1
|
Wang Y, Jia H, Gao K, Du MF, Chu C, Wang D, Ma Q, Hu GL, Zhang X, Sun Y, Man ZY, Mu JJ. Renalase alleviates salt-induced kidney necroptosis and inflammation. Hypertens Res 2024; 47:2811-2825. [PMID: 39117946 DOI: 10.1038/s41440-024-01814-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 06/06/2024] [Accepted: 07/03/2024] [Indexed: 08/10/2024]
Abstract
Recent evidence suggests that necroptosis may contribute to the development of kidney injury. Renalase is a novel secretory protein that exerts potent prosurvival and anti-inflammatory effects. We hypothesized that renalase could protect the kidney from salt-induced injury by modulating necroptosis. High salt and renalase treatments were administered to Dahl salt-sensitive (SS) rats, renalase knockout (KO) mice, and HK-2 cells. Furthermore, a cohort of 514 eligible participants was utilized to investigate the association between single nucleotide polymorphisms (SNPs) in the genes RIPK1, RIPK3, and MLKL, and the risk of subclinical renal damage (SRD) over 14 years. A high-salt diet significantly increased the expression of key components of necroptosis, namely RIPK1, RIPK3, and MLKL, as well as the release of inflammatory factors in SS rats. Treatment with recombinant renalase reduced both necroptosis and inflammation. In renalase KO mice, salt-induced kidney injury was more severe than in wild-type mice, but supplementation with renalase attenuated the kidney injury. In vitro experiments with HK-2 cells revealed high salt increased necroptosis and inflammation. Renalase exhibited a dose-dependent decrease in salt-induced necroptosis, and this cytoprotective effect was negated by the knockdown of PMCA4b, which is the receptor of renalase. Furthermore, the cohort study showed that SNP rs3736724 in RIPK1 and rs11640974 in MLKL were significantly associated with the risk of SRD over 14 years. Our analysis shows that necroptosis plays a significant role in the development of salt-induced kidney injury and that renalase confers its cytoprotective effects by inhibiting necroptosis and inflammation.
Collapse
Affiliation(s)
- Yang Wang
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Key Laboratory of Molecular Cardiology of Shaanxi Province, Xi'an, China
| | - Hao Jia
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Key Laboratory of Molecular Cardiology of Shaanxi Province, Xi'an, China
| | - Ke Gao
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ming-Fei Du
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Key Laboratory of Molecular Cardiology of Shaanxi Province, Xi'an, China
| | - Chao Chu
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Key Laboratory of Molecular Cardiology of Shaanxi Province, Xi'an, China
| | - Dan Wang
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Key Laboratory of Molecular Cardiology of Shaanxi Province, Xi'an, China
| | - Qiong Ma
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Gui-Lin Hu
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Key Laboratory of Molecular Cardiology of Shaanxi Province, Xi'an, China
| | - Xi Zhang
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Key Laboratory of Molecular Cardiology of Shaanxi Province, Xi'an, China
| | - Yue Sun
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Key Laboratory of Molecular Cardiology of Shaanxi Province, Xi'an, China
| | - Zi-Yue Man
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jian-Jun Mu
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
- Key Laboratory of Molecular Cardiology of Shaanxi Province, Xi'an, China.
| |
Collapse
|
2
|
Xiao C, Wu X, Gallagher CS, Rasooly D, Jiang X, Morton CC. Genetic contribution of reproductive traits to risk of uterine leiomyomata: a large-scale, genome-wide, cross-trait analysis. Am J Obstet Gynecol 2024; 230:438.e1-438.e15. [PMID: 38191017 DOI: 10.1016/j.ajog.2023.12.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 12/03/2023] [Accepted: 12/26/2023] [Indexed: 01/10/2024]
Abstract
BACKGROUND Although phenotypic associations between female reproductive characteristics and uterine leiomyomata have long been observed in epidemiologic investigations, the shared genetic architecture underlying these complex phenotypes remains unclear. OBJECTIVE We aimed to investigate the shared genetic basis, pleiotropic effects, and potential causal relationships underlying reproductive traits (age at menarche, age at natural menopause, and age at first birth) and uterine leiomyomata. STUDY DESIGN With the use of large-scale, genome-wide association studies conducted among women of European ancestry for age at menarche (n=329,345), age at natural menopause (n=201,323), age at first birth (n=418,758), and uterine leiomyomata (ncases/ncontrols=35,474/267,505), we performed a comprehensive, genome-wide, cross-trait analysis to examine systematically the common genetic influences between reproductive traits and uterine leiomyomata. RESULTS Significant global genetic correlations were identified between uterine leiomyomata and age at menarche (rg, -0.17; P=3.65×10-10), age at natural menopause (rg, 0.23; P=3.26×10-07), and age at first birth (rg, -0.16; P=1.96×10-06). Thirteen genomic regions were further revealed as contributing significant local correlations (P<.05/2353) to age at natural menopause and uterine leiomyomata. A cross-trait meta-analysis identified 23 shared loci, 3 of which were novel. A transcriptome-wide association study found 15 shared genes that target tissues of the digestive, exo- or endocrine, nervous, and cardiovascular systems. Mendelian randomization suggested causal relationships between a genetically predicted older age at menarche (odds ratio, 0.88; 95% confidence interval, 0.85-0.92; P=1.50×10-10) or older age at first birth (odds ratio, 0.95; 95% confidence interval, 0.90-0.99; P=.02) and a reduced risk for uterine leiomyomata and between a genetically predicted older age at natural menopause and an increased risk for uterine leiomyomata (odds ratio, 1.08; 95% confidence interval, 1.06-1.09; P=2.30×10-27). No causal association in the reverse direction was found. CONCLUSION Our work highlights that there are substantial shared genetic influences and putative causal links that underlie reproductive traits and uterine leiomyomata. The findings suggest that early identification of female reproductive risk factors may facilitate the initiation of strategies to modify potential uterine leiomyomata risk.
Collapse
Affiliation(s)
- Changfeng Xiao
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xueyao Wu
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | | | - Danielle Rasooly
- Division of Aging, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Xia Jiang
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China; Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Solna, Stockholm, Sweden.
| | - Cynthia Casson Morton
- Department of Obstetrics and Gynecology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Broad Institute of MIT and Harvard, Cambridge, MA; Manchester Centre for Audiology and Deafness, Manchester Academic Health Science Center, University of Manchester, Manchester, United Kingdom.
| |
Collapse
|
3
|
Zhang L, Zang CS, Chen B, Wang Y, Xue S, Wu MY. Renalase regulates renal tubular injury in diabetic nephropathy via the p38MAPK signaling pathway. FASEB J 2023; 37:e23188. [PMID: 37732586 DOI: 10.1096/fj.202300708r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 07/24/2023] [Accepted: 08/25/2023] [Indexed: 09/22/2023]
Abstract
Diabetic nephropathy (DN) is an important complication of diabetes and the leading cause of end-stage renal disease globally. Renal tubular damage occurs to varying degrees in the early stages of DN prior to glomerular damage. Renalase (RNLS) is an amine oxidase, which is produced and secreted by the renal tubular epithelial cells. RNLS is reportedly closely related to renal tubular injury in acute and chronic kidney diseases. Herein, we aimed to evaluate the changes in tubular RNLS expression in DN and its correlation with DN-associated renal tubular injury. Conditional permanent renal tubular epithelial rat-cell line NRK-52E was transfected with pcDNA3-RNLS plasmid or administered recombinant rat RNLS protein and high glucose (HG) dose. A total of 22 adult Sprague-Dawley rats were randomly divided into the control (CON, n = 10) or diabetic nephrology (DN, n = 12) group. Random blood glucose levels of the rats were measured by sampling of the caudal vein weekly. After 8 weeks, the rat's body weight, 24-h urinary albumin concentration, and right kidney were evaluated. Our study suggested the decreased expression levels of RNLS in renal tissue and renal tubular epithelial cells in DN rats, accompanied by renal tubulointerstitial fibrosis, apoptosis of renal tubular epithelial cells, and activation of the p38MAPK signal pathway. Reversing the low RNLS expression can reduce the level of p38MAPK phosphorylation and delay renal tubular injury. Thus, the reduction of renal tubular RNLS expression in DN mediates tubulointerstitial fibrosis and cell apoptosis via the activation of the p38MAPK signal pathway. RNLS plays a key mediating role in DN-associated tubular injury via p38MAPK, which provides new therapeutic targets and a theoretical basis for early prevention and treatment of DN.
Collapse
Affiliation(s)
- Li Zhang
- Department of Thyroid Surgery, General surgery center, The First Hospital of Jilin University, Changchun, China
| | - Chong-Sen Zang
- Department of Thyroid Surgery, General surgery center, The First Hospital of Jilin University, Changchun, China
| | - Bin Chen
- Department of Thyroid Surgery, General surgery center, The First Hospital of Jilin University, Changchun, China
| | - Yu Wang
- Department of Thyroid Surgery, General surgery center, The First Hospital of Jilin University, Changchun, China
| | - Shuai Xue
- Department of Nephrology, The First Hospital of Jilin University, Changchun, China
| | - Mei-Yan Wu
- Department of Thyroid Surgery, General surgery center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
4
|
Stojanovic D, Stojanovic M, Milenkovic J, Velickov A, Ignjatovic A, Milojkovic M. The Multi-Faceted Nature of Renalase for Mitochondrial Dysfunction Improvement in Cardiac Disease. Cells 2023; 12:1607. [PMID: 37371077 DOI: 10.3390/cells12121607] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 05/24/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
The cellular mechanisms and signaling network that guide the cardiac disease pathophysiology are inextricably intertwined, which explains the current scarcity of effective therapy and to date remains the greatest challenge in state-of-the-art cardiovascular medicine. Accordingly, a novel concept has emerged in which cardiomyocytes are the centerpiece of therapeutic targeting, with dysregulated mitochondria as a critical point of intervention. Mitochondrial dysfunction pluralism seeks a multi-faceted molecule, such as renalase, to simultaneously combat the pathophysiologic heterogeneity of mitochondria-induced cardiomyocyte injury. This review provides some original perspectives and, for the first time, discusses the functionality spectrum of renalase for mitochondrial dysfunction improvement within cardiac disease, including its ability to preserve mitochondrial integrity and dynamics by suppressing mitochondrial ΔΨm collapse; overall ATP content amelioration; a rise of mtDNA copy numbers; upregulation of mitochondrial genes involved in oxidative phosphorylation and cellular vitality promotion; mitochondrial fission inhibition; NAD+ supplementation; sirtuin upregulation; and anti-oxidant, anti-apoptotic, and anti-inflammatory traits. If verified that renalase, due to its multi-faceted nature, behaves like the "guardian of mitochondria" by thwarting pernicious mitochondrial dysfunction effects and exerting therapeutic potential to target mitochondrial abnormalities in failing hearts, it may provide large-scale benefits for cardiac disease patients, regardless of the underlying causes.
Collapse
Affiliation(s)
- Dijana Stojanovic
- Department of Pathophysiology, Faculty of Medicine, University of Nis, 18000 Nis, Serbia
| | - Miodrag Stojanovic
- Department of Medical Statistics and Informatics, Faculty of Medicine, University of Nis, 18000 Nis, Serbia
- Center of Informatics and Biostatistics in Healthcare, Institute for Public Health, 18000 Nis, Serbia
| | - Jelena Milenkovic
- Department of Pathophysiology, Faculty of Medicine, University of Nis, 18000 Nis, Serbia
| | - Aleksandra Velickov
- Department of Histology and Embryology, Faculty of Medicine, University of Nis, 18000 Nis, Serbia
| | - Aleksandra Ignjatovic
- Department of Medical Statistics and Informatics, Faculty of Medicine, University of Nis, 18000 Nis, Serbia
- Center of Informatics and Biostatistics in Healthcare, Institute for Public Health, 18000 Nis, Serbia
| | - Maja Milojkovic
- Department of Pathophysiology, Faculty of Medicine, University of Nis, 18000 Nis, Serbia
| |
Collapse
|
5
|
Wang Y, Bai L, Wen J, Zhang F, Gu S, Wang F, Yin J, Wang N. Cardiac-specific renalase overexpression alleviates CKD-induced pathological cardiac remodeling in mice. Front Cardiovasc Med 2022; 9:1061146. [PMID: 36588579 PMCID: PMC9798007 DOI: 10.3389/fcvm.2022.1061146] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 11/30/2022] [Indexed: 12/23/2022] Open
Abstract
Introduction CKD-induced pathological cardiac remodeling is characterized by myocardial hypertrophy and cardiac fibrosis. The available therapeutic options are limited, it is thus urgently needed to identify novel therapeutic targets. Renalase (RNLS) is a newly discovered protein secreted by the kidney and was found beneficial in many renal diseases. But whether it exerts protective effects on cardiac remodeling in CKD remains unclear. Methods RNLS knockout (KO) and wild-type (WT) mice were both used to build CKD models and the adeno-associated virus (AAV9) system was used to overexpress RNLS cardiac specifically. Echocardiography was performed to detect cardiac structural changes every 6 weeks until 18 weeks post-surgery. High throughput sequencing was performed to understand the underlying mechanisms and the effects of RNLS on cardiac fibroblasts were validated in vitro. Results Knockout of RNLS aggravated cardiac remodeling in CKD, while RNLS cardiac-specific overexpression significantly reduced left ventricular hypertrophy and cardiac fibrosis induced by CKD. The following RNA-sequencing analysis revealed that RNLS significantly downregulated the extracellular matrix (ECM) receptor interaction pathway, ECM organization, and several ECM-related proteins. GSEA results showed RNLS significantly downregulated several profibrotic biological processes of cardiac fibroblasts which were upregulated by CKD, including fibroblast proliferation, leukocyte migration, antigen presentation, cytokine production, and epithelial-mesenchymal transition (EMT). In vitro, we validated that RNLS reduced the primary cardiac fibroblast proliferation and α-SMA expression stimulated by TGF-β. Conclusion In this study, we examined the cardioprotective role of RNLS in CKD-induced cardiac remodeling. RNLS may be a potential therapeutic factor that exerts an anti-fibrotic effect in pathological cardiac remodeling.
Collapse
Affiliation(s)
- Yi Wang
- Department of Nephrology, Shanghai Sixth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Linnan Bai
- Department of Nephrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiejun Wen
- Department of Nephrology, Shanghai Sixth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fangfei Zhang
- Department of Nephrology, Shanghai Sixth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Sijie Gu
- Department of Nephrology, Shanghai Sixth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng Wang
- Department of Nephrology, Shanghai Sixth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianyong Yin
- Department of Nephrology, Shanghai Sixth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China,*Correspondence: Jianyong Yin,
| | - Niansong Wang
- Department of Nephrology, Shanghai Sixth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China,Niansong Wang,
| |
Collapse
|
6
|
Ameer OZ. Hypertension in chronic kidney disease: What lies behind the scene. Front Pharmacol 2022; 13:949260. [PMID: 36304157 PMCID: PMC9592701 DOI: 10.3389/fphar.2022.949260] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 09/26/2022] [Indexed: 12/04/2022] Open
Abstract
Hypertension is a frequent condition encountered during kidney disease development and a leading cause in its progression. Hallmark factors contributing to hypertension constitute a complexity of events that progress chronic kidney disease (CKD) into end-stage renal disease (ESRD). Multiple crosstalk mechanisms are involved in sustaining the inevitable high blood pressure (BP) state in CKD, and these play an important role in the pathogenesis of increased cardiovascular (CV) events associated with CKD. The present review discusses relevant contributory mechanisms underpinning the promotion of hypertension and their consequent eventuation to renal damage and CV disease. In particular, salt and volume expansion, sympathetic nervous system (SNS) hyperactivity, upregulated renin–angiotensin–aldosterone system (RAAS), oxidative stress, vascular remodeling, endothelial dysfunction, and a range of mediators and signaling molecules which are thought to play a role in this concert of events are emphasized. As the control of high BP via therapeutic interventions can represent the key strategy to not only reduce BP but also the CV burden in kidney disease, evidence for major strategic pathways that can alleviate the progression of hypertensive kidney disease are highlighted. This review provides a particular focus on the impact of RAAS antagonists, renal nerve denervation, baroreflex stimulation, and other modalities affecting BP in the context of CKD, to provide interesting perspectives on the management of hypertensive nephropathy and associated CV comorbidities.
Collapse
Affiliation(s)
- Omar Z. Ameer
- Department of Pharmaceutical Sciences, College of Pharmacy, Alfaisal University, Riyadh, Saudi Arabia
- Department of Biomedical Sciences, Faculty of Medicine, Macquarie University, Sydney, NSW, Australia
- *Correspondence: Omar Z. Ameer,
| |
Collapse
|
7
|
GÜLER Ö, TUĞAN YILDIZ B, HAKKOYMAZ H, AYDIN S, YARDIM M. Levels of Serum and Urine Catecholaminergic and Apelinergic System Members in Acute Ischemic Stroke Patients. KAHRAMANMARAŞ SÜTÇÜ İMAM ÜNIVERSITESI TIP FAKÜLTESI DERGISI 2022. [DOI: 10.17517/ksutfd.1168625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023] Open
Abstract
Objective: To compare levels of catecholaminergic system members, renalase, cerebellin, and their substrates, epinephrine, norepinephrine, and dopamine, and apelinergic system members, apelin, elabela, and nitric oxide in the blood and urine of patients with acute ischemic stroke and healthy controls.
Materials and Methods: 42 patients with acute ischemic stroke and 42 age and sex-matched healthy controls were included in the study. Blood and urine samples were collected simultaneously and within the first 24 hours after the onset of acute stroke clinical manifestations and were measured using an ELISA method.
Results: The levels of serum and urine cerebellin, renalase, epinephrine, norepinephrine, dopamine, apelin, elebela, and nitric oxide were similar in ischemic stroke and in control groups (P>0.05). Strong correlations were found between renalase, cerebellin, and catecholamine levels in serum and urine (p
Collapse
|
8
|
Renalase Challenges the Oxidative Stress and Fibroproliferative Response in COVID-19. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4032704. [PMID: 36132227 PMCID: PMC9484957 DOI: 10.1155/2022/4032704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 06/03/2022] [Accepted: 08/24/2022] [Indexed: 01/08/2023]
Abstract
The hallmark of the coronavirus disease 2019 (COVID-19) pathophysiology was reported to be an inappropriate and uncontrolled immune response, evidenced by activated macrophages, and a robust surge of proinflammatory cytokines, followed by the release of reactive oxygen species, that synergistically result in acute respiratory distress syndrome, fibroproliferative lung response, and possibly even death. For these reasons, all identified risk factors and pathophysiological processes of COVID-19, which are feasible for the prevention and treatment, should be addressed in a timely manner. Accordingly, the evolving anti-inflammatory and antifibrotic therapy for severe COVID-19 and hindering post-COVID-19 fibrosis development should be comprehensively investigated. Experimental evidence indicates that renalase, a novel amino-oxidase, derived from the kidneys, exhibits remarkable organ protection, robustly addressing the most powerful pathways of cell trauma: inflammation and oxidative stress, necrosis, and apoptosis. As demonstrated, systemic renalase administration also significantly alleviates experimentally induced organ fibrosis and prevents adverse remodeling. The recognition that renalase exerts cytoprotection via sirtuins activation, by raising their NAD+ levels, provides a “proof of principle” for renalase being a biologically impressive molecule that favors cell protection and survival and maybe involved in the pathogenesis of COVID-19. This premise supports the rationale that renalase's timely supplementation may prove valuable for pathologic conditions, such as cytokine storm and related acute respiratory distress syndrome. Therefore, the aim for this review is to acknowledge the scientific rationale for renalase employment in the experimental model of COVID-19, targeting the acute phase mechanisms and halting fibrosis progression, based on its proposed molecular pathways. Novel therapies for COVID-19 seek to exploit renalase's multiple and distinctive cytoprotective mechanisms; therefore, this review should be acknowledged as the thorough groundwork for subsequent research of renalase's employment in the experimental models of COVID-19.
Collapse
|
9
|
Renalase: a novel regulator of cardiometabolic and renal diseases. Hypertens Res 2022; 45:1582-1598. [PMID: 35941358 PMCID: PMC9358379 DOI: 10.1038/s41440-022-00986-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 04/26/2022] [Accepted: 06/05/2022] [Indexed: 11/13/2022]
Abstract
Renalase is a ~38 kDa flavin-adenine dinucleotide (FAD) domain-containing protein that can function as a cytokine and an anomerase. It is emerging as a novel regulator of cardiometabolic diseases. Expressed mainly in the kidneys, renalase has been reported to have a hypotensive effect and may control blood pressure through regulation of sympathetic tone. Furthermore, genetic variations in the renalase gene, such as a functional missense polymorphism (Glu37Asp), have implications in the cardiovascular and renal systems and can potentially increase the risk of cardiometabolic disorders. Research on the physiological functions and biochemical actions of renalase over the years has indicated a role for renalase as one of the key proteins involved in various disease states, such as diabetes, impaired lipid metabolism, and cancer. Recent studies have identified three transcription factors (viz., Sp1, STAT3, and ZBP89) as key positive regulators in modulating the expression of the human renalase gene. Moreover, renalase is under the post-transcriptional regulation of two microRNAs (viz., miR-29b, and miR-146a), which downregulate renalase expression. While renalase supplementation may be useful for treating hypertension, inhibition of renalase signaling may be beneficial to patients with cancerous tumors. However, more incisive investigations are required to unravel the potential therapeutic applications of renalase. Based on the literature pertaining to the function and physiology of renalase, this review attempts to consolidate and comprehend the role of renalase in regulating cardiometabolic and renal disorders. ![]()
Collapse
|
10
|
Renalase may be cleaved in blood. Are blood chymotrypsin-like enzymes involved? Med Hypotheses 2022. [DOI: 10.1016/j.mehy.2022.110895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
11
|
Stojanovic D, Mitic V, Stojanovic M, Milenkovic J, Ignjatovic A, Milojkovic M. The Scientific Rationale for the Introduction of Renalase in the Concept of Cardiac Fibrosis. Front Cardiovasc Med 2022; 9:845878. [PMID: 35711341 PMCID: PMC9193824 DOI: 10.3389/fcvm.2022.845878] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 04/25/2022] [Indexed: 12/17/2022] Open
Abstract
Cardiac fibrosis represents a redundant accumulation of extracellular matrix proteins, resulting from a cascade of pathophysiological events involved in an ineffective healing response, that eventually leads to heart failure. The pathophysiology of cardiac fibrosis involves various cellular effectors (neutrophils, macrophages, cardiomyocytes, fibroblasts), up-regulation of profibrotic mediators (cytokines, chemokines, and growth factors), and processes where epithelial and endothelial cells undergo mesenchymal transition. Activated fibroblasts and myofibroblasts are the central cellular effectors in cardiac fibrosis, serving as the main source of matrix proteins. The most effective anti-fibrotic strategy will have to incorporate the specific targeting of the diverse cells, pathways, and their cross-talk in the pathogenesis of cardiac fibroproliferation. Additionally, renalase, a novel protein secreted by the kidneys, is identified. Evidence demonstrates its cytoprotective properties, establishing it as a survival element in various organ injuries (heart, kidney, liver, intestines), and as a significant anti-fibrotic factor, owing to its, in vitro and in vivo demonstrated pleiotropy to alleviate inflammation, oxidative stress, apoptosis, necrosis, and fibrotic responses. Effective anti-fibrotic therapy may seek to exploit renalase’s compound effects such as: lessening of the inflammatory cell infiltrate (neutrophils and macrophages), and macrophage polarization (M1 to M2), a decrease in the proinflammatory cytokines/chemokines/reactive species/growth factor release (TNF-α, IL-6, MCP-1, MIP-2, ROS, TGF-β1), an increase in anti-apoptotic factors (Bcl2), and prevention of caspase activation, inflammasome silencing, sirtuins (1 and 3) activation, and mitochondrial protection, suppression of epithelial to mesenchymal transition, a decrease in the pro-fibrotic markers expression (’α-SMA, collagen I, and III, TIMP-1, and fibronectin), and interference with MAPKs signaling network, most likely as a coordinator of pro-fibrotic signals. This review provides the scientific rationale for renalase’s scrutiny regarding cardiac fibrosis, and there is great anticipation that these newly identified pathways are set to progress one step further. Although substantial progress has been made, indicating renalase’s therapeutic promise, more profound experimental work is required to resolve the accurate underlying mechanisms of renalase, concerning cardiac fibrosis, before any potential translation to clinical investigation.
Collapse
Affiliation(s)
- Dijana Stojanovic
- Institute of Pathophysiology, Faculty of Medicine, University of Niš, Niš, Serbia
| | - Valentina Mitic
- Department of Cardiovascular Rehabilitation, Institute for Treatment and Rehabilitation "Niska Banja", Niska Banja, Serbia
| | - Miodrag Stojanovic
- Department of Medical Statistics and Informatics, Faculty of Medicine, University of Niš, Niš, Serbia.,Center of Informatics and Biostatistics in Healthcare, Institute for Public Health, Niš, Serbia
| | - Jelena Milenkovic
- Institute of Pathophysiology, Faculty of Medicine, University of Niš, Niš, Serbia
| | - Aleksandra Ignjatovic
- Department of Medical Statistics and Informatics, Faculty of Medicine, University of Niš, Niš, Serbia.,Center of Informatics and Biostatistics in Healthcare, Institute for Public Health, Niš, Serbia
| | - Maja Milojkovic
- Institute of Pathophysiology, Faculty of Medicine, University of Niš, Niš, Serbia
| |
Collapse
|
12
|
Guo X, Jessel S, Qu R, Kluger Y, Chen TM, Hollander L, Safirstein R, Nelson B, Cha C, Bosenberg M, Jilaveanu LB, Rimm D, Rothlin CV, Kluger HM, Desir GV. Inhibition of renalase drives tumour rejection by promoting T cell activation. Eur J Cancer 2022; 165:81-96. [PMID: 35219026 PMCID: PMC8940682 DOI: 10.1016/j.ejca.2022.01.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 12/30/2021] [Accepted: 01/10/2022] [Indexed: 12/25/2022]
Abstract
BACKGROUND Although programmed cell death protein 1 (PD-1) inhibitors have revolutionised treatment for advanced melanoma, not all patients respond. We previously showed that inhibition of the flavoprotein renalase (RNLS) in preclinical melanoma models decreases tumour growth. We hypothesised that RNLS inhibition promotes tumour rejection by effects on the tumour microenvironment (TME). METHODS We used two distinct murine melanoma models, studied in RNLS knockout (KO) or wild-type (WT) mice. WT mice were treated with the anti-RNLS antibody, m28, with or without anti-PD-1. 10X single-cell RNA-sequencing was used to identify transcriptional differences between treatment groups, and tumour cell content was interrogated by flow cytometry. Samples from patients treated with immunotherapy were examined for RNLS expression by quantitative immunofluorescence. RESULTS RNLS KO mice injected with wild-type melanoma cells reject their tumours, supporting the importance of RNLS in cells in the TME. This effect was blunted by anti-cluster of differentiation 3. However, MØ-specific RNLS ablation was insufficient to abrogate tumour formation. Anti-RNLS antibody treatment of melanoma-bearing mice resulted in enhanced T cell infiltration and activation and resulted in immune memory on rechallenging mice with injection of melanoma cells. At the single-cell level, treatment with anti-RNLS antibodies resulted in increased tumour density of MØ, neutrophils and lymphocytes and increased expression of IFNγ and granzyme B in natural killer cells and T cells. Intratumoural Forkhead Box P3 + CD4 cells were decreased. In two distinct murine melanoma models, we showed that melanoma-bearing mice treated with anti-RNLS antibodies plus anti-PD-1 had superior tumour shrinkage and survival than with either treatment alone. Importantly, in pretreatment samples from patients treated with PD-1 inhibitors, high RNLS expression was associated with decreased survival (log-rank P = 0.006), independent of other prognostic variables. CONCLUSIONS RNLS KO results in melanoma tumour regression in a T-cell-dependent fashion. Anti-RNLS antibodies enhance anti-PD-1 activity in two distinct aggressive murine melanoma models resistant to PD-1 inhibitors, supporting the development of anti-RNLS antibodies with PD-1 inhibitors as a novel approach for melanomas poorly responsive to anti-PD-1.
Collapse
Affiliation(s)
- Xiaojia Guo
- Department of Medicine Section of Nephrology, Yale University, New Haven, CT, USA
| | - Shlomit Jessel
- Department of Medicine Section of Medical Oncology, Yale University, New Haven, CT, USA
| | - Rihao Qu
- Department of Medicine Pathology, Yale University, New Haven, CT, USA
| | - Yuval Kluger
- Department of Medicine Pathology, Yale University, New Haven, CT, USA
| | - Tian-Min Chen
- Department of Medicine Section of Nephrology, Yale University, New Haven, CT, USA
| | - Lindsay Hollander
- Department of Medicine Section of Nephrology, Yale University, New Haven, CT, USA
| | - Robert Safirstein
- Department of Medicine Section of Nephrology, Yale University, New Haven, CT, USA; Department of Medicine VACHS, Yale University, New Haven, CT, USA
| | - Bryce Nelson
- Department of Medicine Pharmacology, Yale University, New Haven, CT, USA
| | - Charles Cha
- Department of Medicine Surgery, Yale University, New Haven, CT, USA
| | - Marcus Bosenberg
- Department of Medicine Section of Medical Oncology, Yale University, New Haven, CT, USA
| | - Lucia B Jilaveanu
- Department of Medicine Section of Medical Oncology, Yale University, New Haven, CT, USA
| | - David Rimm
- Department of Medicine Pathology, Yale University, New Haven, CT, USA
| | - Carla V Rothlin
- Department of Medicine Immunology, Yale University, New Haven, CT, USA
| | - Harriet M Kluger
- Department of Medicine Section of Medical Oncology, Yale University, New Haven, CT, USA
| | - Gary V Desir
- Department of Medicine Section of Nephrology, Yale University, New Haven, CT, USA; Department of Medicine VACHS, Yale University, New Haven, CT, USA; Department of Medicine Yale School of Medicine, Yale University, New Haven, CT, USA.
| |
Collapse
|
13
|
Wang M, Silva T, Toothaker JM, McCourt BT, Shugrue C, Desir G, Gorelick F, Konnikova L. Renalase and its receptor, PMCA4b, are expressed in the placenta throughout the human gestation. Sci Rep 2022; 12:4953. [PMID: 35322081 PMCID: PMC8943056 DOI: 10.1038/s41598-022-08817-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 03/07/2022] [Indexed: 01/03/2023] Open
Abstract
Placental function requires organized growth, transmission of nutrients, and an anti-inflammatory milieu between the maternal and fetal interface, but placental factors important for its function remain unclear. Renalase is a pro-survival, anti-inflammatory flavoprotein found to be critical in other tissues. We examined the potential role of renalase in placental development. PCR, bulk RNA sequencing, immunohistochemistry, and immunofluorescence for renalase and its binding partners, PMCA4b and PZP, were performed on human placental tissue from second-trimester and full-term placentas separated into decidua, placental villi and chorionic plates. Quantification of immunohistochemistry was used to localize renalase across time course from 17 weeks to term. Endogenous production of renalase was examined in placental tissue and organoids. Renalase and its receptor PMCA4b transcripts and proteins were present in all layers of the placenta. Estimated RNLS protein levels did not change with gestation in the decidual samples. However, placental villi contained more renalase immunoreactive cells in fetal than full-term placental samples. RNLS co-labeled with markers for Hofbauer cells and trophoblasts within the placental villi. Endogenous production of RNLS, PMCA4b, and PZP by trophoblasts was validated in placental organoids. Renalase is endogenously expressed throughout placental tissue and specifically within Hofbauer cells and trophoblasts, suggesting a potential role for renalase in placental development and function. Future studies should assess renalase's role in normal and diseased human placenta.
Collapse
Affiliation(s)
- Melinda Wang
- Yale University School of Medicine, 375 Congress Ave, LSOG 405B, New Haven, CT, 06519, USA
| | - Tatiana Silva
- Yale University School of Medicine, 375 Congress Ave, LSOG 405B, New Haven, CT, 06519, USA
| | - Jessica M Toothaker
- Yale University School of Medicine, 375 Congress Ave, LSOG 405B, New Haven, CT, 06519, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Blake T McCourt
- Yale University School of Medicine, 375 Congress Ave, LSOG 405B, New Haven, CT, 06519, USA
- Department of Pediatrics, Yale University, New Haven, CT, 06520, USA
| | - Christine Shugrue
- Yale University School of Medicine, 375 Congress Ave, LSOG 405B, New Haven, CT, 06519, USA
- Department of Internal Medicine, Yale University, New Haven, CT, 06520, USA
| | - Gary Desir
- Yale University School of Medicine, 375 Congress Ave, LSOG 405B, New Haven, CT, 06519, USA
- Department of Internal Medicine, Yale University, New Haven, CT, 06520, USA
- VA CT Medical Center, Yale University, New Haven, CT, 06520, USA
| | - Fred Gorelick
- Yale University School of Medicine, 375 Congress Ave, LSOG 405B, New Haven, CT, 06519, USA
- Department of Internal Medicine, Yale University, New Haven, CT, 06520, USA
- VA CT Medical Center, Yale University, New Haven, CT, 06520, USA
- Department of Cell Biology, Yale University, New Haven, CT, 06520, USA
| | - Liza Konnikova
- Yale University School of Medicine, 375 Congress Ave, LSOG 405B, New Haven, CT, 06519, USA.
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
- Department of Pediatrics, Yale University, New Haven, CT, 06520, USA.
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University, New Haven, CT, 06520, USA.
- Program in Human and Translational Immunology, Yale University, New Haven, CT, 06520, USA.
| |
Collapse
|
14
|
Safdar B, Wang M, Guo X, Cha C, Chun HJ, Deng Y, Dziura J, El-Khoury JM, Gorelick F, Ko AI, Lee AI, Safirstein R, Simonov M, Zhou B, Desir GV. Association of renalase with clinical outcomes in hospitalized patients with COVID-19. PLoS One 2022; 17:e0264178. [PMID: 35259186 PMCID: PMC8903289 DOI: 10.1371/journal.pone.0264178] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 02/04/2022] [Indexed: 12/27/2022] Open
Abstract
Renalase is a secreted flavoprotein with anti-inflammatory and pro-cell survival properties. COVID-19 is associated with disordered inflammation and apoptosis. We hypothesized that blood renalase levels would correspond to severe COVID-19 and survival. In this retrospective cohort study, clinicopathologic data and blood samples were collected from hospitalized COVID-19 subjects (March—June 2020) at a single institution tertiary hospital. Plasma renalase and cytokine levels were measured and clinical data abstracted from health records. Of 3,450 COVID-19 patients, 458 patients were enrolled. Patients were excluded if <18 years, or opted out of research. The primary composite outcome was intubation or death within 180 days. Secondary outcomes included mortality alone, intensive care unit admission, use of vasopressors, and CPR. Enrolled patients had mean age 64 years (SD±17), were 53% males, and 48% non-whites. Mean renalase levels was 14,108·4 ng/ml (SD±8,137 ng/ml). Compared to patients with high renalase, those with low renalase (< 8,922 ng/ml) were more likely to present with hypoxia, increased ICU admission (54% vs. 33%, p < 0.001), and cardiopulmonary resuscitation (10% vs. 4%, p = 0·023). In Cox proportional hazard model, every 1000 ng/ml increase in renalase decreased the risk of death or intubation by 5% (HR 0·95; 95% CI 0·91–0·98) and increased survival alone by 6% (HR 0·95; CI 0·90–0·98), after adjusting for socio-demographics, initial disease severity, comorbidities and inflammation. Patients with high renalase-low IL-6 levels had the best survival compared to other groups (p = 0·04). Renalase was independently associated with reduced intubation and mortality in hospitalized COVID-19 patients. Future studies should assess the pathophysiological relevance of renalase in COVID-19 disease.
Collapse
Affiliation(s)
- Basmah Safdar
- Department of Emergency Medicine, Yale School of Medicine, New Haven, Connecticut, United States of America
- * E-mail:
| | - Melinda Wang
- Department of Medicine, Yale School of Medicine, New Haven, Connecticut, United States of America
- Department of Cell Biology, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Xiaojia Guo
- Department of Medicine, Yale School of Medicine, New Haven, Connecticut, United States of America
- VA CT HealthCare, West Haven, Connecticut, United States of America
| | - Charles Cha
- Department of Surgery, Hartford HealthCare, Hartford, Connecticut, United States of America
| | - Hyung J. Chun
- Department of Medicine, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Yanhong Deng
- Yale Center of Analytics Sciences, New Haven, Connecticut, United States of America
| | - James Dziura
- Department of Emergency Medicine, Yale School of Medicine, New Haven, Connecticut, United States of America
- Yale Center of Analytics Sciences, New Haven, Connecticut, United States of America
| | - Joe M. El-Khoury
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Fred Gorelick
- Department of Medicine, Yale School of Medicine, New Haven, Connecticut, United States of America
- Department of Cell Biology, Yale School of Medicine, New Haven, Connecticut, United States of America
- Yale Center of Analytics Sciences, New Haven, Connecticut, United States of America
| | - Albert I. Ko
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, United States of America
| | - Alfred I. Lee
- Department of Medicine, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Robert Safirstein
- Department of Medicine, Yale School of Medicine, New Haven, Connecticut, United States of America
- VA CT HealthCare, West Haven, Connecticut, United States of America
| | - Michael Simonov
- Yale Center of Analytics Sciences, New Haven, Connecticut, United States of America
| | - Bin Zhou
- Yale Center of Analytics Sciences, New Haven, Connecticut, United States of America
| | - Gary V. Desir
- Department of Medicine, Yale School of Medicine, New Haven, Connecticut, United States of America
- VA CT HealthCare, West Haven, Connecticut, United States of America
| |
Collapse
|
15
|
Wang Y, Chen C, Hu GL, Chu C, Zhang XY, Du MF, Zou T, Zhou Q, Liao YY, Ma Q, Wang KK, Sun Y, Wang D, Yan Y, Li Y, Jia H, Niu ZJ, Zhang X, Wang L, Man ZY, Gao WH, Li CH, Zhang J, Gao K, Li HX, Chang J, Desir GV, Lu WH, Mu JJ. Associations of Renalase With Blood Pressure and Hypertension in Chinese Adults. Front Cardiovasc Med 2022; 9:800427. [PMID: 35282385 PMCID: PMC8907541 DOI: 10.3389/fcvm.2022.800427] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 01/31/2022] [Indexed: 12/11/2022] Open
Abstract
Objective Renalase, a novel secretory flavoprotein with amine oxidase activity, is secreted into the blood by the kidneys and is hypothesized to participate in blood pressure (BP) regulation. We investigated the associations of renalase with BP and the risk of hypertension by examining renalase single nucleopeptide polymorphism (SNPs), serum renalase levels, and renal expression of renalase in humans. Methods ① Subjects (n = 514) from the original Baoji Salt-Sensitive Study cohort were genotyped to investigate the association of renalase SNPs with longitudinal BP changes and the risk of hypertension during 14 years of follow-up. ② Two thousand three hundred and ninety two participants from the Hanzhong Adolescent Hypertension Study cohort were used to examine the association of serum renalase levels with hypertension. Renalase expression in renal biopsy specimens from 193 patients were measured by immunohistochemistry. ③ Renalase expression was compared in hypertensive vs. normotensive patients. Results ① SNP rs7922058 was associated with 14-year change in systolic BP, and rs10887800, rs796945, rs1935582, rs2296545, and rs2576178 were significantly associated with 14-year change in diastolic BP while rs1935582 and rs2576178 were associated with mean arterial pressure change over 14 years. In addition, SNPs rs796945, rs1935582, and rs2576178 were significantly associated with hypertension incidence. Gene-based analysis found that renalase gene was significantly associated with hypertension incidence over 14-year follow-up after adjustment for multiple measurements. ② Hypertensive subjects had higher serum renalase levels than normotensive subjects (27.2 ± 0.4 vs. 25.1 ± 0.2 μg/mL). Serum renalase levels and BPs showed a linear correlation. In addition, serum renalase was significantly associated with the risk of hypertension [OR = 1.018 (1.006–1.030)]. ③ The expression of renalase in human renal biopsy specimens significantly decreased in hypertensive patients compared to non-hypertensive patients (0.030 ± 0.001 vs. 0.038 ± 0.004). Conclusions These findings indicate that renalase may play an important role in BP progression and development of hypertension.
Collapse
Affiliation(s)
- Yang Wang
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Key Laboratory of Molecular Cardiology of Shaanxi Province, Xi'an, China
| | - Chen Chen
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Key Laboratory of Molecular Cardiology of Shaanxi Province, Xi'an, China
| | - Gui-Lin Hu
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Chao Chu
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Key Laboratory of Molecular Cardiology of Shaanxi Province, Xi'an, China
| | - Xiao-Yu Zhang
- Department of Cardiology, Northwest Women's and Children's Hospital of Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Ming-Fei Du
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ting Zou
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Qing Zhou
- National Engineering Research Center for Beijing Biochip Technology, Beijing, China
| | - Yue-Yuan Liao
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Key Laboratory of Molecular Cardiology of Shaanxi Province, Xi'an, China
| | - Qiong Ma
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Key Laboratory of Molecular Cardiology of Shaanxi Province, Xi'an, China
| | - Ke-Ke Wang
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Key Laboratory of Molecular Cardiology of Shaanxi Province, Xi'an, China
| | - Yue Sun
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Key Laboratory of Molecular Cardiology of Shaanxi Province, Xi'an, China
| | - Dan Wang
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yu Yan
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Key Laboratory of Molecular Cardiology of Shaanxi Province, Xi'an, China
| | - Yan Li
- Department of Nephrology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hao Jia
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ze-Jiaxin Niu
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xi Zhang
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Lan Wang
- Department of Cardiology, Xi'an International Medical Center Hospital, Xi'an, China
| | - Zi-Yue Man
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Key Laboratory of Molecular Cardiology of Shaanxi Province, Xi'an, China
| | - Wei-Hua Gao
- Department of Cardiology, Xi'an No.1 Hospital, Xi'an, China
| | - Chun-Hua Li
- Department of Ophthalmology, Xi'an People's Hospital, Xi'an, China
| | - Jie Zhang
- Department of Cardiology, Xi'an People's Hospital, Xi'an, China
| | - Ke Gao
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hui-Xian Li
- Department of Nephrology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - John Chang
- Department of Medicine, Yale University School of Medicine, New Haven, CT, United States
- Department of Medicine, Veterans Administration Healthcare System, West Haven, CT, United States
| | - Gary V. Desir
- Department of Medicine, Yale University School of Medicine, New Haven, CT, United States
- Department of Medicine, Veterans Administration Healthcare System, West Haven, CT, United States
| | - Wan-Hong Lu
- Department of Nephrology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- *Correspondence: Wan-Hong Lu
| | - Jian-Jun Mu
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Key Laboratory of Molecular Cardiology of Shaanxi Province, Xi'an, China
- Jian-Jun Mu
| |
Collapse
|
16
|
Gao Y, Wang M, Guo X, Hu J, Chen TM, Finn SMB, Lacy J, Kunstman JW, Cha CH, Bellin MD, Robert ME, Desir GV, Gorelick FS. Renalase is a novel tissue and serological biomarker in pancreatic ductal adenocarcinoma. PLoS One 2021; 16:e0250539. [PMID: 34587190 PMCID: PMC8480607 DOI: 10.1371/journal.pone.0250539] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 08/10/2021] [Indexed: 12/24/2022] Open
Abstract
Dysregulated expression of the secretory protein renalase can promote pancreatic ductal adenocarcinoma (PDAC) growth in animal models. We characterized renalase expression in premalignant and malignant PDAC tissue and investigated whether plasma renalase levels corresponded to clinical PDAC characteristics. Renalase immunohistochemistry was used to determine the presence and distribution of renalase in normal pancreas, chronic pancreatitis, PDAC precursor lesions, and PDAC tissues. Associations between pretreatment plasma renalase and PDAC clinical status were assessed in patients with varied clinical stages of PDAC and included tumor characteristics, surgical resection in locally advanced/borderline resectable PDAC, and overall survival. Data were retrospectively obtained and correlated using non-parametric analysis. Little to no renalase was detected by histochemistry in the normal pancreatic head in the absence of abdominal trauma. In chronic pancreatitis, renalase immunoreactivity localized to peri-acinar spindle-shaped cells in some samples. It was also widely present in PDAC precursor lesions and PDAC tissue. Among 240 patients with PDAC, elevated plasma renalase levels were associated with worse tumor characteristics, including greater angiolymphatic invasion (80.0% vs. 58.1%, p = 0.012) and greater node positive disease (76.5% vs. 56.5%, p = 0.024). Overall survival was worse in patients with high plasma renalase levels with median follow-up of 27.70 months vs. 65.03 months (p < 0.001). Renalase levels also predicted whether patients with locally advanced/borderline resectable PDAC underwent resection (AUC 0.674; 95%CI 0.42-0.82, p = 0.04). Overall tissue renalase was increased in both premalignant and malignant PDAC tissues compared to normal pancreas. Elevated plasma renalase levels were associated with advanced tumor characteristics, decreased overall survival, and reduced resectability in patients with locally advanced/borderline resectable PDAC. These studies show that renalase levels are increased in premalignant pancreatic tissues and that its levels in plasma correspond to the clinical behavior of PDAC.
Collapse
Affiliation(s)
- Yasheen Gao
- Yale University, New Haven, Connecticut, United States of America
- Department of Medicine, Veterans Affairs Connecticut Health System, Yale University School of Medicine, West Haven, Connecticut, United States of America
| | - Melinda Wang
- Department of Medicine, Veterans Affairs Connecticut Health System, Yale University School of Medicine, West Haven, Connecticut, United States of America
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Xiaojia Guo
- Department of Medicine, Veterans Affairs Connecticut Health System, Yale University School of Medicine, West Haven, Connecticut, United States of America
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Joanna Hu
- Yale Cancer Center, New Haven, Connecticut, United States of America
| | - Tian-min Chen
- Department of Medicine, Veterans Affairs Connecticut Health System, Yale University School of Medicine, West Haven, Connecticut, United States of America
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Sade´ M. B. Finn
- Department of Surgery, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Jill Lacy
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - John W. Kunstman
- Department of Surgery, Yale University School of Medicine and VA Connecticut, New Haven, Connecticut, United States of America
| | - Charles H. Cha
- Department of Surgery, Hartford Healthcare Saint Vincent’s Medical Center, Bridgeport, Connecticut, United States of America
| | - Melena D. Bellin
- Department of Surgery, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Marie E. Robert
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Gary V. Desir
- Department of Medicine, Veterans Affairs Connecticut Health System, Yale University School of Medicine, West Haven, Connecticut, United States of America
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Fred S. Gorelick
- Department of Medicine, Veterans Affairs Connecticut Health System, Yale University School of Medicine, West Haven, Connecticut, United States of America
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| |
Collapse
|
17
|
Czerwińska K, Poręba R, Gać P. Renalase-A new understanding of its enzymatic and non-enzymatic activity and its implications for future research. Clin Exp Pharmacol Physiol 2021; 49:3-9. [PMID: 34545616 DOI: 10.1111/1440-1681.13594] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 09/16/2021] [Accepted: 09/19/2021] [Indexed: 01/09/2023]
Abstract
Renalase was first described in 2005 and since then it became an object of scientific interest because of its proposed ability to catalyse circulating neurotransmitters and its promising antihypertensive effects. However, further research on the enzymatic activity of renalase did not confirm these initial findings and yielded that renalase serves to oxidize isomeric forms of β-NAD(P)H and recycle them by forming β-NAD(P)+. Moreover, in contrast to initial assumptions, it is indicated that renalase's enzymatic activity is confined to the cell and that extracellular renalase loses its enzymatic properties. These new reports led scientists to question as to whether renalase, as an enzyme, still has the potential to influence various systemic physiological responses (e.g. blood pressure). It was also put into question whether many physiological discoveries published based on the notion that renalase is secreted into the blood and acts by oxidation of catecholamines can still be considered valid. In this article, we attempt to review the literature to confront these doubts and find further possible directions of research on the importance of renalase. Our aim was to evaluate recent reports of non-enzymatic activity for renalase.
Collapse
Affiliation(s)
| | - Rafał Poręba
- Department of Internal and Occupational Diseases, Hypertension and Clinical Oncology, Wroclaw Medical University, Wroclaw, Poland
| | - Paweł Gać
- Department of Hygiene, Wroclaw Medical University, Wroclaw, Poland
| |
Collapse
|
18
|
Pointer TC, Gorelick FS, Desir GV. Renalase: A Multi-Functional Signaling Molecule with Roles in Gastrointestinal Disease. Cells 2021; 10:2006. [PMID: 34440775 PMCID: PMC8391834 DOI: 10.3390/cells10082006] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/21/2021] [Accepted: 07/28/2021] [Indexed: 01/11/2023] Open
Abstract
The survival factor renalase (RNLS) is a recently discovered secretory protein with potent prosurvival and anti-inflammatory effects. Several evolutionarily conserved RNLS domains are critical to its function. These include a 20 aa site that encodes for its prosurvival effects. Its prosurvival effects are shown in GI disease models including acute cerulein pancreatitis. In rodent models of pancreatic cancer and human cancer tissues, increased RNLS expression promotes cancer cell survival but shortens life expectancy. This 37 kD protein can regulate cell signaling as an extracellular molecule and probably also at intracellular sites. Extracellular RNLS signals through a specific plasma membrane calcium export transporter; this interaction appears most relevant to acute injury and cancer. Preliminary studies using RNLS agonists and antagonists, as well as various preclinical disease models, suggest that the immunologic and prosurvival effects of RNLS will be relevant to diverse pathologies that include acute organ injuries and select cancers. Future studies should define the roles of RNLS in intestinal diseases, characterizing the RNLS-activated pathways linked to cell survival and developing therapeutic agents that can increase or decrease RNLS in relevant clinical settings.
Collapse
Affiliation(s)
- Thomas C. Pointer
- Department of Medicine, Yale School of Medicine, 333 Cedar St., New Haven, CT 06510, USA; (T.C.P.); (F.S.G.)
| | - Fred S. Gorelick
- Department of Medicine, Yale School of Medicine, 333 Cedar St., New Haven, CT 06510, USA; (T.C.P.); (F.S.G.)
- VA Connecticut Health Care System, 950 Campbell Avenue, West Haven, CT 06516, USA
| | - Gary V. Desir
- Department of Medicine, Yale School of Medicine, 333 Cedar St., New Haven, CT 06510, USA; (T.C.P.); (F.S.G.)
- VA Connecticut Health Care System, 950 Campbell Avenue, West Haven, CT 06516, USA
| |
Collapse
|
19
|
Stojanovic D, Mitic V, Stojanovic M, Petrovic D, Ignjatovic A, Milojkovic M, Dunjic O, Milenkovic J, Bojanic V, Deljanin Ilic M. The Discriminatory Ability of Renalase and Biomarkers of Cardiac Remodeling for the Prediction of Ischemia in Chronic Heart Failure Patients With the Regard to the Ejection Fraction. Front Cardiovasc Med 2021; 8:691513. [PMID: 34395559 PMCID: PMC8358392 DOI: 10.3389/fcvm.2021.691513] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 06/10/2021] [Indexed: 12/28/2022] Open
Abstract
Background: Renalase has been implicated in chronic heart failure (CHF); however, nothing is known about renalase discriminatory ability and prognostic evaluation. The aims of the study were to assess whether plasma renalase may be validated as a predictor of ischemia in CHF patients stratified to the left ventricular ejection fraction (LVEF) and to determine its discriminatory ability coupled with biomarkers representing a range of heart failure (HF) pathophysiology: brain natriuretic peptide (BNP), soluble suppressor of tumorigenicity (sST2), galectin-3, growth differentiation factor 15 (GDF-15), syndecan-1, and cystatin C. Methods: A total of 77 CHF patients were stratified according to the LVEF and were subjected to exercise stress testing. Receiver operating characteristic curves were constructed, and the areas under curves (AUC) were determined, whereas the calibration was evaluated using the Hosmer-Lemeshow statistic. A DeLong test was performed to compare the AUCs of biomarkers. Results: Independent predictors for ischemia in the total HF cohort were increased plasma concentrations: BNP (p = 0.008), renalase (p = 0.012), sST2 (p = 0.020), galectin-3 (p = 0.018), GDF-15 (p = 0.034), and syndecan-1 (p = 0.024), whereas after adjustments, only BNP (p = 0.010) demonstrated predictive power. In patients with LVEF <45% (HFrEF), independent predictors of ischemia were BNP (p = 0.001), renalase (p < 0.001), sST2 (p = 0.004), galectin-3 (p = 0.003), GDF-15 (p = 0.001), and syndecan-1 (p < 0.001). The AUC of BNP (0.837) was statistically higher compared to those of sST2 (DeLong test: p = 0.042), syndecan-1 (DeLong: p = 0.022), and cystatin C (DeLong: p = 0.022). The AUCs of renalase (0.753), galectin-3 (0.726), and GDF-15 (0.735) were similar and were non-inferior compared to BNP, regarding ischemia prediction. In HFrEF patients, the AUC of BNP (0.980) was statistically higher compared to those of renalase (DeLong: p < 0.001), sST2 (DeLong: p < 0.004), galectin-3 (DeLong: p < 0.001), GDF-15 (DeLong: p = 0.001), syndecan-1 (DeLong: p = 0.009), and cystatin C (DeLong: p = 0.001). The AUC of renalase (0.814) was statistically higher compared to those of galectin-3 (DeLong: p = 0.014) and GDF-15 (DeLong: p = 0.046) and similar to that of sST2. No significant results were obtained in the patients with LVEF >45%. Conclusion: Plasma renalase concentration provided significant discrimination for the prediction of ischemia in patients with CHF and appeared to have similar discriminatory potential to that of BNP. Although further confirmatory studies are warranted, renalase seems to be a relevant biomarker for ischemia prediction, implying its potential contribution to ischemia-risk stratification.
Collapse
Affiliation(s)
- Dijana Stojanovic
- Institute of Pathophysiology, Faculty of Medicine, University of Nis, Nis, Serbia
| | - Valentina Mitic
- Department of Cardiovascular Rehabilitation, Institute for Treatment and Rehabilitation "Niska Banja", Niska Banja, Serbia
| | - Miodrag Stojanovic
- Department of Medical Statistics and Informatics, Faculty of Medicine, University of Nis, Nis, Serbia.,Center of Informatics and Biostatistics in Healthcare, Institute for Public Health, Nis, Serbia
| | - Dejan Petrovic
- Department of Cardiovascular Rehabilitation, Institute for Treatment and Rehabilitation "Niska Banja", Niska Banja, Serbia.,Department of Internal Medicine, Faculty of Medicine, University of Nis, Nis, Serbia
| | - Aleksandra Ignjatovic
- Department of Medical Statistics and Informatics, Faculty of Medicine, University of Nis, Nis, Serbia.,Center of Informatics and Biostatistics in Healthcare, Institute for Public Health, Nis, Serbia
| | - Maja Milojkovic
- Institute of Pathophysiology, Faculty of Medicine, University of Nis, Nis, Serbia
| | - Olivera Dunjic
- Institute of Pathophysiology, Faculty of Medicine, University of Nis, Nis, Serbia
| | - Jelena Milenkovic
- Institute of Pathophysiology, Faculty of Medicine, University of Nis, Nis, Serbia
| | - Vladmila Bojanic
- Institute of Pathophysiology, Faculty of Medicine, University of Nis, Nis, Serbia
| | - Marina Deljanin Ilic
- Department of Cardiovascular Rehabilitation, Institute for Treatment and Rehabilitation "Niska Banja", Niska Banja, Serbia.,Department of Internal Medicine, Faculty of Medicine, University of Nis, Nis, Serbia
| |
Collapse
|
20
|
Tokinoya K, Sekine N, Aoki K, Ono S, Kuji T, Sugasawa T, Yoshida Y, Takekoshi K. Effects of renalase deficiency on liver fibrosis markers in a nonalcoholic steatohepatitis mouse model. Mol Med Rep 2021; 23:210. [PMID: 33495844 PMCID: PMC7830932 DOI: 10.3892/mmr.2021.11849] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 10/06/2020] [Indexed: 01/07/2023] Open
Abstract
Progression of nonalcoholic steatohepatitis (NASH) is attributed to several factors, including inflammation and oxidative stress. In recent years, renalase has been reported to suppress oxidative stress, apoptosis and inflammation. A number of studies have suggested that renalase may be associated with protecting the liver from injury. The present study aimed to clarify the effects of renalase knockout (KO) in mice with NASH that were induced with a choline-deficient high-fat diet (CDAHFD) supplemented with 0.1% methionine. Wild type (WT) and KO mice (6-week-old) were fed a normal diet (ND) or CDAHFD for 6 weeks, followed by analysis of the blood liver function markers and liver tissues. CDAHFD intake was revealed to increase blood hepatic function markers, lipid accumulation and oxidative stress compared with ND, but no significant differences were observed between the WT and KO mice. However, in the KO-CDAHFD group, the Adgre1 and Tgfb1 mRNA levels were significantly higher, and α-SMA expression was significantly lower compared with the WT-CDAHFD group. Furthermore, the Gclc mRNA and phosphorylated protein kinase B (Akt) levels were significantly lower in the KO-ND group compared with the WT-ND group. The results of the current study indicated that as NASH progressed in the absence of renalase, oxidative stress, macrophage infiltration and TGF-β expression were enhanced, while α-SMA expression in NASH may be partly suppressed due to the decreased phosphorylation of Akt level.
Collapse
Affiliation(s)
- Katsuyuki Tokinoya
- Doctoral Program in Sports Medicine, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki 305‑8577, Japan
| | - Nanami Sekine
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki 305‑8574, Japan
| | - Kai Aoki
- Doctoral Program in Sports Medicine, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki 305‑8577, Japan
| | - Seiko Ono
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki 305‑8574, Japan
| | - Tomoaki Kuji
- Doctoral Program in Sports Medicine, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki 305‑8577, Japan
| | - Takehito Sugasawa
- Division of Clinical Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305‑8577, Japan
| | - Yasuko Yoshida
- Department of Clinical Laboratory Science, Faculty of Health Sciences, Tsukuba International University, Tsuchiura, Ibaraki 300‑0051, Japan
| | - Kazuhiro Takekoshi
- Division of Clinical Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305‑8577, Japan
| |
Collapse
|
21
|
Renalase improves pressure overload-induced heart failure in rats by regulating extracellular signal-regulated protein kinase 1/2 signaling. Hypertens Res 2021; 44:481-488. [PMID: 33420473 DOI: 10.1038/s41440-020-00599-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 10/13/2020] [Accepted: 11/10/2020] [Indexed: 01/11/2023]
Abstract
Renalase, a novel flavoprotein that is mainly expressed in the kidney and heart, plays a crucial role in hypertension. Recent studies have shown that renalase is expressed at low levels in the serum of patients with heart failure, while the role of renalase and its mechanism in cardiac failure is unclear. Adult Sprague-Dawley (SD) rats were used to investigate the role and function of renalase in the pathological process of transverse aortic constriction (TAC)-induced heart failure. Renalase-human protein chip analysis showed that renalase was directly associated with P38 and extracellular signal-regulated protein kinase 1/2 (ERK1/2) signaling. We further used lentivirus-mediated RNA interference to study the role of renalase in the progression of pathological ventricular hypertrophy and found that renalase inhibition attenuated the noradrenaline-induced hypertrophic response in vitro or the pressure overload-induced hypertrophic response in vivo. Recombinant renalase protein significantly alleviated pressure overload-induced cardiac failure and was associated with P38 and ERK1/2 signaling. These findings demonstrate that renalase is a potential biomarker of hypertrophy and that exogenous recombinant renalase is a potential and novel drug for heart failure.
Collapse
|
22
|
Stojanovic D, Mitic V, Stojanovic M, Petrovic D, Ignjatovic A, Stefanovic N, Cvetkovic T, Kocic G, Bojanic V, Deljanin Ilic M. The partnership between renalase and ejection fraction as a risk factor for increased cardiac remodeling biomarkers in chronic heart failure patients. Curr Med Res Opin 2020; 36:909-919. [PMID: 32297799 DOI: 10.1080/03007995.2020.1756233] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Objective: Heart failure (HF) represents a huge socio-economic burden. It has been demonstrated, experimentally, that renalase, a newly discovered protein, prevents cardiac hypertrophy and adverse remodeling, which is seen in HF. We postulated the following aims: to investigate associations of renalase with biomarkers of cardiac remodeling: galectin-3, soluble suppression of tumorigenicity, (sST2), growth differentiation factor 15 (GDF-15) and syndecan-1, myocardial stretch (BNP) and cardio-renal axis (cystatin C) in HF patients with reduced ejection fraction (HFrEF) and preserved ejection fraction (HFpEF) to determine whether renalase, in combination with left ventricular ejection fraction (LVEF), represents a risk factor for plasma elevation in biomarkers.Methods: We classified HF patients (n = 76) according to LVEF (preserved/reduced), applied a median plasma renalase (113 ng/mL) as a cut-off value (low/high) and created four subgroups of HF patients: HFpEF/low renalase (n = 19), HFrEF/low renalase (n = 19), HFrEF/high renalase (n = 32) and HFpEF/high renalase (n = 6). A control group (n = 35) consisted of healthy volunteers.Results: Plasma concentrations of evaluated biomarkers were determined using an ELISA technique and were highest in HF patients with reduced EF (p < .001, respectively), and renalase's positive correlations were obtained relating to all biomarkers: galectin-3 (r = 0.913; p < .001), sST2 (r = 0.965; p < .001), GDF-15 (r = 0.887; p < .001), syndecan-1 (r = 0.922; p < .001), BNP (r = 0.527; p < .001) and cystatin C (r = 0.844; p < .001) and strong and negative correlation with LVEF (r = -0.456, p < .001). Increased renalase, regardless of the EF (preserved/reduced), was shown to be an independent risk factor for an increase in all evaluated cardiac remodeling biomarkers, p < .001, respectively. However, increased renalase and reduced EF was the only independent risk factor for BNP and cystatin C elevation, p < .001, respectively. Results after multivariable adjustments (age/gender) were identical.Conclusion: When elevated plasma renalase and HF are present, regardless of EF being reduced or preserved, that represents a significant risk factor for increase in cardiac remodeling biomarker plasma concentrations. However, only elevated renalase and reduced EF demonstrated significance as a risk factor for BNP and cystatin C plasma elevation. Renalase may be considered a promising molecule for the improved predictive abilities of conventional biomarkers and is worthy of further investigation.
Collapse
Affiliation(s)
- Dijana Stojanovic
- Faculty of Medicine, Institute of Pathophysiology, University of Nis, Nis, Serbia
| | - Valentina Mitic
- Institute for Treatment and Rehabilitation "Niska Banja", Niška Banja, Serbia
| | - Miodrag Stojanovic
- Department of Medical Statistics and Informatics, Faculty of Medicine, University of Nis, Nis, Serbia
- Institute for Public Health, Nis, Serbia
| | - Dejan Petrovic
- Institute for Treatment and Rehabilitation "Niska Banja", Niška Banja, Serbia
- Department of Internal Medicine, Faculty of Medicine, University of Nis, Nis, Serbia
| | - Aleksandra Ignjatovic
- Department of Medical Statistics and Informatics, Faculty of Medicine, University of Nis, Nis, Serbia
- Institute for Public Health, Nis, Serbia
| | - Nikola Stefanovic
- Department of Pharmacy, Faculty of Medicine, University of Nis, Nis, Serbia
| | - Tatjana Cvetkovic
- Faculty of Medicine, Institute of Biochemistry, University of Nis, Nis, Serbia
| | - Gordana Kocic
- Faculty of Medicine, Institute of Biochemistry, University of Nis, Nis, Serbia
| | - Vladmila Bojanic
- Faculty of Medicine, Institute of Pathophysiology, University of Nis, Nis, Serbia
| | - Marina Deljanin Ilic
- Institute for Treatment and Rehabilitation "Niska Banja", Niška Banja, Serbia
- Department of Internal Medicine, Faculty of Medicine, University of Nis, Nis, Serbia
| |
Collapse
|
23
|
Potts L, Phillips C, Hwang M, Fulcher S, Choi H. Rescue of human corneal epithelial cells after alkaline insult using renalase derived peptide, RP-220. Int J Ophthalmol 2019; 12:1667-1673. [PMID: 31741852 DOI: 10.18240/ijo.2019.11.01] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 08/12/2019] [Indexed: 12/18/2022] Open
Abstract
AIM To study the effect of renalase peptide, RP-220, on cell viability of human corneal epithelial cells after alkali insult. METHODS A dose-response relationship between cell viability and exposure to NaOH solution were characterized using cultured human corneal epithelial cells. Viability of corneal epithelial cells was determined using commercially available MTT and CyQUANT® assays. RESULTS At a concentration of 6 mmol/L, insult with NaOH leads to reduced corneal epithelial cell viability by approximately 30%. This reduced viability was prevented by treating the cells after initial insult with the 20-amino acid renalase derived peptide (RP-220). CONCLUSION RP-220 has a pro-survival role for RP-220 following alkaline insult to corneal epithelial cells.
Collapse
Affiliation(s)
- Luke Potts
- Department of Ophthalmology and Surgery, Scott and White Eye Institute, Temple, Texas 76508, USA
| | - Casie Phillips
- Central Texas Veterans Research Foundation, Temple, Texas 76504, USA
| | - Munok Hwang
- Department of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Samuel Fulcher
- Department of Surgery, Central Texas Veterans Health Care System, Temple, Texas 76054, USA.,Department of Medicine, College of Medicine, Texas A&M Health Science Center, Bryan, TX 77807, USA
| | - Hosoon Choi
- Central Texas Veterans Research Foundation, Temple, Texas 76504, USA.,Department of Medicine, College of Medicine, Texas A&M Health Science Center, Bryan, TX 77807, USA
| |
Collapse
|
24
|
Association of Plasma Renalase and Left Ventricle Mass Index in Heart Failure Patients Stratified to the Category of the Ejection Fraction: A Pilot Study. DISEASE MARKERS 2019; 2019:7265160. [PMID: 31737132 PMCID: PMC6815612 DOI: 10.1155/2019/7265160] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 08/28/2019] [Indexed: 12/28/2022]
Abstract
Heart failure represents a growing health problem, with increasing morbidity and mortality globally. According to the mechanisms involved in the pathogenesis of heart failure, many biomarkers have been proposed for the timely diagnosis and prognostication of patients with heart failure, but other than natriuretic peptides, none of them has gained enough clinical significance. Renalase, a new protein derived from kidneys was demonstrated to metabolize catecholamines and to have a cardioprotective role. The aim of the study was to determine whether renalase and brain natriuretic peptide (BNP) concentration could be used to differentiate heart failure patients stratified to the category of the ejection fraction and whether plasma renalase could be used as a biomarker for left ventricle hypertrophy in all subgroups of heart failure patients. We included patients diagnosed with heart failure and stratified them to the three subgroups according to the ejection fraction. Regarding echocardiographic parameters, HFmrEF had an intermediate profile in between HFrEF and HFpEF, with statistical significance in most evaluated parameters. BNP concentration was significantly different in all three subgroups (p < 0.001), and renalase was statistically higher in HFrEF (p = 0.007) compared to the HFmrEF and HFpEF, where its results were similar, without statistical significance. Renalase plasma concentration was demonstrated to be highly and positively associated with left ventricle mass index in HFrEF (p = 0.029), as well as increased plasma concentration of BNP (p = 0.006). In the HFmrEF group of patients, body mass index was positively associated with LVMI (p = 0.05), while in the patients with HFpEF, diabetes mellitus was demonstrated to have a positive association with LVMI (p = 0.043). These findings suggest that renalase concentration may be measured in order to differentiate patients with reduced ejection fraction. Plasma renalase concentrations positively correlated with left ventricle hypertrophy in patients with reduced ejection fraction, being strongly associated with increased left ventricular mass index.
Collapse
|
25
|
Renalase attenuates mitochondrial fission in cisplatin-induced acute kidney injury via modulating sirtuin-3. Life Sci 2019; 222:78-87. [DOI: 10.1016/j.lfs.2019.02.042] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 02/04/2019] [Accepted: 02/20/2019] [Indexed: 02/01/2023]
|
26
|
Schmitz B, Kleber ME, Lenders M, Delgado GE, Engelbertz C, Huang J, Pavenstädt H, Breithardt G, Brand SM, März W, Brand E. Genome-wide association study suggests impact of chromosome 10 rs139401390 on kidney function in patients with coronary artery disease. Sci Rep 2019; 9:2750. [PMID: 30809046 PMCID: PMC6391429 DOI: 10.1038/s41598-019-39055-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 01/15/2019] [Indexed: 12/14/2022] Open
Abstract
Chronic kidney disease (CKD) is an independent risk factor for onset and progression of coronary artery disease (CAD). Discovery of predisposing loci for kidney function in CAD patients was performed using a genome-wide association approach. Inclusion criteria were CAD with ≥50% stenosis (≥1 coronary artery) and a creatinine-based estimated glomerular filtration rate (eGFR) of 30–75 ml/min/1.73 m2. An association of rs139401390 located to a region 58.8 kb upstream of renalase (RNLS) with eGFR was detected in the Ludwigshafen Risk and Cardiovascular Health (LURIC) study (n = 499, p = 7.88 × 10−9, mean eGFR 60.7 ml/min/1.73 m2). Direct genotyping of rs139401390A > G suggested increased eGFR by 12.0 ml/min/1.73 m2 per A allele (p = 0.000004). Genome-wide replication of rs139401390A > G in the Coronary Artery Disease and Renal Failure (CAD-REF) registry with a mean eGFR of 47.8 ml/min/1.73 m2 (n = 574, p = 0.033) was only nominally significant. Comparison of rs139401390 genotypes for risk of reduced kidney function in the overall LURIC study revealed higher adjusted odds ratios (OR) for eGFR <60 ml/min/1.73 m2 for CAD patients (n = 1992, OR = 2.36, p = 0.008, G/A + G/G vs A/A) compared to patients with/without CAD (n = 2908, OR = 1.97, p = 0.014, G/A + G/G vs A/A). No significant risk elevation was detected in patients without CAD (n = 948, p = 0.571). rs139401390 may affect kidney function in CAD patients with mild reduction in eGFR.
Collapse
Affiliation(s)
- Boris Schmitz
- Institute of Sports Medicine, Molecular Genetics of Cardiovascular Disease, University Hospital Muenster, Muenster, Germany
| | - Marcus E Kleber
- Medical Clinic V, Mannheim Medical Faculty, University of Heidelberg, Mannheim, Germany.,Institute of Nutrition, Friedrich Schiller University Jena, Jena, Germany
| | - Malte Lenders
- Internal Medicine D, Department of Nephrology, Hypertension and Rheumatology, University Hospital Muenster, Muenster, Germany
| | - Graciela E Delgado
- Medical Clinic V, Mannheim Medical Faculty, University of Heidelberg, Mannheim, Germany
| | - Christiane Engelbertz
- Internal Medicine D, Department of Nephrology, Hypertension and Rheumatology, University Hospital Muenster, Muenster, Germany.,Division of Vascular Medicine, Department of Cardiovascular Medicine, University Hospital Muenster, Muenster, Germany
| | - Jie Huang
- Department of Human Genetics, Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK
| | - Hermann Pavenstädt
- Internal Medicine D, Department of Nephrology, Hypertension and Rheumatology, University Hospital Muenster, Muenster, Germany
| | - Günter Breithardt
- Division of Vascular Medicine, Department of Cardiovascular Medicine, University Hospital Muenster, Muenster, Germany
| | - Stefan-Martin Brand
- Institute of Sports Medicine, Molecular Genetics of Cardiovascular Disease, University Hospital Muenster, Muenster, Germany
| | - Winfried März
- Medical Clinic V, Mannheim Medical Faculty, University of Heidelberg, Mannheim, Germany.,Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria.,Synlab Academy, Synlab Holding Deutschland GmbH, Mannheim, Germany
| | - Eva Brand
- Internal Medicine D, Department of Nephrology, Hypertension and Rheumatology, University Hospital Muenster, Muenster, Germany.
| |
Collapse
|
27
|
Yu X, Han P, Wang J, Sun H, Shao M. Renalase overexpression in ER-positive breast cancer. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2018; 11:1297-1307. [PMID: 31938225 PMCID: PMC6958126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 01/27/2018] [Indexed: 06/10/2023]
Abstract
OBJECTIVES To explore the expression and pathologic significance of renalase in tumor tissues of different molecular subtypes of breast cancer. DESIGN Immunofluorescence methods and laser confocal scanning microscope observations were used to detect expression of renalase, estrogen receptor (ER), phospho-extracellular signal-regulated kinase 1 and 2 (p-ERK1/2), and phospho-signal transducer and activator of transcription (p-STAT3) in 58 cases of breast cancer tissue, 11 normal tissues, and 14 benign fibroadenomas. Statistical analysis of its expression in different molecular subtypes of breast cancer was employed. RESULTS Compared with control tissue (benign lesions and normal breast tissue), renalase was highly expressed in invasive breast cancer and the difference was significant (P<0.0001). Renalase was also expressed significantly higher in ER-positive breast cancer, compared with control tissue (P<0.0001). There was a positive correlation between renalase and ER expression in breast cancer tissues (R=0.7246, P<0.0001) and a positive correlation between renalase and p-ERK 1/2 expression (R=0.6599, P<0.0001). Renalase had no significant correlation with p-STAT3 protein expression. CONCLUSION Renalase is a new molecular marker for ER-positive breast cancer and may become a potential therapeutic target for the ER-positive/HER2-negative subtype breast cancer. Renalase may promote high ER expression and breast cancer cell proliferation and growth through the p-ERK1/2 pathway.
Collapse
Affiliation(s)
- Xuewen Yu
- Department of Pathology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese MedicineChina
| | - Pengxun Han
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese MedicineChina
| | - Jiachuan Wang
- Department of Pathology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese MedicineChina
| | - Huili Sun
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese MedicineChina
| | - Mumin Shao
- Department of Pathology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese MedicineChina
| |
Collapse
|
28
|
Renalase contributes to protection against renal fibrosis via inhibiting oxidative stress in rats. Int Urol Nephrol 2018; 50:1347-1354. [DOI: 10.1007/s11255-018-1820-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 02/04/2018] [Indexed: 12/13/2022]
|
29
|
Kolodecik TR, Reed AM, Date K, Shugrue CA, Patel V, Chung SL, Desir GV, Gorelick FS. The serum protein renalase reduces injury in experimental pancreatitis. J Biol Chem 2017; 292:21047-21059. [PMID: 29042438 DOI: 10.1074/jbc.m117.789776] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 10/12/2017] [Indexed: 12/11/2022] Open
Abstract
Acute pancreatitis is a disease associated with inflammation and tissue damage. One protein that protects against acute injury, including ischemic injury to both the kidney and heart, is renalase, which is secreted into the blood by the kidney and other tissues. However, whether renalase reduces acute injury associated with pancreatitis is unknown. Here, we used both in vitro and in vivo murine models of acute pancreatitis to study renalase's effects on this condition. In isolated pancreatic lobules, pretreatment with recombinant human renalase (rRNLS) blocked zymogen activation caused by cerulein, carbachol, and a bile acid. Renalase also blocked cerulein-induced cell injury and histological changes. In the in vivo cerulein model of pancreatitis, genetic deletion of renalase resulted in more severe disease, and administering rRNLS to cerulein-exposed WT mice after pancreatitis onset was protective. Because pathological increases in acinar cell cytosolic calcium levels are central to the initiation of acute pancreatitis, we also investigated whether rRNLS could function through its binding protein, plasma membrane calcium ATPase 4b (PMCA4b), which excretes calcium from cells. We found that PMCA4b is expressed in both murine and human acinar cells and that a PMCA4b-selective inhibitor worsens pancreatitis-induced injury and blocks the protective effects of rRNLS. These findings suggest that renalase is a protective plasma protein that reduces acinar cell injury through a plasma membrane calcium ATPase. Because exogenous rRNLS reduces the severity of acute pancreatitis, it has potential as a therapeutic agent.
Collapse
Affiliation(s)
- Thomas R Kolodecik
- From the Yale University School of Medicine, New Haven, Connecticut 06510.,Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut 06516, and
| | - Anamika M Reed
- From the Yale University School of Medicine, New Haven, Connecticut 06510.,Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut 06516, and
| | - Kimie Date
- Ochanomizu University, Tokyo 112-8610, Japan
| | - Christine A Shugrue
- From the Yale University School of Medicine, New Haven, Connecticut 06510.,Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut 06516, and
| | - Vikhil Patel
- From the Yale University School of Medicine, New Haven, Connecticut 06510.,Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut 06516, and
| | - Shang-Lin Chung
- From the Yale University School of Medicine, New Haven, Connecticut 06510.,Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut 06516, and
| | - Gary V Desir
- From the Yale University School of Medicine, New Haven, Connecticut 06510.,Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut 06516, and
| | - Fred S Gorelick
- From the Yale University School of Medicine, New Haven, Connecticut 06510, .,Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut 06516, and
| |
Collapse
|