1
|
Shen H, Yang J, Xue W, Wei Z, Li L, Guan J, Li X, Wu X. Renalase rs2296545 variant improve hypertension susceptibility by modifying binding affinity to catecholamines in obstructive sleep apnea. Hypertens Res 2024; 47:3200-3213. [PMID: 39232213 PMCID: PMC11534681 DOI: 10.1038/s41440-024-01850-0] [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: 02/06/2024] [Revised: 07/18/2024] [Accepted: 07/30/2024] [Indexed: 09/06/2024]
Abstract
Obstructive sleep apnea (OSA), a condition often linked with hypertension, has an undefined relationship with renalase, a protein known for regulating blood pressure. This study aimed to investigate the relationship between serum renalase levels as well as renalase functional single nucleotide polymorphism (SNP) rs2296545 variant and hypertension in a Han Chinese OSA population. 126 subjects underwent serum renalase detection, with linear regression being performed to evaluate the relationship between serum renalase levels and OSA-related traits. Additional 4275 subjects were obtained rs2296545 genotype information by SNP microarray. And binary logistic regression was used to assess the effect of rs2296545 on hypertension risk. Molecular dynamics simulation and molecular docking were utilized to access the protein structures and the interplay between protein and catecholamines of wild-type and rs2296545 mutant renalase. The results showed that serum renalase levels were significantly higher in the severe OSA group. Further analysis showed renalase levels were positively correlated with blood pressure in the non-OSA group and negatively correlated in the severe OSA group. For rs2296545 polymorphism analysis, the hypertension risk significantly increased for the recessive model CC/GG + CG (OR = 1.211, 95% CI: 1.025-1.431) and the additive model CC/CG (OR = 1.223, 95% CI: 1.025-1.458) in the severe OSA. The rs2296545 polymorphism affected protein structure, and led to increase binding free energy, weakening interactions between renalase and catecholamines. In conclusion, serum renalase levels had independent association with blood pressure. And rs2296545 polymorphism may influence on susceptibility to hypertension by altering protein ability to bind to catecholamines, which might contribute to the intervention of hypertension in the OSA population.
Collapse
Affiliation(s)
- Hangdong Shen
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
- Otorhinolaryngology Institute of Shanghai Jiao Tong University, Shanghai, China
| | - Jundong Yang
- Department of Clinical Laboratory, Shanghai Children's Hospital, Shanghai Jiaotong University, Shanghai, China
- Faculty of Medical Laboratory Science, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenjun Xue
- Central Laboratory of Shanghai Eighth People's Hospital, Xuhui Branch of Shanghai Sixth People's Hospital, Caobao Road 8, Shanghai, 200235, China
| | - Zhicheng Wei
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
- Otorhinolaryngology Institute of Shanghai Jiao Tong University, Shanghai, China
| | - Lilin Li
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
- Otorhinolaryngology Institute of Shanghai Jiao Tong University, Shanghai, China
| | - Jian Guan
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
- Otorhinolaryngology Institute of Shanghai Jiao Tong University, Shanghai, China
| | - Xinyi Li
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China.
- Otorhinolaryngology Institute of Shanghai Jiao Tong University, Shanghai, China.
| | - Xiaolin Wu
- Central Laboratory of Shanghai Eighth People's Hospital, Xuhui Branch of Shanghai Sixth People's Hospital, Caobao Road 8, Shanghai, 200235, China.
| |
Collapse
|
2
|
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
|
3
|
Walker V. The Intricacies of Renal Phosphate Reabsorption-An Overview. Int J Mol Sci 2024; 25:4684. [PMID: 38731904 PMCID: PMC11083860 DOI: 10.3390/ijms25094684] [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: 03/24/2024] [Revised: 04/17/2024] [Accepted: 04/19/2024] [Indexed: 05/13/2024] Open
Abstract
To maintain an optimal body content of phosphorus throughout postnatal life, variable phosphate absorption from food must be finely matched with urinary excretion. This amazing feat is accomplished through synchronised phosphate transport by myriads of ciliated cells lining the renal proximal tubules. These respond in real time to changes in phosphate and composition of the renal filtrate and to hormonal instructions. How they do this has stimulated decades of research. New analytical techniques, coupled with incredible advances in computer technology, have opened new avenues for investigation at a sub-cellular level. There has been a surge of research into different aspects of the process. These have verified long-held beliefs and are also dramatically extending our vision of the intense, integrated, intracellular activity which mediates phosphate absorption. Already, some have indicated new approaches for pharmacological intervention to regulate phosphate in common conditions, including chronic renal failure and osteoporosis, as well as rare inherited biochemical disorders. It is a rapidly evolving field. The aim here is to provide an overview of our current knowledge, to show where it is leading, and where there are uncertainties. Hopefully, this will raise questions and stimulate new ideas for further research.
Collapse
Affiliation(s)
- Valerie Walker
- Department of Clinical Biochemistry, University Hospital Southampton NHS Foundation Trust, Southampton General Hospital, Southampton S016 6YD, UK
| |
Collapse
|
4
|
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: 1.7] [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
|
5
|
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: 0.3] [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
|
6
|
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: 2.0] [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
|
7
|
Li Y, Wu W, Liu W, Zhou M. Roles and mechanisms of renalase in cardiovascular disease: A promising therapeutic target. Biomed Pharmacother 2020; 131:110712. [PMID: 32916539 DOI: 10.1016/j.biopha.2020.110712] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/28/2020] [Accepted: 08/30/2020] [Indexed: 12/16/2022] Open
Abstract
Cardiovascular disease (CVD) is prevalent worldwide and remains a leading cause of death. Although substantial progress has been made in the diagnosis and treatment of CVD, the prognosis remains unsatisfactory. Renalase is a newly discovered cytokine that is synthesized by the kidney and then secreted into blood. Numerous studies have suggested the efficacy of renalase in treating CVD by metabolizing catecholamines in the circulatory system. As a new biomarker of heart disease, renalase is normally recognized as a signalling molecule that activates cytoprotective intracellular signals to lower blood pressure, protect ischaemic heart muscle and promote atherosclerotic plaque stability in CVD, which subsequently improves cardiac function. Due to its important regulatory role in the circulatory system, renalase has gradually become a potential target in the treatment of CVD. This review summarizes the structure, mechanism and function of renalase in CVD, thereby providing preclinical evidence for alternative approaches and new prospects in the development of renalase-related drugs against CVD.
Collapse
Affiliation(s)
- Yue Li
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing, 100010, China
| | - Weidong Wu
- London Metropolitan University, London, N7 8DB, United Kingdom
| | - Weihong Liu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing, 100010, China
| | - Mingxue Zhou
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing, 100010, China.
| |
Collapse
|
8
|
Matt SM, Gaskill PJ. Where Is Dopamine and how do Immune Cells See it?: Dopamine-Mediated Immune Cell Function in Health and Disease. J Neuroimmune Pharmacol 2020; 15:114-164. [PMID: 31077015 PMCID: PMC6842680 DOI: 10.1007/s11481-019-09851-4] [Citation(s) in RCA: 155] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 04/07/2019] [Indexed: 02/07/2023]
Abstract
Dopamine is well recognized as a neurotransmitter in the brain, and regulates critical functions in a variety of peripheral systems. Growing research has also shown that dopamine acts as an important regulator of immune function. Many immune cells express dopamine receptors and other dopamine related proteins, enabling them to actively respond to dopamine and suggesting that dopaminergic immunoregulation is an important part of proper immune function. A detailed understanding of the physiological concentrations of dopamine in specific regions of the human body, particularly in peripheral systems, is critical to the development of hypotheses and experiments examining the effects of physiologically relevant dopamine concentrations on immune cells. Unfortunately, the dopamine concentrations to which these immune cells would be exposed in different anatomical regions are not clear. To address this issue, this comprehensive review details the current information regarding concentrations of dopamine found in both the central nervous system and in many regions of the periphery. In addition, we discuss the immune cells present in each region, and how these could interact with dopamine in each compartment described. Finally, the review briefly addresses how changes in these dopamine concentrations could influence immune cell dysfunction in several disease states including Parkinson's disease, multiple sclerosis, rheumatoid arthritis, inflammatory bowel disease, as well as the collection of pathologies, cognitive and motor symptoms associated with HIV infection in the central nervous system, known as NeuroHIV. These data will improve our understanding of the interactions between the dopaminergic and immune systems during both homeostatic function and in disease, clarify the effects of existing dopaminergic drugs and promote the creation of new therapeutic strategies based on manipulating immune function through dopaminergic signaling. Graphical Abstract.
Collapse
Affiliation(s)
- S M Matt
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - P J Gaskill
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102, USA.
| |
Collapse
|
9
|
Abstract
Calcium kidney stones are common worldwide. Most are idiopathic and composed of calcium oxalate. Calcium phosphate is present in around 80% and may initiate stone formation. Stone production is multifactorial with a polygenic genetic contribution. Phosphaturia is found frequently among stone formers but until recently received scant attention. This review examines possible mechanisms for the phosphaturia and its relevance to stone formation from a wide angle. There is a striking lack of clinical data. Phosphaturia is associated, but not correlated, with hypercalciuria, increased 1,25 dihydroxy-vitamin D [1,25 (OH)2D], and sometimes evidence of disturbances in proximal renal tubular function. Phosphate reabsorption in the proximal renal tubules requires tightly regulated interaction of many proteins. Paracellular flow through intercellular tight junctions is the major route of phosphate absorption from the intestine and can be reduced therapeutically in hyperphosphatemic patients. In monogenic defects stones develop when phosphaturia is associated with hypercalciuria, generally explained by increased 1,25 (OH)2D production in response to hypophosphatemia. Calcification does not occur in disorders with increased FGF23 when phosphaturia occurs in isolation and 1,25 (OH)2D is suppressed. Candidate gene studies have identified mutations in the phosphate transporters, but in few individuals. One genome-wide study identified a polymorphism of the phosphate transporter gene SLC34A4 associated with stones. Others did not find mutations obviously linked to phosphate reabsorption. Future genetic studies should have a wide trawl and should focus initially on groups of patients with clearly defined phenotypes. The global data should be pooled.
Collapse
Affiliation(s)
- Valerie Walker
- Department of Clinical Biochemistry, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom.
| |
Collapse
|
10
|
Moran GR, Hoag MR. The enzyme: Renalase. Arch Biochem Biophys 2017; 632:66-76. [PMID: 28558965 DOI: 10.1016/j.abb.2017.05.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Revised: 05/22/2017] [Accepted: 05/25/2017] [Indexed: 02/06/2023]
Abstract
Within the last two years catalytic substrates for renalase have been identified, some 10 years after its initial discovery. 2- and 6-dihydronicotinamide (2- and 6-DHNAD) isomers of β-NAD(P)H (4-dihydroNAD(P)) are rapidly oxidized by renalase to form β-NAD(P)+. The two electrons liberated are then passed to molecular oxygen by the renalase FAD cofactor forming hydrogen peroxide. This activity would appear to serve an intracellular detoxification/metabolite repair function that alleviates inhibition of primary metabolism dehydrogenases by 2- and 6-DHNAD molecules. This activity is supported by the complete structural assignment of the substrates, comprehensive kinetic analyses, defined species specific substrate specificity profiles and X-ray crystal structures that reveal ligand complexation consistent with this activity. This apparently intracellular function for the renalase enzyme is not allied with the majority of the renalase research that holds renalase to be a secreted mammalian protein that functions in blood to elicit a broad array of profound physiological changes. In this review a description of renalase as an enzyme is presented and an argument is offered that its enzymatic function can now reasonably be assumed to be uncoupled from whole organism physiological influences.
Collapse
Affiliation(s)
- Graham R Moran
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, 3210 N. Cramer St, Milwaukee, WI 53211-3209, United States.
| | - Matthew R Hoag
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, 3210 N. Cramer St, Milwaukee, WI 53211-3209, United States
| |
Collapse
|
11
|
Wu Y, Yin Q, Lin S, Huang X, Xia Q, Chen Z, Zhang X, Yang D. Increased SLC7A8 expression mediates L-DOPA uptake by renal tubular epithelial cells. Mol Med Rep 2017; 16:887-893. [DOI: 10.3892/mmr.2017.6620] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Accepted: 01/17/2017] [Indexed: 11/05/2022] Open
|
12
|
Wang Y, Safirstein R, Velazquez H, Guo XJ, Hollander L, Chang J, Chen TM, Mu JJ, Desir GV. Extracellular renalase protects cells and organs by outside-in signalling. J Cell Mol Med 2017; 21:1260-1265. [PMID: 28238213 PMCID: PMC5487909 DOI: 10.1111/jcmm.13062] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 11/18/2016] [Indexed: 01/07/2023] Open
Abstract
Renalase was discovered as a protein synthesized by the kidney and secreted in blood where it circulates at a concentration of approximately 3-5 μg/ml. Initial reports suggested that it functioned as an NAD(P)H oxidase and could oxidize catecholamines. Administration of renalase lowers blood pressure and heart rate and also protects cells and organs against ischaemic and toxic injury. Although renalase's protective effect was initially ascribed to its oxidase properties, a paradigm shift in our understanding of the cellular actions of renalase is underway. We now understand that, independent of its enzymatic properties, renalase functions as a cytokine that provides protection to cells, tissues and organs by interacting with its receptor to activate protein kinase B, JAK/STAT, and the mitogen-activated protein kinase pathways. In addition, recent studies suggest that dysregulated renalase signalling may promote survival of several tumour cells due to its capacity to augment expression of growth-related genes. In this review, we focus on the cytoprotective actions of renalase and its capacity to sustain cancer cell growth and also the translational opportunities these findings represent for the development of novel therapeutic strategies for organ injury and cancer.
Collapse
Affiliation(s)
- Yang Wang
- Department of Medicine, Veterans Affairs Connecticut Healthcare System, Yale University, New Haven, CT, USA.,Department of Cardiology, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, Xi'an, China
| | - Robert Safirstein
- Department of Medicine, Veterans Affairs Connecticut Healthcare System, Yale University, New Haven, CT, USA
| | - Heino Velazquez
- Department of Medicine, Veterans Affairs Connecticut Healthcare System, Yale University, New Haven, CT, USA
| | - Xiao-Jia Guo
- Department of Medicine, Veterans Affairs Connecticut Healthcare System, Yale University, New Haven, CT, USA
| | - Lindsay Hollander
- Department of Medicine, Veterans Affairs Connecticut Healthcare System, Yale University, New Haven, CT, USA.,Department of Surgery, University of Connecticut, Farmington, CT, USA
| | - John Chang
- Department of Medicine, Veterans Affairs Connecticut Healthcare System, Yale University, New Haven, CT, USA
| | - Tian-Min Chen
- Department of Medicine, Veterans Affairs Connecticut Healthcare System, Yale University, New Haven, CT, USA
| | - Jian-Jun Mu
- Department of Cardiology, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, Xi'an, China
| | - Gary V Desir
- Department of Medicine, Veterans Affairs Connecticut Healthcare System, Yale University, New Haven, CT, USA
| |
Collapse
|
13
|
Shawar SM, Ramadan AR, Ali BR, Alghamdi MA, John A, Hudaib FM. FGF23-S129F mutant bypasses ER/Golgi to the circulation of hyperphosphatemic familial tumoral calcinosis patients. Bone 2016; 93:187-195. [PMID: 26620085 DOI: 10.1016/j.bone.2015.11.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 11/15/2015] [Accepted: 11/21/2015] [Indexed: 02/05/2023]
Abstract
FGF23 is essential for the homeostasis of phosphate, and vitamin D. Loss-of-function mutations in this hormone cause hyperphosphatemic familial tumoral calcinosis (HFTC). Earlier reports suggested that intact FGF23 from loss of function mutants such as FGF23/S129F (iFGF23/S129F) is retained intracellularly while the carboxy-terminal fragment is secreted. We sought to investigate the fate of iFGF23/S129F mutant hormone in vivo and in vitro. Five patients clinically diagnosed with HFTC and confirmed by DNA sequencing to carry the c.386 C>T; p.S129F mutation in the homozygous state were studied. Healthy and heterozygous individuals were used as controls in the study. Using ELISA assays, we showed that iFGF23/S129F was 2-5 folds higher in patients' plasma, compared to heterozygous or healthy controls. Importantly, the mutant hormone could not be detected in the patients' sera. However, using proteinase inhibition profiling, we found that a serum metalloproteinase degraded the iFGF23/S129F explaining our failure to detect it in sera. The serum metalloproteinase degrades the WT and the mutant at different rates. Also, confocal microscopy imaging using wild-type (WT) FGF23 or FGF23/S129F mutant in transiently transfected HEK293 and HeLa cells showed weak staining of the Golgi complex with some vesicular staining resembling the ER. Additionally, FGF23 variants (FGF23/WT, FGF23/S129F, FGF23/S71G, and FGF23/R176Q) from stably transfected HEK293 cells secreted high levels into a serum-free medium that can be detected by ELISA and Western blot. Our results suggest that iFGF23/S129F mutant bypasses the ER/Golgi quality control system to the circulation of HFTC patients by an unknown pathway. Finally, we hypothesize that either the mutant hormone is unable to bind α-Klotho-FGFR1c, or it binds the dyad receptor with low affinity and, therefore, incapable of initiating maximal intracellular signaling. Our findings raise the potential use of the WT hormone in therapies of some HFTC patients.
Collapse
Affiliation(s)
- Said M Shawar
- Life Sciences Department, Medical Biotechnology, Arabian Gulf University, Manama, Bahrain.
| | - Ahmad R Ramadan
- Life Sciences Department, Medical Biotechnology, Arabian Gulf University, Manama, Bahrain
| | - Bassam R Ali
- Pathology Department, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Manal A Alghamdi
- Life Sciences Department, Medical Biotechnology, Arabian Gulf University, Manama, Bahrain
| | - Anne John
- Pathology Department, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Ferial M Hudaib
- Life Sciences Department, Medical Biotechnology, Arabian Gulf University, Manama, Bahrain
| |
Collapse
|
14
|
Abstract
PURPOSE OF REVIEW This review will highlight recent findings concerning the regulation and signalling of the intrarenal dopaminergic system and the emerging evidence for its importance in blood pressure regulation. RECENT FINDINGS There is an increasing evidence that the intrarenal dopaminergic system plays an important role in the regulation of blood pressure, and defects in dopamine signalling appear to be involved in the development of hypertension. Recent experimental models have definitively demonstrated that abnormalities in intrarenal dopamine production or receptor signalling can predispose to salt-sensitive hypertension and a dysregulated renin-angiotensin system. There are also new results indicating the importance of dopamine receptor mediated regulation of salt and water homeostasis along the nephron, and new studies indicating the role that the intrarenal dopaminergic system plays to mitigate the production of reactive oxygen species and progression of chronic renal disease. SUMMARY New studies underscore the importance of the intrarenal dopaminergic system in the regulation of renal function and indicate how alterations in dopamine production or signalling may underlie the development of hypertension and kidney injury.
Collapse
|
15
|
Abstract
BACKGROUND Contrast-induced acute kidney injury (CI-AKI) remains one of the crucial issues related to the development of invasive cardiology. The massive use of contrast media exposes patients to a great risk of contrast-induced nephropathy and chronic kidney disease development, and increases morbidity and mortality rates. The serum creatinine concentration does not allow for a timely and accurate CI-AKI diagnosis; hence numerous other biomarkers of renal injury have been proposed. Renalase, a novel catecholamine-metabolizing amine oxidase, is synthesized mainly in proximal tubular cells and secreted into urine and blood. It is primarily engaged in the degradation of circulating catecholamines. Notwithstanding its key role in blood pressure regulation, renalase remains a potential CI-AKI biomarker, which was shown to be markedly downregulated in the aftermath of renal injury. In this sense, renalase appears to be the first CI-AKI marker revealing an actual loss of renal function and indicating disease severity. SUMMARY The purpose of this review is to summarize the contemporary knowledge about the application of novel biomarkers of CI-AKI and to highlight the potential role of renalase as a functional marker of contrast-induced renal injury. KEY MESSAGES Renalase may constitute a missing biochemical link in the mutual interplay between kidney and cardiac pathology known as the cardiorenal syndrome.
Collapse
Affiliation(s)
- Maciej T Wybraniec
- First Department of Cardiology, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| | - Katarzyna Mizia-Stec
- First Department of Cardiology, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| |
Collapse
|
16
|
Renalase does not catalyze the oxidation of catecholamines. Arch Biochem Biophys 2015; 579:62-6. [PMID: 26049000 DOI: 10.1016/j.abb.2015.05.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Revised: 05/28/2015] [Accepted: 05/29/2015] [Indexed: 11/21/2022]
Abstract
It is widely accepted that the function of human renalase is to oxidize catecholamines in blood. However, this belief is based on experiments that did not account for slow, facile catecholamine autoxidation reactions. Recent evidence has shown that renalase has substrates with which it reacts rapidly. The reaction catalyzed defines renalase as an oxidase, one that harvests two electrons from either 2-dihydroNAD(P) or 6-dihydroNAD(P) to form β-NAD(P)(+) and hydrogen peroxide. The apparent metabolic purpose of such a reaction is to avoid inhibition of primary dehydrogenase enzymes by these β-NAD(P)H isomers. This article demonstrates that renalase does not catalyze the oxidation of neurotransmitter catecholamines. Using high-performance liquid chromatography we show that there is no evidence of consumption of epinephrine by renalase. Using time-dependent spectrophotometry we show that the renalase FAD cofactor spectrum is unresponsive to added catecholamines, that adrenochromes are not observed to accumulate in the presence of renalase and that the kinetics of single turnover reactions with 6-dihydroNAD are unaltered by the addition of catecholamines. Lastly we show using an oxygen electrode assay that plasma renalase activity is below the level of detection and only when exogenous renalase and 6-dihydroNAD are added can dioxygen be observed to be consumed.
Collapse
|
17
|
Renalase: Another puzzle piece between hypertension and simple renal cysts? Int Urol Nephrol 2015; 47:1181-6. [DOI: 10.1007/s11255-015-1008-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 05/09/2015] [Indexed: 10/23/2022]
|
18
|
Renalase: its role as a cytokine, and an update on its association with type 1 diabetes and ischemic stroke. Curr Opin Nephrol Hypertens 2015; 23:513-8. [PMID: 24992568 DOI: 10.1097/mnh.0000000000000044] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
PURPOSE OF REVIEW Remarkable progress has been achieved over the past 2 years in understanding the cellular actions of renalase, its pathophysiology and potential therapeutic utility. RECENT FINDINGS There has been a paradigm shift in our thinking about the mechanisms underlying the cellular actions of renalase. We now understand that, independent of its enzymatic properties, renalase functions as a signaling molecule, a cytokine that interacts with a yet-to-be identified plasma membrane receptor(s) to activate protein kinase B and the mitogen-activated protein kinase pathway. These signaling properties are critical to its cytoprotective effects. New information regarding renalase's enzymatic function as an α-nicotinamide adenine dinucleotide oxidase/anomerase will be reviewed. Lastly, we will discuss the association of certain single nucleotide polymorphisms in the renalase gene with type 1 diabetes and with ischemic stroke, and the clinical implications of these findings. SUMMARY The consistent association of renalase single nucleotide polymorphisms and the development of type 1 diabetes is a great interest particularly because we now understand that renalase functions as a cytokine. Future work on renalase should focus on exploring the identity of its receptor(s), and its potential role as an immune modulator.
Collapse
|
19
|
The catalytic function of renalase: A decade of phantoms. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2015; 1864:177-86. [PMID: 25900362 DOI: 10.1016/j.bbapap.2015.04.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 04/08/2015] [Accepted: 04/10/2015] [Indexed: 12/31/2022]
Abstract
Ten years after the initial identification of human renalase the first genuinely catalytic substrates have been identified. Throughout the prior decade a consensus belief that renalase is produced predominantly by the kidney and catalytically oxidizes catecholamines in order to lower blood pressure and slow the heart has prevailed. This belief was, however, based on fundamentally flawed scientific observations that did not include control reactions to account for the well-known autoxidation of catecholamines in oxygenated solutions. Nonetheless, the initial claims have served as the kernel for a rapidly expanding body of research largely predicated on the belief that catecholamines are substrates for this enzyme. The proliferation of scientific studies pertaining to renalase as a hormone has proceeded unabated despite well-reasoned expressions of dissent that have indicated the deficiencies of the initial observations and other inconsistencies. Our group has very recently identified isomeric forms of β-NAD(P)H as substrates for renalase. These substrates arise from non-specific reduction of β-NAD(P)(+) that forms β-4-dihydroNAD(P) (β-NAD(P)H), β-2-dihydroNAD(P) and β-6-dihydroNAD(P); the latter two being substrates for renalase. Renalase oxidizes these substrates with rate constants that are up to 10(4)-fold faster than any claimed for catecholamines. The electrons harvested are delivered to dioxygen via the enzyme's FAD cofactor forming both H2O2 and β-NAD(P)(+) as products. It would appear that the metabolic purpose of this chemistry is to alleviate the inhibitory effect of β-2-dihydroNAD(P) and β-6-dihydroNAD(P) on primary metabolism dehydrogenase enzymes. The identification of this genuinely catalytic activity for renalase calls for re-evaluation of much of the research of this enzyme, in which definitive links between renalase catecholamine consumption and physiological responses were reported. This article is part of a Special Issue entitled: Physiological enzymology and protein functions.
Collapse
|
20
|
Malyszko J, Bachorzewska-Gajewska H, Dobrzycki S. Renalase, kidney and cardiovascular disease: are they related or just coincidentally associated? Adv Med Sci 2015; 60:41-9. [PMID: 25461379 DOI: 10.1016/j.advms.2014.10.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 09/15/2014] [Accepted: 10/10/2014] [Indexed: 12/15/2022]
Abstract
Cardiovascular diseases, including hypertension are the leading cause of death in the developed countries. Diabetes and chronic kidney disease became also more prevalent reaching almost the level of epidemy. Researchers are looking eagerly for the new risk and/or pathogenetic factors, as well as therapeutic option in these disease. It has been suggested that human kidney releases a protein named renalase into the bloodstream. It is supposed to be an enzyme which breaks down catecholamines in the blood circulation and regulate blood pressure. However, there were several doubts whether renalase exerts monoaminooxidase activity, or if it is monoaminooxidase at all. Recently, a hypothesis that it is also a cytokine was postulated. Studies on renalase polymorphisms in hypertension, cardiovascular disease or diabetes are inconsistent. Similarly, there are several discrepancies in the animal on the possible role of renalase in hypertension and cardiovascular diseases. Some studies report a protective role of renalase in acute kidney injury, whereas others showed that renalase levels were mainly dependent on kidney function, indicating rather a role of kidney in excretion of this substance. Moreover, validated assays are needed to evaluate renalase levels and activity. On one hand a deeper and more accurate link between renalase and cardiovascular diseases require further profound research, on the other hand whether or not renalase protein could be a new therapeutic target in these pathologies should also be considered. Whether renalase, discovered in 2005, might be a Holy Grail of hypertension, linking kidney and cardiovascular diseases, remains to be proven.
Collapse
Affiliation(s)
- Jolanta Malyszko
- 2nd Department of Nephrology and Hypertension with Dialysis Unit, Medical University of Bialystok, Bialystok, Poland.
| | | | - Slawomir Dobrzycki
- Invasive Cardiology Department, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
21
|
Beaupre BA, Hoag MR, Roman J, Försterling FH, Moran GR. Metabolic function for human renalase: oxidation of isomeric forms of β-NAD(P)H that are inhibitory to primary metabolism. Biochemistry 2015; 54:795-806. [PMID: 25531177 DOI: 10.1021/bi5013436] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Renalase is a recently identified flavoprotein that has been associated with numerous physiological maladies. There remains a prevailing belief that renalase functions as a hormone, imparting an influence on vascular tone and heart rate by oxidizing circulating catecholamines, chiefly epinephrine. This activity, however, has not been convincingly demonstrated in vitro, nor has the stoichiometry of this transformation been shown. In prior work we demonstrated that renalase induced rapid oxidation of low-level contaminants of β-NAD(P)H solutions ( Beaupre, B. A. et al. (2013) Biochemistry 52 , 8929 - 8937 ; Beaupre, B. A. et al. (2013) J. Am. Chem. Soc . 135 , 13980 - 13987 ). Slow aqueous speciation of β-NAD(P)H resulted in the production of renalase substrate molecules whose spectrophotometric characteristics and equilibrium fractional accumulation closely matched those reported for α-anomers of NAD(P)H. The fleeting nature of these substrates precluded structural assignment. Here we structurally assign and identify two substrates for renalase. These molecules are 2- and 6-dihydroNAD(P), isomeric forms of β-NAD(P)H that arise either by nonspecific reduction of β-NAD(P)(+) or by tautomerization of β-NAD(P)H (4-dihydroNAD(P)). The pure preparations of these molecules induce rapid reduction of the renalase flavin cofactor (230 s(-1) for 6-dihydroNAD, 850 s(-1) for 2-dihydroNAD) but bind only a few fold more tightly than β-NADH. We also show that 2- and 6-dihydroNAD(P) are potent inhibitors of primary metabolism dehydrogenases and therefore conclude that the metabolic function of renalase is to oxidize these isomeric NAD(P)H molecules to β-NAD(P)(+), eliminating the threat they pose to normal respiratory activity.
Collapse
Affiliation(s)
- Brett A Beaupre
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee , 3210 North Cramer Street, Milwaukee, Wisconsin 53211-3209, United States
| | | | | | | | | |
Collapse
|
22
|
Quelhas-Santos J, Serrão MP, Soares-Silva I, Fernandes-Cerqueira C, Simões-Silva L, Pinho MJ, Remião F, Sampaio-Maia B, Desir GV, Pestana M. Renalase regulates peripheral and central dopaminergic activities. Am J Physiol Renal Physiol 2014; 308:F84-91. [PMID: 25411385 DOI: 10.1152/ajprenal.00274.2014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Renalase is a recently identified FAD/NADH-dependent amine oxidase mainly expressed in kidney that is secreted into blood and urine where it was suggested to metabolize catecholamines. The present study evaluated central and peripheral dopaminergic activities in the renalase knockout (KO) mouse model and examined the changes induced by recombinant renalase (RR) administration on plasma and urine catecholamine levels. Compared with wild-type (WT) mice, KO mice presented increased plasma levels of epinephrine (Epi), norepinephrine (NE), and dopamine (DA) that were accompanied by increases in the urinary excretion of Epi, NE, DA. In addition, the KO mice presented an increase in urinary DA-to-l-3,4-dihydroxyphenylalanine (l-DOPA) ratios without changes in renal tubular aromatic-l-amino acid decarboxylase (AADC) activity. By contrast, the in vivo administration of RR (1.5 mg/kg sc) to KO mice was accompanied by significant decreases in plasma levels of Epi, DA, and l-DOPA as well as in urinary excretion of Epi, DA, and DA-to-l-DOPA ratios notwithstanding the accompanied increase in renal AADC activity. In addition, the increase in renal DA output observed in renalase KO mice was accompanied by an increase in the expression of the L-type amino acid transporter like (LAT) 1 that is reversed by the administration of RR in these animals. These results suggest that the overexpression of LAT1 in the renal cortex of the renalase KO mice might contribute to the enhanced l-DOPA availability/uptake and consequently to the activation of the renal dopaminergic system in the presence of renalase deficiency.
Collapse
Affiliation(s)
- Janete Quelhas-Santos
- Nephrology Research and Development Unit, Faculty of Medicine, University of Porto, Porto, Portugal; Nephrology and Infectious Diseases Research and Development Group, Instituto Nacional de Engenharia Biomédica-(I3S);
| | - Maria Paula Serrão
- Faculdade de Medicina da Universidade do Porto, Department of Pharmacology and Therapeutics, Porto, Portugal
| | - Isabel Soares-Silva
- Nephrology Research and Development Unit, Faculty of Medicine, University of Porto, Porto, Portugal; Nephrology and Infectious Diseases Research and Development Group, Instituto Nacional de Engenharia Biomédica-(I3S)
| | | | - Liliana Simões-Silva
- Nephrology Research and Development Unit, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Maria João Pinho
- Faculdade de Medicina da Universidade do Porto, Department of Pharmacology and Therapeutics, Porto, Portugal
| | - Fernando Remião
- Centro de Química da Universidade do Porto/Serviço de Toxicologia, Faculdade de Farmácia, University of Porto, Porto, Portugal
| | - Benedita Sampaio-Maia
- Nephrology Research and Development Unit, Faculty of Medicine, University of Porto, Porto, Portugal; Faculty of Dental Medicine, University of Porto, Porto, Portugal
| | - Gary V Desir
- Department of Medicine, Veterans Affairs Connecticut Healthcree System, Yale University, New Haven, Connecticut
| | - Manuel Pestana
- Nephrology and Infectious Diseases Research and Development Group, Instituto Nacional de Engenharia Biomédica-(I3S); Faculdade de Medicina da Universidade do Porto, Department of Renal, Urological, and Infectious Diseases, Porto, Portugal; and Department of Nephrology, São João Hospital Center, Entidade Pública Empresarial, Porto, Portugal
| |
Collapse
|
23
|
Abstract
The regulation of serum phosphate, an acknowledged risk factor for chronic kidney disease and cardiovascular mortality, is poorly understood. The discovery of fibroblast growth factor 23 (FGF23) as a key regulator of renal phosphate handling and activation of vitamin D has revolutionized our comprehension of phosphate homeostasis. Through as yet undetermined mechanisms, circulating and dietary phosphate appear to have a direct effect on FGF23 release by bone cells that, in turn, causes renal phosphate excretion and decreases intestinal phosphate absorption through a decrease in vitamin D production. Thus, the two major phosphaturic hormones, PTH and FGF23, have opposing effects on vitamin D production, placing vitamin D at the nexus of phosphate homeostasis. While our understanding of phosphate homeostasis has advanced, the factors determining regulation of serum phosphate level remain enigmatic. Diet, time of day, season, gender, age and genetics have all been identified as significant contributors to serum phosphate level. The effects of these factors on serum phosphate have major implications for what is understood as 'normal' and for studies of phosphate homeostasis and metabolism. Moreover, other hormonal mediators such as dopamine, insulin-like growth factor, and angiotensin II also affect renal handling of phosphate. How the major hormone effects on phosphate handling are regulated and how the effect of these other factors are integrated to yield the measurable serum phosphate are only now beginning to be studied.
Collapse
Affiliation(s)
- Eleanor Lederer
- Medical Services, Robley Rex VA Medical Center, Department of Medicine, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| |
Collapse
|