions timely, avoiding fatal complications, and improving prognosis. Therefore, we sought to review some common biomarkers of liver fibrosis along with some emerging candidates, including the oxidative stress-mediated biomarkers, epigenetic and genetic markers, exosomes, and miRNAs that needs further evaluation and would have better sensitivity and specificity. We also aim to elucidate the potential role of cardiotonic steroids (CTS) and evaluate the pro-inflammatory and profibrotic effects of CTS in exacerbating hepatic fibrosis. By understanding the underlying pathogenic processes, the efficacy of these biomarkers could allow for early diagnosis and treatment of hepatic fibrosis in chronic liver diseases, once validated.
Collapse
|
12
|
Kaesler N, Babler A, Floege J, Kramann R. Cardiac Remodeling in Chronic Kidney Disease. Toxins (Basel) 2020; 12:toxins12030161. [PMID: 32150864 PMCID: PMC7150902 DOI: 10.3390/toxins12030161] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/02/2020] [Accepted: 03/03/2020] [Indexed: 12/19/2022] Open
Abstract
Cardiac remodeling occurs frequently in chronic kidney disease patients and affects quality of life and survival. Current treatment options are highly inadequate. As kidney function declines, numerous metabolic pathways are disturbed. Kidney and heart functions are highly connected by organ crosstalk. Among others, altered volume and pressure status, ischemia, accelerated atherosclerosis and arteriosclerosis, disturbed mineral metabolism, renal anemia, activation of the renin-angiotensin system, uremic toxins, oxidative stress and upregulation of cytokines stress the sensitive interplay between different cardiac cell types. The fatal consequences are left-ventricular hypertrophy, fibrosis and capillary rarefaction, which lead to systolic and/or diastolic left-ventricular failure. Furthermore, fibrosis triggers electric instability and sudden cardiac death. This review focuses on established and potential pathophysiological cardiorenal crosstalk mechanisms that drive uremia-induced senescence and disease progression, including potential known targets and animal models that might help us to better understand the disease and to identify novel therapeutics.
Collapse
Affiliation(s)
- Nadine Kaesler
- Clinic for Renal and Hypertensive Disorders, Rheumatological and Immunological Disease, University Hospital of the RWTH Aachen, 52074 Aachen, Germany
| | - Anne Babler
- Clinic for Renal and Hypertensive Disorders, Rheumatological and Immunological Disease, University Hospital of the RWTH Aachen, 52074 Aachen, Germany
| | - Jürgen Floege
- Clinic for Renal and Hypertensive Disorders, Rheumatological and Immunological Disease, University Hospital of the RWTH Aachen, 52074 Aachen, Germany
| | - Rafael Kramann
- Clinic for Renal and Hypertensive Disorders, Rheumatological and Immunological Disease, University Hospital of the RWTH Aachen, 52074 Aachen, Germany
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands
- Correspondence:
| |
Collapse
|
13
|
Liu J, Nie Y, Chaudhry M, Bai F, Chuang J, Sodhi K, Shapiro JI. The Redox-Sensitive Na/K-ATPase Signaling in Uremic Cardiomyopathy. Int J Mol Sci 2020; 21:ijms21041256. [PMID: 32069992 PMCID: PMC7072896 DOI: 10.3390/ijms21041256] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 02/10/2020] [Accepted: 02/10/2020] [Indexed: 02/07/2023] Open
Abstract
In recent years, Na/K-ATPase signaling has been implicated in different physiological and pathophysiological conditions, including cardiac hypertrophy and uremic cardiomyopathy. Cardiotonic steroids (CTS), specific ligands of Na/K-ATPase, regulate its enzymatic activity (at higher concentrations) and signaling function (at lower concentrations without significantly affecting its enzymatic activity) and increase reactive oxygen species (ROS) generation. On the other hand, an increase in ROS alone also regulates the Na/K-ATPase enzymatic activity and signaling function. We termed this phenomenon the Na/K-ATPase-mediated oxidant-amplification loop, in which oxidative stress regulates both the Na/K-ATPase activity and signaling. Most recently, we also demonstrated that this amplification loop is involved in the development of uremic cardiomyopathy. This review aims to evaluate the redox-sensitive Na/K-ATPase-mediated oxidant amplification loop and uremic cardiomyopathy.
Collapse
Affiliation(s)
- Jiang Liu
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA; (Y.N.); (M.C.); (F.B.)
- Correspondence:
| | - Ying Nie
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA; (Y.N.); (M.C.); (F.B.)
| | - Muhammad Chaudhry
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA; (Y.N.); (M.C.); (F.B.)
| | - Fang Bai
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA; (Y.N.); (M.C.); (F.B.)
| | - Justin Chuang
- Department of Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA; (J.C.); (K.S.); (J.I.S.)
| | - Komal Sodhi
- Department of Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA; (J.C.); (K.S.); (J.I.S.)
| | - Joseph I. Shapiro
- Department of Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA; (J.C.); (K.S.); (J.I.S.)
| |
Collapse
|
14
|
Khalaf FK, Tassavvor I, Mohamed A, Chen Y, Malhotra D, Xie Z, Tian J, Haller ST, Westfall K, Tang WHW, Kennedy DJ. Epithelial and Endothelial Adhesion of Immune Cells Is Enhanced by Cardiotonic Steroid Signaling Through Na +/K +-ATPase-α-1. J Am Heart Assoc 2020; 9:e013933. [PMID: 32013704 PMCID: PMC7033897 DOI: 10.1161/jaha.119.013933] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Background Recent studies have highlighted a critical role for a group of natriuretic hormones, cardiotonic steroid (CTS), in mediating renal inflammation and fibrosis associated with volume expanded settings, such as chronic kidney disease. Immune cell adhesion is a critical step in the inflammatory response; however, little is currently understood about the potential regulatory role of CTS signaling in this setting. Herein, we tested the hypothesis that CTS signaling through Na+/K+‐ATPase α‐1 (NKA α‐1) enhances immune cell recruitment and adhesion to renal epithelium that ultimately advance renal inflammation. Methods and Results We demonstrate that knockdown of the α‐1 isoform of Na/K‐ATPase causes a reduction in CTS‐induced macrophage infiltration in renal tissue as well reduces the accumulation of immune cells in the peritoneal cavity in vivo. Next, using functional adhesion assay, we demonstrate that CTS‐induced increases in the adhesion of macrophages to renal epithelial cells were significantly diminished after reduction of NKA α‐1 in either macrophages or renal epithelial cells as well after inhibition of NKA α‐1‐Src signaling cascade with a specific peptide inhibitor, pNaKtide in vitro. Finally, CTS‐induced expression of adhesion markers in both endothelial and immune cells was significantly inhibited in an NKA α‐1‐Src signaling dependent manner in vitro. Conclusions These findings suggest that CTS potentiates immune cell migration and adhesion to renal epithelium through an NKA α‐1–dependent mechanism; our new findings suggest that pharmacological inhibition of this feed‐forward loop may be useful in the treatment of renal inflammation associated with renal disease.
Collapse
Affiliation(s)
- Fatimah K Khalaf
- University of Toledo College of Medicine and Life Sciences Toledo OH
| | - Iman Tassavvor
- University of Toledo College of Medicine and Life Sciences Toledo OH
| | - Amal Mohamed
- University of Toledo College of Medicine and Life Sciences Toledo OH
| | - Yiliang Chen
- Blood Research Institute Blood Center of Wisconsin Milwaukee WI
| | - Deepak Malhotra
- University of Toledo College of Medicine and Life Sciences Toledo OH
| | - Zijian Xie
- Marshall Institute for Interdisciplinary Research Marshall University Huntington WV
| | - Jiang Tian
- University of Toledo College of Medicine and Life Sciences Toledo OH
| | - Steven T Haller
- University of Toledo College of Medicine and Life Sciences Toledo OH
| | - Kristen Westfall
- Department of Cardiovascular and Metabolic Sciences Lerner Research Institute Cleveland Clinic Cleveland OH
| | - W H Wilson Tang
- Department of Cardiovascular and Metabolic Sciences Lerner Research Institute Cleveland Clinic Cleveland OH.,Center for Cardiovascular Diagnostics and Prevention Lerner Research Institute Cleveland Clinic Cleveland OH.,Department of Cardiovascular Medicine, Heart and Vascular Institute Cleveland Clinic Cleveland OH
| | - David J Kennedy
- University of Toledo College of Medicine and Life Sciences Toledo OH
| |
Collapse
|
15
|
Abstract
The term uraemic cardiomyopathy refers to the cardiac abnormalities that are seen in patients with chronic kidney disease (CKD). Historically, this term was used to describe a severe cardiomyopathy that was associated with end-stage renal disease and characterized by severe functional abnormalities that could be reversed following renal transplantation. In a modern context, uraemic cardiomyopathy describes the clinical phenotype of cardiac disease that accompanies CKD and is perhaps best characterized as diastolic dysfunction seen in conjunction with left ventricular hypertrophy and fibrosis. A multitude of factors may contribute to the pathogenesis of uraemic cardiomyopathy, and current treatments only modestly improve outcomes. In this Review, we focus on evolving concepts regarding the roles of fibroblast growth factor 23 (FGF23), inflammation and systemic oxidant stress and their interactions with more established mechanisms such as pressure and volume overload resulting from hypertension and anaemia, respectively, activation of the renin-angiotensin and sympathetic nervous systems, activation of the transforming growth factor-β (TGFβ) pathway, abnormal mineral metabolism and increased levels of endogenous cardiotonic steroids.
Collapse
Affiliation(s)
- Xiaoliang Wang
- Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, USA
| | - Joseph I Shapiro
- Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, USA.
| |
Collapse
|
16
|
Xie JX, Zhang S, Cui X, Zhang J, Yu H, Khalaf FK, Malhotra D, Kennedy DJ, Shapiro JI, Tian J, Haller ST. Na/K-ATPase/src complex mediates regulation of CD40 in renal parenchyma. Nephrol Dial Transplant 2019; 33:1138-1149. [PMID: 29294050 DOI: 10.1093/ndt/gfx334] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 11/07/2017] [Indexed: 02/07/2023] Open
Abstract
Background Recent studies have highlighted a critical role for CD40 in the pathogenesis of renal injury and fibrosis. However, little is currently understood about the regulation of CD40 in this setting. Methods We use novel Na/K-ATPase cell lines and inhibitors in order to demonstrate the regulatory function of Na/K-ATPase with regards to CD40 expression and function. We utilize 5/6 partial nephrectomy as well as direct infusion of a Na/K-ATPase ligand to demonstrate this mechanism exists in vivo. Results We demonstrate that knockdown of the α1 isoform of Na/K-ATPase causes a reduction in CD40 while rescue of the α1 but not the α2 isoform restores CD40 expression in renal epithelial cells. Second, because the major functional difference between α1 and α2 is the ability of α1 to form a functional signaling complex with Src, we examined whether the Na/K-ATPase/Src complex is important for CD40 expression. We show that a gain-of-Src binding α2 mutant restores CD40 expression while loss-of-Src binding α1 reduces CD40 expression. Furthermore, loss of a functional Na/K-ATPase/Src complex also disrupts CD40 signaling. Importantly, we show that use of a specific Na/K-ATPase/Src complex antagonist, pNaKtide, can attenuate cardiotonic steroid (CTS)-induced induction of CD40 expression in vitro. Conclusions Because the Na/K-ATPase/Src complex is also a key player in the pathogenesis of renal injury and fibrosis, our new findings suggest that Na/K-ATPase and CD40 may comprise a pro-fibrotic feed-forward loop in the kidney and that pharmacological inhibition of this loop may be useful in the treatment of renal fibrosis.
Collapse
Affiliation(s)
- Jeffrey X Xie
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Shungang Zhang
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Xiaoyu Cui
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV, USA
| | - Jue Zhang
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Yu
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fatimah K Khalaf
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Deepak Malhotra
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - David J Kennedy
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Joseph I Shapiro
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV, USA
| | - Jiang Tian
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Steven T Haller
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| |
Collapse
|
17
|
Yan Y, Wang J, Chaudhry MA, Nie Y, Sun S, Carmon J, Shah PT, Bai F, Pratt R, Brickman C, Sodhi K, Kim JH, Pierre S, Malhotra D, Rankin GO, Xie ZJ, Shapiro JI, Liu J. Metabolic Syndrome and Salt-Sensitive Hypertension in Polygenic Obese TALLYHO/JngJ Mice: Role of Na/K-ATPase Signaling. Int J Mol Sci 2019; 20:ijms20143495. [PMID: 31315267 PMCID: PMC6678942 DOI: 10.3390/ijms20143495] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 07/06/2019] [Accepted: 07/13/2019] [Indexed: 12/12/2022] Open
Abstract
We have demonstrated that Na/K-ATPase acts as a receptor for reactive oxygen species (ROS), regulating renal Na+ handling and blood pressure. TALLYHO/JngJ (TH) mice are believed to mimic the state of obesity in humans with a polygenic background of type 2 diabetes. This present work is to investigate the role of Na/K-ATPase signaling in TH mice, focusing on susceptibility to hypertension due to chronic excess salt ingestion. Age-matched male TH and the control C57BL/6J (B6) mice were fed either normal diet or high salt diet (HS: 2, 4, and 8% NaCl) to construct the renal function curve. Na/K-ATPase signaling including c-Src and ERK1/2 phosphorylation, as well as protein carbonylation (a commonly used marker for enhanced ROS production), were assessed in the kidney cortex tissues by Western blot. Urinary and plasma Na+ levels were measured by flame photometry. When compared to B6 mice, TH mice developed salt-sensitive hypertension and responded to a high salt diet with a significant rise in systolic blood pressure indicative of a blunted pressure-natriuresis relationship. These findings were evidenced by a decrease in total and fractional Na+ excretion and a right-shifted renal function curve with a reduced slope. This salt-sensitive hypertension correlated with changes in the Na/K-ATPase signaling. Specifically, Na/K-ATPase signaling was not able to be stimulated by HS due to the activated baseline protein carbonylation, phosphorylation of c-Src and ERK1/2. These findings support the emerging view that Na/K-ATPase signaling contributes to metabolic disease and suggest that malfunction of the Na/K-ATPase signaling may promote the development of salt-sensitive hypertension in obesity. The increased basal level of renal Na/K-ATPase-dependent redox signaling may be responsible for the development of salt-sensitive hypertension in polygenic obese TH mice.
Collapse
Affiliation(s)
- Yanling Yan
- Departments of Clinical & Translational Sciences, Biomedical Sciences, and Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA.
| | - Jiayan Wang
- Departments of Clinical & Translational Sciences, Biomedical Sciences, and Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Muhammad A Chaudhry
- Departments of Clinical & Translational Sciences, Biomedical Sciences, and Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Ying Nie
- Departments of Clinical & Translational Sciences, Biomedical Sciences, and Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Shuyan Sun
- Departments of Clinical & Translational Sciences, Biomedical Sciences, and Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
- Hebei Medical University, Shijiazhuang 50017, China
| | - Jazmin Carmon
- Departments of Clinical & Translational Sciences, Biomedical Sciences, and Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Preeya T Shah
- Departments of Clinical & Translational Sciences, Biomedical Sciences, and Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Fang Bai
- Departments of Clinical & Translational Sciences, Biomedical Sciences, and Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Rebecca Pratt
- Departments of Clinical & Translational Sciences, Biomedical Sciences, and Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Cameron Brickman
- Departments of Clinical & Translational Sciences, Biomedical Sciences, and Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Komal Sodhi
- Departments of Clinical & Translational Sciences, Biomedical Sciences, and Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Jung Han Kim
- Departments of Clinical & Translational Sciences, Biomedical Sciences, and Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Sandrine Pierre
- Departments of Clinical & Translational Sciences, Biomedical Sciences, and Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Deepak Malhotra
- Department of Medicine, The University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Gary O Rankin
- Departments of Clinical & Translational Sciences, Biomedical Sciences, and Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Zi-Jian Xie
- Departments of Clinical & Translational Sciences, Biomedical Sciences, and Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Joseph I Shapiro
- Departments of Clinical & Translational Sciences, Biomedical Sciences, and Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Jiang Liu
- Departments of Clinical & Translational Sciences, Biomedical Sciences, and Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| |
Collapse
|
18
|
Sciarretta S, Forte M, Frati G, Sadoshima J. New Insights Into the Role of mTOR Signaling in the Cardiovascular System. Circ Res 2019; 122:489-505. [PMID: 29420210 DOI: 10.1161/circresaha.117.311147] [Citation(s) in RCA: 342] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The mTOR (mechanistic target of rapamycin) is a master regulator of several crucial cellular processes, including protein synthesis, cellular growth, proliferation, autophagy, lysosomal function, and cell metabolism. mTOR interacts with specific adaptor proteins to form 2 multiprotein complexes, called mTORC1 (mTOR complex 1) and mTORC2 (mTOR complex 2). In the cardiovascular system, the mTOR pathway regulates both physiological and pathological processes in the heart. It is needed for embryonic cardiovascular development and for maintaining cardiac homeostasis in postnatal life. Studies involving mTOR loss-of-function models revealed that mTORC1 activation is indispensable for the development of adaptive cardiac hypertrophy in response to mechanical overload. mTORC2 is also required for normal cardiac physiology and ensures cardiomyocyte survival in response to pressure overload. However, partial genetic or pharmacological inhibition of mTORC1 reduces cardiac remodeling and heart failure in response to pressure overload and chronic myocardial infarction. In addition, mTORC1 blockade reduces cardiac derangements induced by genetic and metabolic disorders and has been reported to extend life span in mice. These studies suggest that pharmacological targeting of mTOR may represent a therapeutic strategy to confer cardioprotection, although clinical evidence in support of this notion is still scarce. This review summarizes and discusses the new evidence on the pathophysiological role of mTOR signaling in the cardiovascular system.
Collapse
Affiliation(s)
- Sebastiano Sciarretta
- From the Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy (S.S., G.F.); Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli, Italy (S.S., M.F., G.F.); and Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (J.S.)
| | - Maurizio Forte
- From the Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy (S.S., G.F.); Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli, Italy (S.S., M.F., G.F.); and Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (J.S.)
| | - Giacomo Frati
- From the Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy (S.S., G.F.); Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli, Italy (S.S., M.F., G.F.); and Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (J.S.)
| | - Junichi Sadoshima
- From the Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy (S.S., G.F.); Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli, Italy (S.S., M.F., G.F.); and Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (J.S.).
| |
Collapse
|
19
|
Xu T, Lin T, Xie J, Ren H, Chen N, Wang W. Comparison of anti-peritoneal fibrotic effects between an mTORC1-specific blocker and a PI3K/mTOR dual-blocker. Ren Fail 2019; 41:267-277. [PMID: 30982374 PMCID: PMC6484467 DOI: 10.1080/0886022x.2019.1596818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
OBJECTIVE To compare the anti-peritoneal fibrotic effects between a mammalian target of rapamycin complex 1-specific blocker and a phosphatidyl-inositol 3-kinase/mammalian target of rapamycin dual-blocker. METHODS A total of 40 male Sprague-Dawley rats were randomly divided into five groups with eight animals per group. The normal group (N group) did not receive any intervention. The normal saline group (NS group) received an intraperitoneal injection of normal saline at 1 ml/100 g daily. The model group (3 W group), rapamycin (RAPA) group and BEZ235 (PI3K/mTOR dual-blocker) group all received an intraperitoneal injection of 0.1% chlorhexidine gluconate at 1 ml/100g daily. And the RAPA and BEZ235 groups also received a 0.5 mg/d RAPA or 2.5 mg/d BEZ235 gavage every day, respectively. Rats in each group were sacrificed after 3 weeks. RESULTS Immunohistochemistry, real-time PCR and western blotting analysis of fibrosis-related indicators (FN, Col 1, and α-SMA) confirmed that RAPA and BEZ235 significantly inhibited peritoneal fibrosis and that these two drugs had similar effects. The p-Akt, p-mTOR, p-p70S6K expression levels were significantly up-regulated in the 3 W group compared to the NS group, confirming that the mTOR pathway was significantly activated during peritoneal fibrosis. RAPA significantly inhibited the phosphorylation of mTOR and p70S6K but did not have significant effects on p-Akt upstream of mTOR. BEZ235 had significant inhibitory effects on all signaling molecules (p-Akt, p-mTOR, and p-p70S6K) in the mTOR pathway. CONCLUSION RAPA did not up-regulate p-Akt in a negative feedback fashion. Both drugs effectively inhibited peritoneal fibrosis.
Collapse
Affiliation(s)
- Tian Xu
- a Department of Nephrology, Institute of Nephrology, Ruijin Hospital , Shanghai Jiaotong University School of Medicine , Shanghai , China
| | - Tao Lin
- a Department of Nephrology, Institute of Nephrology, Ruijin Hospital , Shanghai Jiaotong University School of Medicine , Shanghai , China
| | - Jingyuan Xie
- a Department of Nephrology, Institute of Nephrology, Ruijin Hospital , Shanghai Jiaotong University School of Medicine , Shanghai , China
| | - Hong Ren
- a Department of Nephrology, Institute of Nephrology, Ruijin Hospital , Shanghai Jiaotong University School of Medicine , Shanghai , China
| | - Nan Chen
- a Department of Nephrology, Institute of Nephrology, Ruijin Hospital , Shanghai Jiaotong University School of Medicine , Shanghai , China
| | - Weiming Wang
- a Department of Nephrology, Institute of Nephrology, Ruijin Hospital , Shanghai Jiaotong University School of Medicine , Shanghai , China
| |
Collapse
|
20
|
Zanoli L, Lentini P, Briet M, Castellino P, House AA, London GM, Malatino L, McCullough PA, Mikhailidis DP, Boutouyrie P. Arterial Stiffness in the Heart Disease of CKD. J Am Soc Nephrol 2019; 30:918-928. [PMID: 31040188 DOI: 10.1681/asn.2019020117] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
CKD frequently leads to chronic cardiac dysfunction. This complex relationship has been termed as cardiorenal syndrome type 4 or cardio-renal link. Despite numerous studies and reviews focused on the pathophysiology and therapy of this syndrome, the role of arterial stiffness has been frequently overlooked. In this regard, several pathogenic factors, including uremic toxins (i.e., uric acid, phosphates, endothelin-1, advanced glycation end-products, and asymmetric dimethylarginine), can be involved. Their effect on the arterial wall, direct or mediated by chronic inflammation and oxidative stress, results in arterial stiffening and decreased vascular compliance. The increase in aortic stiffness results in increased cardiac workload and reduced coronary artery perfusion pressure that, in turn, may lead to microvascular cardiac ischemia. Conversely, reduced arterial stiffness has been associated with increased survival. Several approaches can be considered to reduce vascular stiffness and improve vascular function in patients with CKD. This review primarily discusses current understanding of the mechanisms concerning uremic toxins, arterial stiffening, and impaired cardiac function, and the therapeutic options to reduce arterial stiffness in patients with CKD.
Collapse
Affiliation(s)
| | - Paolo Lentini
- Division of Nephrology and Dialysis, St. Bassiano Hospital, Bassano del Grappa, Italy
| | - Marie Briet
- Institut National de la Santé et de la Recherche Médicale U1083, National Center for Scientific Research Joint Research Unit 6214, Centre Hospitalo-Universitaire d'Angers, Université d'Angers, Angers, France
| | - Pietro Castellino
- Internal Medicine, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Andrew A House
- Department of Medicine, University of Western Ontario, London, Ontario, Canada
| | - Gerard M London
- Institut National de la Santé et de la Recherche Médicale U970, Paris, France
| | - Lorenzo Malatino
- Internal Medicine, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Peter A McCullough
- Department of Medicine, Baylor University Medical Center, Baylor Heart and Vascular Institute, Baylor Jack and Jane Hamilton Heart and Vascular Hospital, Dallas, Texas
| | | | - Pierre Boutouyrie
- Institut National de la Santé et de la Recherche Médicale U970, Paris, France.,Faculté de Médecine, Université Paris Descartes, Sorbonne Paris Cité, Paris, France; and.,Department of Pharmacology, Hôpital Européen Georges-Pompidou, Assistance Publique-Hôpitaux de Paris, Paris, France
| |
Collapse
|
21
|
Li S, Peng F, Gong W, Wu J, Wang Y, Xu Z, Liu W, Li H, Yin B, Zhang Y, Chen S, Luo C, Li P, Chen Y, Huang Q, Zhou W, Long H. Dimethylaminomicheliolide ameliorates peritoneal fibrosis through the activation of autophagy. J Mol Med (Berl) 2019; 97:659-674. [PMID: 30854581 DOI: 10.1007/s00109-019-01757-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 01/22/2019] [Accepted: 02/11/2019] [Indexed: 12/29/2022]
Abstract
Peritoneal fibrosis (PF) is a major cause of ultrafiltration failure in patients receiving long-term peritoneal dialysis (PD), and effective prevention and treatment strategies are urgently needed. The dimethylamino Michael adduct of a natural product-derived micheliolide (MCL), dimethylaminomicheliolide (DMAMCL), is a new lead compound with the advantages of high stability, low toxicity, and sustainable release of MCL. This study aimed to investigate the protective effect of DMAMCL against PD-related PF and the mechanisms involved. In this study, we found that DMAMCL significantly decreased PD-induced extracellular matrix (ECM) deposition in a mouse model of PD, and that delayed DMAMCL administration halted the progression of PF in an established PD model. In addition, rapamycin administration induced autophagy and significantly ameliorated PF. The protective effect of DMAMCL against PF was weakened when co-administered with DMAMCL and 3-methyladenine. Inducing autophagy by rapamycin decreased transforming growth factor-β1-induced ECM accumulation in vitro. MCL promoted autophagy and inhibited ECM deposition. The anti-fibrotic effect of MCL was eliminated when knocking down ATG7 by siRNA. Taken together, DMAMCL might prevent against PF through activating autophagy. The anti-fibrotic effect of DMAMCL may be a new candidate for the treatment in patients with PD-related PF. KEY MESSAGES: Dimethylaminomicheliolide, the pro-drug of micheliolide, protects against peritoneal fibrosis in a mouse peritoneal dialysis model. Micheliolide inhibits TGF-β1-induced extracellular matrix accumulation in vitro. Autophagy plays a protective role against peritoneal fibrosis. The antifibrogenic effect of dimethylaminomicheliolide may be due to the activation of autophagy.
Collapse
Affiliation(s)
- Shuting Li
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Fenfen Peng
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Wangqiu Gong
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Jiayu Wu
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Yuxian Wang
- Department of Gerontology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Zhaozhong Xu
- Department of Emergency, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Wenting Liu
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Hongyu Li
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Bohui Yin
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Ying Zhang
- Department of Nephrology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, China
| | - Sijia Chen
- Department of Nephrology, The First Hospital of Changsha, Changsha, 410000, China
| | - Congwei Luo
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Peilin Li
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Yihua Chen
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Qianyin Huang
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Weidong Zhou
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| | - Haibo Long
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| |
Collapse
|
22
|
Asleh R, Briasoulis A, Pereira NL, Edwards BS, Frantz RP, Daly RC, Lerman A, Kushwaha SS. Hypercholesterolemia after conversion to sirolimus as primary immunosuppression and cardiac allograft vasculopathy in heart transplant recipients. J Heart Lung Transplant 2018; 37:1372-1380. [PMID: 30174165 DOI: 10.1016/j.healun.2018.07.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 07/01/2018] [Accepted: 07/05/2018] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Sirolimus (SRL) attenuates cardiac allograft vasculopathy (CAV) progression after heart transplantation (HT) but often results in hyperlipidemia. In this study we investigated the differential effects of SRL-based and calcineurin inhibitor (CNI)-based immunosuppression on CAV progression and clinical outcomes in HT recipients. METHODS CAV progression was assessed by coronary intravascular ultrasound (IVUS) as changes in volumetric measurements after correction to time between the first and last follow-up IVUS exams. CAV progression rate and CAV-associated events were compared between patients with mean follow-up low-density lipoprotein (LDL) <100 mg/dl (lower level or LL) and ≥100 mg/dl (higher level or HL) in the SRL and CNI groups. RESULTS We identified 227 patients on SRL (LL: 118; HL: 109) and 96 on CNI (LL: 56; HL: 40), with a median follow-up of 6.7 years. Clinical characteristics did not differ between the LL and HL groups and all patients were on statins. In the SRL arm, there were no significant differences in CAV progression rate and there were no differences in all-cause mortality and CAV-associated events between the LL and HL groups. In the CNI arm, the Δ change in plaque volume normalized to segment length and time of follow-up (PV/SL/year) (0.55 ± 0.53 vs 1.53 ± 2.32, p = 0.003) and Δ change in plaque index per year (defined as PV/vessel volume ratio) (3.1 ± 3.7% vs 6.3 ± 10.4%; p = 0.034) were significantly lower in the LL than the HL group. After adjusting for patient characteristics, HL was associated with higher rates of advanced CAV requiring coronary angioplasty (hazard ratio [HR] 3.0, 95% confidence interval [CI] 1.05 to 9.40, p = 0.040) and higher rates of all CAV-associated events (HR 2.2, 95% CI 1.10 to 4.54, p = 0.026) in these CNI-treated subjects. CONCLUSION Unlike CNI-based immunosuppression, the effects of SRL on attenuating CAV progression are independent of LDL cholesterol levels post-HT.
Collapse
Affiliation(s)
- Rabea Asleh
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Naveen L Pereira
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, USA
| | - Brooks S Edwards
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, USA
| | - Robert P Frantz
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, USA
| | - Richard C Daly
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, USA
| | - Amir Lerman
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, USA
| | - Sudhir S Kushwaha
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, USA.
| |
Collapse
|
23
|
Khalaf FK, Dube P, Mohamed A, Tian J, Malhotra D, Haller ST, Kennedy DJ. Cardiotonic Steroids and the Sodium Trade Balance: New Insights into Trade-Off Mechanisms Mediated by the Na⁺/K⁺-ATPase. Int J Mol Sci 2018; 19:E2576. [PMID: 30200235 PMCID: PMC6165267 DOI: 10.3390/ijms19092576] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 08/24/2018] [Accepted: 08/26/2018] [Indexed: 02/06/2023] Open
Abstract
In 1972 Neal Bricker presented the "trade-off" hypothesis in which he detailed the role of physiological adaptation processes in mediating some of the pathophysiology associated with declines in renal function. In the late 1990's Xie and Askari published seminal studies indicating that the Na⁺/K⁺-ATPase (NKA) was not only an ion pump, but also a signal transducer that interacts with several signaling partners. Since this discovery, numerous studies from multiple laboratories have shown that the NKA is a central player in mediating some of these long-term "trade-offs" of the physiological adaptation processes which Bricker originally proposed in the 1970's. In fact, NKA ligands such as cardiotonic steroids (CTS), have been shown to signal through NKA, and consequently been implicated in mediating both adaptive and maladaptive responses to volume overload such as fibrosis and oxidative stress. In this review we will emphasize the role the NKA plays in this "trade-off" with respect to CTS signaling and its implication in inflammation and fibrosis in target organs including the heart, kidney, and vasculature. As inflammation and fibrosis exhibit key roles in the pathogenesis of a number of clinical disorders such as chronic kidney disease, heart failure, atherosclerosis, obesity, preeclampsia, and aging, this review will also highlight the role of newly discovered NKA signaling partners in mediating some of these conditions.
Collapse
Affiliation(s)
- Fatimah K Khalaf
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Health Education Building RM 205, 3000 Arlington Ave, Toledo, OH 43614, USA.
| | - Prabhatchandra Dube
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Health Education Building RM 205, 3000 Arlington Ave, Toledo, OH 43614, USA.
| | - Amal Mohamed
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Health Education Building RM 205, 3000 Arlington Ave, Toledo, OH 43614, USA.
| | - Jiang Tian
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Health Education Building RM 205, 3000 Arlington Ave, Toledo, OH 43614, USA.
| | - Deepak Malhotra
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Health Education Building RM 205, 3000 Arlington Ave, Toledo, OH 43614, USA.
| | - Steven T Haller
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Health Education Building RM 205, 3000 Arlington Ave, Toledo, OH 43614, USA.
| | - David J Kennedy
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Health Education Building RM 205, 3000 Arlington Ave, Toledo, OH 43614, USA.
| |
Collapse
|
24
|
Kennedy DJ, Khalaf FK, Sheehy B, Weber ME, Agatisa-Boyle B, Conic J, Hauser K, Medert CM, Westfall K, Bucur P, Fedorova OV, Bagrov AY, Tang WHW. Telocinobufagin, a Novel Cardiotonic Steroid, Promotes Renal Fibrosis via Na⁺/K⁺-ATPase Profibrotic Signaling Pathways. Int J Mol Sci 2018; 19:ijms19092566. [PMID: 30158457 PMCID: PMC6164831 DOI: 10.3390/ijms19092566] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 08/26/2018] [Accepted: 08/27/2018] [Indexed: 12/24/2022] Open
Abstract
Cardiotonic steroids (CTS) are Na+/K+-ATPase (NKA) ligands that are elevated in volume-expanded states and associated with cardiac and renal dysfunction in both clinical and experimental settings. We test the hypothesis that the CTS telocinobufagin (TCB) promotes renal dysfunction in a process involving signaling through the NKA α-1 in the following studies. First, we infuse TCB (4 weeks at 0.1 µg/g/day) or a vehicle into mice expressing wild-type (WT) NKA α-1, as well as mice with a genetic reduction (~40%) of NKA α-1 (NKA α-1+/−). Continuous TCB infusion results in increased proteinuria and cystatin C in WT mice which are significantly attenuated in NKA α-1+/− mice (all p < 0.05), despite similar increases in blood pressure. In a series of in vitro experiments, 24-h treatment of HK2 renal proximal tubular cells with TCB results in significant dose-dependent increases in both Collagens 1 and 3 mRNA (2-fold increases at 10 nM, 5-fold increases at 100 nM, p < 0.05). Similar effects are seen in primary human renal mesangial cells. TCB treatment (100 nM) of SYF fibroblasts reconstituted with cSrc results in a 1.5-fold increase in Collagens 1 and 3 mRNA (p < 0.05), as well as increases in both Transforming Growth factor beta (TGFb, 1.5 fold, p < 0.05) and Connective Tissue Growth Factor (CTGF, 2 fold, p < 0.05), while these effects are absent in SYF cells without Src kinase. In a patient study of subjects with chronic kidney disease, TCB is elevated compared to healthy volunteers. These studies suggest that the pro-fibrotic effects of TCB in the kidney are mediated though the NKA-Src kinase signaling pathway and may have relevance to volume-overloaded conditions, such as chronic kidney disease where TCB is elevated.
Collapse
Affiliation(s)
- David J Kennedy
- Department of Medicine, University of Toledo College of Medicine, Toledo, OH 43614, USA.
| | - Fatimah K Khalaf
- Department of Medicine, University of Toledo College of Medicine, Toledo, OH 43614, USA.
| | - Brendan Sheehy
- Department of Cellular and Molecular Medicine, Lerner Research Institute Cleveland Clinic, Cleveland, OH 44106, USA.
| | - Malory E Weber
- Department of Cellular and Molecular Medicine, Lerner Research Institute Cleveland Clinic, Cleveland, OH 44106, USA.
| | - Brendan Agatisa-Boyle
- Department of Cellular and Molecular Medicine, Lerner Research Institute Cleveland Clinic, Cleveland, OH 44106, USA.
| | - Julijana Conic
- Department of Cellular and Molecular Medicine, Lerner Research Institute Cleveland Clinic, Cleveland, OH 44106, USA.
| | - Kayla Hauser
- Department of Cellular and Molecular Medicine, Lerner Research Institute Cleveland Clinic, Cleveland, OH 44106, USA.
| | - Charles M Medert
- Department of Cellular and Molecular Medicine, Lerner Research Institute Cleveland Clinic, Cleveland, OH 44106, USA.
| | - Kristen Westfall
- Department of Cellular and Molecular Medicine, Lerner Research Institute Cleveland Clinic, Cleveland, OH 44106, USA.
| | - Philip Bucur
- Department of Cellular and Molecular Medicine, Lerner Research Institute Cleveland Clinic, Cleveland, OH 44106, USA.
| | - Olga V Fedorova
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| | - Alexei Y Bagrov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, St. Petersburg 194223, Russia.
| | - W H Wilson Tang
- Department of Cellular and Molecular Medicine, Lerner Research Institute Cleveland Clinic, Cleveland, OH 44106, USA.
- Center for Cardiovascular Diagnostics and Prevention, Lerner Research Institute Cleveland Clinic, Cleveland, OH 44106, USA.
- Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
| |
Collapse
|
25
|
Piecha G, Kujawa-Szewieczek A, Kuczera P, Skiba K, Sikora-Grabka E, Więcek A. Plasma marinobufagenin immunoreactivity in patients with chronic kidney disease: a case control study. Am J Physiol Renal Physiol 2018; 315:F637-F643. [PMID: 30043626 DOI: 10.1152/ajprenal.00046.2018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Experimental data have shown increased plasma levels of marinobufagenin in kidney failure. In this case-controlled retrospective analysis, we evaluated plasma marinobufagenin immunoreactivity in hemodialysis patients compared with subjects with normal kidney function. Sixty-eight adult hemodialysis patients with chronic kidney disease (34 females and 34 males) as well as 68 age-, gender-, and blood pressure-matched subjects without chronic kidney disease were enrolled. Patients on stable hemodialysis regimen for at least 3 mo before the study were included. Exclusion criteria were: age <18 yr, severe liver or heart insufficiency, and overhydration. Subjects without chronic kidney disease must have had an estimated glomerular filtration rate ≥60 ml·min-1·1.72 m-2 according to the Modification of Diet in Renal Disease formula. Plasma marinobufagenin immunoreactivity was significantly ( P < 0.001) higher in hemodialysis patients (1.66 ± 1.13 nmol/l) compared with subjects with normal kidney function (0.46 ± 0.23). In hemodialysis patients, plasma marinobufagenin immunoreactivity was higher in men compared with women. A significant positive correlation has been found between plasma marinobufagenin immunoreactivity and serum NT-proBNP, NT-proANP, or aldosterone concentrations in all analyzed subjects. In hemodialyzed patients with plasma marinobufagenin immunoreactivity above median value 5-yr, all-cause mortality was higher compared with those with plasma marinobufagenin concentration below median. We have shown that plasma marinobufagenin immunoreactivity is increased in patients with end-stage kidney failure treated with hemodialysis parallel to the increase in serum NT-proBNP, NT-proANP, and aldosterone concentrations. Higher marinobufagenin immunoreactivity has been associated with worse survival in hemodialyzed patients.
Collapse
Affiliation(s)
- Grzegorz Piecha
- Department of Nephrology, Transplantation and Internal Medicine, Medical University of Silesia , Katowice , Poland
| | - Agata Kujawa-Szewieczek
- Department of Nephrology, Transplantation and Internal Medicine, Medical University of Silesia , Katowice , Poland
| | - Piotr Kuczera
- Department of Nephrology, Transplantation and Internal Medicine, Medical University of Silesia , Katowice , Poland
| | - Katarzyna Skiba
- Department of Nephrology, Transplantation and Internal Medicine, Medical University of Silesia , Katowice , Poland
| | - Ewelina Sikora-Grabka
- Department of Nephrology, Transplantation and Internal Medicine, Medical University of Silesia , Katowice , Poland
| | - Andrzej Więcek
- Department of Nephrology, Transplantation and Internal Medicine, Medical University of Silesia , Katowice , Poland
| |
Collapse
|
26
|
Dexmedetomidine attenuates renal fibrosis via α2-adrenergic receptor-dependent inhibition of cellular senescence after renal ischemia/reperfusion. Life Sci 2018; 207:1-8. [PMID: 29729264 DOI: 10.1016/j.lfs.2018.05.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 04/23/2018] [Accepted: 05/02/2018] [Indexed: 12/28/2022]
Abstract
BACKGROUND Renal ischemia/reperfusion (IR) can induce acute kidney injury (AKI), which often progresses to chronic kidney disease (CKD). Dexmedetomidine (Dex), a highly selective α2 adrenergic receptor (α2-AR) agonist, protects against acute renal IR-induced injury. However, the effects of Dex on the transition of AKI to CKD remain unclear. Therefore, we investigated the mechanisms of Dex on renal fibrosis. METHODS Adult male C57BL/6 mice were pretreated with Dex, a specific α2A-adrenergic receptor (AR) blocker (BRL-44408), or a cell senescence inhibitor (rapamycin) in a surgical bilateral renal IR model. The diagnoses of AKI and chronic renal fibrosis were performed by histopathological staining and western blotting. Histopathological changes, cell senescence, tubular fibrotic markers, and the expression of inflammatory factors were studied. RESULTS Pretreatment with Dex alleviated renal IR-induced AKI and chronic tubulointerstitial fibrosis in later stages. Similar to the effects of rapamycin, pretreatment with Dex also decreased the number of senescent tubular cells and weakened the protein expression of senescence-associated markers such as p53, p21, and p16. Furthermore, the expression of inflammatory markers was also decreased in Dex-treated IR mice; and these protective effects of Dex could be abolished by treatment with the specific α2A-AR blocker, BRL-44408. CONCLUSIONS The administration of a single dose of Dex protects against AKI and CKD. Dex inhibits tubular cell senescence and inflammation as well as improves renal fibrosis to moderate the AKI-to-CKD transition. The renal protective potential of Dex may provide a novel treatment strategy for high-risk renal injury patients.
Collapse
|
27
|
Reifsnyder PC, Ryzhov S, Flurkey K, Anunciado-Koza RP, Mills I, Harrison DE, Koza RA. Cardioprotective effects of dietary rapamycin on adult female C57BLKS/J-Lepr db mice. Ann N Y Acad Sci 2018; 1418:106-117. [PMID: 29377150 PMCID: PMC5934313 DOI: 10.1111/nyas.13557] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 10/24/2017] [Accepted: 10/30/2017] [Indexed: 02/06/2023]
Abstract
Rapamycin (RAPA), an inhibitor of mTORC signaling, has been shown to extend life span in mice and other organisms. Recently, animal and human studies have suggested that inhibition of mTORC signaling can alleviate or prevent the development of cardiomyopathy. In view of this, we used a murine model of type 2 diabetes (T2D), BKS-Leprdb , to determine whether RAPA treatment can mitigate the development of T2D-induced cardiomyopathy in adult mice. Female BKS-Leprdb mice fed diet supplemented with RAPA from 11 to 27 weeks of age showed reduced weight gain and significant reductions of fat and lean mass compared with untreated mice. No differences in plasma glucose or insulin levels were observed between groups; however, RAPA-treated mice were more insulin sensitive (P < 0.01) than untreated mice. Urine albumin/creatinine ratio was lower in RAPA-treated mice, suggesting reduced diabetic nephropathy and improved kidney function. Echocardiography showed significantly reduced left ventricular wall thickness in mice treated with RAPA compared with untreated mice (P = 0.02) that was consistent with reduced heart weight/tibia length ratios, reduced myocyte size and cardiac fibrosis measured by histomorphology, and reduced mRNA expression of Col1a1, a marker for cardiomyopathy. Our results suggest that inhibition of mTORC signaling is a plausible strategy for ameliorating complications of obesity and T2D, including cardiomyopathy.
Collapse
Affiliation(s)
| | - Sergey Ryzhov
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine
| | | | - Rea P Anunciado-Koza
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine
| | - Ian Mills
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine
| | | | - Robert A Koza
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine
| |
Collapse
|
28
|
Ma L, Ma X, Kong F, Guo J, Shi H, Zhu J, Zou Y, Ge J. Mammalian target of rapamycin inhibition attenuates myocardial ischaemia-reperfusion injury in hypertrophic heart. J Cell Mol Med 2018; 22:1708-1719. [PMID: 29314656 PMCID: PMC5824378 DOI: 10.1111/jcmm.13451] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 10/06/2017] [Indexed: 01/08/2023] Open
Abstract
Pathological cardiac hypertrophy aggravated myocardial infarction and is causally related to autophagy dysfunction and increased oxidative stress. Rapamycin is an inhibitor of serine/threonine kinase mammalian target of rapamycin (mTOR) involved in the regulation of autophagy as well as oxidative/nitrative stress. Here, we demonstrated that rapamycin ameliorates myocardial ischaemia reperfusion injury by rescuing the defective cytoprotective mechanisms in hypertrophic heart. Our results showed that chronic rapamycin treatment markedly reduced the phosphorylated mTOR and ribosomal protein S6 expression, but not Akt in both normal and aortic-banded mice. Moreover, chronic rapamycin treatment significantly mitigated TAC-induced autophagy dysfunction demonstrated by prompted Beclin-1 activation, elevated LC3-II/LC3-I ratio and increased autophagosome abundance. Most importantly, we found that MI/R-induced myocardial injury was markedly reduced by rapamycin treatment manifested by the inhibition of myocardial apoptosis, the reduction of myocardial infarct size and the improvement of cardiac function in hypertrophic heart. Mechanically, rapamycin reduced the MI/R-induced iNOS/gp91phox protein expression and decreased the generation of NO and superoxide, as well as the cytotoxic peroxynitrite. Moreover, rapamycin significantly mitigated MI/R-induced endoplasmic reticulum stress and mitochondrial impairment demonstrated by reduced Caspase-12 activity, inhibited CHOP activation, decreased cytoplasmic Cyto-C release and preserved intact mitochondria. In addition, inhibition of mTOR also enhanced the phosphorylated ERK and eNOS, and inactivated GSK3β, a pivotal downstream target of Akt and ERK signallings. Taken together, these results suggest that mTOR signalling protects against MI/R injury through autophagy induction and ERK-mediated antioxidative and anti-nitrative stress in mice with hypertrophic myocardium.
Collapse
Affiliation(s)
- Lei‐Lei Ma
- Shanghai Institute of Cardiovascular DiseasesZhongshan Hospital and Institute of Biomedical ScienceFudan UniversityShanghaiChina
- Department of Critical Care MedicineZhejiang Provincial People's Hospital and People's Hospital of Hangzhou Medical CollegeHangzhouChina
| | - Xin Ma
- Shanghai Institute of Cardiovascular DiseasesZhongshan Hospital and Institute of Biomedical ScienceFudan UniversityShanghaiChina
| | - Fei‐Juan Kong
- Department of Endocrinology and MetabolismShanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
| | - Jun‐Jie Guo
- Department of CardiologyAffiliated Hospital of Qingdao UniversityQingdaoChina
| | - Hong‐Tao Shi
- Shanghai Institute of Cardiovascular DiseasesZhongshan Hospital and Institute of Biomedical ScienceFudan UniversityShanghaiChina
| | - Jian‐Bing Zhu
- Shanghai Institute of Cardiovascular DiseasesZhongshan Hospital and Institute of Biomedical ScienceFudan UniversityShanghaiChina
| | - Yun‐Zeng Zou
- Shanghai Institute of Cardiovascular DiseasesZhongshan Hospital and Institute of Biomedical ScienceFudan UniversityShanghaiChina
| | - Jun‐Bo Ge
- Shanghai Institute of Cardiovascular DiseasesZhongshan Hospital and Institute of Biomedical ScienceFudan UniversityShanghaiChina
| |
Collapse
|
29
|
Hypertrophied myocardium is vulnerable to ischemia/reperfusion injury and refractory to rapamycin-induced protection due to increased oxidative/nitrative stress. Clin Sci (Lond) 2018; 132:93-110. [PMID: 29175946 DOI: 10.1042/cs20171471] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 11/22/2017] [Accepted: 11/24/2017] [Indexed: 11/17/2022]
Abstract
Left ventricular hypertrophy (LVH) is causally related to increased morbidity and mortality following acute myocardial infarction (AMI) via still unknown mechanisms. Although rapamycin exerts cardioprotective effects against myocardial ischemia/reperfusion (MI/R) injury in normal animals, whether rapamycin-elicited cardioprotection is altered in the presence of LVH has yet to be determined. Pressure overload induced cardiac hypertrophied mice and sham-operated controls were exposed to AMI by coronary artery ligation, and treated with vehicle or rapamycin 10 min before reperfusion. Rapamycin produced marked cardioprotection in normal control mice, whereas pressure overload induced cardiac hypertrophied mice manifested enhanced myocardial injury, and was refractory to rapamycin-elicited cardioprotection evidenced by augmented infarct size, aggravated cardiomyocyte apoptosis, and worsening cardiac function. Rapamycin alleviated MI/R injury via ERK-dependent antioxidative pathways in normal mice, whereas cardiac hypertrophied mice manifested markedly exacerbated oxidative/nitrative stress after MI/R evidenced by the increased iNOS/gp91phox expression, superoxide production, total NO metabolites, and nitrotyrosine content. Moreover, scavenging superoxide or peroxynitrite by selective gp91phox assembly inhibitor gp91ds-tat or ONOO- scavenger EUK134 markedly ameliorated MI/R injury, as shown by reduced myocardial oxidative/nitrative stress, alleviated myocardial infarction, hindered cardiomyocyte apoptosis, and improved cardiac function in aortic-banded mice. However, no additional cardioprotective effects were achieved when we combined rapamycin and gp91ds-tat or EUK134 in ischemic/reperfused hearts with or without LVH. These results suggest that cardiac hypertrophy attenuated rapamycin-induced cardioprotection by increasing oxidative/nitrative stress and scavenging superoxide/peroxynitrite protects the hypertrophied heart from MI/R.
Collapse
|
30
|
Melilli E, Manonelles A, Montero N, Grinyo J, Martinez-Castelao A, Bestard O, Cruzado J. Impact of immunosuppressive therapy on arterial stiffness in kidney transplantation: are all treatments the same? Clin Kidney J 2017; 11:413-421. [PMID: 29988241 PMCID: PMC6007381 DOI: 10.1093/ckj/sfx120] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 09/06/2017] [Indexed: 02/07/2023] Open
Abstract
Arterial stiffness is a biologic process related to ageing and its relationship with cardiovascular risk is well established. Several methods are currently available for non-invasive measurement of arterial stiffness that provide valuable information to further assess patients’ vascular status in real time. In kidney transplantation recipients, several factors could accelerate the stiffness process, such as the use of calcineurin inhibitors (CNIs), the presence of chronic kidney disease and other classical cardiovascular factors, which would explain, at least in part, the high cardiovascular mortality and morbidity. Despite the importance of arterial stiffness as a biomarker of cardiovascular risk, and unlike other cardiovascular risk factors (e.g. left ventricular hypertrophy), only a few clinical trials or retrospective studies of kidney recipients have evaluated its impact. In this review we describe the clinical impact of arterial stiffness as a prognostic marker of cardiovascular disease and the effects of different immunosuppressive regimens on its progression, focusing on the potential benefits of CNI-sparing protocols and supporting the rationale for individualization of immunosuppression in patients with lower arterial elasticity. Among the immunosuppressive drugs, a belatacept-based regimen seems to offer better vascular protection compared with CNIs, although further studies are needed to confirm the preliminary positive results.
Collapse
Affiliation(s)
- Edoardo Melilli
- Department of Nephrology, Bellvitge University Hospital, L’Hospitalet de Llobregat, Cataluny, Spain
- Correspondence and offprint requests to: Edoardo Melilli; E-mail:
| | - Anna Manonelles
- Department of Nephrology, Bellvitge University Hospital, L’Hospitalet de Llobregat, Cataluny, Spain
| | - Nuria Montero
- Department of Nephrology, Bellvitge University Hospital, L’Hospitalet de Llobregat, Cataluny, Spain
| | - Josep Grinyo
- Department of Nephrology, Bellvitge University Hospital, L’Hospitalet de Llobregat, Cataluny, Spain
| | | | - Oriol Bestard
- Department of Nephrology, Bellvitge University Hospital, L’Hospitalet de Llobregat, Cataluny, Spain
| | - Josep Cruzado
- Department of Nephrology, Bellvitge University Hospital, L’Hospitalet de Llobregat, Cataluny, Spain
| |
Collapse
|
31
|
Lombardi A, Gambardella J, Du XL, Sorriento D, Mauro M, Iaccarino G, Trimarco B, Santulli G. Sirolimus induces depletion of intracellular calcium stores and mitochondrial dysfunction in pancreatic beta cells. Sci Rep 2017; 7:15823. [PMID: 29158477 PMCID: PMC5696524 DOI: 10.1038/s41598-017-15283-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 10/20/2017] [Indexed: 12/19/2022] Open
Abstract
Sirolimus (rapamycin) is an immunosuppressive drug used in transplantation. One of its major side effects is the increased risk of diabetes mellitus; however, the exact mechanisms underlying such association have not been elucidated. Here we show that sirolimus impairs glucose-stimulated insulin secretion both in human and murine pancreatic islets and in clonal β cells in a dose- and time-dependent manner. Importantly, we demonstrate that sirolimus markedly depletes calcium (Ca2+) content in the endoplasmic reticulum and significantly decreases glucose-stimulated mitochondrial Ca2+ uptake. Crucially, the reduced mitochondrial Ca2+ uptake is mirrored by a significant impairment in mitochondrial respiration. Taken together, our findings indicate that sirolimus causes depletion of intracellular Ca2+ stores and alters mitochondrial fitness, eventually leading to decreased insulin release. Our results provide a novel molecular mechanism underlying the increased incidence of diabetes mellitus in patients treated with this drug.
Collapse
Affiliation(s)
- Angela Lombardi
- Department of Medicine, Albert Einstein College of Medicine, New York, NY, USA
| | - Jessica Gambardella
- Department of Advanced Biomedical Sciences, "Federico II" University of Naples, Naples, Italy
- Department of Medicine, Surgery and Dentistry, "Scuola Medica Salernitana", University of Salerno, Salerno, Italy
| | - Xue-Liang Du
- Department of Medicine, Albert Einstein College of Medicine, New York, NY, USA
| | - Daniela Sorriento
- Department of Advanced Biomedical Sciences, "Federico II" University of Naples, Naples, Italy
| | - Maurizio Mauro
- Department of Medicine, Albert Einstein College of Medicine, New York, NY, USA
| | - Guido Iaccarino
- Department of Medicine, Surgery and Dentistry, "Scuola Medica Salernitana", University of Salerno, Salerno, Italy
| | - Bruno Trimarco
- Department of Advanced Biomedical Sciences, "Federico II" University of Naples, Naples, Italy
| | - Gaetano Santulli
- Department of Medicine, Albert Einstein College of Medicine, New York, NY, USA.
- Department of Advanced Biomedical Sciences, "Federico II" University of Naples, Naples, Italy.
| |
Collapse
|
32
|
Dmitrieva RI, Cranford SM, Doris PA. Genetic Control of Serum Marinobufagenin in the Spontaneously Hypertensive Rat and the Relationship to Blood Pressure. J Am Heart Assoc 2017; 6:e006704. [PMID: 28982675 PMCID: PMC5721872 DOI: 10.1161/jaha.117.006704] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 08/23/2017] [Indexed: 11/16/2022]
Abstract
BACKGROUND We have investigated serum levels of immunoreactive marinobufagenin (MBG) in 16- to 20-week-old spontaneously hypertensive rats (SHRs)-A3 and in the normotensive Wistar-Kyoto (WKY) rat strain in the absence of salt loading, and we have investigated the genetic control of serum MBG. METHODS AND RESULTS We genotyped the F2 progeny of an SHR-A3×WKY intercross using a genome-wide panel of 253 single-nucleotide polymorphism markers that were dimorphic between SHR-A3 and WKY and measured serum MBG by ELISA. Serum MBG levels were lower in SHR-A3 than WKY rats (0.39±0.07 and 1.27±0.40 nmol/L, respectively), suggesting that MBG may not play a role in the markedly divergent blood pressure measured by telemetry in rats of these 2 strains (SHR-A3 and WKY, 198.3±4.43 and 116.8±1.51 mm Hg, respectively). The strain difference in serum MBG was investigated to determine whether genomic regions influencing MBG might be identified by genetic mapping. Quantitative trait locus mapping indicated a single locus influencing serum MBG in the region of chromosome 6q12. Homozygosity of WKY alleles at this locus was associated with increased serum MBG levels. We surveyed whole genome sequences from our SHR-A3 and WKY lines, seeking coding sequence variation between SHR-A3 and WKY within the mapped locus that might explain the inherited strain difference in serum MBG. CONCLUSIONS We identified amino acid substitution in the sterol transport protein Abcg5, present in SHR-A3, but absent in WKY, that is a potential mechanism influencing MBG levels.
Collapse
Affiliation(s)
- Renata I Dmitrieva
- Institute of Molecular Medicine, University of Texas Health Science Center at Houston, TX
| | - Stacy M Cranford
- Institute of Molecular Medicine, University of Texas Health Science Center at Houston, TX
| | - Peter A Doris
- Institute of Molecular Medicine, University of Texas Health Science Center at Houston, TX
| |
Collapse
|
33
|
Wang X, Liu J, Drummond CA, Shapiro JI. Sodium potassium adenosine triphosphatase (Na/K-ATPase) as a therapeutic target for uremic cardiomyopathy. Expert Opin Ther Targets 2017; 21:531-541. [PMID: 28338377 PMCID: PMC5590225 DOI: 10.1080/14728222.2017.1311864] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 03/23/2017] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Clinically, patients with significant reductions in renal function present with cardiovascular dysfunction typically termed, uremic cardiomyopathy. It is a progressive series of cardiac pathophysiological changes, including left ventricular diastolic dysfunction and hypertrophy (LVH) which sometimes progress to left ventricular dilation (LVD) and systolic dysfunction in the setting of chronic kidney disease (CKD). Uremic cardiomyopathy is almost ubiquitous in patients afflicted with end stage renal disease (ESRD). Areas covered: This article reviews recent epidemiology, pathophysiology of uremic cardiomyopathy and provide a board overview of Na/K-ATPase research with detailed discussion on the mechanisms of Na/K-ATPase/Src/ROS amplification loop. We also present clinical and preclinical evidences as well as molecular mechanism of this amplification loop in the development of uremic cardiomyopathy. A potential therapeutic peptide that targets on this loop is discussed. Expert opinion: Current clinical treatment for uremic cardiomyopathy remains disappointing. Targeting the ROS amplification loop mediated by the Na/K-ATPase signaling function may provide a novel therapeutic target for uremic cardiomyopathy and related diseases. Additional studies of Na/K-ATPase and other strategies that regulate this loop will lead to new therapeutics.
Collapse
Affiliation(s)
- Xiaoliang Wang
- a Joan C Edwards School of Medicine at Marshall University , Huntington , WV , United States
- b University of Toledo College of Medicine and Life Sciences , Toledo , OH , United States
| | - Jiang Liu
- a Joan C Edwards School of Medicine at Marshall University , Huntington , WV , United States
| | - Christopher A Drummond
- b University of Toledo College of Medicine and Life Sciences , Toledo , OH , United States
| | - Joseph I Shapiro
- a Joan C Edwards School of Medicine at Marshall University , Huntington , WV , United States
| |
Collapse
|
34
|
Fan X, Xie J, Tian J. Reducing Cardiac Fibrosis: Na/K-ATPase Signaling Complex as a Novel Target. ACTA ACUST UNITED AC 2017; 6. [PMID: 29034264 DOI: 10.4172/2329-6607.1000204] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cardiac fibrosis is a common pathological process in cardiac disease and may lead to heart failure. It can also cause sudden death even in those without cardiac symptoms. Tissue fibrosis can be categorized into two categories: replacement fibrosis (also called reparative fibrosis) and reactive fibrosis. In replacement fibrosis, infiltration of inflammatory cells and accumulation of Extracellular Matrix (ECM) proteins are the initial steps in forming scarlike fibrotic tissue after acute cardiac injury and cardiac cell necrosis. Reactive fibrosis can be formed in response to hormonal change and pressure or volume overload. Experimental studies in animals have identified important pathways such as the Renin-Angiotensin-Aldosterone System (RAAS) and the endothelin pathway that contribute to fibrosis formation. Despite the fact that clinical trials using RAAS inhibitors as therapies for reducing cardiac fibrosis and improving cardiac function have been promising, heart failure is still the leading cause of deaths in the United States. Intensive efforts have been made to find novel targets and to develop new treatments for cardiac fibrosis and heart failure in the past few decades. The Na/K-ATPase, a canonical ion transporter, has been shown to also function as a signal transducer and prolonged activation of Na/K-ATPase signaling has been found to promote the formation of cardiac fibrosis. Novel tools that block the activation of Na/K-ATPase signaling have been developed and have shown promise in reducing cardiac fibrosis. This review will discuss the recent development of novel molecular targets, focusing on the Na/K-ATPase signaling complex as a therapeutic target in treatment of cardiac fibrosis.
Collapse
Affiliation(s)
- X Fan
- Department of Medicine, Center for Hypertension and Personalized Medicine, University of Toledo, Ohio 43614, USA
| | - J Xie
- Department of Medicine, Center for Hypertension and Personalized Medicine, University of Toledo, Ohio 43614, USA
| | - J Tian
- Department of Medicine, Center for Hypertension and Personalized Medicine, University of Toledo, Ohio 43614, USA
| |
Collapse
|