1
|
Kounatidis D, Vallianou NG, Stratigou T, Voukali M, Karampela I, Dalamaga M. The Kidney in Obesity: Current Evidence, Perspectives and Controversies. Curr Obes Rep 2024; 13:680-702. [PMID: 39141201 DOI: 10.1007/s13679-024-00583-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/01/2024] [Indexed: 08/15/2024]
Abstract
PURPOSE OF REVIEW As obesity and chronic kidney disease (CKD) remain a public health issue, we aim to elaborate on their complex relationship regarding pathogenetic mechanisms and therapeutic potential as well. The purpose of this review is to enhance our understanding of the interplay between obesity and CKD in order to timely diagnose and treat obesity-related CKD. RECENT FINDINGS Obesity and CKD pose significant intertwined challenges to global health, affecting a substantial portion of the population worldwide. Obesity is recognized as an independent risk factor, intricately contributing to CKD pathogenesis through mechanisms such as lipotoxicity, chronic inflammation, and insulin resistance. Recent evidence highlights additional factors including hemodynamic changes and intestinal dysbiosis that exacerbate kidney dysfunction in obese individuals, leading to histologic alterations known as obesity-related glomerulopathy (ORG). This narrative review synthesizes current knowledge on the prevalence, pathophysiology, clinical manifestations, and diagnostic strategies of obesity-related kidney disease. Furthermore, it explores mechanistic insights to delineate current therapeutic approaches, future directions for managing this condition and controversies. By elucidating the multifaceted interactions between obesity and kidney health, this review aims to inform clinical practice and stimulate further research to address this global health epidemic effectively.
Collapse
Affiliation(s)
- Dimitris Kounatidis
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, 11527, Athens, Greece
| | - Natalia G Vallianou
- First Department of Internal Medicine, Sismanogleio General Hospital, 15126, Athens, Greece.
| | - Theodora Stratigou
- Department of Endocrinology, Diabetes and Metabolism, European and National Expertise Center for Rare Endocrine Disorders, Evangelismos General Hospital, 10676, Athens, Greece
| | - Maria Voukali
- First Department of Internal Medicine, Sismanogleio General Hospital, 15126, Athens, Greece
| | - Irene Karampela
- Second Department of Critical Care, Medical School, Attikon General University Hospital, National and Kapodistrian University of Athens, 12462, Athens, Greece
| | - Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527, Athens, Greece
| |
Collapse
|
2
|
Ruan Y, Yu Y, Wu M, Jiang Y, Qiu Y, Ruan S. The renin-angiotensin-aldosterone system: An old tree sprouts new shoots. Cell Signal 2024; 124:111426. [PMID: 39306263 DOI: 10.1016/j.cellsig.2024.111426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 08/25/2024] [Accepted: 09/17/2024] [Indexed: 09/27/2024]
Abstract
The intricate physiological and pathological diversity of the Renin-Angiotensin-Aldosterone System (RAAS) underpins its role in maintaining bodily equilibrium. This paper delves into the classical axis (Renin-ACE-Ang II-AT1R axis), the protective arm (ACE2-Ang (1-7)-MasR axis), the prorenin-PRR-MAP kinases ERK1/2 axis, and the Ang IV-AT4R-IRAP cascade of RAAS, examining their functions in both physiological and pathological states. The dysregulation or hyperactivation of RAAS is intricately linked to numerous diseases, including cardiovascular disease (CVD), renal damage, metabolic disease, eye disease, Gastrointestinal disease, nervous system and reproductive system diseases. This paper explores the pathological mechanisms of RAAS in detail, highlighting its significant role in disease progression. Currently, in addition to traditional drugs like ACEI, ARB, and MRA, several novel therapeutics have emerged, such as angiotensin receptor-enkephalinase inhibitors, nonsteroidal mineralocorticoid receptor antagonists, aldosterone synthase inhibitors, aminopeptidase A inhibitors, and angiotensinogen inhibitors. These have shown potential efficacy and application prospects in various clinical trials for related diseases. Through an in-depth analysis of RAAS, this paper aims to provide crucial insights into its complex physiological and pathological mechanisms and offer valuable guidance for developing new therapeutic approaches. This comprehensive discussion is expected to advance the RAAS research field and provide innovative ideas and directions for future clinical treatment strategies.
Collapse
Affiliation(s)
- Yaqing Ruan
- The Affiliated People's Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou 350004, China; Fujian University of Traditional Chinese Medicine, Fuzhou 350000, China
| | - Yongxin Yu
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Meiqin Wu
- The Affiliated People's Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou 350004, China; Fujian University of Traditional Chinese Medicine, Fuzhou 350000, China
| | - Yulang Jiang
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yuliang Qiu
- The Affiliated People's Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou 350004, China; Fujian University of Traditional Chinese Medicine, Fuzhou 350000, China.
| | - Shiwei Ruan
- The Affiliated People's Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou 350004, China; Fujian University of Traditional Chinese Medicine, Fuzhou 350000, China.
| |
Collapse
|
3
|
Uhrbom M, Muhl L, Genové G, Liu J, Palmgren H, Alexandersson I, Karlsson F, Zhou AX, Lunnerdal S, Gustafsson S, Buyandelger B, Petkevicius K, Ahlstedt I, Karlsson D, Aasehaug L, He L, Jeansson M, Betsholtz C, Peng XR. Adipose stem cells are sexually dimorphic cells with dual roles as preadipocytes and resident fibroblasts. Nat Commun 2024; 15:7643. [PMID: 39223126 PMCID: PMC11369120 DOI: 10.1038/s41467-024-51867-9] [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: 07/31/2023] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
Cell identities are defined by intrinsic transcriptional networks and spatio-temporal environmental factors. Here, we explored multiple factors that contribute to the identity of adipose stem cells, including anatomic location, microvascular neighborhood, and sex. Our data suggest that adipose stem cells serve a dual role as adipocyte precursors and fibroblast-like cells that shape the adipose tissue's extracellular matrix in an organotypic manner. We further find that adipose stem cells display sexual dimorphism regarding genes involved in estrogen signaling, homeobox transcription factor expression and the renin-angiotensin-aldosterone system. These differences could be attributed to sex hormone effects, developmental origin, or both. Finally, our data demonstrate that adipose stem cells are distinct from mural cells, and that the state of commitment to adipogenic differentiation is linked to their anatomic position in the microvascular niche. Our work supports the importance of sex and microvascular function in adipose tissue physiology.
Collapse
Affiliation(s)
- Martin Uhrbom
- Department of Medicine, Huddinge, Karolinska Institutet Campus Flemingsberg, Neo building, 141 52, Huddinge, Sweden.
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden.
| | - Lars Muhl
- Department of Medicine, Huddinge, Karolinska Institutet Campus Flemingsberg, Neo building, 141 52, Huddinge, Sweden
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, University of Bergen, 5020, Bergen, Norway
| | - Guillem Genové
- Department of Medicine, Huddinge, Karolinska Institutet Campus Flemingsberg, Neo building, 141 52, Huddinge, Sweden
| | - Jianping Liu
- Department of Medicine, Huddinge, Karolinska Institutet Campus Flemingsberg, Neo building, 141 52, Huddinge, Sweden
| | - Henrik Palmgren
- Bioscience Renal, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Ida Alexandersson
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Fredrik Karlsson
- Data Sciences & Quantitative Biology, Discovery Sciences, R&D AstraZeneca, Gothenburg, Sweden
| | - Alex-Xianghua Zhou
- Bioscience Renal, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Sandra Lunnerdal
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Sonja Gustafsson
- Department of Medicine, Huddinge, Karolinska Institutet Campus Flemingsberg, Neo building, 141 52, Huddinge, Sweden
| | - Byambajav Buyandelger
- Department of Medicine, Huddinge, Karolinska Institutet Campus Flemingsberg, Neo building, 141 52, Huddinge, Sweden
| | - Kasparas Petkevicius
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Ingela Ahlstedt
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Daniel Karlsson
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Leif Aasehaug
- Bioscience Cardiovascular, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Liqun He
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 23, Uppsala, Sweden
| | - Marie Jeansson
- Department of Medicine, Huddinge, Karolinska Institutet Campus Flemingsberg, Neo building, 141 52, Huddinge, Sweden
| | - Christer Betsholtz
- Department of Medicine, Huddinge, Karolinska Institutet Campus Flemingsberg, Neo building, 141 52, Huddinge, Sweden.
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 23, Uppsala, Sweden.
| | - Xiao-Rong Peng
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden.
| |
Collapse
|
4
|
Zhao Y, Feng Y, Zeng Y, Di W, Luo X, Wu X, Guan R, Xu L, Yang X, Li Y, Wu Y, Wu X, Zhang Y, Li X, Qin P, Hu F, Hu D, Li H, Zhang M. Sugar intake and risk of hypertension: a systematic review and dose-response meta-analysis of cohort and cross-sectional studies. Crit Rev Food Sci Nutr 2024; 64:9483-9494. [PMID: 37218681 DOI: 10.1080/10408398.2023.2213330] [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] [Indexed: 05/24/2023]
Abstract
Several epidemiological studies have investigated the association between sugar intake, the levels of systolic blood pressure (SBP) and diastolic blood pressure (DBP) and the risk of hypertension, but findings have been inconsistent. We carried out a systematic review and meta-analysis of observational studies to examine the associations between sugar intake, hypertension risk, and BP levels. Articles published up to February 2, 2021 were sourced through PubMed, EMBASE and Web of Science. Pooled relative risks (RRs) and 95% confidence intervals (CIs) were estimated using a fixed- or random-effects model. Restricted cubic splines were used to evaluate dose-response associations. Overall, 35 studies were included in the present meta-analysis (23 for hypertension and 12 for BP). Sugar-sweetened beverages (SSBs) and artificially sweetened beverages (ASBs) were positively associated with hypertension risk: 1.26 (95% CI, 1.15-1.37) and 1.10 (1.07-1.13) per 250-g/day increment, respectively. For SBP, only SSBs were significant with a pooled β value of 0.24 mmHg (95% CI, 0.12-0.36) per 250 g increase. Fructose, sucrose, and added sugar, however, were shown to be associated with elevated DBP with 0.83 mmHg (0.07-1.59), 1.10 mmHg (0.12-2.08), and 5.15 mmHg (0.09-10.21), respectively. Current evidence supports the harmful effects of sugar intake for hypertension and BP level, especially SSBs, ASBs, and total sugar intake.
Collapse
Affiliation(s)
- Yang Zhao
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Medical School, Shenzhen, People's Republic of China
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Shenzhen University Medical School, Shenzhen, People's Republic of China
| | - Yifei Feng
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Medical School, Shenzhen, People's Republic of China
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Shenzhen University Medical School, Shenzhen, People's Republic of China
| | - Yunhong Zeng
- Center for Health Management, The Affiliated Shenzhen Hospital of University of Chinese Academy of Sciences, Shenzhen, People's Republic of China
| | - Wencheng Di
- Department of Cardiology, The Third People's Hospital of Shenzhen, Shenzhen, People's Republic of China
| | - Xinping Luo
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Medical School, Shenzhen, People's Republic of China
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Shenzhen University Medical School, Shenzhen, People's Republic of China
| | - Xiaojing Wu
- Department of Cardiology, Shenzhen University General Hospital, Shenzhen, People's Republic of China
| | - Ruiyun Guan
- Department of Pharmacy, Shenzhen University General Hospital, Shenzhen, People's Republic of China
| | - Lidan Xu
- Department of Nutrition, The Second Affiliated Hospital, Shenzhen University Medical School, Shenzhen, People's Republic of China
| | - Xingjin Yang
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, People's Republic of China
| | - Yang Li
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Medical School, Shenzhen, People's Republic of China
| | - Yuying Wu
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Medical School, Shenzhen, People's Republic of China
| | - Xiaoyan Wu
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Medical School, Shenzhen, People's Republic of China
| | - Yanyan Zhang
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Medical School, Shenzhen, People's Republic of China
| | - Xi Li
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, People's Republic of China
| | - Pei Qin
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Medical School, Shenzhen, People's Republic of China
| | - Fulan Hu
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Medical School, Shenzhen, People's Republic of China
| | - Dongsheng Hu
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Medical School, Shenzhen, People's Republic of China
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, People's Republic of China
- Department of General Practice, The Affiliated Luohu Hospital of Shenzhen University Medical School, Shenzhen, People's Republic of China
| | - Honghui Li
- Department of Endocrinology, The Affiliated Luohu Hospital of Shenzhen University Health Science Center, Shenzhen, People's Republic of China
| | - Ming Zhang
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Medical School, Shenzhen, People's Republic of China
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Shenzhen University Medical School, Shenzhen, People's Republic of China
| |
Collapse
|
5
|
Proença AB, Medeiros GR, Reis GDS, Losito LDF, Ferraz LM, Bargut TCL, Soares NP, Alexandre-Santos B, Campagnole-Santos MJ, Magliano DC, Nobrega ACLD, Santos RAS, Frantz EDC. Adipose tissue plasticity mediated by the counterregulatory axis of the renin-angiotensin system: Role of Mas and MrgD receptors. J Cell Physiol 2024; 239:e31265. [PMID: 38577921 DOI: 10.1002/jcp.31265] [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: 12/07/2023] [Revised: 02/28/2024] [Accepted: 03/18/2024] [Indexed: 04/06/2024]
Abstract
The renin-angiotensin system (RAS) is an endocrine system composed of two main axes: the classical and the counterregulatory, very often displaying opposing effects. The classical axis, primarily mediated by angiotensin receptors type 1 (AT1R), is linked to obesity-associated metabolic effects. On the other hand, the counterregulatory axis appears to exert antiobesity effects through the activation of two receptors, the G protein-coupled receptor (MasR) and Mas-related receptor type D (MrgD). The local RAS in adipose organ has prompted extensive research into white adipose tissue and brown adipose tissue (BAT), with a key role in regulating the cellular and metabolic plasticity of these tissues. The MasR activation favors the brown plasticity signature in the adipose organ by improve the thermogenesis, adipogenesis, and lipolysis, decrease the inflammatory state, and overall energy homeostasis. The MrgD metabolic effects are related to the maintenance of BAT functionality, but the signaling remains unexplored. This review provides a summary of RAS counterregulatory actions triggered by Mas and MrgD receptors on adipose tissue plasticity. Focus on the effects related to the morphology and function of adipose tissue, especially from animal studies, will be given targeting new avenues for treatment of obesity-associated metabolic effects.
Collapse
Affiliation(s)
- Ana Beatriz Proença
- Department of Physiology, Laboratory of Exercise Sciences, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
- Department of Morphology, Research Center on Morphology and Metabolism, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
| | - Gabriela Rodrigues Medeiros
- Department of Physiology, Laboratory of Exercise Sciences, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
- Department of Morphology, Research Center on Morphology and Metabolism, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
| | - Guilherme Dos Santos Reis
- Department of Physiology, Laboratory of Exercise Sciences, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
- Department of Morphology, Research Center on Morphology and Metabolism, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
| | - Luiza da França Losito
- Department of Physiology, Laboratory of Exercise Sciences, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
- Department of Morphology, Research Center on Morphology and Metabolism, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
| | - Luiza Mazzali Ferraz
- Department of Physiology, Laboratory of Exercise Sciences, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
- Department of Morphology, Research Center on Morphology and Metabolism, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
| | - Thereza Cristina Lonzetti Bargut
- Department of Basic Sciences, Nova Friburgo Health Institute, Fluminense Federal University, Nova Friburgo, Rio de Janeiro, Brazil
| | - Nícia Pedreira Soares
- Department of Physiology and Biophysics, National Institute of Science and Technology in Nanobiopharmaceutics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Beatriz Alexandre-Santos
- Department of Physiology, Laboratory of Exercise Sciences, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
- Department of Morphology, Research Center on Morphology and Metabolism, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
| | - Maria Jose Campagnole-Santos
- Department of Physiology and Biophysics, National Institute of Science and Technology in Nanobiopharmaceutics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - D'Angelo Carlo Magliano
- Department of Morphology, Research Center on Morphology and Metabolism, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
| | - Antonio Claudio Lucas da Nobrega
- Department of Physiology, Laboratory of Exercise Sciences, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
| | - Robson Augusto Souza Santos
- Department of Physiology and Biophysics, National Institute of Science and Technology in Nanobiopharmaceutics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Eliete Dalla Corte Frantz
- Department of Physiology, Laboratory of Exercise Sciences, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
- Department of Morphology, Research Center on Morphology and Metabolism, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
| |
Collapse
|
6
|
Takeda Y, Yoshikawa T, Dai P. Angiotensin II participates in mitochondrial thermogenic functions via the activation of glycolysis in chemically induced human brown adipocytes. Sci Rep 2024; 14:10789. [PMID: 38734719 PMCID: PMC11088625 DOI: 10.1038/s41598-024-61774-0] [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: 02/22/2024] [Accepted: 05/09/2024] [Indexed: 05/13/2024] Open
Abstract
Brown adipocytes are potential therapeutic targets for the prevention of obesity-associated metabolic diseases because they consume circulating glucose and fatty acids for heat production. Angiotensin II (Ang II) peptide is involved in the pathogenesis of obesity- and cold-induced hypertension; however, the mechanism underlying the direct effects of Ang II on human brown adipocytes remains unclear. Our transcriptome analysis of chemical compound-induced brown adipocytes (ciBAs) showed that the Ang II type 1 receptor (AGTR1), but not AGTR2 and MAS1 receptors, was expressed. The Ang II/AGTR1 axis downregulated the expression of mitochondrial uncoupling protein 1 (UCP1). The simultaneous treatment with β-adrenergic receptor agonists and Ang II attenuated UCP1 expression, triglyceride lipolysis, and cAMP levels, although cAMP response element-binding protein (CREB) phosphorylation was enhanced by Ang II mainly through the protein kinase C pathway. Despite reduced lipolysis, both coupled and uncoupled mitochondrial respiration was enhanced in Ang II-treated ciBAs. Instead, glycolysis and glucose uptake were robustly activated upon treatment with Ang II without a comprehensive transcriptional change in glucose metabolic genes. Elevated mitochondrial energy status induced by Ang II was likely associated with UCP1 repression. Our findings suggest that the Ang II/AGTR1 axis participates in mitochondrial thermogenic functions via glycolysis.
Collapse
Affiliation(s)
- Yukimasa Takeda
- Department of Cellular Regenerative Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan.
| | - Toshikazu Yoshikawa
- Louis Pasteur Center for Medical Research, 103-5 Tanaka-Monzen-cho, Sakyo-ku, Kyoto, 606-8225, Japan
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Ping Dai
- Department of Cellular Regenerative Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan.
| |
Collapse
|
7
|
Labandeira-Garcia JL, Labandeira CM, Guerra MJ, Rodriguez-Perez AI. The role of the brain renin-angiotensin system in Parkinson´s disease. Transl Neurodegener 2024; 13:22. [PMID: 38622720 PMCID: PMC11017622 DOI: 10.1186/s40035-024-00410-3] [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: 12/13/2023] [Accepted: 03/22/2024] [Indexed: 04/17/2024] Open
Abstract
The renin-angiotensin system (RAS) was classically considered a circulating hormonal system that regulates blood pressure. However, different tissues and organs, including the brain, have a local paracrine RAS. Mutual regulation between the dopaminergic system and RAS has been observed in several tissues. Dysregulation of these interactions leads to renal and cardiovascular diseases, as well as progression of dopaminergic neuron degeneration in a major brain center of dopamine/angiotensin interaction such as the nigrostriatal system. A decrease in the dopaminergic function induces upregulation of the angiotensin type-1 (AT1) receptor activity, leading to recovery of dopamine levels. However, AT1 receptor overactivity in dopaminergic neurons and microglial cells upregulates the cellular NADPH-oxidase-superoxide axis and Ca2+ release, which mediate several key events in oxidative stress, neuroinflammation, and α-synuclein aggregation, involved in Parkinson's disease (PD) pathogenesis. An intraneuronal antioxidative/anti-inflammatory RAS counteracts the effects of the pro-oxidative AT1 receptor overactivity. Consistent with this, an imbalance in RAS activity towards the pro-oxidative/pro-inflammatory AT1 receptor axis has been observed in the substantia nigra and striatum of several animal models of high vulnerability to dopaminergic degeneration. Interestingly, autoantibodies against angiotensin-converting enzyme 2 and AT1 receptors are increased in PD models and PD patients and contribute to blood-brain barrier (BBB) dysregulation and nigrostriatal pro-inflammatory RAS upregulation. Therapeutic strategies addressed to the modulation of brain RAS, by AT1 receptor blockers (ARBs) and/or activation of the antioxidative axis (AT2, Mas receptors), may be neuroprotective for individuals with a high risk of developing PD or in prodromal stages of PD to reduce progression of the disease.
Collapse
Affiliation(s)
- Jose Luis Labandeira-Garcia
- Cellular and Molecular Neurobiology of Parkinson´S Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain.
- Networking Research Center On Neurodegenerative Diseases (CIBERNED), Madrid, Spain.
| | | | - Maria J Guerra
- Cellular and Molecular Neurobiology of Parkinson´S Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
- Networking Research Center On Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Ana I Rodriguez-Perez
- Cellular and Molecular Neurobiology of Parkinson´S Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain.
- Networking Research Center On Neurodegenerative Diseases (CIBERNED), Madrid, Spain.
| |
Collapse
|
8
|
Pedrosa MA, Labandeira CM, Lago-Baameiro N, Valenzuela R, Pardo M, Labandeira-Garcia JL, Rodriguez-Perez AI. Extracellular Vesicles and Their Renin-Angiotensin Cargo as a Link between Metabolic Syndrome and Parkinson's Disease. Antioxidants (Basel) 2023; 12:2045. [PMID: 38136165 PMCID: PMC10741149 DOI: 10.3390/antiox12122045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 11/20/2023] [Accepted: 11/23/2023] [Indexed: 12/24/2023] Open
Abstract
Several studies showed an association between metabolic syndrome (MetS) and Parkinson's disease (PD). The linking mechanisms remain unclear. MetS promotes low-grade peripheral oxidative stress and inflammation and dysregulation of the adipose renin-angiotensin system (RAS). Interestingly, brain RAS dysregulation is involved in the progression of dopaminergic degeneration and PD. Circulating extracellular vesicles (EVs) from MetS fat tissue can cross the brain-blood barrier and may act as linking signals. We isolated and characterized EVs from MetS and control rats and analyzed their mRNA and protein cargo using RT-PCR and the ExoView R200 platform, respectively. Furthermore, cultures of the N27 dopaminergic cell line and the C6 astrocytic cell line were treated with EVs from MetS rats. EVs were highly increased in MetS rat serum, which was inhibited by treatment of the rats with the angiotensin type-1-receptor blocker candesartan. Furthermore, EVs from MetS rats showed increased pro-oxidative/pro-inflammatory and decreased anti-oxidative/anti-inflammatory RAS components, which were inhibited in candesartan-treated MetS rats. In cultures, EVs from MetS rats increased N27 cell death and modulated C6 cell function, upregulating markers of neuroinflammation and oxidative stress, which were inhibited by the pre-treatment of cultures with candesartan. The results from rat models suggest EVs and their RAS cargo as a mechanism linking Mets and PD.
Collapse
Affiliation(s)
- Maria A. Pedrosa
- Cellular and Molecular Neurobiology of Parkinson’s Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (M.A.P.); (R.V.)
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain
| | | | - Nerea Lago-Baameiro
- Grupo Obesidómica, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela/SERGAS, 15706 Santiago de Compostela, Spain; (N.L.-B.); (M.P.)
| | - Rita Valenzuela
- Cellular and Molecular Neurobiology of Parkinson’s Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (M.A.P.); (R.V.)
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain
| | - Maria Pardo
- Grupo Obesidómica, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela/SERGAS, 15706 Santiago de Compostela, Spain; (N.L.-B.); (M.P.)
- CIBER Fisiopatología Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Jose Luis Labandeira-Garcia
- Cellular and Molecular Neurobiology of Parkinson’s Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (M.A.P.); (R.V.)
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain
| | - Ana I. Rodriguez-Perez
- Cellular and Molecular Neurobiology of Parkinson’s Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (M.A.P.); (R.V.)
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain
| |
Collapse
|
9
|
Cheng X, Jiang S, Pan B, Xie W, Meng J. Ectopic and visceral fat deposition in aging, obesity, and idiopathic pulmonary fibrosis: an interconnected role. Lipids Health Dis 2023; 22:201. [PMID: 38001499 PMCID: PMC10668383 DOI: 10.1186/s12944-023-01964-3] [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: 09/25/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is considered an age-related disease. Age-related changes, along with other factors such as obesity, hormonal imbalances, and various metabolic disorders, lead to ectopic fat deposition (EFD). This accumulation of fat outside of its normal storage sites is associated with detrimental effects such as lipotoxicity, oxidative stress, inflammation, and insulin resistance. This narrative review provides an overview of the connection between ectopic and visceral fat deposition in aging, obesity, and IPF. It also elucidates the mechanism by which ectopic fat deposition in the airways and lungs, pericardium, skeletal muscles, and pancreas contributes to lung injury and fibrosis in patients with IPF, directly or indirectly. Moreover, the review discusses the impact of EFD on the severity of the disease, quality of life, presence of comorbidities, and overall prognosis in IPF patients. The review provides detailed information on recent research regarding representative lipid-lowering drugs, hypoglycemic drugs, and lipid-targeting drugs in animal experiments and clinical studies. This may offer new therapeutic directions for patients with IPF.
Collapse
Affiliation(s)
- Xiaoyun Cheng
- Department of Pulmonary and Critical Care Medicine, The Third Xiangya Hospital of Central South University, Tongzipo Road 138, Yuelu District, Changsha, 410000, China
- Hunan Key Laboratory of Organ Fibrosis, Tongzipo Road 138, Yuelu District, Changsha, 410000, China
| | - Shuhan Jiang
- Department of Pulmonary and Critical Care Medicine, The Third Xiangya Hospital of Central South University, Tongzipo Road 138, Yuelu District, Changsha, 410000, China
- Hunan Key Laboratory of Organ Fibrosis, Tongzipo Road 138, Yuelu District, Changsha, 410000, China
| | - Boyu Pan
- Departments of Orthopedics, The Third Hospital of Changsha, Laodong West Road 176, Tianxin District, Changsha, 410000, China
| | - Wei Xie
- Department of Cardiology, Xiangya Hospital of Central South University, Furong Middle Road 36, Kaifu District, Changsha, 410000, China
| | - Jie Meng
- Department of Pulmonary and Critical Care Medicine, The Third Xiangya Hospital of Central South University, Tongzipo Road 138, Yuelu District, Changsha, 410000, China.
- Hunan Key Laboratory of Organ Fibrosis, Tongzipo Road 138, Yuelu District, Changsha, 410000, China.
| |
Collapse
|
10
|
Ma JI, Zern EK, Parekh JK, Owunna N, Jiang N, Wang D, Rambarat PK, Pomerantsev E, Picard MH, Ho JE. Obesity Modifies Clinical Outcomes of Right Ventricular Dysfunction. Circ Heart Fail 2023; 16:e010524. [PMID: 37886836 PMCID: PMC10841712 DOI: 10.1161/circheartfailure.123.010524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Accepted: 08/18/2023] [Indexed: 10/28/2023]
Abstract
BACKGROUND Right ventricular (RV) dysfunction is associated with increased mortality across a spectrum of cardiovascular diseases. The role of obesity in RV dysfunction and adverse outcomes is unclear. METHODS We examined patients undergoing right heart catheterization between 2005 and 2016 in a hospital-based cohort. Linear regression was used to examine the association of obesity with hemodynamic indices of RV dysfunction (pulmonary artery pulsatility index, right atrial pressure:pulmonary capillary wedge pressure ratio, RV stroke work index). Cox models were used to examine the association of RV function measures with clinical outcomes. RESULTS Among 8285 patients (mean age, 63 years; 40% women), higher body mass index was associated with worse indices of RV dysfunction, including lower pulmonary artery pulsatility index (β, -0.23; SE, 0.01; P<0.001), higher right atrium:pulmonary capillary wedge pressure ratio (β, 0.25; SE, 0.01; P<0.001), and lower RV stroke work index (β, -0.05; SE, 0.01; P<0.001). Over median of 7.3 years of follow-up, we observed 3006 mortality and 2004 heart failure hospitalization events. RV dysfunction was associated with a greater risk of mortality (eg, pulmonary artery pulsatility index:hazard ratio, 1.11 per 1-SD increase [95% CI, 1.04-1.18]), with similar associations with risk of heart failure hospitalization. Body mass index modified the effect of RV dysfunction on all-cause mortality (Pinteraction≤0.005 for PAPi and RA:PCWP ratio), such that the effect of RV dysfunction was more pronounced at higher body mass index. CONCLUSIONS Patients with obesity had worse hemodynamic measured indices of RV function across a broad hospital-based sample. While RV dysfunction was associated with worse clinical outcomes including mortality and heart failure hospitalization, this association was especially pronounced among individuals with higher body mass index.
Collapse
Affiliation(s)
- Janet I. Ma
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Emily K. Zern
- Providence Heart Institute, Center for Cardiovascular Analytics, Research, and Data Science (CARDS), Providence St. Joseph Health, Portland, Oregon
| | - Juhi K. Parekh
- Cardiovascular Institute and Division of Cardiology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Ndidi Owunna
- Cardiovascular Institute and Division of Cardiology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Nona Jiang
- Cardiovascular Institute and Division of Cardiology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Dongyu Wang
- Cardiovascular Institute and Division of Cardiology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - Paula K. Rambarat
- Division of Cardiology, Duke University Medical Center, Durham, North Carolina
| | - Eugene Pomerantsev
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Michael H. Picard
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Jennifer E. Ho
- Cardiovascular Institute and Division of Cardiology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| |
Collapse
|
11
|
Woo H, Han A, Park JE, Cha YS. Korean fermented soybean paste (Doenjang) has anti-obesity and anti-hypertensive effects via the renin-angiotensin system (RAS) in high-fat diet-induced obese rats. PLoS One 2023; 18:e0291762. [PMID: 37862361 PMCID: PMC10588895 DOI: 10.1371/journal.pone.0291762] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 09/05/2023] [Indexed: 10/22/2023] Open
Abstract
The health-beneficial outcomes of doenjang, a Korean fermented food have been questioned due to its high salt content; moreover, the detailed underlying mechanisms of its health beneficial effects are not fully investigated. Thus, this study aimed to investigate doenjang's anti-obesity effects, anti-hypertensive effects, and its underlying mechanisms in high-fat diet -induced obesity. Sprague-Dawley rats fed with normal diet (ND), high-fat diet (HD), HD with 8% additive salt (HDS), or HD with doenjang containing 8% salt (HDJ) for 13 weeks. Compared to HD and HDS groups, the HDJ group had lower body and epididymal fat tissue weight gain and showed hypotrophy and hypoplasia. The RAS-related mRNA levels in the adipose tissue, including Renin and Ace were downregulated in the HDJ group compared to HD and HDS groups. Additionally, HDJ groups had significant improvements in systolic blood pressure, serum RAS-associated parameters (e.g., angiotensin II and aldosterone), renal mRNA levels related to RAS (e.g., angiotensin II receptor type 1 and 2), and aldosterone-associated mRNA expressions (e.g., mineralocorticoid receptor) in the kidney of HD-induced obese rats. Most importantly, HDS and HDJ groups showed distinct outcomes regarding adipogenesis and electrolytes metabolism, even though both diets contain a high level of salt. HDS group showed a higher epididymal fat tissue weight, mass, and adipocyte size than HDJ group. In addition, compared with HDJ group, HDS group significantly decreased the release of Na+ and K+ throughout the urine and feces. The present study addresses that doenjang has anti-obesity effects and anti-hypertensive effects by activating RAS in the adipose tissue and kidney, respectively. Additionally, this study also demonstrates that salt in doenjang and the additive salt differently influences adipogenesis and electrolytes metabolism, supporting doenjang has health advantageous effects regardless of its high salt contents.
Collapse
Affiliation(s)
- Hayoung Woo
- Department of Food Science and Human Nutrition, Jeonbuk National University, Jeonju-si, Jeollabuk-do, Republic of Korea
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT, United States of America
| | - Anna Han
- Department of Food Science and Human Nutrition, Jeonbuk National University, Jeonju-si, Jeollabuk-do, Republic of Korea
- K-Food Research Center, Jeonbuk National University, Jeonju-si, Jeollabuk-do, Republic of Korea
| | - Jung Eun Park
- Department of Food Science and Human Nutrition, Jeonbuk National University, Jeonju-si, Jeollabuk-do, Republic of Korea
- Nutracore Co., Ltd., Suwon, Republic of Korea
| | - Youn-Soo Cha
- Department of Food Science and Human Nutrition, Jeonbuk National University, Jeonju-si, Jeollabuk-do, Republic of Korea
- K-Food Research Center, Jeonbuk National University, Jeonju-si, Jeollabuk-do, Republic of Korea
| |
Collapse
|
12
|
Kreiner FF, Schytz PA, Heerspink HJL, von Scholten BJ, Idorn T. Obesity-Related Kidney Disease: Current Understanding and Future Perspectives. Biomedicines 2023; 11:2498. [PMID: 37760939 PMCID: PMC10526045 DOI: 10.3390/biomedicines11092498] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/07/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Obesity is a serious chronic disease and an independent risk factor for the new onset and progression of chronic kidney disease (CKD). CKD prevalence is expected to increase, at least partly due to the continuous rise in the prevalence of obesity. The concept of obesity-related kidney disease (OKD) has been introduced to describe the still incompletely understood interplay between obesity, CKD, and other cardiometabolic conditions, including risk factors for OKD and cardiovascular disease, such as diabetes and hypertension. Current therapeutics target obesity and CKD individually. Non-pharmacological interventions play a major part, but the efficacy and clinical applicability of lifestyle changes and metabolic surgery remain debatable, because the strategies do not benefit everyone, and it remains questionable whether lifestyle changes can be sustained in the long term. Pharmacological interventions, such as sodium-glucose co-transporter 2 inhibitors and the non-steroidal mineralocorticoid receptor antagonist finerenone, provide kidney protection but have limited or no impact on body weight. Medicines based on glucagon-like peptide-1 (GLP-1) induce clinically relevant weight loss and may also offer kidney benefits. An urgent medical need remains for investigations to better understand the intertwined pathophysiologies in OKD, paving the way for the best possible therapeutic strategies in this increasingly prevalent disease complex.
Collapse
Affiliation(s)
| | | | - Hiddo J. L. Heerspink
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, 9700 RB Groningen, The Netherlands;
| | | | - Thomas Idorn
- Novo Nordisk A/S, DK-2860 Søborg, Denmark; (F.F.K.); (P.A.S.); (B.J.v.S.)
| |
Collapse
|
13
|
Pan S, Worker CJ, Feng Earley Y. The hypothalamus as a key regulator of glucose homeostasis: emerging roles of the brain renin-angiotensin system. Am J Physiol Cell Physiol 2023; 325:C141-C154. [PMID: 37273237 PMCID: PMC10312332 DOI: 10.1152/ajpcell.00533.2022] [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: 11/29/2022] [Revised: 05/26/2023] [Accepted: 05/26/2023] [Indexed: 06/06/2023]
Abstract
The regulation of plasma glucose levels is a complex and multifactorial process involving a network of receptors and signaling pathways across numerous organs that act in concert to ensure homeostasis. However, much about the mechanisms and pathways by which the brain regulates glycemic homeostasis remains poorly understood. Understanding the precise mechanisms and circuits employed by the central nervous system to control glucose is critical to resolving the diabetes epidemic. The hypothalamus, a key integrative center within the central nervous system, has recently emerged as a critical site in the regulation of glucose homeostasis. Here, we review the current understanding of the role of the hypothalamus in regulating glucose homeostasis, with an emphasis on the paraventricular nucleus, the arcuate nucleus, the ventromedial hypothalamus, and lateral hypothalamus. In particular, we highlight the emerging role of the brain renin-angiotensin system in the hypothalamus in regulating energy expenditure and metabolic rate, as well as its potential importance in the regulation of glucose homeostasis.
Collapse
Affiliation(s)
- Shiyue Pan
- Department of Pharmacology, School of Medicine, University of Nevada, Reno, Reno, Nevada, United States
- Department of Physiology & Cell Biology, School of Medicine, University of Nevada, Reno, Reno, Nevada, United States
- Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada, United States
| | - Caleb J Worker
- Department of Pharmacology, School of Medicine, University of Nevada, Reno, Reno, Nevada, United States
- Department of Physiology & Cell Biology, School of Medicine, University of Nevada, Reno, Reno, Nevada, United States
- Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada, United States
| | - Yumei Feng Earley
- Department of Pharmacology, School of Medicine, University of Nevada, Reno, Reno, Nevada, United States
- Department of Physiology & Cell Biology, School of Medicine, University of Nevada, Reno, Reno, Nevada, United States
- Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada, United States
| |
Collapse
|
14
|
Nemoto W, Yamagata R, Nakagawasai O, Tan-No K. Angiotensin-Related Peptides and Their Role in Pain Regulation. BIOLOGY 2023; 12:biology12050755. [PMID: 37237567 DOI: 10.3390/biology12050755] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 05/19/2023] [Accepted: 05/20/2023] [Indexed: 05/28/2023]
Abstract
Angiotensin (Ang)-generating system has been confirmed to play an important role in the regulation of fluid balance and blood pressure and is essential for the maintenance of biological functions. Ang-related peptides and their receptors are found throughout the body and exhibit diverse physiological effects. Accordingly, elucidating novel physiological roles of Ang-generating system has attracted considerable research attention worldwide. Ang-generating system consists of the classical Ang-converting enzyme (ACE)/Ang II/AT1 or AT2 receptor axis and the ACE2/Ang (1-7)/MAS1 receptor axis, which negatively regulates AT1 receptor-mediated responses. These Ang system components are expressed in various tissues and organs, forming a local Ang-generating system. Recent findings indicate that changes in the expression of Ang system components under pathological conditions are involved in the development of neuropathy, inflammation, and their associated pain. Here, we summarized the effects of changes in the Ang system on pain transmission in various organs and tissues involved in pain development process.
Collapse
Affiliation(s)
- Wataru Nemoto
- Division of Pharmacology, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, Sendai 981-8558, Japan
| | - Ryota Yamagata
- Division of Pharmacology, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, Sendai 981-8558, Japan
| | - Osamu Nakagawasai
- Division of Pharmacology, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, Sendai 981-8558, Japan
| | - Koichi Tan-No
- Division of Pharmacology, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, Sendai 981-8558, Japan
| |
Collapse
|
15
|
Ashkar F, Bhullar KS, Jiang X, Wu J. Tripeptide IRW Improves AMPK/eNOS Signaling Pathway via Activating ACE2 in the Aorta of High-Fat-Diet-Fed C57BL/6 Mice. BIOLOGY 2023; 12:biology12040556. [PMID: 37106756 PMCID: PMC10135585 DOI: 10.3390/biology12040556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/30/2023] [Accepted: 04/04/2023] [Indexed: 04/08/2023]
Abstract
This study aims to investigate the effect of tripeptide IRW on the local renin–angiotensin system (RAS), particularly angiotensin-converting enzyme 2 (ACE2), and their association with signaling pathways in the aorta of a high-fat-diet (HFD)-induced insulin-resistant mouse model. C57BL/6 mice were fed HFD (45% of the total calories) for six weeks, and then IRW was added to the diet (45 mg/kg body weight (BW)) for another eight weeks. ACE2 mRNA expression and protein level(s) were increased (p < 0.05), while angiotensin II receptor (AT1R) and angiotensin-converting enzyme (ACE) protein abundance was significantly reduced (p < 0.05) in the aorta of HFD mice treated by IRW. IRW supplementation also improved glucose transporter 4 (GLUT4) abundance (p < 0.05) alongside AMP-activated protein kinase (AMPK) (p < 0.05), Sirtuin 1 (SIRT1) (p < 0.05), and endothelial nitric oxide synthase (eNOS) (p < 0.05) expression. IRW downregulated the levels of endothelin 1 (ET-1) and p38 mitogen-activated protein kinases (p38 MAPK, p < 0.05). Furthermore, the levels of AMPK and eNOS in vascular smooth muscle cells (VSMCs) were significantly reduced in ACE2 knockdown cells treated with or without IRW (p < 0.01). In conclusion, this study provided new evidence of the regulatory role of IRW on the aortic ACE2 against metabolic syndrome (MetS) in an HFD-induced insulin-resistant model.
Collapse
Affiliation(s)
- Fatemeh Ashkar
- Department of Agricultural Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Khushwant S. Bhullar
- Department of Agricultural Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Xu Jiang
- Department of Agricultural Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Jianping Wu
- Department of Agricultural Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2R3, Canada
| |
Collapse
|
16
|
Barrea L, Verde L, Camajani E, Šojat AS, Marina L, Savastano S, Colao A, Caprio M, Muscogiuri G. Effects of very low-calorie ketogenic diet on hypothalamic-pituitary-adrenal axis and renin-angiotensin-aldosterone system. J Endocrinol Invest 2023:10.1007/s40618-023-02068-6. [PMID: 37017918 DOI: 10.1007/s40618-023-02068-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 03/10/2023] [Indexed: 04/06/2023]
Abstract
BACKGROUND The hypothalamic-pituitary-adrenal (HPA) axis is a neuroendocrine system involved in controlling stress responses in humans under physiological and pathological conditions; cortisol is the main hormone produced by the HPA axis. It is known that calorie restriction acts as a stressor and can lead to an increase in cortisol production. Renin-angiotensin-aldosterone system (RAAS) is a complex endocrine network regulating blood pressure and hydrosaline metabolism, whose final hormonal effector is aldosterone. RAAS activation is linked to cardiometabolic diseases, such as heart failure and obesity. Obesity has become a leading worldwide pandemic, associated with serious health outcomes. Calorie restriction represents a pivotal strategy to tackle obesity. On the other hand, it is well known that an increased activity of the HPA may favour visceral adipose tissue expansion, which may jeopardize a successful diet-induced weight loss. Very low-calorie ketogenic diet (VLCKD) is a normoprotein diet with a drastic reduction of the carbohydrate content and total calorie intake. Thanks to its sustained protein content, VLCKD is extremely effective to reduce adipose tissue while preserving lean body mass and resting metabolic rate. PURPOSE The purpose of this narrative review is to gain more insights on the effects of VLCKD on the HPA axis and RAAS, in different phases of weight loss and in different clinical settings.
Collapse
Affiliation(s)
- L Barrea
- Dipartimento di Scienze Umanistiche, Università Telematica Pegaso, Via Porzio, Centro Direzionale, Isola F2, 80143, Naples, Italy
- Department of Clinical Medicine and Surgery, Endocrinology Unit, Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), University Medical School of Naples, Via Sergio Pansini 5, 80131, Naples, Italy
| | - L Verde
- Department of Clinical Medicine and Surgery, Endocrinology Unit, Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), University Medical School of Naples, Via Sergio Pansini 5, 80131, Naples, Italy
- Department of Public Health, Federico II University, Naples, Italy
| | - E Camajani
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166, Rome, Italy
| | - A S Šojat
- Department for Obesity, Metabolic and Reproductive Disorders, Clinic for Endocrinology, Diabetes and Metabolic Diseases, University Clinical Centre of Serbia, Belgrade, Serbia
| | - L Marina
- Department for Obesity, Metabolic and Reproductive Disorders, Clinic for Endocrinology, Diabetes and Metabolic Diseases, University Clinical Centre of Serbia, Belgrade, Serbia
| | - S Savastano
- Department of Clinical Medicine and Surgery, Endocrinology Unit, Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), University Medical School of Naples, Via Sergio Pansini 5, 80131, Naples, Italy
- Dipartimento di Medicina Clinica e Chirurgia, Diabetologia ed Andrologia, Unità di Endocrinologia, Università Federico II, Via Sergio Pansini 5, 80131, Naples, Italy
| | - A Colao
- Department of Clinical Medicine and Surgery, Endocrinology Unit, Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), University Medical School of Naples, Via Sergio Pansini 5, 80131, Naples, Italy
- Dipartimento di Medicina Clinica e Chirurgia, Diabetologia ed Andrologia, Unità di Endocrinologia, Università Federico II, Via Sergio Pansini 5, 80131, Naples, Italy
- Cattedra Unesco "Educazione Alla Salute E Allo Sviluppo Sostenibile", University Federico II, Naples, Italy
| | - M Caprio
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166, Rome, Italy
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Roma, 00166, Rome, Italy
| | - G Muscogiuri
- Department of Clinical Medicine and Surgery, Endocrinology Unit, Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), University Medical School of Naples, Via Sergio Pansini 5, 80131, Naples, Italy.
- Dipartimento di Medicina Clinica e Chirurgia, Diabetologia ed Andrologia, Unità di Endocrinologia, Università Federico II, Via Sergio Pansini 5, 80131, Naples, Italy.
- Cattedra Unesco "Educazione Alla Salute E Allo Sviluppo Sostenibile", University Federico II, Naples, Italy.
| |
Collapse
|
17
|
Guimarães VHD, Marinho BM, Motta-Santos D, Mendes GDRL, Santos SHS. Nutritional implications in the mechanistic link between the intestinal microbiome, renin-angiotensin system, and the development of obesity and metabolic syndrome. J Nutr Biochem 2023; 113:109252. [PMID: 36509338 DOI: 10.1016/j.jnutbio.2022.109252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 11/12/2022] [Accepted: 12/07/2022] [Indexed: 12/13/2022]
Abstract
Obesity and metabolic disorders represent a significant global health problem and the gut microbiota plays an important role in modulating systemic homeostasis. Recent evidence shows that microbiota and its signaling pathways may affect the whole metabolism and the Renin-Angiotensin System (RAS), which in turn seems to modify microbiota. The present review aimed to investigate nutritional implications in the mechanistic link between the intestinal microbiome, renin-angiotensin system, and the development of obesity and metabolic syndrome components. A description of metabolic changes was obtained based on relevant scientific literature. The molecular and physiological mechanisms that impact the human microbiome were addressed, including the gut microbiota associated with obesity, diabetes, and hepatic steatosis. The RAS interaction signaling and modulation were analyzed. Strategies including the use of prebiotics, symbiotics, probiotics, and biotechnology may affect the gut microbiota and its impact on human health.
Collapse
Affiliation(s)
- Victor Hugo Dantas Guimarães
- Laboratory of Health Science, Postgraduate Program in Health Science, Universidade Estadual de Montes Claros (Unimontes), Montes Claros, Minas Gerais, Brazil
| | - Barbhara Mota Marinho
- Laboratory of Health Science, Postgraduate Program in Health Science, Universidade Estadual de Montes Claros (Unimontes), Montes Claros, Minas Gerais, Brazil
| | - Daisy Motta-Santos
- School of Physical Education, Physiotherapy, and Occupational Therapy - EEFFTO, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | - Gabriela da Rocha Lemos Mendes
- Food Engineering, Institute of Agricultural Sciences (ICA), Universidade Federal de Minas Gerais (UFMG), Montes Claros, Minas Gerais, Brazil
| | - Sérgio Henrique Sousa Santos
- Laboratory of Health Science, Postgraduate Program in Health Science, Universidade Estadual de Montes Claros (Unimontes), Montes Claros, Minas Gerais, Brazil; Food Engineering, Institute of Agricultural Sciences (ICA), Universidade Federal de Minas Gerais (UFMG), Montes Claros, Minas Gerais, Brazil.
| |
Collapse
|
18
|
Ma JI, Zern E, Jiang N, Wang D, Rambarat P, Pomerantsev E, Picard MH, Ho JE. Obesity Modifies Clinical Outcomes of Right Ventricular Dysfunction. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.01.18.23284734. [PMID: 36711542 PMCID: PMC9882441 DOI: 10.1101/2023.01.18.23284734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Introduction Right ventricular (RV) dysfunction is associated with increased mortality across a spectrum of cardiovascular diseases. The role of obesity in RV dysfunction and adverse outcomes is unclear. Methods We examined patients undergoing right heart catheterization between 2005-2016 in a hospital-based cohort. Linear regression was used to examine the association of obesity with hemodynamic indices of RV dysfunction [pulmonary artery pulsatility index (PAPi), right atrial pressure: pulmonary capillary wedge pressure ratio (RAP:PCWP), RV stroke work index (RVSWI)]. Cox models were used to examine the association of RV function measures with clinical outcomes. Results Among 8285 patients (mean age 63 years, 40% women), higher BMI was associated with worse indices of RV dysfunction, including lower PAPi (β -0.26, SE 0.01, p <0.001), higher RA:PCWP ratio (β 0.25, SE 0.01, p-value <0.001), and lower RVSWI (β -0.05, SE 0.01, p-value <0.001). Over 7.3 years of follow-up, we observed 3006 mortality and 2004 heart failure (HF) hospitalization events. RV dysfunction was associated with greater risk of mortality (eg PAPi: HR 1.11 per 1-SD increase, 95% CI 1.04-1.18), with similar associations with risk of HF hospitalization. BMI modified the effect of RV dysfunction on outcomes (P-interaction <=0.005 for both), such that the effect of RV dysfunction was more pronounced at higher BMI. Conclusions Patients with obesity had worse hemodynamic measured indices of RV function across a broad hospital-based sample. While RV dysfunction was associated with worse clinical outcomes including mortality and HF hospitalization, this association was especially pronounced among individuals with higher BMI.
Collapse
|
19
|
Teles MC, Oliveira Portes AM, Campos Coelho BI, Resende LT, Isoldi MC. Cardiac changes in spontaneously hypertensive rats: Modulation by aerobic exercise. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2023; 177:109-124. [PMID: 36347337 DOI: 10.1016/j.pbiomolbio.2022.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 10/10/2022] [Accepted: 11/01/2022] [Indexed: 11/08/2022]
Abstract
Systemic arterial hypertension is a multifactorial clinical condition characterized by high and sustained levels of blood pressure. For a better understanding of the pathophysiology of hypertension, studies are conducted with spontaneously hypertensive animals, which allow the investigation of physiological changes that in most cases cannot be studied in humans. In these animals, myocardial remodeling, increased pro-inflammatory markers, redox imbalance and contractile dysfunctions that lead to changes in cardiac function can be observed. However, it can be inferring that aerobic training improves cardiac function and cardiomyocyte contractility, in addition to controlling inflammation and reducing oxidative stress in cardiac muscle, despite this, the precise mechanisms by which physical exercise improves cardiovascular control are not fully understood. In this review, we provide an overview of the pathophysiological changes that affect the heart of spontaneously hypertensive animals and their modulation by aerobic exercise.
Collapse
Affiliation(s)
- Maria Cecília Teles
- Laboratory of Cell Signaling, Department Pharmacy, Federal University of Ouro Preto, Ouro Preto, 35400-000, MG, Brazil.
| | | | - Bianca Iara Campos Coelho
- Laboratory of Cell Signaling, Department Nutrition, Federal University of Ouro Preto, Ouro Preto, 35400-000, MG, Brazil
| | - Letícia Teresinha Resende
- Laboratory of Cell Signaling, Department of General Biology, Federal University of Ouro Preto, Ouro Preto, 35400-000, MG, Brazil
| | - Mauro Cesar Isoldi
- Laboratory of Cell Signaling, Department of General Biology, Federal University of Ouro Preto, Ouro Preto, 35400-000, MG, Brazil
| |
Collapse
|
20
|
Abstract
The prevalence of obesity has increased dramatically during the past decades, which has been a major health problem. Since 1975, the number of people with obesity worldwide has nearly tripled. An increasing number of studies find obesity as a driver of chronic kidney disease (CKD) progression, and the mechanisms are complex and include hemodynamic changes, inflammation, oxidative stress, and activation of the renin-angiotensin-aldosterone system (RAAS). Obesity-related kidney disease is characterized by glomerulomegaly, which is often accompanied by localized and segmental glomerulosclerosis lesions. In these patients, the early symptoms are atypical, with microproteinuria being the main clinical manifestation and nephrotic syndrome being rare. Weight loss and RAAS blockers have a protective effect on obesity-related CKD, but even so, a significant proportion of patients eventually progress to end-stage renal disease despite treatment. Thus, it is critical to comprehend the mechanisms underlying obesity-related CKD to create new tactics for slowing or stopping disease progression. In this review, we summarize current knowledge on the mechanisms of obesity-related kidney disease, its pathological changes, and future perspectives on its treatment.
Collapse
Affiliation(s)
- Zongmiao Jiang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, China
| | - Yao Wang
- Department of Orthopedics, The Second Hospital Jilin University, Changchun, China
| | - Xue Zhao
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, China
| | - Haiying Cui
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, China
| | - Mingyue Han
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, China
| | - Xinhua Ren
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, China
| | - Xiaokun Gang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, China
| | - Guixia Wang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
21
|
Da Eira D, Jani S, Stefanovic M, Ceddia RB. Obesogenic versus ketogenic diets in the regulation of the renin-angiotensin system in rat white and brown adipose tissues. Nutrition 2023; 105:111862. [PMID: 36356378 DOI: 10.1016/j.nut.2022.111862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 06/13/2022] [Accepted: 09/26/2022] [Indexed: 11/09/2022]
Abstract
OBJECTIVE The ketogenic diet (KD) has been reported to reverse metabolic dysfunction in obesity. However, it remains unknown how the KD affects the balance between the classical and counterregulatory renin-angiotensin system (RAS) arms in adipose tissue, which carries important implications for metabolic function in adipocytes. The aim of this study was to compare the effects of the obesogenic diet and the KD on RAS balance in white and brown fat. METHODS Nine male Wistar rats were fed a standard chow (SC), 11 fed a high-fat sucrose-enriched (HFS) obesogenic diet, and 12 a KD. At the end of the 8-wk feeding period, subcutaneous inguinal (Sc Ing), epididymal (Epid), and interscapular brown adipose tissue (iBAT) fat depots were extracted and subsequently used for the measurement of RAS proteins and MasR gene expression. RESULTS In SC-fed rats, the Sc Ing fat displayed the highest levels of angiotensin-converting enzyme (ACE)1, but very low levels of angiotensin II types 1 and 2 receptors (AT1R and AT2R) and ACE2. Conversely, the highest levels of ACE2, AT1R, and AT2R were found in iBAT. The HFS diet increased AT1R protein in Sc Ing fat and iBAT, whereas the KD maintained low AT1R levels in these fat depots. However, in Sc Ing and Epid fat depots, the KD elevated AT2R levels and significantly reduced Epid ACE1 levels. CONCLUSION Despite fat depot-specific differences in RAS components, the obesogenic diet promoted the classical RAS arm, whereas the KD attenuated it and enhanced the counterregulatory arm.
Collapse
Affiliation(s)
- Daniel Da Eira
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, North York, Ontario, Canada
| | - Shailee Jani
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, North York, Ontario, Canada
| | - Mateja Stefanovic
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, North York, Ontario, Canada
| | - Rolando B Ceddia
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, North York, Ontario, Canada.
| |
Collapse
|
22
|
Effects of captopril on glucose metabolism and autophagy in liver and muscle from mice with type 1 diabetes and diet-induced obesity. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166477. [PMID: 35780942 DOI: 10.1016/j.bbadis.2022.166477] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 06/17/2022] [Accepted: 06/24/2022] [Indexed: 11/23/2022]
Abstract
Impaired metabolic functions underlie the pathophysiology of diabetes and obesity. The renin-angiotensin system (RAS) is one pathway related to the pathophysiology of both diseases. RAS activation in metabolically active tissues exerts pro-inflammatory effects via angiotensin II (Ang II), linked to dysfunction in cellular processes such as autophagy, which is associated with obesity and diabetes. Here, we determined whether RAS is involved in metabolic dysregulations in a Type 1 Diabetes (T1D) mouse model, treated with captopril, and in an obesity mouse model (Agt-Tg) that overexpresses angiotensinogen (Agt) in adipose tissue. T1D mice had lower plasma leptin, resistin and higher non-esterified fatty acids (NEFA) compared to wild type (Wt) mice, even under captopril treatment. Further, mRNA levels for Agt, At1, Insr, and Beclin1 were upregulated in muscle and liver of T1D mice with captopril compared to Wt. Moreover, autophagy markers LC3 and p62 proteins were decreased, regardless of captopril treatment in the liver from T1D mice. In obese Wt mice, captopril increased muscle Irs1 gene levels. Further, captopril reduced mRNA levels of At1, Insr, Ampk, Beclin1, Atg12, and Lc3 in the liver from both Wt and Agt-Tg mice, while Agt, At1, Insr, and Atg12 expression was reduced in Agt-Tg mice without captopril treatment. Irs1 expression was decreased in the liver from obese Wt mice treated with captopril. Our results suggest that captopril treatment upregulates components of RAS, insulin signaling, and autophagy in both muscle and liver, indicating potential utility of captopril in targeting both insulin sensitivity and autophagy in diabetes and obesity.
Collapse
|
23
|
Wu J, Wang M, Han L, Zhang H, Lei S, Zhang Y, Mo X. RNA modification-related variants in genomic loci associated with body mass index. Hum Genomics 2022; 16:25. [PMID: 35879730 PMCID: PMC9316745 DOI: 10.1186/s40246-022-00403-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 07/16/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Genome-wide association studies (GWASs) have identified hundreds of loci for body mass index (BMI), but functional variants in these loci are less known. The purpose of this study was to identify RNA modification-related SNPs (RNAm-SNPs) for BMI in GWAS loci. BMI-associated RNAm-SNPs were identified in a GWAS of approximately 700,000 individuals. Gene expression and circulating protein levels affected by the RNAm-SNPs were identified by QTL analyses. Mendelian randomization (MR) methods were applied to test whether the gene expression and protein levels were associated with BMI. RESULTS A total of 78 RNAm-SNPs associated with BMI (P < 5.0 × 10-8) were identified, including 65 m6A-, 10 m1A-, 3 m7G- and 1 A-to-I-related SNPs. Two functional loss, high confidence level m6A-SNPs, rs6713978 (P = 6.4 × 10-60) and rs13410999 (P = 8.2 × 10-59), in the intron of ADCY3 were the top significant SNPs. These two RNAm-SNPs were associated with ADCY3 gene expression in adipose tissues, whole blood cells, the tibial nerve, the tibial artery and lymphocytes, and the expression levels in these tissues were associated with BMI. Proteins enriched in specific KEGG pathways, such as natural killer cell-mediated cytotoxicity, the Rap1 signaling pathway and the Ras signaling pathway, were affected by the RNAm-SNPs, and circulating levels of some of these proteins (ADH1B, DOCK9, MICB, PRDM1, STOM, TMPRSS11D and TXNDC12) were associated with BMI in MR analyses. CONCLUSIONS Our study identified RNAm-SNPs in BMI-related genomic loci and suggested that RNA modification may affect BMI by affecting the expression levels of corresponding genes and proteins.
Collapse
Affiliation(s)
- Jingyun Wu
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Department of Epidemiology, School of Public Health, Soochow University, 199 Renai Road, Suzhou, 215123, Jiangsu, People's Republic of China.,Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Center for Genetic Epidemiology and Genomics, School of Public Health, Soochow University, Suzhou, China
| | - Mimi Wang
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Department of Epidemiology, School of Public Health, Soochow University, 199 Renai Road, Suzhou, 215123, Jiangsu, People's Republic of China.,Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Center for Genetic Epidemiology and Genomics, School of Public Health, Soochow University, Suzhou, China
| | - Limin Han
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Department of Epidemiology, School of Public Health, Soochow University, 199 Renai Road, Suzhou, 215123, Jiangsu, People's Republic of China.,Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Center for Genetic Epidemiology and Genomics, School of Public Health, Soochow University, Suzhou, China
| | - Huan Zhang
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Department of Epidemiology, School of Public Health, Soochow University, 199 Renai Road, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Shufeng Lei
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Department of Epidemiology, School of Public Health, Soochow University, 199 Renai Road, Suzhou, 215123, Jiangsu, People's Republic of China.,Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Center for Genetic Epidemiology and Genomics, School of Public Health, Soochow University, Suzhou, China
| | - Yonghong Zhang
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Department of Epidemiology, School of Public Health, Soochow University, 199 Renai Road, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Xingbo Mo
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Department of Epidemiology, School of Public Health, Soochow University, 199 Renai Road, Suzhou, 215123, Jiangsu, People's Republic of China. .,Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Center for Genetic Epidemiology and Genomics, School of Public Health, Soochow University, Suzhou, China.
| |
Collapse
|
24
|
Detection of SARS-CoV-2 in subcutaneous fat but not visceral fat, and the disruption of fat lymphocyte homeostasis in both fat tissues in the macaque. Commun Biol 2022; 5:542. [PMID: 35661814 PMCID: PMC9166782 DOI: 10.1038/s42003-022-03503-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 05/19/2022] [Indexed: 12/13/2022] Open
Abstract
The well documented association between obesity and the severity of SARS-CoV-2 infection raises the question of whether adipose tissue (AT) is impacted during this infection. Using a model of SARS-CoV-2 infection in cynomolgus macaques, we detected the virus within subcutaneous AT (SCAT) but not in visceral AT (VAT) or epicardial AT on day 7 post-infection. We sought to determine the mechanisms responsible for this selective detection and observed higher levels of angiotensin-converting-enzyme-2 mRNA expression in SCAT than in VAT. Lastly, we evaluated the immunological consequences of SARS-CoV-2 infection on AT: both SCAT and VAT T cells showed a drastic reduction in CD69 expression, a standard marker of resident memory T cell in tissue, that is also involved in the migratory and metabolic properties of T cells. Our results demonstrate that in a model of mild infection, SCAT is selectively infected by SARS-CoV-2 although changes in the immune properties of AT are observed in both SCAT and VAT. Subcutaneous fat tissue expresses higher angiotensin-converting-enzyme 2 mRNA than visceral fat tissue and is selectively infected by SARS-Cov-2, while both fat tissues show drastic reduction in CD69 expression in T cells.
Collapse
|
25
|
Pan C, Yang C, Ma Y, Sheng H, Lei Z, Wang S, Hu H, Feng X, Zhang J, Ma Y. Identification of Key Genes Associated With Early Calf-Hood Nutrition in Subcutaneous and Visceral Adipose Tissues by Co-Expression Analysis. Front Vet Sci 2022; 9:831129. [PMID: 35619603 PMCID: PMC9127810 DOI: 10.3389/fvets.2022.831129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 03/29/2022] [Indexed: 12/31/2022] Open
Abstract
Background Substantive evidence has confirmed that nutrition state is associated with health risk and the onset of pubertal and metabolic profile. Due to heterogeneity, adipose tissues in different anatomical positions tend to show various metabolic mechanisms for nutrition. To date, the complicated molecular mechanisms of early calf-hood nutrition on bovine adipose tissue are still largely unknown. This study aimed to identify key genes and functionally enriched pathways associated with early calf-hood nutrition in visceral and subcutaneous adipose tissue. Results The RNA-seq data of visceral and subcutaneous adipose tissues of calves feeding on low and high dietary nutrition for more than 100 days were downloaded and analyzed by weighted gene co-expression network analysis (WGCNA). Two modules that positively associated with a low plane of nutrition diet and two modules with a high plane of nutrition diet were identified in the subcutaneous adipose tissue. The blue and yellow modules, most closely associated with low and high nutrition, were selected for the functional enrichment analysis and exploration of hub genes. The results showed that genes in the blue module were significantly enriched in pathways that related to fat metabolism, reproduction, and cell communication. Genes in the yellow module were enriched in pathways related to fat metabolism, reproduction, cell proliferation, and senescence. Meanwhile, the blue and brown modules in visceral adipose tissue were most closely associated with low and high nutrition, respectively. Notably, genes of the blue module were significantly enriched in pathways related to substance metabolism, and genes in the brown module were significantly enriched in energy metabolism and disease pathways. Finally, key genes in subcutaneous adipose tissue for low nutrition (PLCG1, GNA11, and ANXA5) and high nutrition (BUB1B, ASPM, RRM2, PBK, NCAPG, and MKI67), and visceral adipose tissue for low nutrition (RPS5, RPL4, RPL14, and RPLP0) and high nutrition (SDHA and AKT1) were obtained and verified. Conclusion The study applied WGCNA to identify hub genes and functionally enriched pathways in subcutaneous and visceral adipose tissue and provided a basis for studying the effect of early calf-hood nutrition on the two adipose tissue types.
Collapse
|
26
|
Remodeling and Fibrosis of the Cardiac Muscle in the Course of Obesity-Pathogenesis and Involvement of the Extracellular Matrix. Int J Mol Sci 2022; 23:ijms23084195. [PMID: 35457013 PMCID: PMC9032681 DOI: 10.3390/ijms23084195] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 04/09/2022] [Indexed: 12/16/2022] Open
Abstract
Obesity is a growing epidemiological problem, as two-thirds of the adult population are carrying excess weight. It is a risk factor for the development of cardiovascular diseases (hypertension, ischemic heart disease, myocardial infarct, and atrial fibrillation). It has also been shown that chronic obesity in people may be a cause for the development of heart failure with preserved ejection fraction (HFpEF), whose components include cellular hypertrophy, left ventricular diastolic dysfunction, and increased extracellular collagen deposition. Several animal models with induced obesity, via the administration of a high-fat diet, also developed increased heart fibrosis as a result of extracellular collagen accumulation. Excessive collagen deposition in the extracellular matrix (ECM) in the course of obesity may increase the stiffness of the myocardium and thereby deteriorate the heart diastolic function and facilitate the occurrence of HFpEF. In this review, we include a rationale for that process, including a discussion about possible putative factors (such as increased renin–angiotensin–aldosterone activity, sympathetic overdrive, hemodynamic alterations, hypoadiponectinemia, hyperleptinemia, and concomitant heart diseases). To address the topic clearly, we include a description of the fundamentals of ECM turnover, as well as a summary of studies assessing collagen deposition in obese individuals.
Collapse
|
27
|
Kalenga CZ, Ramesh S, Dumanski SM, MacRae JM, Nerenberg K, Metcalfe A, Sola DY, Ahmed SB. Sex influences the effect of adiposity on arterial stiffness and renin‐angiotensin aldosterone system activity in young adults. Endocrinol Diabetes Metab 2022; 5:e00317. [PMID: 34954909 PMCID: PMC8917865 DOI: 10.1002/edm2.317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 10/23/2021] [Accepted: 12/04/2021] [Indexed: 11/06/2022] Open
Abstract
Introduction Methods Results Conclusion
Collapse
Affiliation(s)
- Cindy Z. Kalenga
- Cumming School of Medicine University of Calgary Calgary Alberta Canada
- Libin Cardiovascular Institute University of Calgary Calgary Alberta Canada
| | - Sharanya Ramesh
- Temerty Faculty of Medicine University of Toronto Toronto Ontario Canada
| | - Sandra M. Dumanski
- Cumming School of Medicine University of Calgary Calgary Alberta Canada
- Libin Cardiovascular Institute University of Calgary Calgary Alberta Canada
- Alberta Kidney Disease Network Calgary Alberta Canada
| | - Jennifer M. MacRae
- Cumming School of Medicine University of Calgary Calgary Alberta Canada
- Libin Cardiovascular Institute University of Calgary Calgary Alberta Canada
| | - Kara Nerenberg
- Cumming School of Medicine University of Calgary Calgary Alberta Canada
- Libin Cardiovascular Institute University of Calgary Calgary Alberta Canada
- O’Brien Institute for Public Health University of Calgary Calgary Alberta Canada
| | - Amy Metcalfe
- Cumming School of Medicine University of Calgary Calgary Alberta Canada
- Libin Cardiovascular Institute University of Calgary Calgary Alberta Canada
- O’Brien Institute for Public Health University of Calgary Calgary Alberta Canada
- Alberta Children's Hospital Research Institute Calgary Alberta Canada
| | - Darlene Y. Sola
- Cumming School of Medicine University of Calgary Calgary Alberta Canada
- Libin Cardiovascular Institute University of Calgary Calgary Alberta Canada
| | - Sofia B. Ahmed
- Cumming School of Medicine University of Calgary Calgary Alberta Canada
- Libin Cardiovascular Institute University of Calgary Calgary Alberta Canada
- Alberta Kidney Disease Network Calgary Alberta Canada
- O’Brien Institute for Public Health University of Calgary Calgary Alberta Canada
| |
Collapse
|
28
|
Demerdash HM. Weight regain after bariatric surgery: Promoters and potential predictors. World J Meta-Anal 2021; 9:438-454. [DOI: 10.13105/wjma.v9.i5.438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 06/07/2021] [Accepted: 09/17/2021] [Indexed: 02/06/2023] Open
Abstract
Obesity is globally viewed as chronic relapsing disease. Bariatric surgery offers the most efficient and durable weight loss approach. However, weight regain after surgery is a distressing issue as obesity can revert. Surgical procedures were originally designed to reduce food intake and catalyze weight loss, provided that its role is marginalized in long-term weight maintenance. Consequently, it is essential to establish a scientifically standardized applicable definitions for weight regain, which necessitates enhanced comprehension of the clinical situation, as well as have realistic expectations concerning weight loss. Moreover, several factors are proposed to influence weight regain as psychological, behavioral factors, hormonal, metabolic, anatomical lapses, as well as genetic predisposition. Recently, there is a growing evidence of utilization of scoring system to anticipate excess body weight loss, along with characterizing certain biomarkers that identify subjects at risk of suboptimal weight loss after surgery. Furthermore, personalized counseling is warranted to help select bariatric procedure, reinforce self-monitoring skills, motivate patient, encourage mindful eating practices, to avoid recidivism.
Collapse
Affiliation(s)
- Hala Mourad Demerdash
- Department of Clinical Pathology, Alexandria University Hospitals, Alexandria 21311, Egypt
| |
Collapse
|
29
|
Goette A, Lendeckel U. Atrial Cardiomyopathy: Pathophysiology and Clinical Consequences. Cells 2021; 10:cells10102605. [PMID: 34685585 PMCID: PMC8533786 DOI: 10.3390/cells10102605] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 09/26/2021] [Indexed: 12/18/2022] Open
Abstract
Around the world there are 33.5 million patients suffering from atrial fibrillation (AF) with an annual increase of 5 million cases. Most AF patients have an established form of an atrial cardiomyopathy. The concept of atrial cardiomyopathy was introduced in 2016. Thus, therapy of underlying diseases and atrial tissue changes appear as a cornerstone of AF therapy. Furthermore, therapy or prevention of atrial endocardial changes has the potential to reduce atrial thrombogenesis and thereby cerebral stroke. The present manuscript will summarize the underlying pathophysiology and remodeling processes observed in the development of an atrial cardiomyopathy, thrombogenesis, and atrial fibrillation. In particular, the impact of oxidative stress, inflammation, diabetes, and obesity will be addressed.
Collapse
Affiliation(s)
- Andreas Goette
- Department of Cardiology and Intensive Care Medicine, St. Vincenz Hospital, 33098 Paderborn, Germany
- MAESTRIA Consortium/AFNET, 48149 Münster, Germany
- Correspondence:
| | - Uwe Lendeckel
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, 17475 Greifswald, Germany;
| |
Collapse
|
30
|
Vikøren LA, Drotningsvik A, Midttun Ø, McCann A, Bergseth MT, Austgulen MH, Mellgren G, Ueland PM, Gudbrandsen OA. Baked cod consumption delayed the development of kidney and liver dysfunction and affected plasma amino acid concentrations, but did not affect blood pressure, blood glucose or liver triacylglycerol concentrations in obese fa/fa Zucker rats. Nutr Res 2021; 92:72-83. [PMID: 34274556 DOI: 10.1016/j.nutres.2021.05.009] [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: 11/26/2020] [Revised: 05/13/2021] [Accepted: 05/23/2021] [Indexed: 10/21/2022]
Abstract
Obesity is associated with changes in amino acid metabolism, and studies show that ingestion of fish proteins influence amino acid composition in plasma and urine, in addition to affecting risk factors for metabolic syndrome. Since the majority of fish proteins consumed by humans are as fish fillet, it is of interest to investigate if cod fillet intake affects amino acid composition and metabolic disorders. We hypothesized that a modified AIN-93G diet containing cod fillet would affect amino acid compositions in plasma and urine in obese rats, and also affect risk factors for metabolic syndrome when compared to rats fed a regular AIN-93G diet with casein as the protein source. Obese Zucker fa/fa rats, a rat model of metabolic syndrome, received diets containing 25% protein from lyophilized baked cod fillet and 75% protein from casein (Baked cod diet), or a Control diet with casein for four weeks. The Baked cod diet affected the amino acid composition in plasma, with e.g., lower glycine, histidine, homoarginine, homocysteine, methionine, proline and tyrosine concentrations, but did not affect amino acid concentrations in urine. The concentrations of markers for kidney and liver dysfunction were lower in the Baked cod group, however blood pressure development, fasting and postprandial glucose, and hepatic triacylglycerol concentrations were similar to the Control group. To conclude, substituting 25% of dietary protein with baked cod fillet affected concentrations of some amino acids in plasma and delayed development of kidney and liver dysfunction, but did not affect blood pressure, glucose concentration or fatty liver.
Collapse
Affiliation(s)
- Linn A Vikøren
- Dietary Protein Research Group, Department of Clinical Medicine, University of Bergen, 5021 Bergen, Norway; Department of Clinical Science, University of Bergen, 5021 Bergen, Norway
| | - Aslaug Drotningsvik
- Dietary Protein Research Group, Department of Clinical Medicine, University of Bergen, 5021 Bergen, Norway
| | | | | | - Marthe T Bergseth
- Dietary Protein Research Group, Department of Clinical Medicine, University of Bergen, 5021 Bergen, Norway
| | - Maren H Austgulen
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway
| | - Gunnar Mellgren
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Haukeland University Hospital, 5020 Bergen, Norway; Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, 5021 Bergen, Norway
| | | | - Oddrun A Gudbrandsen
- Dietary Protein Research Group, Department of Clinical Medicine, University of Bergen, 5021 Bergen, Norway.
| |
Collapse
|
31
|
ILRUN Downregulates ACE2 Expression and Blocks Infection of Human Cells by SARS-CoV-2. J Virol 2021; 95:e0032721. [PMID: 33963054 DOI: 10.1128/jvi.00327-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The human protein-coding gene ILRUN (inflammation and lipid regulator with UBA-like and NBR1-like domains; previously C6orf106) was identified as a proviral factor for Hendra virus infection and was recently characterized to function as an inhibitor of type I interferon expression. Here, we have utilized transcriptome sequencing (RNA-seq) to define cellular pathways regulated by ILRUN in the context of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection of Caco-2 cells. We find that inhibition of ILRUN expression by RNA interference alters transcription profiles of numerous cellular pathways, including upregulation of the SARS-CoV-2 entry receptor ACE2 and several other members of the renin-angiotensin aldosterone system. In addition, transcripts of the SARS-CoV-2 coreceptors TMPRSS2 and CTSL were also upregulated. Inhibition of ILRUN also resulted in increased SARS-CoV-2 replication, while overexpression of ILRUN had the opposite effect, identifying ILRUN as a novel antiviral factor for SARS-CoV-2 replication. This represents, to our knowledge, the first report of ILRUN as a regulator of the renin-angiotensin-aldosterone system (RAAS). IMPORTANCE There is no doubt that the current rapid global spread of COVID-19 has had significant and far-reaching impacts on our health and economy and will continue to do so. Research in emerging infectious diseases, such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is growing rapidly, with new breakthroughs in the understanding of host-virus interactions to assist with the development of innovative and exciting therapeutic strategies. Here, we present the first evidence that modulation of the human protein-coding gene ILRUN functions as an antiviral factor for SARS-CoV-2 infection, likely through its newly identified role in regulating the expression of SARS-CoV-2 entry receptors ACE2, TMPRSS2, and CTSL. These data improve our understanding of biological pathways that regulate host factors critical to SARS-CoV-2 infection, contributing to the development of antiviral strategies to deal with the current SARS-CoV-2 pandemic.
Collapse
|
32
|
Bourgeois C, Gorwood J, Olivo A, Le Pelletier L, Capeau J, Lambotte O, Béréziat V, Lagathu C. Contribution of Adipose Tissue to the Chronic Immune Activation and Inflammation Associated With HIV Infection and Its Treatment. Front Immunol 2021; 12:670566. [PMID: 34220817 PMCID: PMC8250865 DOI: 10.3389/fimmu.2021.670566] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 05/24/2021] [Indexed: 12/12/2022] Open
Abstract
White adipose tissue (AT) contributes significantly to inflammation – especially in the context of obesity. Several of AT’s intrinsic features favor its key role in local and systemic inflammation: (i) large distribution throughout the body, (ii) major endocrine activity, and (iii) presence of metabolic and immune cells in close proximity. In obesity, the concomitant pro-inflammatory signals produced by immune cells, adipocytes and adipose stem cells help to drive local inflammation in a vicious circle. Although the secretion of adipokines by AT is a prime contributor to systemic inflammation, the lipotoxicity associated with AT dysfunction might also be involved and could affect distant organs. In HIV-infected patients, the AT is targeted by both HIV infection and antiretroviral therapy (ART). During the primary phase of infection, the virus targets AT directly (by infecting AT CD4 T cells) and indirectly (via viral protein release, inflammatory signals, and gut disruption). The initiation of ART drastically changes the picture: ART reduces viral load, restores (at least partially) the CD4 T cell count, and dampens inflammatory processes on the whole-body level but also within the AT. However, ART induces AT dysfunction and metabolic side effects, which are highly dependent on the individual molecules and the combination used. First generation thymidine reverse transcriptase inhibitors predominantly target mitochondrial DNA and induce oxidative stress and adipocyte death. Protease inhibitors predominantly affect metabolic pathways (affecting adipogenesis and adipocyte homeostasis) resulting in insulin resistance. Recently marketed integrase strand transfer inhibitors induce both adipocyte adipogenesis, hypertrophy and fibrosis. It is challenging to distinguish between the respective effects of viral persistence, persistent immune defects and ART toxicity on the inflammatory profile present in ART-controlled HIV-infected patients. The host metabolic status, the size of the pre-established viral reservoir, the quality of the immune restoration, and the natural ageing with associated comorbidities may mitigate and/or reinforce the contribution of antiretrovirals (ARVs) toxicity to the development of low-grade inflammation in HIV-infected patients. Protecting AT functions appears highly relevant in ART-controlled HIV-infected patients. It requires lifestyle habits improvement in the absence of effective anti-inflammatory treatment. Besides, reducing ART toxicities remains a crucial therapeutic goal.
Collapse
Affiliation(s)
- Christine Bourgeois
- CEA - Université Paris Saclay - INSERM U1184, Center for Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, Fontenay-aux-Roses, France
| | - Jennifer Gorwood
- Sorbonne Université, INSERM UMR_S 938, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire de Cardio-métabolisme et Nutrition (ICAN), FRM EQU201903007868, Paris, France
| | - Anaelle Olivo
- CEA - Université Paris Saclay - INSERM U1184, Center for Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, Fontenay-aux-Roses, France
| | - Laura Le Pelletier
- Sorbonne Université, INSERM UMR_S 938, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire de Cardio-métabolisme et Nutrition (ICAN), FRM EQU201903007868, Paris, France
| | - Jacqueline Capeau
- Sorbonne Université, INSERM UMR_S 938, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire de Cardio-métabolisme et Nutrition (ICAN), FRM EQU201903007868, Paris, France
| | - Olivier Lambotte
- CEA - Université Paris Saclay - INSERM U1184, Center for Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, Fontenay-aux-Roses, France.,AP-HP, Groupe Hospitalier Universitaire Paris Saclay, Hôpital Bicêtre, Service de Médecine Interne et Immunologie Clinique, Le Kremlin-Bicêtre, France
| | - Véronique Béréziat
- Sorbonne Université, INSERM UMR_S 938, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire de Cardio-métabolisme et Nutrition (ICAN), FRM EQU201903007868, Paris, France
| | - Claire Lagathu
- Sorbonne Université, INSERM UMR_S 938, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire de Cardio-métabolisme et Nutrition (ICAN), FRM EQU201903007868, Paris, France
| |
Collapse
|
33
|
Mascolo A, Urbanek K, De Angelis A, Sessa M, Scavone C, Berrino L, Rosano GMC, Capuano A, Rossi F. Angiotensin II and angiotensin 1-7: which is their role in atrial fibrillation? Heart Fail Rev 2021; 25:367-380. [PMID: 31375968 DOI: 10.1007/s10741-019-09837-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Atrial fibrillation (AF) is a significant cause of morbidity and mortality as well as a public health burden considering the high costs of AF-related hospitalizations. Pre-clinical and clinical evidence showed a potential role of the renin angiotensin system (RAS) in the etiopathogenesis of AF. Among RAS mediators, angiotensin II (AII) and angiotensin 1-7 (A1-7) have been mostly investigated in AF. Specifically, the stimulation of the pathway mediated by AII or the inhibition of the pathway mediated by A1-7 may participate in inducing and sustaining AF. In this review, we summarize the evidence showing that both RAS pathways may balance the onset of AF through different biological mechanisms involving inflammation, epicardial adipose tissue (EAT) accumulation, and electrical cardiac remodeling. EAT is a predictor for AF as it may induce its onset through direct (infiltration of epicardial adipocytes into the underlying atrial myocardium) and indirect (release of inflammatory adipokines, the stimulation of oxidative stress, macrophage phenotype switching, and AF triggers) mechanisms. Classic RAS blockers such as angiotensin converting enzyme inhibitors (ACE-I) and angiotensin receptor blockers (ARB) may prevent AF by affecting the accumulation of the EAT, representing a useful therapeutic strategy for preventing AF especially in patients with heart failure and known left ventricular dysfunction. Further studies are necessary to prove this benefit in patients with other cardiovascular diseases. Finally, the possibility of using the A1-7 or ACE2 analogues, to enlarge current therapeutic options for AF, may represent an important field of research.
Collapse
Affiliation(s)
- Annamaria Mascolo
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, University of Campania "Luigi Vanvitelli", Via Santa Maria di Costantinopoli 16, 80138, Naples, Italy.
| | - Konrad Urbanek
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, University of Campania "Luigi Vanvitelli", Via Santa Maria di Costantinopoli 16, 80138, Naples, Italy
| | - Antonella De Angelis
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, University of Campania "Luigi Vanvitelli", Via Santa Maria di Costantinopoli 16, 80138, Naples, Italy
| | - Maurizio Sessa
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, University of Campania "Luigi Vanvitelli", Via Santa Maria di Costantinopoli 16, 80138, Naples, Italy
| | - Cristina Scavone
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, University of Campania "Luigi Vanvitelli", Via Santa Maria di Costantinopoli 16, 80138, Naples, Italy
| | - Liberato Berrino
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, University of Campania "Luigi Vanvitelli", Via Santa Maria di Costantinopoli 16, 80138, Naples, Italy
| | - Giuseppe Massimo Claudio Rosano
- IRCCS San Raffaele Pisana, Rome, Italy.,Cardiovascular and Cell Sciences Research Institute, St. George's, University of London, London, UK
| | - Annalisa Capuano
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, University of Campania "Luigi Vanvitelli", Via Santa Maria di Costantinopoli 16, 80138, Naples, Italy
| | - Francesco Rossi
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, University of Campania "Luigi Vanvitelli", Via Santa Maria di Costantinopoli 16, 80138, Naples, Italy
| |
Collapse
|
34
|
Hammoud SH, AlZaim I, Al-Dhaheri Y, Eid AH, El-Yazbi AF. Perirenal Adipose Tissue Inflammation: Novel Insights Linking Metabolic Dysfunction to Renal Diseases. Front Endocrinol (Lausanne) 2021; 12:707126. [PMID: 34408726 PMCID: PMC8366229 DOI: 10.3389/fendo.2021.707126] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 07/19/2021] [Indexed: 12/15/2022] Open
Abstract
A healthy adipose tissue (AT) is indispensable to human wellbeing. Among other roles, it contributes to energy homeostasis and provides insulation for internal organs. Adipocytes were previously thought to be a passive store of excess calories, however this view evolved to include an endocrine role. Adipose tissue was shown to synthesize and secrete adipokines that are pertinent to glucose and lipid homeostasis, as well as inflammation. Importantly, the obesity-induced adipose tissue expansion stimulates a plethora of signals capable of triggering an inflammatory response. These inflammatory manifestations of obese AT have been linked to insulin resistance, metabolic syndrome, and type 2 diabetes, and proposed to evoke obesity-induced comorbidities including cardiovascular diseases (CVDs). A growing body of evidence suggests that metabolic disorders, characterized by AT inflammation and accumulation around organs may eventually induce organ dysfunction through a direct local mechanism. Interestingly, perirenal adipose tissue (PRAT), surrounding the kidney, influences renal function and metabolism. In this regard, PRAT emerged as an independent risk factor for chronic kidney disease (CKD) and is even correlated with CVD. Here, we review the available evidence on the impact of PRAT alteration in different metabolic states on the renal and cardiovascular function. We present a broad overview of novel insights linking cardiovascular derangements and CKD with a focus on metabolic disorders affecting PRAT. We also argue that the confluence among these pathways may open several perspectives for future pharmacological therapies against CKD and CVD possibly by modulating PRAT immunometabolism.
Collapse
Affiliation(s)
- Safaa H. Hammoud
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Beirut Arab University, Beirut, Lebanon
| | - Ibrahim AlZaim
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Departmment of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Yusra Al-Dhaheri
- Department of Biology, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Ali H. Eid
- Department of Basic Medical Sciences, College of Medicine, Qatar University, Doha, Qatar
- Biomedical and Pharmaceutical Research Unit, Qatar University (QU) Health, Qatar University, Doha, Qatar
| | - Ahmed F. El-Yazbi
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
- Faculty of Pharmacy, Alalamein International University, Alalamein, Egypt
- *Correspondence: Ahmed F. El-Yazbi,
| |
Collapse
|
35
|
Gencer S, Lacy M, Atzler D, van der Vorst EPC, Döring Y, Weber C. Immunoinflammatory, Thrombohaemostatic, and Cardiovascular Mechanisms in COVID-19. Thromb Haemost 2020; 120:1629-1641. [PMID: 33124029 PMCID: PMC7869061 DOI: 10.1055/s-0040-1718735] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 09/14/2020] [Indexed: 12/15/2022]
Abstract
The global coronavirus disease 2019 (COVID-19) pandemic has deranged the recent history of humankind, afflicting more than 27 million individuals to date. While the majority of COVID-19 patients recuperate, a considerable number of patients develop severe complications. Bilateral pneumonia constitutes the hallmark of severe COVID-19 disease but an involvement of other organ systems, namely the cardiovascular system, kidneys, liver, and central nervous system, occurs in at least half of the fatal COVID-19 cases. Besides respiratory failure requiring ventilation, patients with severe COVID-19 often display manifestations of systemic inflammation and thrombosis as well as diffuse microvascular injury observed postmortem. In this review, we survey the mechanisms that may explain how viral entry and activation of endothelial cells by severe acute respiratory syndrome coronavirus 2 can give rise to a series of events including systemic inflammation, thrombosis, and microvascular dysfunction. This pathophysiological scenario may be particularly harmful in patients with overt cardiovascular disease and may drive the fatal aspects of COVID-19. We further shed light on the role of the renin-angiotensin aldosterone system and its inhibitors in the context of COVID-19 and discuss the potential impact of antiviral and anti-inflammatory treatment options. Acknowledging the comorbidities and potential organ injuries throughout the course of severe COVID-19 is crucial in the clinical management of patients affecting treatment approaches and recovery rate.
Collapse
Affiliation(s)
- Selin Gencer
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität, Munich, Germany
| | - Michael Lacy
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität, Munich, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Dorothee Atzler
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität, Munich, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
- Walther Straub Institute for Pharmacology and Toxicology, Ludwig-Maximilians-Universität, Munich, Germany
| | - Emiel P. C. van der Vorst
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität, Munich, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
- Interdisciplinary Center for Clinical Research (IZKF), Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Yvonne Döring
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität, Munich, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
- Divison of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Christian Weber
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität, Munich, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, The Netherlands
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
36
|
Maldonado V, Hernandez-Ramírez C, Oliva-Pérez EA, Sánchez-Martínez CO, Pimentel-González JF, Molina-Sánchez JR, Jiménez-Villalba YZ, Chávez-Alderete J, Loza-Mejía MA. Pentoxifylline decreases serum LDH levels and increases lymphocyte count in COVID-19 patients: Results from an external pilot study. Int Immunopharmacol 2020; 90:107209. [PMID: 33278747 PMCID: PMC7690298 DOI: 10.1016/j.intimp.2020.107209] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 11/11/2020] [Accepted: 11/11/2020] [Indexed: 02/08/2023]
Abstract
Pentoxifylline could be beneficial for the treatment of COVID-19. Serum lactate dehydrogenase and lymphocyte count are accessible biomarkers that correlate with the severity of COVID-19. Pentoxifylline treatment was associated with an increase in the lymphocyte count and decreased LDH levels.
We have previously hypothesized that pentoxifylline could be beneficial for the treatment of COVID-19 given its potential to restore the immune response equilibrium, reduce the impact of the disease on the endothelium and alveolar epithelial cells, and improve the circulatory function. Serum lactate dehydrogenase (LDH) and lymphocyte count are accessible biomarkers that correlate with the severity of COVID-19, the need for hospitalization, and mortality, reflecting the host immune response’s contribution to the seriousness of SARS-CoV-2 infection. We carried out this external pilot study on 38 patients with moderate and severe COVID-19 to test the effect pentoxifylline on parameters such as LDH, lymphocyte count, days of hospitalization, mortality, and proportion of patients requiring intubation. Twenty-six patients were randomized to receive 400 mg of pentoxifylline t.i.d. plus standard therapy (pentoxifylline group), while the rest received the standard treatment (control group). Linear regression models were built for statistically significant parameters. Pentoxifylline treatment was associated with a 64.25% increase (CI95% 11.83, 116.68) in lymphocyte count and a 29.61% decrease (CI95% 15.11, 44.10) in serum LDH. Although a trend towards reduced days of hospitalization, mortality, and proportion of patients requiring intubation was observed, no statistically significant difference was found for these parameters. Our findings open the possibility of pentoxifylline being repositioned as a drug for COVID-19 treatment with the advantages of a proven safety profile, availability, and no risk of immunosuppression; however, this evidence needs to be confirmed in a pragmatic randomized controlled trial.
Collapse
Affiliation(s)
- Valente Maldonado
- Faculty of Chemical Sciences, Universidad La Salle-México, Cuauhtémoc, Mexico City 06140, Mexico; Department of Allergy and Clinical Immunology Internal Medicine, General Hospital of Zone 27 Mexican Institute of Social Security, Lerdo 311, Nonoalco Tlatelolco, Cuauhtémoc, Mexico City 6390, Mexico.
| | - Claudia Hernandez-Ramírez
- Department of Allergy and Clinical Immunology Internal Medicine, General Hospital of Zone 27 Mexican Institute of Social Security, Lerdo 311, Nonoalco Tlatelolco, Cuauhtémoc, Mexico City 6390, Mexico
| | - Eniel Alonso Oliva-Pérez
- Department of Internal Medicine, General Hospital of Zone 27 Mexican Institute of Social Security, Nonoalco Tlatelolco, Cuauhtémoc, Mexico City 6390, Mexico
| | - César Omar Sánchez-Martínez
- Department of Internal Medicine, General Hospital of Zone 27 Mexican Institute of Social Security, Nonoalco Tlatelolco, Cuauhtémoc, Mexico City 6390, Mexico
| | - Jorge Fabián Pimentel-González
- Department of Internal Medicine, General Hospital of Zone 27 Mexican Institute of Social Security, Nonoalco Tlatelolco, Cuauhtémoc, Mexico City 6390, Mexico
| | - José Raúl Molina-Sánchez
- Department of Internal Medicine, General Hospital of Zone 27 Mexican Institute of Social Security, Nonoalco Tlatelolco, Cuauhtémoc, Mexico City 6390, Mexico
| | - Yeimmy Zuyenn Jiménez-Villalba
- Department of Internal Medicine, General Hospital of Zone 27 Mexican Institute of Social Security, Nonoalco Tlatelolco, Cuauhtémoc, Mexico City 6390, Mexico
| | - Jaime Chávez-Alderete
- Department of Bronchial Hyperreactivity, National Institute of Respiratory Diseases Ismael Cosío Villegas, Tlalpan, Mexico City 14080, Mexico
| | - Marco A Loza-Mejía
- Faculty of Chemical Sciences, Universidad La Salle-México, Cuauhtémoc, Mexico City 06140, Mexico
| |
Collapse
|
37
|
Maldonado V, Loza-Mejía MA, Chávez-Alderete J. Repositioning of pentoxifylline as an immunomodulator and regulator of the renin-angiotensin system in the treatment of COVID-19. Med Hypotheses 2020; 144:109988. [PMID: 32540603 PMCID: PMC7282759 DOI: 10.1016/j.mehy.2020.109988] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/04/2020] [Accepted: 06/07/2020] [Indexed: 02/07/2023]
Abstract
Pentoxifylline (PTX) is a phosphodiesterase inhibitor that increases cyclic adenosine monophosphate levels, which in turn activate protein kinase, leading to a reduction in the synthesis of proinflammatory cytokines to ultimately influence the renin-angiotensin system (RAS) in vitro by inhibiting angiotensin 1 receptor (AT1R) expression. The rheological, anti-inflammatory, and renin-angiotensin axis properties of PTX highlight this drug as a therapeutic treatment alternative for patients with COVID-19 by helping reduce the production of the inflammatory cytokines without deleterious effects on the immune system to delay viral clearance. Moreover, PTX can restore the balance of the immune response, reduce damage to the endothelium and alveolar epithelial cells, improve circulation, and prevent microvascular thrombosis. There is further evidence that PTX can improve ventilatory parameters. Therefore, we propose repositioning PTX in the treatment of COVID-19. The main advantage of repositioning PTX is that it is an affordable drug that is already available worldwide with an established safety profile, further offering the possibility of immediately analysing the result of its use and associated success rates. Another advantage is that PTX selectively reduces the concentration of TNF-α mRNA in cells, which, in the case of an acute infectious state such as COVID-19, would seem to offer a more strategic approach.
Collapse
Affiliation(s)
- Valente Maldonado
- Faculty of Chemical Sciences, Universidad La Salle-México, Cuauhtémoc, Mexico City 06140, Mexico; Department of Allergy and Clinical Immunology Internal Medicine, General Hospital of Zone 27 Mexican Institute of Social Security, Col. Nonoalco Tlatelolco Cuauhtémoc, Mexico City 6390, Mexico.
| | - Marco A Loza-Mejía
- Faculty of Chemical Sciences, Universidad La Salle-México, Cuauhtémoc, Mexico City 06140, Mexico
| | - Jaime Chávez-Alderete
- Laboratory of Bronchial Hyperreactivity, National Institute of Respiratory Diseases Ismael Cosío Villegas, Tlalpan, Mexico City 14080, Mexico
| |
Collapse
|
38
|
Kim DY, Choi MJ, Ko TK, Lee NH, Kim OH, Cheon HG. Angiotensin AT 1 receptor antagonism by losartan stimulates adipocyte browning via induction of apelin. J Biol Chem 2020; 295:14878-14892. [PMID: 32839272 DOI: 10.1074/jbc.ra120.013834] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 08/04/2020] [Indexed: 12/31/2022] Open
Abstract
Adipocyte browning appears to be a potential therapeutic strategy to combat obesity and related metabolic disorders. Recent studies have shown that apelin, an adipokine, stimulates adipocyte browning and has negative cross-talk with angiotensin II receptor type 1 (AT1 receptor) signaling. Here, we report that losartan, a selective AT1 receptor antagonist, induces browning, as evidenced by an increase in browning marker expression, mitochondrial biogenesis, and oxygen consumption in murine adipocytes. In parallel, losartan up-regulated apelin expression, concomitant with increased phosphorylation of protein kinase B and AMP-activated protein kinase. However, the siRNA-mediated knockdown of apelin expression attenuated losartan-induced browning. Angiotensin II cotreatment also inhibited losartan-induced browning, suggesting that AT1 receptor antagonism-induced activation of apelin signaling may be responsible for adipocyte browning induced by losartan. The in vivo browning effects of losartan were confirmed using both C57BL/6J and ob/ob mice. Furthermore, in vivo apelin knockdown by adeno-associated virus carrying-apelin shRNA significantly inhibited losartan-induced adipocyte browning. In summary, these data suggested that AT1 receptor antagonism by losartan promotes the browning of white adipocytes via the induction of apelin expression. Therefore, apelin modulation may be an effective strategy for the treatment of obesity and its related metabolic disorders.
Collapse
Affiliation(s)
- Dong Young Kim
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Republic of Korea
| | - Mi Jin Choi
- Department of Pharmacology, College of Medicine, Gachon University, Incheon, Republic of Korea
| | - Tae Kyung Ko
- Department of Pharmacology, College of Medicine, Gachon University, Incheon, Republic of Korea
| | - Na Hyun Lee
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Republic of Korea
| | - Ok-Hee Kim
- Department of Pharmacology, College of Medicine, Gachon University, Incheon, Republic of Korea
| | - Hyae Gyeong Cheon
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Republic of Korea; Department of Pharmacology, College of Medicine, Gachon University, Incheon, Republic of Korea.
| |
Collapse
|
39
|
Hoevenaar M, Goossens D, Roorda J. Angiotensin-converting enzyme 2, the complement system, the kallikrein-kinin system, type-2 diabetes, interleukin-6, and their interactions regarding the complex COVID-19 pathophysiological crossroads. J Renin Angiotensin Aldosterone Syst 2020; 21:1470320320979097. [PMID: 33283602 PMCID: PMC7724427 DOI: 10.1177/1470320320979097] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 11/11/2020] [Indexed: 12/15/2022] Open
Abstract
Because of the current COVID-19-pandemic, the world is currently being held hostage in various lockdowns. ACE2 facilitates SARS-CoV-2 cell-entry, and is at the very center of several pathophysiological pathways regarding the RAAS, CS, KKS, T2DM, and IL-6. Their interactions with severe COVID-19 complications (e.g. ARDS and thrombosis), and potential therapeutic targets for pharmacological intervention, will be reviewed.
Collapse
Affiliation(s)
| | | | - Janne Roorda
- Medical Doctor, General Practice
van Dijk, Oisterwijk, The Netherlands
| |
Collapse
|
40
|
Macedo AVS. Targeting the Renin-Angiotensin-Aldosterone System in Obesity. Arq Bras Cardiol 2020; 115:29-30. [PMID: 32785498 PMCID: PMC8384313 DOI: 10.36660/abc.20200345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
41
|
Giori IG, Magliano DC, Alexandre-Santos B, Fernandes T, Oliveira EM, Vieira CP, Conte-Junior CA, Ceddia RB, Nobrega ACL, Frantz EDC. Enalapril and treadmill running reduce adiposity, but only the latter causes adipose tissue browning in mice. J Cell Physiol 2020; 236:900-910. [PMID: 32617979 DOI: 10.1002/jcp.29900] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 06/12/2020] [Accepted: 06/13/2020] [Indexed: 12/18/2022]
Abstract
This study investigated whether regulation of the renin-angiotensin system (RAS) by enalapril and/or aerobic exercise training (AET) causes browning of the subcutaneous white adipose tissue (sWAT). C57BL/6 mice were fed either a standard chow or a high-fat (HF) diet for 16 weeks. At Week 8, HF-fed animals were divided into sedentary (HF), enalapril (HF-E), AET (HF-T), and enalapril plus AET (HF-ET) groups. Subsequently, sWAT was extracted for morphometry, determination of RAS expression, and biomarkers of WAT browning. The HF group displayed adipocyte hypertrophy and induction of the classical RAS axis. Conversely, all interventions reduced adiposity and induced the counterregulatory RAS axis. However, only AET raised plasma irisin, increased peroxisome proliferator-activated receptor-γ coactivator-1α, and uncoupling protein-1 levels, and the expression of PR-domain containing 16 in sWAT. Therefore, we concluded that AET-induced sWAT browning was independent of the counterregulatory axis shifting of RAS in HF diet-induced obesity.
Collapse
Affiliation(s)
- Isabele G Giori
- Laboratory of Exercise Sciences, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
| | - D'Angelo C Magliano
- Laboratory of Morphological and Metabolic Analyses, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
| | - Beatriz Alexandre-Santos
- Laboratory of Exercise Sciences, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil.,Laboratory of Morphological and Metabolic Analyses, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
| | - Tiago Fernandes
- National Institute for Science and Technology, INCT Physical (In)activity and Exercise, CNPq, Niteroi, Rio de Janeiro, Brazil.,Laboratory of Biochemistry and Molecular Biology of Exercise, School of Physical Education and Sport, University of Sao Paulo, São Paulo, State of São Paulo, Brazil
| | - Edilamar M Oliveira
- National Institute for Science and Technology, INCT Physical (In)activity and Exercise, CNPq, Niteroi, Rio de Janeiro, Brazil.,Laboratory of Biochemistry and Molecular Biology of Exercise, School of Physical Education and Sport, University of Sao Paulo, São Paulo, State of São Paulo, Brazil
| | - Carla P Vieira
- Department of Food Technology, Faculty of Veterinary, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
| | - Carlos A Conte-Junior
- Department of Food Technology, Faculty of Veterinary, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
| | - Rolando B Ceddia
- Muscle Health Research Center, School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
| | - Antonio C L Nobrega
- Laboratory of Exercise Sciences, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil.,National Institute for Science and Technology, INCT Physical (In)activity and Exercise, CNPq, Niteroi, Rio de Janeiro, Brazil
| | - Eliete D C Frantz
- Laboratory of Exercise Sciences, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil.,Laboratory of Morphological and Metabolic Analyses, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil.,National Institute for Science and Technology, INCT Physical (In)activity and Exercise, CNPq, Niteroi, Rio de Janeiro, Brazil
| |
Collapse
|
42
|
Santos IB, de Bem GF, da Costa CA, de Carvalho LCRM, de Medeiros AF, Silva DLB, Romão MH, de Andrade Soares R, Ognibene DT, de Moura RS, Resende AC. Açaí seed extract prevents the renin-angiotensin system activation, oxidative stress and inflammation in white adipose tissue of high-fat diet-fed mice. Nutr Res 2020; 79:35-49. [PMID: 32610256 DOI: 10.1016/j.nutres.2020.05.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 04/15/2020] [Accepted: 05/13/2020] [Indexed: 12/18/2022]
Abstract
The role of the renin-angiotensin system (RAS), oxidative stress, and inflammation on the development of obesity and its comorbidities has been extensively addressed. Euterpe oleracea Mart. (açaí) seed extract (ASE), with antioxidant and anti-inflammatory properties and capable to modulate plasma renin levels, has been evidenced as a potential regulator of body mass. We hypothesized that the supplementation with ASE might exert beneficial effects on obesity-related white adipose tissue changes and metabolic disorders by interfering with the local adipose tissue overexpression of RAS, inflammation, and oxidative stress in C57BL/6 mice fed a high-fat (HF) diet. The animals were fed a standard diet (10% fat, control), 60% fat (HF), HF + ASE (300 mg/kg per day) and HF + ENA (enalapril, 30 mg/kg per day) for 12 weeks. ASE and ENA prevented weight gain and adiposity, adipocyte hypertrophy, dyslipidemia, and insulin resistance. In adipose tissue, ASE increased the insulin receptor expression and reduced renin and AT1 receptor expression, which was associated with decreased plasma levels of renin and angiotensin II. Differently, ENA increased the expression of angiotensin-conversing enzyme 2, AT2, B2, and Mas receptors in adipose tissue. Also, ASE but not ENA decreased malondialdehyde and 8-isoprostane levels in adipose tissue. Finally, ASE and ENA reduced the adipose tissue inflammatory markers tumor necrosis factor alpha and interleukin 6. These results demonstrate that ASE prevented the adipocyte hypertrophy, obesity, hyperlipidemia, hyperglycemia, and insulin resistance in HF diet-fed mice. The downregulation of RAS in adipose tissue, reducing oxidative stress and inflammation, may contribute to the prevention of obesity-related disorders.
Collapse
Affiliation(s)
- Izabelle Barcellos Santos
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil.
| | - Graziele Freitas de Bem
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil.
| | - Cristiane Aguiar da Costa
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil.
| | | | - Amanda Faria de Medeiros
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil.
| | - Dafne Lopes Beserra Silva
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil.
| | - Matheus Henrique Romão
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil.
| | - Ricardo de Andrade Soares
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil.
| | - Dayane Teixeira Ognibene
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil.
| | - Roberto Soares de Moura
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil.
| | - Angela Castro Resende
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil.
| |
Collapse
|
43
|
Polonis K, Becari C, Chahal CAA, Zhang Y, Allen AM, Kellogg TA, Somers VK, Singh P. Chronic Intermittent Hypoxia Triggers a Senescence-like Phenotype in Human White Preadipocytes. Sci Rep 2020; 10:6846. [PMID: 32321999 PMCID: PMC7176724 DOI: 10.1038/s41598-020-63761-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 04/03/2020] [Indexed: 12/13/2022] Open
Abstract
Obstructive sleep apnea (OSA) is a common sleep disorder associated with obesity. Emerging evidence suggest that OSA increases the risk of cardiovascular morbidity and mortality partly via accelerating the process of cellular aging. Thus, we sought to examine the effects of intermittent hypoxia (IH), a hallmark of OSA, on senescence in human white preadipocytes. We demonstrate that chronic IH is associated with an increased generation of mitochondrial reactive oxygen species along with increased prevalence of cells with nuclear localization of γH2AX & p16. A higher prevalence of cells positive for senescence-associated β-galactosidase activity was also evident with chronic IH exposure. Intervention with aspirin, atorvastatin or renin-angiotensin system (RAS) inhibitors effectively attenuated IH-mediated senescence-like phenotype. Importantly, the validity of in vitro findings was confirmed by examination of the subcutaneous abdominal adipose tissue which showed that OSA patients had a significantly higher percentage of cells with nuclear localization of γH2AX & p16 than non-OSA individuals (20.1 ± 10.8% vs. 10.3 ± 2.7%, Padjusted < 0.001). Furthermore, the frequency of dual positive γH2AX & p16 nuclei in adipose tissue of OSA patients receiving statin, aspirin, and/or RAS inhibitors was comparable to non-OSA individuals. This study identifies chronic IH as a trigger of senescence-like phenotype in preadipocytes. Together, our data suggest that OSA may be considered as a senescence-related disorder.
Collapse
Affiliation(s)
- Katarzyna Polonis
- Department of Cardiovascular Medicine, Mayo Clinic, MN, Rochester, USA
| | - Christiane Becari
- Department of Cardiovascular Medicine, Mayo Clinic, MN, Rochester, USA
- Department of Surgery and Anatomy, Ribeirao Preto Medical School, Ribeirão Preto, SP, Brazil
| | - C Anwar A Chahal
- Department of Cardiovascular Medicine, Mayo Clinic, MN, Rochester, USA
- Mayo Clinic Graduate School of Biomedical Sciences, MN, Rochester, USA
| | - Yuebo Zhang
- Department of Cardiovascular Medicine, Mayo Clinic, MN, Rochester, USA
| | - Alina M Allen
- Division of Gastroenterology and Hepatology, Mayo Clinic, MN, Rochester, USA
| | | | - Virend K Somers
- Department of Cardiovascular Medicine, Mayo Clinic, MN, Rochester, USA
| | - Prachi Singh
- Department of Cardiovascular Medicine, Mayo Clinic, MN, Rochester, USA.
- Pennington Biomedical Research Center, LA, Baton Rouge, USA.
| |
Collapse
|
44
|
Romão MH, de Bem GF, Santos IB, de Andrade Soares R, Ognibene DT, de Moura RS, da Costa CA, Resende ÂC. Açaí (Euterpe oleracea Mart.) seed extract protects against hepatic steatosis and fibrosis in high-fat diet-fed mice: Role of local renin-angiotensin system, oxidative stress and inflammation. J Funct Foods 2020. [DOI: 10.1016/j.jff.2019.103726] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
45
|
Haylett WL, Ferris WF. Adipocyte-progenitor cell communication that influences adipogenesis. Cell Mol Life Sci 2020; 77:115-128. [PMID: 31352534 PMCID: PMC11104918 DOI: 10.1007/s00018-019-03256-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 07/05/2019] [Accepted: 07/24/2019] [Indexed: 12/12/2022]
Abstract
Adipose tissue is located in discrete depots that are differentially associated with elevated risk of metabolic complications, with fat accretion in visceral depots being most detrimental to metabolic health. Currently, the regulation of specific adipose depot expansion, by adipocyte hypertrophy and hyperplasia and consequently fat distribution, is not well understood. However, a growing body of evidence from in vitro investigations indicates that mature adipocytes secrete factors that modulate the proliferation and differentiation of progenitor, adipose-derived stem cells (ADSCs). It is therefore plausible that endocrine communication between adipocytes and ADSCs located in different depots influences fat distribution, and may therefore contribute to the adverse health outcomes associated with visceral adiposity. This review will explore the available evidence of paracrine and endocrine crosstalk between mature adipocytes and ADSCs that affects adipogenesis, as a better understanding of the regulatory roles of the extracellular signalling mechanisms within- and between adipose depots may profoundly change the way we view adipose tissue growth in obesity and related comorbidities.
Collapse
Affiliation(s)
- William Lloyd Haylett
- Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - William Frank Ferris
- Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa.
| |
Collapse
|
46
|
Den Hartogh DJ, Gabriel A, Tsiani E. Antidiabetic Properties of Curcumin I: Evidence from In Vitro Studies. Nutrients 2020; 12:nu12010118. [PMID: 31906278 PMCID: PMC7019345 DOI: 10.3390/nu12010118] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 12/24/2019] [Accepted: 12/27/2019] [Indexed: 12/22/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a growing metabolic disease characterized by insulin resistance and hyperglycemia. Current preventative and treatment strategies for T2DM and insulin resistance lack in efficacy resulting in the need for new approaches to prevent and manage/treat the disease better. In recent years, epidemiological studies have suggested that diets rich in fruits and vegetables have beneficial health effects including protection against insulin resistance and T2DM. Curcumin, a polyphenol found in turmeric, and curcuminoids have been reported to have antioxidant, anti-inflammatory, hepatoprotective, nephroprotective, neuroprotective, immunomodulatory and antidiabetic properties. The current review (I of II) summarizes the existing in vitro studies examining the antidiabetic effects of curcumin, while a second (II of II) review summarizes evidence from existing in vivo animal studies and clinical trials focusing on curcumin’s antidiabetic properties.
Collapse
Affiliation(s)
- Danja J. Den Hartogh
- Department of Health Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada; (D.J.D.H.); (A.G.)
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON L2S 3A1, Canada
| | - Alessandra Gabriel
- Department of Health Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada; (D.J.D.H.); (A.G.)
| | - Evangelia Tsiani
- Department of Health Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada; (D.J.D.H.); (A.G.)
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON L2S 3A1, Canada
- Correspondence: or ; Tel.: +1-905-688-5550 (ext. 3881)
| |
Collapse
|
47
|
Den Hartogh DJ, Gabriel A, Tsiani E. Antidiabetic Properties of Curcumin II: Evidence from In Vivo Studies. Nutrients 2019; 12:nu12010058. [PMID: 31881654 PMCID: PMC7019668 DOI: 10.3390/nu12010058] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 12/19/2019] [Accepted: 12/24/2019] [Indexed: 12/14/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a growing metabolic disease characterized by insulin resistance and hyperglycemia. Current preventative and treatment approaches to insulin resistance and T2DM lack in efficacy, resulting in the need for new approaches to prevent and treat the disease. In recent years, epidemiological studies have suggested that diets rich in fruits and vegetables have beneficial health effects, including protection against insulin resistance and T2DM. Curcumin, a polyphenol found in turmeric, and curcuminoids have been reported to have antioxidant, anti-inflammatory, hepatoprotective, nephroprotective, neuroprotective, immunomodulatory and antidiabetic properties. The current review (II of II) summarizes the existing in vivo studies examining the antidiabetic effects of curcumin.
Collapse
Affiliation(s)
- Danja J. Den Hartogh
- Department of Health Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada; (D.J.D.H.); (A.G.)
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON L2S 3A1, Canada
| | - Alessandra Gabriel
- Department of Health Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada; (D.J.D.H.); (A.G.)
| | - Evangelia Tsiani
- Department of Health Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada; (D.J.D.H.); (A.G.)
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON L2S 3A1, Canada
- Correspondence: ; Tel.: +1-905-688-5550 (ext. 3881)
| |
Collapse
|
48
|
Abstract
Maintenance of systemic homeostasis and the response to nutritional and environmental challenges require the coordination of multiple organs and tissues. To respond to various metabolic demands, higher organisms have developed a system of inter-organ communication through which one tissue can affect metabolic pathways in a distant tissue. Dysregulation of these lines of communication contributes to human pathologies, including obesity, diabetes, liver disease and atherosclerosis. In recent years, technical advances such as data-driven bioinformatics, proteomics and lipidomics have enabled efforts to understand the complexity of systemic metabolic cross-talk and its underlying mechanisms. Here, we provide an overview of inter-organ signals and their roles in metabolic control, and highlight recent discoveries in the field. We review peptide, small-molecule and lipid mediators secreted by metabolic tissues, as well as the role of the central nervous system in orchestrating peripheral metabolic functions. Finally, we discuss the contributions of inter-organ signalling networks to the features of metabolic syndrome.
Collapse
Affiliation(s)
- Christina Priest
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
- Molecular Biology Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
49
|
Tanaka M. Improving obesity and blood pressure. Hypertens Res 2019; 43:79-89. [PMID: 31649313 DOI: 10.1038/s41440-019-0348-x] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 09/28/2019] [Accepted: 09/30/2019] [Indexed: 01/09/2023]
Abstract
Obesity-associated hypertension is a serious public health concern. Sympathetic nervous system (SNS) overactivity, especially in the kidneys, is an important mechanism linking obesity to hypertension. Some adipokines play important roles in elevating blood pressure (BP). Hyperinsulinemia caused by insulin resistance stimulates sodium reabsorption, enhances sodium retention, and increases circulating plasma volume. Hyperinsulinemia also stimulates both the renin-angiotensin-aldosterone system (RAAS) and the SNS, resulting in the acceleration of atherosclerosis through the hypertrophy of vascular smooth muscle cells, which contributes to increased peripheral vascular resistance. Obesity is associated with increased RAAS activity despite volume overload, as the tissue RAASs are stimulated in obese hypertensive individuals. Mineralocorticoid receptor-associated hypertension must also be considered in obese patients with resistant hypertension. Obstructive sleep apnea syndrome (OSAS) is the most common cause of secondary hypertension. Some components of the gut microbiota contribute to BP control; therefore, gut dysbiosis caused by obesity might lead to increased BP. The ratio of visceral fat to subcutaneous fat is higher in Japanese patients than in Caucasian patients, which may explain why Japanese patients are more susceptible to metabolic disorders even though they are less obese than Caucasian individuals. Obesity-associated kidney dysfunction directly increases BP, leading to further deterioration of kidney function. A bodyweight reduction of more than 3% or 5 kg significantly lowers BP. Gastrointestinal bypass surgery is an effective treatment for morbid obesity and its related metabolic disorders, including hypertension. Because both obesity and hypertension are representative lifestyle-related disorders, lifestyle modification, especially to improve obesity, should be performed first as a treatment for hypertension.
Collapse
Affiliation(s)
- Masami Tanaka
- Department of Endocrinology, Metabolism, and Nephrology, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| |
Collapse
|
50
|
Liu J, Li X, Lu Q, Ren D, Sun X, Rousselle T, Li J, Leng J. AMPK: a balancer of the renin-angiotensin system. Biosci Rep 2019; 39:BSR20181994. [PMID: 31413168 PMCID: PMC6722492 DOI: 10.1042/bsr20181994] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 07/24/2019] [Accepted: 08/12/2019] [Indexed: 02/07/2023] Open
Abstract
The renin-angiotensin system (RAS) is undisputedly well-studied as one of the oldest and most critical regulators for arterial blood pressure, fluid volume, as well as renal function. In recent studies, RAS has also been implicated in the development of obesity, diabetes, hyperlipidemia, and other diseases, and also involved in the regulation of several signaling pathways such as proliferation, apoptosis and autophagy, and insulin resistance. AMP-activated protein kinase (AMPK), an essential cellular energy sensor, has also been discovered to be involved in these diseases and cellular pathways. This would imply a connection between the RAS and AMPK. Therefore, this review serves to draw attention to the cross-talk between RAS and AMPK, then summering the most recent literature which highlights AMPK as a point of balance between physiological and pathological functions of the RAS.
Collapse
Affiliation(s)
- Jia Liu
- Department of Geriatrics, The First Hospital of Jilin University, Changchun 130021, China
- Department of Surgery, University of South Florida, Tampa, FL 33612, U.S.A
| | - Xuan Li
- Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, U.S.A
| | - Qingguo Lu
- Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, U.S.A
| | - Di Ren
- Department of Surgery, University of South Florida, Tampa, FL 33612, U.S.A
| | - Xiaodong Sun
- Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, U.S.A
| | - Thomas Rousselle
- Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, U.S.A
| | - Ji Li
- Department of Surgery, University of South Florida, Tampa, FL 33612, U.S.A
| | - Jiyan Leng
- Department of Geriatrics, The First Hospital of Jilin University, Changchun 130021, China
| |
Collapse
|