1
|
Nishida N, Sugimoto S, Miyagaki S, Cho C, Konishi M, Goda T, Yamaguchi M, Kawabe Y, Morimoto H, Kusuyama J, Okamura T, Hamaguchi M, Mori J, Nakajima H, Fukui M, Iehara T. Anti-inflammatory effect of Angiotensin 1-7 in white adipose tissue. Adipocyte 2025; 14:2449027. [PMID: 39803918 PMCID: PMC11730366 DOI: 10.1080/21623945.2024.2449027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 12/08/2024] [Accepted: 12/29/2024] [Indexed: 01/16/2025] Open
Abstract
Obesity is a global health concern that promotes chronic low-grade inflammation, leading to insulin resistance, a key factor in many metabolic diseases. Angiotensin 1-7 (Ang 1-7), a component of the renin-angiotensin system (RAS), exhibits anti-inflammatory effects in obesity and related disorders, though its mechanisms remain unclear. In this study, we examined the effect of Ang 1-7 on inflammation of white adipose tissue (WAT) in dietary-induced obese mice. Monocyte chemoattractant protein-1 (MCP-1) produced by white adipocytes and tumour necrosis factor-α (TNF-α) produced by macrophages are pro-inflammatory cytokines and interact to form a pathogenic loop to exacerbate obesity-induced inflammation. We found that Ang 1-7 reduced MCP-1 and TNF-α gene expressions and the number of crown-like structures, which are histological hallmarks of the pro-inflammatory process, in visceral epididymal WAT (eWAT) and reduced circulating MCP-1 and TNF-α levels, accompanied by improvement in insulin resistance, in dietary-induced obese mice. Furthermore, Ang 1-7 reduced MCP-1 and TNF-α secretions in 3T3-L1 white adipocytes and RAW 264.7 macrophages, respectively, which are in vitro experimental models mimicking obesity condition. Our results suggest that Ang 1-7 directly acts on WAT to mitigate obesity-induced inflammation. Thus, this study provides novel insights into the underlying mechanism of anti-obesity effects of Ang 1-7.
Collapse
Affiliation(s)
- Nozomi Nishida
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Satoru Sugimoto
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Satoshi Miyagaki
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Chiharu Cho
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Madoka Konishi
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takeshi Goda
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Mihoko Yamaguchi
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yasuhiro Kawabe
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hidechika Morimoto
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Joji Kusuyama
- Department of Biosignals and Inheritance, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
| | - Takuro Okamura
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Masahide Hamaguchi
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Jun Mori
- Division of Pediatric Endocrinology, Metabolism and Nephrology, Children’s Medical Center, Osaka City General Hospital, Osaka, Japan
| | - Hisakazu Nakajima
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Michiaki Fukui
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tomoko Iehara
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
2
|
Dominici FP, Gironacci MM, Narvaez Pardo JA. Therapeutic opportunities in targeting the protective arm of the renin-angiotensin system to improve insulin sensitivity: a mechanistic review. Hypertens Res 2024; 47:3397-3408. [PMID: 39363004 DOI: 10.1038/s41440-024-01909-y] [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: 05/13/2024] [Revised: 08/04/2024] [Accepted: 09/02/2024] [Indexed: 10/05/2024]
Abstract
In recent years, the knowledge of the physiological and pathophysiological roles of the renin-angiotensin system (RAS) in glucose metabolism has advanced significantly. It is now well-established that blockade of the angiotensin AT1 receptor (AT1R) improves insulin sensitivity. Activation of the AT2 receptor (AT2R) and the MAS receptor are significant contributors to this beneficial effect. Elevated availability of angiotensin (Ang) II) for interaction with the AT2R and increased Ang-(1-7) formation during AT1R blockade mediate these effects. The ongoing development of selective AT2R agonists, such as compound 21 and the novel Ang III peptidomimetics, has significantly advanced the exploration of the role of AT2R in metabolism and its potential as a therapeutic target. These agents show promise, particularly when RAS inhibition is contraindicated. Additionally, other RAS peptides, including Ang IV, des-Asp-Ang I, Ang-(1-9), and alamandine, hold therapeutic capability for addressing metabolic disturbances linked to type 2 diabetes. The possibility of AT2R heteromerization with either AT1R or MAS receptor offers an exciting area for future research, particularly concerning therapeutic strategies to improve glycemic control. This review focuses on therapeutic opportunities to improve insulin sensitivity, taking advantage of the protective arm of the RAS.
Collapse
Affiliation(s)
- Fernando P Dominici
- Departamento de Química Biológica and IQUIFIB (UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.
| | - Mariela M Gironacci
- Departamento de Química Biológica and IQUIFIB (UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Jorge A Narvaez Pardo
- Departamento de Química Biológica and IQUIFIB (UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
3
|
Malveira AT, Guimarães VHD, Lima SR, Farias LC, de Paula AMB, Guimarães ALS, Santos SHS. Development of a malnutrition model in mice: Comparative evaluation of food restriction percentage and different diets. J Nutr Biochem 2024; 134:109721. [PMID: 39128608 DOI: 10.1016/j.jnutbio.2024.109721] [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: 12/18/2023] [Revised: 07/24/2024] [Accepted: 08/01/2024] [Indexed: 08/13/2024]
Abstract
Malnutrition is a complicated illness that affects people worldwide and is linked to higher death rates, a heightened vulnerability to infections, and delayed cognitive development. Experimental models have been constructed to comprehend the mechanisms associated with hunger. In this regard, the current study used two different types of food aiming to validate a murine model of malnutrition based on dietary restriction. The study was conducted with fifty-six Swiss male mice (eight-week-old) divided into eight groups (n=7 each) and fed the following experimental diets (10 weeks): Standard Diet (ST) ad libitum; ST 20% dietary restriction; ST 40% dietary restriction; ST 60% dietary restriction; AIN93-M diet ad libitum; AIN93-M 20% dietary restriction; AIN93-M 40% dietary restriction; AIN93-M 60% dietary restriction. Body, biochemical, and histological parameters were measured, and the restriction effects on genes related to oxidative stress (GPX1 and GPX4) in epididymal adipose tissue were evaluated. The results obtained showed that 20%, 40%, and 60% of dietary restrictions were able to reduce body weight when compared to controls, highlighting the accentuated weight loss in animals with 60% restrictions, especially those fed with AIN-93 M, which showed physical changes such as whitish skin and dull coat, voracious eating, and hunched posture. The present animal model also showed biochemical changes with hypoalbuminemia, as well as histological epididymal adipose tissue modulation. The presence of increased oxidative stress was observed when evaluating the GPX4 gene. Given the results, 60% food restriction using the AIN93-M diet was the best protocol for inducing malnutrition.
Collapse
Affiliation(s)
- André Tiago Malveira
- Postgraduate Program in Food and Health. Federal University of Minas Gerais (UFMG), Montes Claros, Minas Gerais, Brazil
| | - Victor Hugo Dantas Guimarães
- Health Sciences Laboratory, Postgraduate Program in Health Sciences, State University of Montes Claros (Unimontes), Minas Gerais, Brazil
| | - Sonielle Rodrigues Lima
- Institute of Agricultural Sciences (ICA), Postgraduate Program in Food and Health, Federal University of Minas Gerais (UFMG), Montes Claros, Minas Gerais, Brazil
| | - Lucyana Conceição Farias
- Health Sciences Laboratory, Postgraduate Program in Health Sciences, State University of Montes Claros (Unimontes), Minas Gerais, Brazil
| | - Alfredo Maurício Batista de Paula
- Health Sciences Laboratory, Postgraduate Program in Health Sciences, State University of Montes Claros (Unimontes), Minas Gerais, Brazil
| | - André Luiz Sena Guimarães
- Health Sciences Laboratory, Postgraduate Program in Health Sciences, State University of Montes Claros (Unimontes), Minas Gerais, Brazil
| | - Sérgio Henrique Sousa Santos
- Postgraduate Program in Food and Health. Federal University of Minas Gerais (UFMG), Montes Claros, Minas Gerais, Brazil; Institute of Agricultural Sciences (ICA), Postgraduate Program in Food and Health, Federal University of Minas Gerais (UFMG), Montes Claros, Minas Gerais, Brazil; Health Sciences Laboratory, Postgraduate Program in Health Sciences, State University of Montes Claros (Unimontes), Minas Gerais, Brazil.
| |
Collapse
|
4
|
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
|
5
|
Nysather J, Kaya E, Manka P, Gudsoorkar P, Syn WK. Nonalcoholic Fatty Liver Disease and Chronic Kidney Disease Cross Talk. ADVANCES IN KIDNEY DISEASE AND HEALTH 2023; 30:315-335. [PMID: 37657879 DOI: 10.1053/j.akdh.2023.04.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 12/14/2022] [Accepted: 04/04/2023] [Indexed: 09/03/2023]
Abstract
Nonalcoholic fatty liver disease is a multisystem condition with effects beyond the liver. The identification of chronic kidney disease as an independent mediator of nonalcoholic fatty liver disease or associated entity with shared cardiometabolic risk factors remains controversial and continues to draw scientific interest. With increasing prevalence of nonalcoholic fatty liver disease and lack of Food and Drug Administration approved therapies, these shared cardiometabolic risk factors have drawn significant attention. In this article, we review shared pathophysiological mechanisms between nonalcoholic fatty liver disease and chronic kidney disease along with current treatment strategies that might be useful for both disease processes.
Collapse
Affiliation(s)
- Jacob Nysather
- Division of Nephrology and Kidney C.A.R.E. Program, University of Cincinnati, OH
| | - Eda Kaya
- Department of Internal Medicine, University Hospital Knappschaftskrankenhaus Bochum, Ruhr-University Bochum, Bochum, Germany
| | - Paul Manka
- Department of Internal Medicine, University Hospital Knappschaftskrankenhaus Bochum, Ruhr-University Bochum, Bochum, Germany
| | - Prakash Gudsoorkar
- Division of Nephrology and Kidney C.A.R.E. Program, University of Cincinnati, OH
| | - Wing-Kin Syn
- Division of Gastroenterology and Hepatology, Saint Louis University School of Medicine, St. Louis, MO; Division of Gastroenterology and Hepatology, Medical University of South Carolina, Charleston, SC; Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country, Euskal Herriko Unibertsitatea/Universidad del País Vasco, Leioa, Spain.
| |
Collapse
|
6
|
Passaglia P, Silva HB, de Jesus AA, Filho MAM, Trajano IP, Batalhão ME, Navegantes LCC, Branco LGS, Cárnio EC. Angiotensin-(1-7) improves tail skin heat loss and increases the survival of rats with polymicrobial sepsis. Peptides 2023; 167:171042. [PMID: 37315714 DOI: 10.1016/j.peptides.2023.171042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 06/02/2023] [Accepted: 06/08/2023] [Indexed: 06/16/2023]
Abstract
Sepsis is a serious syndrome, characterized by the excessive release of inflammatory mediators and thermoregulatory changes, being fever the most common sign. However, despite the importance of Angiotensin (Ang)-(1-7) in controlling the inflammation, the role of the peptide in the febrile response and mortality in animals submitted to experimental model of sepsis is still not clear. In this way, we evaluate the effect of continuous infusion of Ang-(1-7) in inflammatory response, thermoregulation and in mortality of Wistar male rats submitted to colonic ligation puncture (CLP). Before CLP surgery, the infusion pumps (Ang-(1-7), 1.5mg/mL or saline) were inserted into the abdominal cavity and maintained for 24hours. CLP rats showed a febrile response starting from 3h after and persisted until the 24th hour of experiment. Continuous treatment with Ang-(1-7) attenuated the febrile response and reestablished the euthermia 11h after CLP, until the end of experiment, which coincided with an increased heat loss index (HLI). This effect was associated with a decrease in production of pro-inflammatory mediators in liver, white adipose tissue (WAT) and hypothalamus. Moreover, an increase in norepinephrine (NE) content in interscapular brown adipose tissue (iBAT) was observed in CLP animals, which was attenuated with treatment with Ang-(1-7), and decreased mortality in CLP animals treated with Ang-(1-7). Taken together, the present study demonstrates that continuous infusion treatment with Ang-(1-7) can promote a global anti-inflammatory effect, reestablishing the tail skin heat loss as a key thermo-effector function, resulting in an increased survival of animals submitted to experimental sepsis.
Collapse
Affiliation(s)
- Patrícia Passaglia
- Department of Oral and Basic Biology Ribeirão Preto, School of Dentistry of Ribeirão Preto - University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Hadder Batista Silva
- Department of General Nursing, School of Nursing of Ribeirão Preto - University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Aline Alves de Jesus
- Department of Physiology, Ribeirão Preto Medical School - University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Marco Antonio Marangão Filho
- Department of General Nursing, School of Nursing of Ribeirão Preto - University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Isis Paiva Trajano
- Department of Physiology, Ribeirão Preto Medical School - University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Marcelo Eduardo Batalhão
- Department of General Nursing, School of Nursing of Ribeirão Preto - University of São Paulo, Ribeirão Preto, SP, Brazil
| | | | - Luiz Guilherme Siqueira Branco
- Department of Oral and Basic Biology Ribeirão Preto, School of Dentistry of Ribeirão Preto - University of São Paulo, Ribeirão Preto, SP, Brazil; Department of Physiology, Ribeirão Preto Medical School - University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Evelin Capellari Cárnio
- Department of Physiology, Ribeirão Preto Medical School - University of São Paulo, Ribeirão Preto, SP, Brazil; Department of General Nursing, School of Nursing of Ribeirão Preto - University of São Paulo, Ribeirão Preto, SP, Brazil
| |
Collapse
|
7
|
Cerri GC, Santos SHS, Bader M, Santos RAS. Brown adipose tissue transcriptome unveils an important role of the Beta-alanine/alamandine receptor, MrgD, in metabolism. J Nutr Biochem 2023; 114:109268. [PMID: 36641071 DOI: 10.1016/j.jnutbio.2023.109268] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 10/28/2022] [Accepted: 01/05/2023] [Indexed: 01/13/2023]
Abstract
Alamandine is a recently described heptapeptide component of the renin-angiotensin system (RAS), and its effects are mediated by the receptor Mas-related G protein-coupled receptor D (MrgD) RAS represents an important link between obesity and its consequences by directly modulating the thermogenesis and brown adipose tissue (BAT) function. The alamandine/MrgD metabolic effects and signaling remain unexplored. In this context, the main goal of the present study was to assess the metabolic consequences of MrgD genetic ablation in C57BL6/J mice by evaluating brown adipose tissue RNA sequencing. The main results showed that MrgD-KO mice have diminished brown adipose tissue and that a high-glucose diet (HG) decreased both circulating alamandine levels and MrgD expression in BAT from wild-type mice (WT). BAT transcriptome reveals that MrgD-KO HG mice regulated 45 genes, while WT HG mice regulated 1,148 genes. MrgD-KO mice fed a standard diet (ST) compared with WT ST mice regulated 476 genes, of which 445 genes were downregulated. BAT uses the MrgD receptor to display a normal pattern of gene expression and to respond, like WT mice, to an HG diet. In conclusion, the MrgD signaling is important for the metabolic regulation and manutention of BAT functionality.
Collapse
Affiliation(s)
- Gabriela C Cerri
- Laboratory of Hypertension, Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil; National Institute of Science and Technology in Nanobiopharmaceutics (INCT-Nanobiofar), Belo Horizonte, Minas Gerais, Brazil
| | - Sérgio H S Santos
- Institute of Agricultural Sciences, Food Engineering College, Federeal University of Minas Gerais, Montes Claros, Minas Gerais, Brazil
| | - Michael Bader
- Max Delbrück Center for Molecular Medicine, Berlin-Buch, Germany; National Institute of Science and Technology in Nanobiopharmaceutics (INCT-Nanobiofar), Belo Horizonte, Minas Gerais, Brazil
| | - Robson A S Santos
- Laboratory of Hypertension, Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil; National Institute of Science and Technology in Nanobiopharmaceutics (INCT-Nanobiofar), Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
8
|
Guimarães VHD, Lelis DDF, Oliveira LP, Borém LMA, Guimarães FAD, Farias LC, de Paula AMB, Guimarães ALS, Santos SHS. Comparative study of dietary fat: lard and sugar as a better obesity and metabolic syndrome mice model. Arch Physiol Biochem 2023; 129:449-459. [PMID: 33176505 DOI: 10.1080/13813455.2020.1835986] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Diet macronutrient heterogeneity hinders animal studies' data extrapolation from metabolic disorders to human diseases. OBJECTIVE The present study aimed to evaluate different fat-diet compositions' effect on inducing lipid/glucose metabolism alterations in mice. METHODS Swiss male mice were fed for 12 weeks with five different diets: Standard Diet (ST), American Institute of Nutrition 93 for growth (AIN93G) high-butter/high-sugar (HBHS), high-lard/high-sugar (HLHS), and high-oil/high-sugar diet (soybean oil) (HOHS). Several parameters, such as serum biochemistry, histology, and liver mRNA expression, were accessed. RESULTS The main findings revealed that the HLHS diet dramatically altered liver metabolism inducing hepatic steatosis and increased total cholesterol, triglycerides, VLDL, increasing liver CCAAT/enhancer binding protein (CEBP-α), Acetyl-CoA carboxylase (ACC) and Catalase (CAT) mRNA expression. Moreover, the HLHS diet increased glucose intolerance and reduced insulin sensitivity. CONCLUSIONS High-fat/high-sugar diets are efficient to induce obesity and metabolic syndrome-associated alterations, and diets enriched with lard and sugar showed more effective results.
Collapse
Affiliation(s)
- Victor Hugo Dantas Guimarães
- Laboratory of Health Science, Postgraduate Program in Health Science, Universidade Estadual de Montes Claros (Unimontes), Montes Claros, Brasil
| | - Deborah de Farias Lelis
- Laboratory of Health Science, Postgraduate Program in Health Science, Universidade Estadual de Montes Claros (Unimontes), Montes Claros, Brasil
| | - Luis Paulo Oliveira
- Laboratory of Health Science, Postgraduate Program in Health Science, Universidade Estadual de Montes Claros (Unimontes), Montes Claros, Brasil
| | | | - Felipe Alberto Dantas Guimarães
- Laboratory of Health Science, Postgraduate Program in Health Science, Universidade Estadual de Montes Claros (Unimontes), Montes Claros, Brasil
| | - Lucyana Conceição Farias
- Laboratory of Health Science, Postgraduate Program in Health Science, Universidade Estadual de Montes Claros (Unimontes), Montes Claros, Brasil
| | - Alfredo Mauricio Batista de Paula
- Laboratory of Health Science, Postgraduate Program in Health Science, Universidade Estadual de Montes Claros (Unimontes), Montes Claros, Brasil
| | - André Luiz Sena Guimarães
- Laboratory of Health Science, Postgraduate Program in Health Science, Universidade Estadual de Montes Claros (Unimontes), Montes Claros, Brasil
| | - Sérgio Henrique Sousa Santos
- Laboratory of Health Science, Postgraduate Program in Health Science, Universidade Estadual de Montes Claros (Unimontes), Montes Claros, Brasil
- Institute of Agricultural Sciences (ICA), Postgraduate Program in Food and Health, Universidade Federal de Minas Gerais (UFMG), Montes Claros, Brasil
| |
Collapse
|
9
|
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: 5] [Impact Index Per Article: 2.5] [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
|
10
|
Binayi F, Moslemi M, Khodagholi F, Hedayati M, Zardooz H. Long-term high-fat diet disrupts lipid metabolism and causes inflammation in adult male rats: possible intervention of endoplasmic reticulum stress. Arch Physiol Biochem 2023; 129:204-212. [PMID: 32907408 DOI: 10.1080/13813455.2020.1808997] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
This study investigated the effect of long-term high-fat diet (HFD) on plasma lipid profile and probability of inflammation in adult rats. After weaning, male offspring were divided into six groups based on diet type and medication. After 20 weeks of dietary intake, 4-PBA (endoplasmic reticulum (ER) stress inhibitor) was injected for three days. Then, blood samples were taken to measure plasma concentrations of low-density lipoprotein (LDL), triglyceride (TG), high-density lipoprotein (HDL), cholesterol, leptin and interleukin 1-β (IL 1-β). The HFD increased body weight and food intake and intra-abdominal fat and thymus weights, which were associated with elevated plasma leptin level. Moreover, HFD increased plasma concentrations of TG, LDL, cholesterol and IL 1-β and decreased HDL level. Injection of 4-PBA reversed the plasma parameters changes caused by HFD. It seems that long-term HFD feeding through inducing the ER stress, disrupted the lipid metabolism and resulted in inflammation.
Collapse
Affiliation(s)
- Fateme Binayi
- Department of Physiology, School of Medicine, Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Moslemi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- NeuroBiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Hedayati
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Homeira Zardooz
- Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
Godoy-Lugo JA, Mendez DA, Rodriguez R, Nishiyama A, Nakano D, Soñanez-Organis JG, Ortiz RM. Improved lipogenesis gene expression in liver is associated with elevated plasma angiotensin 1-7 after AT1 receptor blockade in insulin-resistant OLETF rats. Mol Cell Endocrinol 2022; 555:111729. [PMID: 35921918 DOI: 10.1016/j.mce.2022.111729] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 07/18/2022] [Accepted: 07/21/2022] [Indexed: 11/19/2022]
Abstract
Increased angiotensin II (Ang II) signaling contributes to insulin resistance and liver steatosis. In addition to ameliorating hypertension, angiotensin receptor blockers (ARBs) improve lipid metabolism and hepatic steatosis, which are impaired with metabolic syndrome (MetS). Chronic blockade of the Ang II receptor type 1 (AT1) increases plasma angiotensin 1-7 (Ang 1-7), which mediates mechanisms counterregulatory to AT1 signaling. Elevated plasma Ang 1-7 is associated with decreased plasma triacylglycerol (TAG), cholesterol, glucose, and insulin; however, the benefits of RAS modulation to prevent non-alcoholic fatty liver disease (NAFLD) are not fully investigated. To better address the relationships among chronic ARB treatment, plasma Ang 1-7, and hepatic steatosis, three groups of 10-week-old-rats were studied: (1) untreated lean Long Evans Tokushima Otsuka (LETO), (2) untreated Otsuka Long Evans Tokushima Fatty (OLETF), and (3) OLETF + ARB (ARB; 10 mg olmesartan/kg/d × 6 weeks). Following overnight fasting, rats underwent an acute glucose load to better understand the dynamic metabolic responses during hepatic steatosis and early MetS. Tissues were collected at baseline (pre-load; T0) and 1 and 2 h post-glucose load. AT1 blockade increased plasma Ang 1-7 and decreased liver lipids, which was associated with decreased fatty acid transporter 5 (FATP5) and fatty acid synthase (FASN) expression. AT1 blockade decreased liver glucose and increased glucokinase (GCK) expression. These results demonstrate that during MetS, overactivation of AT1 promotes hepatic lipid deposition that is stimulated by an acute glucose load and lipogenesis genes, suggesting that the chronic hyperglycemia associated with MetS contributes to fatty liver pathologies via an AT1-mediated mechanism.
Collapse
Affiliation(s)
- Jose A Godoy-Lugo
- School of Natural Sciences, University of California, Merced, CA, USA.
| | - Dora A Mendez
- School of Natural Sciences, University of California, Merced, CA, USA
| | - Ruben Rodriguez
- School of Natural Sciences, University of California, Merced, CA, USA
| | - Akira Nishiyama
- Department of Pharmacology, Kagawa University Medical School, Kagawa, Japan
| | - Daisuke Nakano
- Department of Pharmacology, Kagawa University Medical School, Kagawa, Japan
| | - Jose G Soñanez-Organis
- Universidad de Sonora, Departamento de Ciencias Químico Biológicas y Agropecuarias, Navojoa, Sonora, Mexico
| | - Rudy M Ortiz
- School of Natural Sciences, University of California, Merced, CA, USA
| |
Collapse
|
12
|
Pioglitazone Synthetic Analogue Ameliorates Streptozotocin-Induced Diabetes Mellitus through Modulation of ACE 2/Angiotensin 1–7 via PI3K/AKT/mTOR Signaling Pathway. Pharmaceuticals (Basel) 2022; 15:ph15030341. [PMID: 35337139 PMCID: PMC8955304 DOI: 10.3390/ph15030341] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/04/2022] [Accepted: 03/08/2022] [Indexed: 02/01/2023] Open
Abstract
The renin angiotensin aldosterone system has a localized key regulatory action, especially in liver and body circulation. Furthermore, it accomplishes a significant role in the downregulation of the PI3K/AKT/mTOR signaling pathway that is involved in type II diabetes mellitus pathogenesis. The current study aimed to evaluate the effect of a synthetic pioglitazone analogue (benzenesulfonamide derivative) compared to the standard pioglitazone hypoglycemic drug on enhancing liver insulin sensitivity via ACE 2/Ang (1–7)/PI3K/AKT/mTOR in experimental STZ-induced diabetes. After the model was established, rats were distributed into the normal control group, diabetic group, pioglitazone group (20 mg/kg), and a benzenesulfonamide derivative group (20 mg/kg), with the last 2 groups receiving oral treatment for 14 consecutive days. Our results suggested enhancing liver insulin sensitivity against the ACE2/Ang (1–7)/PI3K/AKT/mTOR pathway. Moreover, the synthetic compound produced a reduction in blood glucose levels, restored hyperinsulinemia back to normal, and enhanced liver glycogen deposition. In addition, it up regulated the ACE2/Ang (1–7)/PI3K/AKT/mTOR signaling pathway via increasing insulin receptor substrate 1 and 2 sensitivity to insulin, while it increased glucose transporter 2 expression in the rat pancreas. The study findings imply that the hypoglycemic effect of the benzenesulfonamide derivative is due to enhancing liver sensitivity to regulate blood glucose level via the ACE2/Ang (1–7)/PI3K/AKT/mTOR pathway.
Collapse
|
13
|
Sabri S, Bourron O, Phan F, Nguyen LS. Interactions between diabetes and COVID-19: A narrative review. World J Diabetes 2021; 12:1674-1692. [PMID: 34754370 PMCID: PMC8554367 DOI: 10.4239/wjd.v12.i10.1674] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/29/2021] [Accepted: 08/31/2021] [Indexed: 02/06/2023] Open
Abstract
Diabetes, whether due to pancreatic beta cells insufficiency or peripheral resistance to insulin, has been suggested as a risk factor of developing severe acute respiratory disease coronavirus-2 (SARS-CoV-2) infections. Indeed, diabetes has been associated with a higher risk of infections and higher risk of developing severe forms of coronavirus disease 2019 (COVID-19) related pneumonia. Diabetic patients often present associated comorbidities such as obesity, hypertension and cardiovascular diseases, and complications of diabetes, including chronic kidney disease, vasculopathy and relative immune dysfunction, all of which make them more susceptible to infectious complications. Moreover, they often present low-grade inflammation with increased circulating interleukin levels, endothelial susceptibility to inflammation and dysfunction, and finally, hyperglycemia, which increases this risk. Additionally, corticosteroids, which count among the few medications which showed benefit on survival and mechanical ventilation requirement in COVID-19 pneumonia in large randomized controlled trials, are associated to new onsets of diabetes, and metabolic disorders in patients with previous history of diabetes. Finally, SARS-CoV-2 via the alternate effects of the renin-angiotensin system, mediated by the angiotensin-converting-enzyme 2, was also associated with insulin resistance in key tissues involved in glucose homeostasis, such as liver, skeletal muscles, and adipose tissue; and also, with impaired insulin secretion by pancreatic β-cells. In this work, we reviewed all elements which may help understand how diabetes affects patients with COVID-19, how treatments affect outcomes in patients with COVID-19, how they may cause new onsets of diabetes, and finally review how SARS-CoV-2 may inherently be a risk factor of developing diabetes, through immune-mediated diabetogenic mechanisms.
Collapse
Affiliation(s)
- Sophia Sabri
- Intensive Care Medicine, CMC Ambroise Paré, Neuilly-Sur-Seine 92200, France
| | - Olivier Bourron
- Sorbonne Université Médecine; Assistance publique Hôpitaux de Paris (APHP), Service de Diabétologie, Hôpital Pitié-Salpêtrière; INSERM UMRS_1138, Centre de recherche des Cordeliers; Institute of CArdiometabolisme and Nutrition (ICAN), Paris 75013, France
| | - Franck Phan
- Sorbonne Université Médecine; Assistance publique Hôpitaux de Paris (APHP), Service de Diabétologie, Hôpital Pitié-Salpêtrière; INSERM UMRS_1138, Centre de recherche des Cordeliers; Institute of CArdiometabolisme and Nutrition (ICAN), Paris 75013, France
| | - Lee S Nguyen
- Research and Innovation, RICAP, CMC Ambroise Paré, Neuilly-Sur-Seine 92200, France
| |
Collapse
|
14
|
Machado AS, Oliveira JR, de F Lelis D, D Guimarães VH, de Paula AMB, Guimarães ALS, Brandi IV, de Carvalho BMA, da Costa DV, Vieira CR, Pereira UA, de Oliveira Costa T, Andrade JMO, Dos Santos RAS, Santos SHS. Oral angiotensin-(1-7) peptide modulates intestinal microbiota improving metabolic profile in obese mice. Protein Pept Lett 2021; 28:1127-1137. [PMID: 34397321 DOI: 10.2174/0929866528666210816115645] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/25/2021] [Accepted: 06/15/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Obesity is a serious health problem which dysregulate Renin-Angiotensin System and intestinal microbiota. OBJECTIVE The present study aimed to evaluate the Angiotensin-(1-7) [ANG-(1-7)] oral formulation effects on obese mice intestinal microbiota. METHODS Mice were divided into four groups: obese and non-obese treated with ANG-(1-7) and obese and non-obese without ANG-(1-7) during four weeks. RESULTS We observed a significant decrease in the fasting plasma glucose, total cholesterol, triglycerides, and Low-density lipoprotein levels and increased High-density lipoprotein in animals treated with ANG-(1-7). The histological analysis showed intestinal villi height reduction in mice treated with ANG-(1-7). Additionally, increased Bacteroidetes and decreased Firmicutes (increased Bacteroidetes/Firmicutes ratio) and Enterobacter cloacae populations were observed in the High-Fat Diet + ANG-(1-7) group. Receptor toll-like 4 (TLR4) intestinal mRNA expression was reduced in the HFD+ ANG-(1-7) group. Finally, the intestinal expression of the neutral amino acid transporter (B0AT1) was increased in animals treated with ANG-(1-7), indicating a possible mechanism associated with tryptophan uptake. CONCLUSION The results of the present study suggest for the first time an interaction between oral ANG-(1-7) and intestinal microbiota modulation.
Collapse
Affiliation(s)
- Amanda S Machado
- Laboratory of Health Science, Postgraduate Program in Health Sciences, Montes Claros, Minas Gerais, Brazil
| | - Janaína R Oliveira
- Laboratory of Health Science, Postgraduate Program in Health Sciences, Montes Claros, Minas Gerais, Brazil
| | - Deborah de F Lelis
- Laboratory of Health Science, Postgraduate Program in Health Sciences, Montes Claros, Minas Gerais, Brazil
| | - Victor Hugo D Guimarães
- Laboratory of Health Science, Postgraduate Program in Health Sciences, Montes Claros, Minas Gerais, Brazil
| | - Alfredo M B de Paula
- Laboratory of Health Science, Postgraduate Program in Health Sciences, Montes Claros, Minas Gerais, Brazil
| | - André L S Guimarães
- Laboratory of Health Science, Postgraduate Program in Health Sciences, Montes Claros, Minas Gerais, Brazil
| | - Igor V Brandi
- Institute of Agricultural Sciences. Food Engineering College, Universidade Federal de Minas Gerais (UFMG), Montes Claros, Minas Gerais, Brazil
| | - Bruna Mara A de Carvalho
- Institute of Agricultural Sciences. Food Engineering College, Universidade Federal de Minas Gerais (UFMG), Montes Claros, Minas Gerais, Brazil
| | - Diego Vicente da Costa
- Institute of Agricultural Sciences. Food Engineering College, Universidade Federal de Minas Gerais (UFMG), Montes Claros, Minas Gerais, Brazil
| | - Cláudia Regina Vieira
- Institute of Agricultural Sciences. Food Engineering College, Universidade Federal de Minas Gerais (UFMG), Montes Claros, Minas Gerais, Brazil
| | - Ulisses Alves Pereira
- Institute of Agricultural Sciences. Food Engineering College, Universidade Federal de Minas Gerais (UFMG), Montes Claros, Minas Gerais, Brazil
| | - Theles de Oliveira Costa
- Institute of Agricultural Sciences. Food Engineering College, Universidade Federal de Minas Gerais (UFMG), Montes Claros, Minas Gerais, Brazil
| | - João M O Andrade
- Institute of Agricultural Sciences. Food Engineering College, Universidade Federal de Minas Gerais (UFMG), Montes Claros, Minas Gerais, Brazil
| | - Robson A S Dos Santos
- Institute of Biological Sciences, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Sérgio H S Santos
- Laboratory of Health Science, Postgraduate Program in Health Sciences, Montes Claros, Minas Gerais, Brazil
| |
Collapse
|
15
|
Gómez-Zorita S, Milton-Laskibar I, García-Arellano L, González M, Portillo MP. An Overview of Adipose Tissue ACE2 Modulation by Diet and Obesity. Potential Implications in COVID-19 Infection and Severity. Int J Mol Sci 2021; 22:7975. [PMID: 34360741 PMCID: PMC8347022 DOI: 10.3390/ijms22157975] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/18/2021] [Accepted: 07/21/2021] [Indexed: 12/11/2022] Open
Abstract
The present review is aimed at analysing the current evidence concerning the potential modulation of obesity and/or diet in adipose tissue ACE2. Additionally, the potential implications of these effects on COVID-19 are also addressed. The results published show that diet and obesity are two factors that effectively influence the expression of Ace2 gene in adipose tissue. However, the shifts in this gene do not always occur in the same direction, nor with the same intensity. Additionally, there is no consensus regarding the implications of increased adipose tissue ACE2 expression in health. Thus, while in some studies a protective role is attributed to ACE2 overexpression, other studies suggest otherwise. Similarly, there is much debate regarding the role played by ACE2 in COVID-19 in terms of degree of infection and disease outcomes. The greater risk of infection that may hypothetically derive from enhanced ACE2 expression is not clear since the functionality of the enzyme seems to be as important as the abundance. Thus, the greater abundance of ACE2 in adipose tissue of obese subjects may be counterbalanced by its lower activation. In addition, a protective role of ACE2 overexpression has also been suggested, associated with the increase in anti-inflammatory factors that it may produce.
Collapse
Affiliation(s)
- Saioa Gómez-Zorita
- Nutrition and Obesity Group, Lucio Lascaray Research Center, Department of Nutrition and Food Science, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), 01006 Vitoria-Gasteiz, Spain; (L.G.-A.); (M.P.P.)
- CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
- BIOARABA Health Research Institute, 01006 Vitoria-Gasteiz, Spain
| | - Iñaki Milton-Laskibar
- CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
- Precision Nutrition and Cardiometabolic Health, IMDEA-Food Institute (Madrid Institute for Advanced Studies), Campus of International Excellence (CEI) UAM+CSIC, Spanish National Research Council, 28049 Madrid, Spain
| | - Laura García-Arellano
- Nutrition and Obesity Group, Lucio Lascaray Research Center, Department of Nutrition and Food Science, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), 01006 Vitoria-Gasteiz, Spain; (L.G.-A.); (M.P.P.)
| | - Marcela González
- Nutrition and Food Science Department, Faculty of Biochemistry and Biological Sciences, National University of Litoral and National Scientific and Technical Research Council (CONICET), Santa Fe 3000, Argentina;
| | - María P. Portillo
- Nutrition and Obesity Group, Lucio Lascaray Research Center, Department of Nutrition and Food Science, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), 01006 Vitoria-Gasteiz, Spain; (L.G.-A.); (M.P.P.)
- CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
- BIOARABA Health Research Institute, 01006 Vitoria-Gasteiz, Spain
| |
Collapse
|
16
|
Laghlam D, Jozwiak M, Nguyen LS. Renin-Angiotensin-Aldosterone System and Immunomodulation: A State-of-the-Art Review. Cells 2021; 10:cells10071767. [PMID: 34359936 PMCID: PMC8303450 DOI: 10.3390/cells10071767] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 06/30/2021] [Accepted: 07/09/2021] [Indexed: 12/11/2022] Open
Abstract
The renin–angiotensin system (RAS) has long been described in the field of cardiovascular physiology as the main player in blood pressure homeostasis. However, other effects have since been described, and include proliferation, fibrosis, and inflammation. To illustrate the immunomodulatory properties of the RAS, we chose three distinct fields in which RAS may play a critical role and be the subject of specific treatments. In oncology, RAS hyperactivation has been associated with tumor migration, survival, cell proliferation, and angiogenesis; preliminary data showed promise of the benefit of RAS blockers in patients treated for certain types of cancer. In intensive care medicine, vasoplegic shock has been associated with severe macro- and microcirculatory imbalance. A relative insufficiency in angiotensin II (AngII) was associated to lethal outcomes and synthetic AngII has been suggested as a specific treatment in these cases. Finally, in solid organ transplantation, both AngI and AngII have been associated with increased rejection events, with a regional specificity in the RAS activity. These elements emphasize the complexity of the direct and indirect interactions of RAS with immunomodulatory pathways and warrant further research in the field.
Collapse
|
17
|
Faria BCD, Sacramento LGG, Filipin CSA, da Cruz AF, Nagata SN, Silva ACSE. An analysis of chronic kidney disease as a prognostic factor in pediatric cases of COVID-19. J Bras Nefrol 2021; 43:400-409. [PMID: 33704348 PMCID: PMC8428649 DOI: 10.1590/2175-8239-jbn-2020-0208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 12/17/2020] [Indexed: 11/24/2022] Open
Abstract
Advanced age is a risk factor for severe infection by acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Children, however, often present with milder manifestations of Coronavirus Disease 2019 (COVID-19). Associations have been found between COVID-19 and multisystem inflammatory syndrome in children (MIS-C). Patients with the latter condition present more severe involvement. Adults with comorbidities such as chronic kidney disease (CKD) are more severely affected. This narrative review aimed to look into whether CKD contributed to more severe involvement in pediatric patients with COVID-19. The studies included in this review did not report severe cases or deaths, and indicated that pediatric patients with CKD and previously healthy children recovered quickly from infection. However, some patients with MIS-C required hospitalization in intensive care units and a few died, although it was not possible to correlate MIS-C and CKD. Conversely, adults with CKD reportedly had increased risk of severe infection by SARS-CoV-2 and higher death rates. The discrepancies seen between age groups may be due to immune system and renin-angiotensin system differences, with more pronounced expression of ACE2 in children. Immunosuppressant therapy has not been related with positive or negative effects in individuals with COVID-19, although current recommendations establish decreases in the dosage of some medications. To sum up with, CKD was not associated with more severe involvement in children diagnosed with COVID-19. Studies enrolling larger populations are still required.
Collapse
Affiliation(s)
| | | | | | - Aniel Feitosa da Cruz
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Belo
Horizonte, MG, Brasil
| | - Sarah Naomi Nagata
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Belo
Horizonte, MG, Brasil
| | - Ana Cristina Simões e Silva
- Universidade Federal de Minas Gerais, Faculdade de Medicina,
Departamento de Pediatria, Belo Horizonte, MG, Brasil
| |
Collapse
|
18
|
Nozato Y, Yamamoto K. Angiotensin-(1-7) as a biomarker of childhood obesity: Is there a causal relationship? Hypertens Res 2021; 44:1233-1235. [PMID: 34155361 PMCID: PMC8215480 DOI: 10.1038/s41440-021-00684-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 04/13/2021] [Accepted: 04/14/2021] [Indexed: 11/09/2022]
Affiliation(s)
- Yoichi Nozato
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.
| | - Koichi Yamamoto
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.
| |
Collapse
|
19
|
ACE2 and energy metabolism: the connection between COVID-19 and chronic metabolic disorders. Clin Sci (Lond) 2021; 135:535-554. [PMID: 33533405 DOI: 10.1042/cs20200752] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/19/2021] [Accepted: 01/21/2021] [Indexed: 12/13/2022]
Abstract
The renin-angiotensin system (RAS) has currently attracted increasing attention due to its potential function in regulating energy homeostasis, other than the actions on cellular growth, blood pressure, fluid, and electrolyte balance. The existence of RAS is well established in metabolic organs, including pancreas, liver, skeletal muscle, and adipose tissue, where activation of angiotensin-converting enzyme (ACE) - angiotensin II pathway contributes to the impairment of insulin secretion, glucose transport, fat distribution, and adipokines production. However, the activation of angiotensin-converting enzyme 2 (ACE2) - angiotensin (1-7) pathway, a novel branch of the RAS, plays an opposite role in the ACE pathway, which could reverse these consequences by improving local microcirculation, inflammation, stress state, structure remolding, and insulin signaling pathway. In addition, new studies indicate the protective RAS arm possesses extraordinary ability to enhance brown adipose tissue (BAT) activity and induces browning of white adipose tissue, and consequently, it leads to increased energy expenditure in the form of heat instead of ATP synthesis. Interestingly, ACE2 is the receptor of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which is threating public health worldwide. The main complications of SARS-CoV-2 infected death patients include many energy metabolism-related chronic diseases, such as diabetes. The specific mechanism leading to this phenomenon is largely unknown. Here, we summarize the latest pharmacological and genetic tools on regulating ACE/ACE2 balance and highlight the beneficial effects of the ACE2 pathway axis hyperactivity on glycolipid metabolism, as well as the thermogenic modulation.
Collapse
|
20
|
Evangelista FS. Physical Exercise and the Renin Angiotensin System: Prospects in the COVID-19. Front Physiol 2020; 11:561403. [PMID: 33178033 PMCID: PMC7593780 DOI: 10.3389/fphys.2020.561403] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 09/16/2020] [Indexed: 12/13/2022] Open
Abstract
Recent reports have shown that the renin angiotensin system (RAS) plays an important role in the Coronavirus disease 2019 (COVID-19) because the angiotensin converting enzyme 2 is the receptor for the severe acute respiratory syndrome coronavirus 2. In addition, the balance of RAS components can be involved in the pathogenesis and progression of COVID-19, especially in patients with metabolic and cardiovascular diseases. On the other hand, physical exercise is effective to prevent and to counteract the consequences of such diseases and one of the biological mediators of the exercise adaptation is the RAS. This review was designed to highlight the connection between COVID-19 and RAS, and to discuss the role of the RAS as a mediator of the benefits of physical exercise in COVID-19 pandemic.
Collapse
|
21
|
Oliveira LP, Guimarães VHD, Oliveira JR, Guimarães ALS, de Paula AMB, Bader M, Santos RASD, Santos SHS. Genetic deletion of the angiotensin-(1-7) receptor Mas leads to alterations in gut villi length modulating TLR4/PI3K/AKT and produces microbiome dysbiosis. Neuropeptides 2020; 82:102056. [PMID: 32505463 DOI: 10.1016/j.npep.2020.102056] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 04/17/2020] [Accepted: 05/12/2020] [Indexed: 02/06/2023]
Abstract
Renin-Angiotensin System (RAS) is an important peptide cascade involved in physiological processes. RAS homeostasis disruption produces several cardiovascular and metabolic disorders, such as arterial hypertension, atherosclerosis, acute myocardial infarct, obesity, diabetes, metabolic syndrome and increases gastrointestinal tract (GIT) cell proliferation. Angiotensin (Ang)-(1-7) peptide is the main RAS counter-regulatory axis effector. It is formed from ACE2 enzyme and acts mainly through Mas receptor (MasR). In this context, the aim of the present study was to evaluate alterations in small intestine morphology and intestinal microbiota composition in MasR knockout C57BL/6 mice. We analyzed glucose tolerance; insulin sensitivity and blood collected for biochemical parameters as well as small intestine tissues samples for immunohistochemistry. mRNA and bacteria gDNA expression evaluation. mRNA expression was evaluated by qRT-PCR for TLR4, PI3K and AKT. The main results showed that Mas-R-knockout mice presented lower body weight. MasR-knockout mice also presented increased fasted blood glucose and total cholesterol with reduced HDL, lower glucose tolerance and impaired insulin sensitivity. Increased intestinal mucosa length, increased intestinal villi, reduced Lieberkühn crypt depth. The increased expression of cell proliferation markers Ki-67 and Cyclin D1 and increased TLR4, PI3K and AKT expressions were observed with augmented Bacteroidetes and decreased amount of Firmicutes. That results suggests that MasR deletion generated changes in intestinal microbiota, possibly due to a lower neutral amino acids absorption followed by a compensatory increase in intestinal villi length associated with disbiosis and LPS overproduction that ultimately lead to proliferation and cell inflammation.
Collapse
Affiliation(s)
- Luis Paulo Oliveira
- Laboratory of Health Science, Postgraduate Program in Health Science, Universidade Estadual de Montes Claros (Unimontes), Minas Gerais, Brazil
| | - Victor Hugo Dantas Guimarães
- Laboratory of Health Science, Postgraduate Program in Health Science, Universidade Estadual de Montes Claros (Unimontes), Minas Gerais, Brazil
| | - Janaina Ribeiro Oliveira
- Laboratory of Health Science, Postgraduate Program in Health Science, Universidade Estadual de Montes Claros (Unimontes), Minas Gerais, Brazil
| | - André Luiz Sena Guimarães
- Laboratory of Health Science, Postgraduate Program in Health Science, Universidade Estadual de Montes Claros (Unimontes), Minas Gerais, Brazil
| | - Alfredo Maurício Batista de Paula
- Laboratory of Health Science, Postgraduate Program in Health Science, Universidade Estadual de Montes Claros (Unimontes), Minas Gerais, Brazil
| | - Michael Bader
- Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | - Robson Augusto Souza Dos Santos
- Institute of Biological Sciences (ICB), Physiology Department, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | - Sérgio Henrique Sousa Santos
- Institute of Agricultural Sciences (ICA), Food Engineering, Universidade Federal de Minas Gerais (UFMG), Montes Claros, Minas Gerais, Brazil.
| |
Collapse
|
22
|
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: 7.5] [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
|
23
|
NAFLD and Extra-Hepatic Comorbidities: Current Evidence on a Multi-Organ Metabolic Syndrome. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:ijerph16183415. [PMID: 31540048 PMCID: PMC6765902 DOI: 10.3390/ijerph16183415] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 09/06/2019] [Accepted: 09/08/2019] [Indexed: 02/06/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common cause of chronic liver disease worldwide and its incidence is definitely increasing. NAFLD is a metabolic disease with extensive multi-organ involvement, whose extra-hepatic manifestations include type 2 diabetes mellitus, cardiovascular disease, obstructive sleep apnea, chronic kidney disease, osteoporosis, and polycystic ovarian syndrome. Recently, further evidence has given attention to pathological correlations not strictly related to metabolic disease, also incorporating in this broad spectrum of systemic involvement hypothyroidism, psoriasis, male sexual dysfunction, periodontitis, and urolithiasis. The most common cause of mortality in NAFLD is represented by cardiovascular disease, followed by liver-related complications. Therefore, clinicians should learn to screen and initiate treatment for these extra-hepatic manifestations, in order to provide appropriate multidisciplinary assessments and rigorous surveillance. This review evaluates the current evidence regarding extra-hepatic associations of NAFLD, focusing on the pathogenic hypothesis and the clinical implications.
Collapse
|
24
|
White MC, Fleeman R, Arnold AC. Sex differences in the metabolic effects of the renin-angiotensin system. Biol Sex Differ 2019; 10:31. [PMID: 31262355 PMCID: PMC6604144 DOI: 10.1186/s13293-019-0247-5] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 06/18/2019] [Indexed: 02/07/2023] Open
Abstract
Obesity is a global epidemic that greatly increases risk for developing cardiovascular disease and type II diabetes. Sex differences in the obese phenotype are well established in experimental animal models and clinical populations. While having higher adiposity and obesity prevalence, females are generally protected from obesity-related metabolic and cardiovascular complications. This protection is, at least in part, attributed to sex differences in metabolic effects of hormonal mediators such as the renin-angiotensin system (RAS). Previous literature has predominantly focused on the vasoconstrictor arm of the RAS and shown that, in contrast to male rodent models of obesity and diabetes, females are protected from metabolic and cardiovascular derangements produced by angiotensinogen, renin, and angiotensin II. A vasodilator arm of the RAS has more recently emerged which includes angiotensin-(1-7), angiotensin-converting enzyme 2 (ACE2), mas receptors, and alamandine. While accumulating evidence suggests that activation of components of this counter-regulatory axis produces positive effects on glucose homeostasis, lipid metabolism, and energy balance in male animal models, female comparison studies and clinical data related to metabolic outcomes are lacking. This review will summarize current knowledge of sex differences in metabolic effects of the RAS, focusing on interactions with gonadal hormones and potential clinical implications.
Collapse
Affiliation(s)
- Melissa C White
- Department of Comparative Medicine, Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA, USA
| | - Rebecca Fleeman
- Department of Neural and Behavioral Sciences, Pennsylvania State University College of Medicine, 500 University Drive, Mail Code H109, Hershey, PA, 17033, USA
| | - Amy C Arnold
- Department of Neural and Behavioral Sciences, Pennsylvania State University College of Medicine, 500 University Drive, Mail Code H109, Hershey, PA, 17033, USA.
| |
Collapse
|
25
|
Lelis DDF, Freitas DFD, Machado AS, Crespo TS, Santos SHS. Angiotensin-(1-7), Adipokines and Inflammation. Metabolism 2019; 95:36-45. [PMID: 30905634 DOI: 10.1016/j.metabol.2019.03.006] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 03/11/2019] [Accepted: 03/20/2019] [Indexed: 12/12/2022]
Abstract
Nowadays the adipose tissue is recognized as one of the most critical endocrine organs releasing many adipokines that regulate metabolism, inflammation and body homeostasis. There are several described adipokines, including the renin-angiotensin system (RAS) components that are especially activated in some diseases with increased production of angiotensin II and several pro-inflammatory hormones. On the other hand, RAS also expresses angiotensin-(1-7), which is now recognized as the main peptide on counteracting Ang II effects. New studies have shown that increased activation of ACE2/Ang-(1-7)/MasR arm can revert and prevent local and systemic dysfunctions improving lipid profile and insulin resistance by modulating insulin actions, and reducing inflammation. In this context, the present review shows the interaction and relevance of Ang-(1-7) effects on regulating adipokines, and as one adipokine itself, modulating body homeostasis, with emphasis on its anti-inflammatory properties, especially in the context of metabolic disorders with focus on obesity and type 2 diabetes mellitus pandemic.
Collapse
Affiliation(s)
- Deborah de Farias Lelis
- Laboratory of Health Sciences, Post Graduate Program in Health Sciences, Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Minas Gerais, Brazil
| | - Daniela Fernanda de Freitas
- Laboratory of Health Sciences, Post Graduate Program in Health Sciences, Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Minas Gerais, Brazil
| | - Amanda Souto Machado
- Laboratory of Health Sciences, Post Graduate Program in Health Sciences, Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Minas Gerais, Brazil
| | - Thaísa Soares Crespo
- Laboratory of Health Sciences, Post Graduate Program in Health Sciences, Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Minas Gerais, Brazil
| | - Sérgio Henrique Sousa Santos
- Institute of Agricultural Sciences, Food Engineering College, Universidade Federal de Minas Gerais (UFMG), Montes Claros, Minas Gerais, Brazil; Laboratory of Health Sciences, Post Graduate Program in Health Sciences, Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Minas Gerais, Brazil.
| |
Collapse
|
26
|
Américo ALV, Muller CR, Vecchiatto B, Martucci LF, Fonseca-Alaniz MH, Evangelista FS. Aerobic exercise training prevents obesity and insulin resistance independent of the renin angiotensin system modulation in the subcutaneous white adipose tissue. PLoS One 2019; 14:e0215896. [PMID: 31022246 PMCID: PMC6483229 DOI: 10.1371/journal.pone.0215896] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 04/10/2019] [Indexed: 12/15/2022] Open
Abstract
We investigate the effects of aerobic exercise training (AET) on the thermogenic response, substrate metabolism and renin angiotensin system (RAS) in the subcutaneous white adipose tissue (SC-WAT) of mice fed cafeteria diet (CAF). Male C57BL/6J mice were assigned into groups CHOW-SED (chow diet, sedentary; n = 10), CHOW-TR (chow diet, trained; n = 10), CAF-SED (CAF, sedentary; n = 10) and CAF-TR (CAF, trained; n = 10). AET consisted in running sessions of 60 min at 60% of maximal speed, five days per week for eight weeks. The CAF-SED group showed higher body weight and adiposity, glucose intolerance and insulin resistance (IR), while AET prevented such damages in CAF-TR group. AET reduced the p-AKT/t-AKT ratio and increased ATGL expression in CHOW-TR and CAF-TR groups and increased t-HSL and p-HSL/t-HSL ratio in CAF-TR. AET prevented adipocyte hypertrophy in CAF-TR group and increased UCP-1 protein expression only in CHOW-TR. Serum ACE2 increased in CHOW-TR and CAF-TR groups, and Ang (1–7) increased in the CHOW-TR group. In the SC-WAT, CAF-TR group increased the expression of AT1, AT2 and Mas receptors, whereas CHOW-TR increased Ang (1–7) and Ang (1–7)/Ang II ratio in SC-WAT. No changes were observed in ACE and Ang II. Positive correlations were observed between UCP-1 and kITT (r = 0.6), between UCP-1 and Ang (1–7) concentration (r = 0.6), and between UCP-1 and Ang (1–7)/Ang II ratio (r = 0.7). In conclusion, the AET prevented obesity and IR, reduced insulin signaling proteins and increased lipolysis signaling proteins in the SC-WAT. In addition, the CAF diet precludes the AET-induced thermogenic response and the partial modulation of the RAS suggests that the protective effect of AET against obesity and IR could not be associated with SC-WAT RAS.
Collapse
Affiliation(s)
- Anna Laura V. Américo
- Department of Experimental Pathophysiology, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Cynthia R. Muller
- Department of Experimental Pathophysiology, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Bruno Vecchiatto
- School of Arts, Science and Humanities, University of Sao Paulo, São Paulo, Brazil
| | - Luiz Felipe Martucci
- Department of Experimental Pathophysiology, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | | | - Fabiana S. Evangelista
- School of Arts, Science and Humanities, University of Sao Paulo, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
27
|
Barbosa MA, de Sousa GG, de Castro UGM, Carneiro CM, Figueiredo VP, Guerra de Sá R, Santos RASD, Alzamora AC. Oral Ang-(1-7) treatment improves white adipose tissue remodeling and hypertension in rats with metabolic syndrome. Nutrition 2019; 67-68S:100004. [PMID: 34332714 DOI: 10.1016/j.nutx.2019.100004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 09/01/2019] [Accepted: 10/05/2019] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Angiotensin (Ang)-(1-7) has preventive effects on metabolic syndrome (MetS). The aim of this study was to evaluate the therapeutic effect of oral Ang-(1-7) on mean arterial pressure (MAP), insulin resistance (IR), inflammatory process, and remodeling of white adipose tissue (WAT) in rats with established MetS. METHODS Rats were subjected to control (CT; AIN-93M) or high-fat (HF) diets for 13 wk to induce MetS and treated with Ang-(1-7) or vehicle (V) for the last 6 wk. At the end of 13 wk, MAP, biochemical and histological parameters, and uncoupling protein (UCP) and inflammatory gene expression were determined by quantitative reverse transcription polymerase chain reaction. RESULTS HF-V rats showed increased visceral fat deposition, inflammatory cytokine expression, hyperplasia, and hypertrophy in retroperitoneal (WAT) and brown adipose tissue (BAT). Additionally, the gastrocnemius muscle reduced UCP-3 and increased the UCP-1 expression in BAT. HF-V also elevated levels of plasma insulin, glucose, homeostatic model assessment (HOMA) of IR and HOMA-β, and increased body mass, adiposity, and MAP. Ang-(1-7) treatment in rats with MetS [HF-Ang-(1-7)] reduced WAT area, number of adipocytes, and expression of proinflammatory adipokines in WAT and BAT and increased UCP-3 in gastrocnemius muscle and UCP-1 expression in BAT compared with the HF-V group. These events prevented body mass gain, reduced adiposity, and normalized fasting plasma glucose, insulin levels, HOMA-IR, HOMA-β, and MAP. CONCLUSION Data from the present study demonstrated that oral Ang-(1-7) treatment is effective in restoring biochemical parameters and hypertension in established MetS by improving hypertrophy and hyperplasia in WAT and inflammation in adipose tissue, and regulating metabolic processes in the gastrocnemius muscle and BAT.
Collapse
Affiliation(s)
- Maria Andréa Barbosa
- Núcleo de Pesquisa em Ciências Biológicas, Universidade Federal de Ouro Preto, MG, Brazil
| | | | | | | | | | - Renata Guerra de Sá
- Núcleo de Pesquisa em Ciências Biológicas, Universidade Federal de Ouro Preto, MG, Brazil; Departamento de Ciências Biológicas, Instituto de Ciências Exatas e Biológicas Ouro Preto, MG, Brazil
| | | | - Andréia Carvalho Alzamora
- Núcleo de Pesquisa em Ciências Biológicas, Universidade Federal de Ouro Preto, MG, Brazil; Departamento de Ciências Biológicas, Instituto de Ciências Exatas e Biológicas Ouro Preto, MG, Brazil.
| |
Collapse
|
28
|
Zhang Y, Somers KR, Becari C, Polonis K, Pfeifer MA, Allen AM, Kellogg TA, Covassin N, Singh P. Comparative Expression of Renin-Angiotensin Pathway Proteins in Visceral Versus Subcutaneous Fat. Front Physiol 2018; 9:1370. [PMID: 30364113 PMCID: PMC6191467 DOI: 10.3389/fphys.2018.01370] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 09/10/2018] [Indexed: 01/12/2023] Open
Abstract
Body fat distribution contributes to obesity-related metabolic and cardiovascular disorders. Visceral fat is more detrimental than subcutaneous fat. However, the mechanisms underlying visceral fat-mediated cardiometabolic dysregulation are not completely understood. Localized increases in expression of the renin angiotensin system (RAS) in adipose tissue (AT) may be implicated. We therefore investigated mRNA and protein expression of RAS components in visceral versus subcutaneous AT using paired samples from individuals undergoing surgery (N = 20, body mass index: 45.6 ± 6.2 kg/m2, and age: 44.6 ± 9.1 years). We also examined RAS-related proteins in AT obtained from individuals on renin angiotensin aldosterone system (RAAS) targeted drugs (N = 10, body mass index: 47.2 ± 9.3 kg/m2, and age: 53.3 ± 10.1 years). Comparison of protein expression between subcutaneous and visceral AT samples showed an increase in renin (p = 0.004) and no change in angiotensinogen (p = 0.987) expression in visceral AT. Among proteins involved in angiotensin peptide generation, angiotensin converting enzyme (p = 0.02) was increased in subcutaneous AT while chymase (p = 0.001) and angiotensin converting enzyme-2 (p = 0.001) were elevated in visceral fat. Furthermore, visceral fat expression of angiotensin II type-2 receptor (p = 0.007) and angiotensin II type-1 receptor (p = 0.031) was higher, and MAS receptor (p < 0.001) was lower. Phosphorylated-p53 (p = 0.147), AT fibrosis (p = 0.138) and average adipocyte size (p = 0.846) were similar in the two depots. Nonetheless, visceral AT showed increased mRNA expression of inflammatory (TNFα, p < 0.001; IL-6, p = 0.001) and oxidative stress markers (NOX2, p = 0.038; NOX4, p < 0.001). Of note, mRNA and protein expression of RAS components did not differ between subjects taking or not taking RAAS related drugs. In summary, several RAS related proteins are differentially expressed in subcutaneous versus visceral AT. This differential expression may not alter AngII but likely increases Ang1-7 generation in visceral fat. These potential differences in active angiotensin peptides and receptor expression in the two depots suggest that localized RAS may not be involved in differences in visceral vs subcutaneous AT function in obese individuals. Our findings do not support a role for localized RAS differences in visceral fat-mediated development of cardiovascular and metabolic pathology.
Collapse
Affiliation(s)
- Yuebo Zhang
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| | - Kiran R Somers
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| | - Christiane Becari
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| | - Katarzyna Polonis
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| | - Michaela A Pfeifer
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| | - Alina M Allen
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, United States
| | - Todd A Kellogg
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
| | - Naima Covassin
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| | - Prachi Singh
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
29
|
Borém LMA, Neto JFR, Brandi IV, Lelis DF, Santos SHS. The role of the angiotensin II type I receptor blocker telmisartan in the treatment of non-alcoholic fatty liver disease: a brief review. Hypertens Res 2018; 41:394-405. [PMID: 29636553 PMCID: PMC7091617 DOI: 10.1038/s41440-018-0040-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 10/31/2017] [Accepted: 11/17/2017] [Indexed: 01/18/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is currently considered an important component of metabolic syndrome (MetS). The spectrum of NAFLD includes conditions that range from simple hepatic steatosis to non-alcoholic steatohepatitis. NAFLD is correlated with liver-related death and is predicted to be the most frequent indication for liver transplantation by 2030. Insulin resistance is directly correlated to the central mechanisms of hepatic steatosis in NAFLD patients, which is strongly correlated to the imbalance of the renin–angiotensin system, that is involved in lipid and glucose metabolism. Among the emerging treatment approaches for NAFLD is the anti-hypertensive agent telmisartan, which has positive effects on liver, lipid, and glucose metabolism, especially through its action on the renin–angiotensin system, by blocking the ACE/AngII/AT1 axis and increasing ACE2/Ang(1–7)/Mas axis activation. However, treatment with this drug is only recommended for patients with an established indication for anti-hypertensive therapy. Thus, there is an increased need for large randomized controlled trials with the aim of elucidating the effects of telmisartan on liver disease, especially NAFLD. From this perspective, the present review aims to provide a brief examination of the pathogenesis of NAFLD/NASH and the role of telmisartan on preventing liver disorders and thus to improve the discussion on potential therapies.
Collapse
Affiliation(s)
- Luciana M A Borém
- Laboratory of Health Science, Postgraduate Program in Health Sciences, Universidade Estadual de Montes Claros (Unimontes), Montes Claros, Minas Gerais, Brazil.,Medicine Department, Faculdades Integradas Pitágoras, Montes Claros, Minas Gerais, Brazil
| | - João F R Neto
- Laboratory of Health Science, Postgraduate Program in Health Sciences, Universidade Estadual de Montes Claros (Unimontes), Montes Claros, Minas Gerais, Brazil
| | - Igor V Brandi
- Institute of Agricultural Sciences, Food Engineering College, Universidade Federal de Minas Gerais (UFMG), Montes Claros, Minas Gerais, Brazil
| | - Deborah F Lelis
- Laboratory of Health Science, Postgraduate Program in Health Sciences, Universidade Estadual de Montes Claros (Unimontes), Montes Claros, Minas Gerais, Brazil
| | - Sergio H S Santos
- Laboratory of Health Science, Postgraduate Program in Health Sciences, Universidade Estadual de Montes Claros (Unimontes), Montes Claros, Minas Gerais, Brazil. .,Institute of Agricultural Sciences, Food Engineering College, Universidade Federal de Minas Gerais (UFMG), Montes Claros, Minas Gerais, Brazil.
| |
Collapse
|
30
|
Jie X, Li X, Song JQ, Wang D, Wang JH. Anti-inflammatory and autonomic effects of electroacupuncture in a rat model of diet-induced obesity. Acupunct Med 2018; 36:103-109. [PMID: 29487062 DOI: 10.1136/acupmed-2016-011223] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2017] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To study the effect of electroacupuncture (EA) on the cholinergic anti-inflammatory pathway (CAP) by measurement of vagal activity in rats with high-fat diet (HFD)-induced obesity. METHODS Diet-induced obesity (DIO) was induced in 30 rats by feeding them a HFD for 12 weeks. A further 10 rats fed normal food comprised the lean diet (LD) control group. DIO rats were further subdivided into three groups that received a HFD only (HFD group, n=10), a HFD plus electroacupuncture (HFD+EA group, n=10) or a HFD plus minimal acupuncture (HFD+MA group, n=10). EA and MA treatments were continued for 8 weeks. Heart rate variability (HRV) was used to measure the function of the autonomic nervous system before and after treatment. ELISA was used to determine acetylcholine (ACh) and tumour necrosis factor (TNF)-α levels in the serum. Real-time PCR was used to assess the mRNA expression of α7-subtype nicotinic acetylcholine cholinergic receptors (α7nAChRs) and TNF-α in the mesenteric white adipose tissues (MWAT). RESULTS EA but not MA significantly reduced rats' bodyweight. No difference was found in the low frequency (LF), high frequency (HF) and the balance between LF and HF (LF/HF) components of HRV before treatment. After the EA intervention, HF was elevated and LF/HF was reduced in the HFD+EA group comparedwith the HFD group. TNF-α in the serum and MWAT were increased in the HFD group, but were reduced in the HFD+EA group. Furthermore, EA promoted expression of α7nAChRs and ACh in the MWAT. There was no difference between the HFD and HFD+MA groups for any indices. CONCLUSIONS EA enhanced vagal activity, promoted ACh release and activated α7nAChRs in the MWAT, leading to inhibition of proinflammatory cytokine production.
Collapse
Affiliation(s)
- Xiaoyan Jie
- Department of Neurosurgical Intensive Care Unit, Nanyang Second General Hospital, Nanyang, Henan, China
| | - Xu Li
- Department of Medicine, Nanyang Second General Hospital, Nanyang, Henan, China
| | - Jian-Qing Song
- Department of Neurosurgical Intensive Care Unit, Nanyang Second General Hospital, Nanyang, Henan, China
| | - Dan Wang
- Department of Neurosurgical Intensive Care Unit, Nanyang Second General Hospital, Nanyang, Henan, China
| | - Jian-Hua Wang
- Department of Neurosurgical Intensive Care Unit, Nanyang Second General Hospital, Nanyang, Henan, China
| |
Collapse
|
31
|
Santos RAS, Sampaio WO, Alzamora AC, Motta-Santos D, Alenina N, Bader M, Campagnole-Santos MJ. The ACE2/Angiotensin-(1-7)/MAS Axis of the Renin-Angiotensin System: Focus on Angiotensin-(1-7). Physiol Rev 2018; 98:505-553. [PMID: 29351514 PMCID: PMC7203574 DOI: 10.1152/physrev.00023.2016] [Citation(s) in RCA: 774] [Impact Index Per Article: 110.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 05/09/2017] [Accepted: 06/18/2017] [Indexed: 12/16/2022] Open
Abstract
The renin-angiotensin system (RAS) is a key player in the control of the cardiovascular system and hydroelectrolyte balance, with an influence on organs and functions throughout the body. The classical view of this system saw it as a sequence of many enzymatic steps that culminate in the production of a single biologically active metabolite, the octapeptide angiotensin (ANG) II, by the angiotensin converting enzyme (ACE). The past two decades have revealed new functions for some of the intermediate products, beyond their roles as substrates along the classical route. They may be processed in alternative ways by enzymes such as the ACE homolog ACE2. One effect is to establish a second axis through ACE2/ANG-(1-7)/MAS, whose end point is the metabolite ANG-(1-7). ACE2 and other enzymes can form ANG-(1-7) directly or indirectly from either the decapeptide ANG I or from ANG II. In many cases, this second axis appears to counteract or modulate the effects of the classical axis. ANG-(1-7) itself acts on the receptor MAS to influence a range of mechanisms in the heart, kidney, brain, and other tissues. This review highlights the current knowledge about the roles of ANG-(1-7) in physiology and disease, with particular emphasis on the brain.
Collapse
Affiliation(s)
- Robson Augusto Souza Santos
- National Institute of Science and Technology in Nanobiopharmaceutics, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais , Belo Horizonte , Brazil ; Department of Biological Sciences, Federal University of Ouro Preto , Ouro Preto , Brazil ; Max-Delbrück-Center for Molecular Medicine (MDC), Berlin , Germany ; Berlin Institute of Health (BIH), Berlin , Germany ; Charité - University Medicine, Berlin , Germany ; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin , Germany ; Institute for Biology, University of Lübeck , Lübeck , Germany
| | - Walkyria Oliveira Sampaio
- National Institute of Science and Technology in Nanobiopharmaceutics, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais , Belo Horizonte , Brazil ; Department of Biological Sciences, Federal University of Ouro Preto , Ouro Preto , Brazil ; Max-Delbrück-Center for Molecular Medicine (MDC), Berlin , Germany ; Berlin Institute of Health (BIH), Berlin , Germany ; Charité - University Medicine, Berlin , Germany ; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin , Germany ; Institute for Biology, University of Lübeck , Lübeck , Germany
| | - Andreia C Alzamora
- National Institute of Science and Technology in Nanobiopharmaceutics, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais , Belo Horizonte , Brazil ; Department of Biological Sciences, Federal University of Ouro Preto , Ouro Preto , Brazil ; Max-Delbrück-Center for Molecular Medicine (MDC), Berlin , Germany ; Berlin Institute of Health (BIH), Berlin , Germany ; Charité - University Medicine, Berlin , Germany ; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin , Germany ; Institute for Biology, University of Lübeck , Lübeck , Germany
| | - Daisy Motta-Santos
- National Institute of Science and Technology in Nanobiopharmaceutics, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais , Belo Horizonte , Brazil ; Department of Biological Sciences, Federal University of Ouro Preto , Ouro Preto , Brazil ; Max-Delbrück-Center for Molecular Medicine (MDC), Berlin , Germany ; Berlin Institute of Health (BIH), Berlin , Germany ; Charité - University Medicine, Berlin , Germany ; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin , Germany ; Institute for Biology, University of Lübeck , Lübeck , Germany
| | - Natalia Alenina
- National Institute of Science and Technology in Nanobiopharmaceutics, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais , Belo Horizonte , Brazil ; Department of Biological Sciences, Federal University of Ouro Preto , Ouro Preto , Brazil ; Max-Delbrück-Center for Molecular Medicine (MDC), Berlin , Germany ; Berlin Institute of Health (BIH), Berlin , Germany ; Charité - University Medicine, Berlin , Germany ; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin , Germany ; Institute for Biology, University of Lübeck , Lübeck , Germany
| | - Michael Bader
- National Institute of Science and Technology in Nanobiopharmaceutics, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais , Belo Horizonte , Brazil ; Department of Biological Sciences, Federal University of Ouro Preto , Ouro Preto , Brazil ; Max-Delbrück-Center for Molecular Medicine (MDC), Berlin , Germany ; Berlin Institute of Health (BIH), Berlin , Germany ; Charité - University Medicine, Berlin , Germany ; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin , Germany ; Institute for Biology, University of Lübeck , Lübeck , Germany
| | - Maria Jose Campagnole-Santos
- National Institute of Science and Technology in Nanobiopharmaceutics, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais , Belo Horizonte , Brazil ; Department of Biological Sciences, Federal University of Ouro Preto , Ouro Preto , Brazil ; Max-Delbrück-Center for Molecular Medicine (MDC), Berlin , Germany ; Berlin Institute of Health (BIH), Berlin , Germany ; Charité - University Medicine, Berlin , Germany ; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin , Germany ; Institute for Biology, University of Lübeck , Lübeck , Germany
| |
Collapse
|
32
|
Obesity and malnutrition similarly alter the renin–angiotensin system and inflammation in mice and human adipose. J Nutr Biochem 2017; 48:74-82. [DOI: 10.1016/j.jnutbio.2017.06.008] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 06/09/2017] [Accepted: 06/19/2017] [Indexed: 12/18/2022]
|
33
|
Frantz EDC, Giori IG, Machado MV, Magliano DC, Freitas FM, Andrade MSB, Vieira AB, Nóbrega ACL, Tibiriçá E. High, but not low, exercise volume shifts the balance of renin-angiotensin system toward ACE2/Mas receptor axis in skeletal muscle in obese rats. Am J Physiol Endocrinol Metab 2017; 313:E473-E482. [PMID: 28679623 DOI: 10.1152/ajpendo.00078.2017] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 06/12/2017] [Accepted: 06/30/2017] [Indexed: 01/10/2023]
Abstract
Metabolic syndrome is a cluster of metabolic risk factors that is linked to central obesity, elevated blood pressure, insulin resistance (IR), and dyslipidemia, where the renin-angiotensin system (RAS) may provide a link among them. This study aimed to evaluate volume exercise effects comparing low vs. high volume of chronic aerobic exercise on RAS axes in skeletal muscle in a diet-induced obesity (DIO) rat model. For this, male Wistar-Kyoto rats were fed a standard chow (SC) diet or a high-fat (HF) diet for 32 wk. Animals receiving the HF diet were randomly divided into low exercise volume (LEV, 150 min/wk) and high exercise volume (HEV, 300 min/wk) at the 20th week. After 12 wk of aerobic treadmill training, the body mass and composition, blood pressure, glucose and lipid metabolism, RAS axes, insulin signaling, and inflammatory pathway were performed. HEV slowed the body mass gain, reduced intra-abdominal fat pad and leptin levels, improved total and peripheral body composition and inflammatory cytokine, reduced angiotensin II type 1 receptor expression, and increased Mas receptor protein expression compared with the HF animals. Sedentary groups (SC and HF) presented lower time to exhaustion and maximal velocity compared with the LEV and HEV groups. Both exercise training groups showed reduced resting systolic blood pressure and heart rate, improved glucose tolerance, IR, insulin signaling, and lipid profile. We conclude that the HEV, but not LEV, shifted the balance of RAS toward the ACE2/Mas receptor axis in skeletal muscle, presenting protective effects against the DIO model.
Collapse
Affiliation(s)
- Eliete Dalla Corte Frantz
- Laboratory of Exercise Sciences, Department of Physiology and Pharmacology, Fluminense Federal University, Niteroi, Brazil; and
| | - Isabele Gomes Giori
- Laboratory of Exercise Sciences, Department of Physiology and Pharmacology, Fluminense Federal University, Niteroi, Brazil; and
| | - Marcus Vinícius Machado
- Laboratory of Exercise Sciences, Department of Physiology and Pharmacology, Fluminense Federal University, Niteroi, Brazil; and
- Laboratory of Cardiovascular Investigation, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
| | - D'Angelo Carlo Magliano
- Laboratory of Exercise Sciences, Department of Physiology and Pharmacology, Fluminense Federal University, Niteroi, Brazil; and
| | - Fernanda Marques Freitas
- Laboratory of Exercise Sciences, Department of Physiology and Pharmacology, Fluminense Federal University, Niteroi, Brazil; and
| | - Mariana Sodré Boêta Andrade
- Laboratory of Exercise Sciences, Department of Physiology and Pharmacology, Fluminense Federal University, Niteroi, Brazil; and
| | - Aline Bomfim Vieira
- Laboratory of Cardiovascular Investigation, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
| | - Antonio Claudio Lucas Nóbrega
- Laboratory of Exercise Sciences, Department of Physiology and Pharmacology, Fluminense Federal University, Niteroi, Brazil; and
| | - Eduardo Tibiriçá
- Laboratory of Cardiovascular Investigation, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
| |
Collapse
|
34
|
Genetic variation in the promoter region of the TNF rs1800629 gene is not associated with adiposity index, but AA genotype is more likely to have low cellular membrane integrity. Meta Gene 2017. [DOI: 10.1016/j.mgene.2017.05.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
35
|
Moreira CCL, Lourenço FC, Mario ÉG, Santos RAS, Botion LM, Chaves VE. Long-term effects of angiotensin-(1-7) on lipid metabolism in the adipose tissue and liver. Peptides 2017; 92:16-22. [PMID: 28438644 DOI: 10.1016/j.peptides.2017.04.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 04/11/2017] [Accepted: 04/12/2017] [Indexed: 12/11/2022]
Abstract
The angiotensin (Ang) converting enzyme 2/Ang-(1-7)/Mas axis has been described to have a beneficial role on metabolic disorders. In the present study, the use of a transgenic rat model that chronically overexpresses Ang-(1-7) enabled us to investigate the chronic effects of this peptide on lipid accumulation in the liver and adipose tissue. The transgenic group showed a marked tendency toward increased expression of peroxisome proliferator-activated receptor-γ (PPARγ) and decreased lipoprotein lipase (LPL) expression and activity in epididymal adipose tissue. We also showed that Mas receptor-knockout mice had decreased PPARγ expression in adipose tissue, accompanied by an increase in LPL activity. These results confirm the regulation of adipose tissue LPL activity by Ang-(1-7) and suggest that this occurs independent of PPARγ expression. The reduced adiposity index of transgenic rats, due to the effect of Ang-(1-7), was accompanied by a decrease in lipogenesis. These findings suggest a direct effect of Ang-(1-7) on lipogenesis, independent of the stimulatory effect of insulin. Furthermore, the decreased concentration of triacylglycerol in the liver of transgenic rats may result from increased activity of cytosolic lipases and decreased fatty acid uptake from the adipose tissue, determined from fatty acid-binding protein expression, and hepatic de novo fatty acid synthesis, evaluated by fatty acid synthase expression. The data clearly show that Ang-(1-7) regulates lipid metabolism in the adipose tissue and liver.
Collapse
Affiliation(s)
- Carolina Campos Lima Moreira
- Department of Physiology and Biophysics, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Fabíola Cesário Lourenço
- Department of Physiology and Biophysics, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Érica Guilhen Mario
- Department of Physiology and Biophysics, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Robson Augusto Souza Santos
- Department of Physiology and Biophysics, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Leida Maria Botion
- Department of Physiology and Biophysics, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Valéria Ernestânia Chaves
- Laboratory of Physiology, Federal University of São João del-Rei, Divinópolis, Minas Gerais, Brazil.
| |
Collapse
|
36
|
Tang A, Li C, Zou N, Zhang Q, Liu M, Zhang X. Angiotensin-(1-7) improves non-alcoholic steatohepatitis through an adiponectin-independent mechanism. Hepatol Res 2017; 47:116-122. [PMID: 26992300 DOI: 10.1111/hepr.12707] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 02/15/2016] [Accepted: 03/11/2016] [Indexed: 01/12/2023]
Abstract
AIM Recent evidence suggests that angiotensin-(1-7) [Ang-(1-7)] could improve non-alcoholic steatohepatitis (NASH) through an adiponectin-dependent mechanism. This study aimed to investigate whether and how Ang-(1-7) influences NASH without adiponectin. METHODS Adiponectin knockout mice were fed with a high fat diet (HFD) or normal chow for 6 months, and were subsequently infused with Ang-(1-7) or saline for 2 weeks. RESULTS We found that HFD-fed mice showed obesity, hyperlipidemia, NASH, and significantly increased levels of serum Ang-(1-7). Chronic infusion of Ang-(1-7) could reduce body weight, absolute and relative liver weight, and serum levels of total cholesterol, triglyceride, and low-density lipoprotein cholesterol in HFD-fed mice. In addition, Ang-(1-7) treatment could attenuate hepatocellular inflammation, steatosis, and ballooning with activation of the hepatic AMP-activated protein kinase signaling pathway in HFD-fed knockout mice. CONCLUSIONS These results showed the protective role of Ang-(1-7) in the development of NASH through an adiponectin-independent mechanism, which may be partially attributed to the activation of hepatic AMP-activated protein kinase pathway.
Collapse
Affiliation(s)
- Ailian Tang
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Can Li
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Nan Zou
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qian Zhang
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Meiling Liu
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xia Zhang
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
37
|
Slamkova M, Zorad S, Krskova K. Alternative renin-angiotensin system pathways in adipose tissue and their role in the pathogenesis of obesity. Endocr Regul 2016; 50:229-240. [DOI: 10.1515/enr-2016-0025] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Abstract
Adipose tissue expresses all the renin-angiotensin system (RAS) components that play an important role in the adipogenesis, lipid and glucose metabolism regulation in an auto/paracrine manner. The classical RAS has been found to be over-activated during the adipose tissue enlargement, thus elevated generation of angiotensin II (Ang II) may contribute to the obesity pathogenesis. The contemporary view on the RAS has become more complex with the discovery of alternative pathways, including angiotensin-converting enzyme 2 (ACE2)/angiotensin (Ang)-(1-7)/Mas receptor, (pro)renin receptor, as well as angiotensin IV(Ang IV)/AT4 receptor. Ang-(1-7) via Mas receptor counteracts with most of the deleterious effects of the Ang II-mediated by AT1 receptor implying its beneficial role in the glucose and lipid metabolism, oxidative stress, inflammation, and insulin resistance. Pro(renin) receptor may play a role (at least partial) in the pathogenesis of the obesity by increasing the local production of Ang II in adipose tissue as well as triggering signal transduction independently of Ang II. In this review, modulation of alternative RAS pathways in adipose tissue during obesity is discussed and the involvement of Ang-(1-7), (pro)renin and AT4 receptors in the regulation of adipose tissue homeostasis and insulin resistance is summarized.
Collapse
Affiliation(s)
- M Slamkova
- Institute of Experimental Endocrinology, Biomedical Research Centre, Slovak Academy of Sciences, Bratislava, Slovakia
| | - S Zorad
- Institute of Experimental Endocrinology, Biomedical Research Centre, Slovak Academy of Sciences, Bratislava, Slovakia
| | - K Krskova
- Institute of Experimental Endocrinology, Biomedical Research Centre, Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
38
|
Metformin Reduces Lipogenesis Markers in Obese Mice Fed a Low-Carbohydrate and High-Fat Diet. Lipids 2016; 51:1375-1384. [DOI: 10.1007/s11745-016-4209-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 10/18/2016] [Indexed: 12/17/2022]
|
39
|
Nguyen Dinh Cat A, Antunes TT, Callera GE, Sanchez A, Tsiropoulou S, Dulak-Lis MG, Anagnostopoulou A, He Y, Montezano AC, Jaisser F, Touyz RM. Adipocyte-Specific Mineralocorticoid Receptor Overexpression in Mice Is Associated With Metabolic Syndrome and Vascular Dysfunction: Role of Redox-Sensitive PKG-1 and Rho Kinase. Diabetes 2016; 65:2392-403. [PMID: 27207514 DOI: 10.2337/db15-1627] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 04/09/2016] [Indexed: 11/13/2022]
Abstract
Mineralocorticoid receptor (MR) expression is increased in adipose tissue from obese individuals and animals. We previously demonstrated that adipocyte-MR overexpression (Adipo-MROE) in mice is associated with metabolic changes. Whether adipocyte MR directly influences vascular function in these mice is unknown. We tested this hypothesis in resistant mesenteric arteries from Adipo-MROE mice using myography and in cultured adipocytes. Molecular mechanisms were probed in vessels/vascular smooth muscle cells and adipose tissue/adipocytes and focused on redox-sensitive pathways, Rho kinase activity, and protein kinase G type-1 (PKG-1) signaling. Adipo-MROE versus control-MR mice exhibited reduced vascular contractility, associated with increased generation of adipocyte-derived hydrogen peroxide, activation of vascular redox-sensitive PKG-1, and downregulation of Rho kinase activity. Associated with these vascular changes was increased elastin content in Adipo-MROE. Inhibition of PKG-1 with Rp-8-Br-PET-cGMPS normalized vascular contractility in Adipo-MROE. In the presence of adipocyte-conditioned culture medium, anticontractile effects of the adipose tissue were lost in Adipo-MROE mice but not in control-MR mice. In conclusion, adipocyte-MR upregulation leads to impaired contractility with preserved endothelial function and normal blood pressure. Increased elasticity may contribute to hypocontractility. We also identify functional cross talk between adipocyte MR and arteries and describe novel mechanisms involving redox-sensitive PKG-1 and Rho kinase. Our results suggest that adipose tissue from Adipo-MROE secrete vasoactive factors that preferentially influence vascular smooth muscle cells rather than endothelial cells. Our findings may be important in obesity/adiposity where adipocyte-MR expression/signaling is amplified and vascular risk increased.
Collapse
Affiliation(s)
- Aurelie Nguyen Dinh Cat
- Cardiovascular Research and Medical Sciences Institute, University of Glasgow, Glasgow, U.K.
| | - Tayze T Antunes
- Kidney Research Centre, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Glaucia E Callera
- Kidney Research Centre, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Ana Sanchez
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense, Madrid, Spain
| | - Sofia Tsiropoulou
- Cardiovascular Research and Medical Sciences Institute, University of Glasgow, Glasgow, U.K
| | - Maria G Dulak-Lis
- Cardiovascular Research and Medical Sciences Institute, University of Glasgow, Glasgow, U.K
| | | | - Ying He
- Kidney Research Centre, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Augusto C Montezano
- Cardiovascular Research and Medical Sciences Institute, University of Glasgow, Glasgow, U.K
| | - Frederic Jaisser
- INSERM Unit 1138 Team 1, Centre de Recherche des Cordeliers, University Pierre and Marie Curie, Paris, France INSERM, Clinical Investigation Centre 1430, APHP, Henri Mondor Hospital, Pole VERDI, Paris East University, Creteil, France
| | - Rhian M Touyz
- Cardiovascular Research and Medical Sciences Institute, University of Glasgow, Glasgow, U.K. Kidney Research Centre, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
40
|
Musso G, Cassader M, Cohney S, Pinach S, Saba F, Gambino R. Emerging Liver-Kidney Interactions in Nonalcoholic Fatty Liver Disease. Trends Mol Med 2016; 21:645-662. [PMID: 26432021 DOI: 10.1016/j.molmed.2015.08.005] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2015] [Revised: 08/07/2015] [Accepted: 08/16/2015] [Indexed: 12/12/2022]
Abstract
Mounting evidence connects non-alcoholic fatty liver disease (NAFLD) to chronic kidney disease (CKD). We review emerging mechanistic links between NAFLD and CKD, including altered activation of angiotensin converting enzyme (ACE)-2, nutrient/energy sensors sirtuin-1 and AMP-activated kinase, as well as impaired antioxidant defense mediated by nuclear factor erythroid 2-related factor-2 (Nrf2). Dietary fructose excess may also contribute to NAFLD and CKD. NAFLD affects renal injury through lipoprotein dysmetabolism and altered secretion of the hepatokines fibroblast growth factor-21, fetuin-A, insulin-like growth factor-1, and syndecan-1. CKD may mutually aggravate NAFLD and associated metabolic disturbances through altered intestinal barrier function and microbiota composition, the accumulation of uremic toxic metabolites, and alterations in pre-receptor glucocorticoid metabolism. We conclude by discussing the implications of these findings for the treatment of NAFLD and CKD.
Collapse
Affiliation(s)
| | - Maurizio Cassader
- Department of Medical Sciences, San Giovanni Battista Hospital, University of Turin, Turin, Italy
| | - Solomon Cohney
- Department of Nephrology, Royal Melbourne and Western Hospital, Victoria, University of Melbourne, Melbourne, Australia
| | - Silvia Pinach
- Department of Medical Sciences, San Giovanni Battista Hospital, University of Turin, Turin, Italy
| | - Francesca Saba
- Department of Medical Sciences, San Giovanni Battista Hospital, University of Turin, Turin, Italy
| | - Roberto Gambino
- Department of Medical Sciences, San Giovanni Battista Hospital, University of Turin, Turin, Italy
| |
Collapse
|
41
|
Angiotensin-converting enzyme 2/angiotensin-(1-7)/Mas axis activates Akt signaling to ameliorate hepatic steatosis. Sci Rep 2016; 6:21592. [PMID: 26883384 PMCID: PMC4756304 DOI: 10.1038/srep21592] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 01/27/2016] [Indexed: 02/08/2023] Open
Abstract
The classical axis of renin-angiotensin system (RAS), angiotensin (Ang)-converting enzyme (ACE)/Ang II/AT1, contributes to the development of non-alcoholic fatty liver disease (NAFLD). However, the role of bypass axis of RAS (Angiotensin-converting enzyme 2 (ACE2)/Ang-(1–7)/Mas) in hepatic steatosis is still unclear. Here we showed that deletion of ACE2 aggravates liver steatosis, which is correlated with the increased expression of hepatic lipogenic genes and the decreased expression of fatty acid oxidation-related genes in the liver of ACE2 knockout (ACE2−/y) mice. Meanwhile, oxidative stress and inflammation were also aggravated in ACE2−/y mice. On the contrary, overexpression of ACE2 improved fatty liver in db/db mice, and the mRNA levels of fatty acid oxidation-related genes were up-regulated. In vitro, Ang-(1–7)/ACE2 ameliorated hepatic steatosis, oxidative stress and inflammation in free fatty acid (FFA)-induced HepG2 cells, and what’s more, Akt inhibitors reduced ACE2-mediated lipid metabolism. Furthermore, ACE2-mediated Akt activation could be attenuated by blockade of ATP/P2 receptor/Calmodulin (CaM) pathway. These results indicated that Ang-(1–7)/ACE2/Mas axis may reduce liver lipid accumulation partly by regulating lipid-metabolizing genes through ATP/P2 receptor/CaM signaling pathway. Our findings support the potential role of ACE2/Ang-(1–7)/Mas axis in prevention and treatment of hepatic lipid metabolism.
Collapse
|
42
|
Blanke K, Schlegel F, Raasch W, Bader M, Dähnert I, Dhein S, Salameh A. Effect of Angiotensin(1-7) on Heart Function in an Experimental Rat Model of Obesity. Front Physiol 2015; 6:392. [PMID: 26733884 PMCID: PMC4685089 DOI: 10.3389/fphys.2015.00392] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 12/02/2015] [Indexed: 12/02/2022] Open
Abstract
Aim: Obesity is a risk factor for the development of cardiovascular diseases. Recently it was shown that overexpression of the Mas-receptor antagonist angiotensin(1-7) could prevent from diet-induced obesity. However, it remained unclear whether diet-induced obesity and angiotensin(1-7) overexpression might also have effects on the cardiovascular system in these rats. Methods:Twenty three male Sprague Dawley rats were fed with standard chow (SD+chow, n = 5) or a cafeteria diet (SD+CD, n = 6) for 5 months. To investigate the effect of angiotensin(1-7) transgenic rats, expressing an angiotensin(1-7)-producing fusion protein in testis were used. These transgenic rats also received a 5 month's feeding period with either chow (TGR+chow, n = 6) or cafeteria diet (TGR+CD, n = 6), respectively. Hemodynamic measurements (pressure-volume loops) were carried out to assess cardiac function and blood pressure. Subsequently, hearts were explanted and investigated according to the Langendorff technique. Furthermore, cardiac remodeling in these animals was investigated histologically. Results:After 5 months cafeteria diet feeding rats showed a significantly increased body weight, which could be prevented in transgenic rats. However, there was no effect on cardiac performance after cafeteria diet in non-transgenic and transgenic rats. Moreover, overexpression of angiotensin(1-7) deteriorated cardiac contractility as indicated by impaired dp/dt. Furthermore, histological analysis revealed that cafeteria diet led to myocardial fibrosis in both, control and transgenic rats and this was not inhibited by an overproduction of angiotensin(1-7). Conclusion:These results indicate that an overexpression of circulating angiotensin(1-7) prevents a cafeteria diet-induced increase in body weight, but does not affect cardiac performance in this experimental rat model of obesity. Furthermore, overexpression of angiotensin(1-7) alone resulted in an impairment of cardiac function.
Collapse
Affiliation(s)
- Katja Blanke
- Department of Pediatric Cardiology, Heart Center Leipzig, University of Leipzig Leipzig, Germany
| | - Franziska Schlegel
- Clinic for Cardiac Surgery, Heart Center Leipzig, University of Leipzig Leipzig, Germany
| | - Walter Raasch
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck Lübeck, Germany
| | - Michael Bader
- Max-Delbrück Center for Molecular Medicine, Helmholtz-Gemeinschaft Berlin, Germany
| | - Ingo Dähnert
- Department of Pediatric Cardiology, Heart Center Leipzig, University of Leipzig Leipzig, Germany
| | - Stefan Dhein
- Clinic for Cardiac Surgery, Heart Center Leipzig, University of Leipzig Leipzig, Germany
| | - Aida Salameh
- Department of Pediatric Cardiology, Heart Center Leipzig, University of Leipzig Leipzig, Germany
| |
Collapse
|
43
|
Kim M, Lim SJ, Lee HJ, Kim SY, Nho CW. Gomisin J Inhibits Oleic Acid-Induced Hepatic Lipogenesis by Activation of the AMPK-Dependent Pathway and Inhibition of the Hepatokine Fetuin-A in HepG2 Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2015; 63:9729-9739. [PMID: 26455261 DOI: 10.1021/acs.jafc.5b04089] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The aim of our study is to investigate the molecular mechanism of gomisin J from Schisandra chinensis on the oleic acid (OA)-induced lipid accumulation in HepG2 cells. Gomisin J attenuated lipid accumulation in OA-induced HepG2 cells. It also suppressed the expression of lipogenic enzymes and inflammatory mediators and increased the expression of lipolytic enzymes in OA-induced HepG2 cells. Furthermore, the use of specific inhibitors and fetuin-A siRNA and liver kinase B1 (LKB1) siRNA transfected cells demonstrated that gomisin J regulated lipogenesis and lipolysis via inhibition of fetuin-A and activation of an AMP-activated protein kinase (AMPK)-dependent pathway in HepG2 cells. Our results showed that gomisin J suppressed lipid accumulation by regulating the expression of lipogenic and lipolytic enzymes and inflammatory molecules through activation of AMPK, LKB1, and Ca(2+)/calmodulin-dependent protein kinase II and inhibition of fetuin-A in HepG2 cells. This suggested that gomisin J has potential benefits in treating nonalcoholic fatty liver disease.
Collapse
Affiliation(s)
- Myungsuk Kim
- Natural Products Research Center, Korea Institute of Science and Technology (KIST) Gangneung Institute , Gangwon 210-340, Korea
| | - Sue Ji Lim
- Natural Products Research Center, Korea Institute of Science and Technology (KIST) Gangneung Institute , Gangwon 210-340, Korea
- Department of Chemistry, Gangneung-Wonju National University , Gangneung, Gangwon-do 210-702, Korea
| | - Hee-Ju Lee
- Natural Products Research Center, Korea Institute of Science and Technology (KIST) Gangneung Institute , Gangwon 210-340, Korea
| | - Sun Young Kim
- Natural Products Research Center, Korea Institute of Science and Technology (KIST) Gangneung Institute , Gangwon 210-340, Korea
| | - Chu Won Nho
- Natural Products Research Center, Korea Institute of Science and Technology (KIST) Gangneung Institute , Gangwon 210-340, Korea
| |
Collapse
|
44
|
Magliano DC, Penna-de-Carvalho A, Vazquez-Carrera M, Mandarim-de-Lacerda CA, Aguila MB. Short-term administration of GW501516 improves inflammatory state in white adipose tissue and liver damage in high-fructose-fed mice through modulation of the renin-angiotensin system. Endocrine 2015; 50:355-67. [PMID: 25854303 DOI: 10.1007/s12020-015-0590-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 03/30/2015] [Indexed: 02/08/2023]
Abstract
High activation of the angiotensin-converting enzyme (ACE)/(angiotensin-II type 1 receptor) AT1r axis is closely linked to pro-inflammatory effects and liver damage. The aim of this study was to evaluate the effects of the short-term administration of GW501516 on pro-inflammatory markers in white adipose tissue (WAT) and hepatic stellate cells (HSCs), lipogenesis and insulin resistance in the liver upon high-fructose diet (HFru)-induced ACE/AT1r axis activation. Three-month-old male C57Bl/6 mice were fed a standard chow diet or a HFru for 8 weeks. Then, the animals were separated randomly into four groups and treated with GW501516 for 3 weeks. Morphological variables, systolic blood pressure, and plasma determinations were analyzed. In the WAT, the ACE/AT1r axis and pro-inflammatory cytokines were assessed, and in the liver, the ACE/AT1r axis, HSCs, fatty acid oxidation, insulin resistance, and AMPK activation were evaluated. The HFru group displayed a high activation of the ACE/AT1r axis in both the WAT and liver; consequently, we detected inflammation and liver damage. Although GW501516 abolished the increased activation of the ACE/AT1r axis in the WAT, no differences were found in the liver. GW501516 blunted the inflammatory state in the WAT and reduced HSC activation in the liver. In addition, GW501516 alleviates damage in the liver by increasing the expression of the genes that regulate beta-oxidation and decreasing the expression of the genes and proteins that are involved in lipogenesis and gluconeogenesis. We conclude that GW501516 may serve as a therapeutic option for the treatment of a highly activated ACE/AT1r axis in WAT and liver.
Collapse
Affiliation(s)
- D'Angelo C Magliano
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Av 28 de Setembro 87 fds, Rio de Janeiro, 20551-030, Brazil
- Department of Pharmacology and Therapeutic Chemistry, Faculty of Pharmacy, Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona, Spain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain
| | - Aline Penna-de-Carvalho
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Av 28 de Setembro 87 fds, Rio de Janeiro, 20551-030, Brazil
| | - Manuel Vazquez-Carrera
- Department of Pharmacology and Therapeutic Chemistry, Faculty of Pharmacy, Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona, Spain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain
| | - Carlos A Mandarim-de-Lacerda
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Av 28 de Setembro 87 fds, Rio de Janeiro, 20551-030, Brazil.
| | - Marcia B Aguila
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Av 28 de Setembro 87 fds, Rio de Janeiro, 20551-030, Brazil
| |
Collapse
|
45
|
Schuchard J, Winkler M, Stölting I, Schuster F, Vogt FM, Barkhausen J, Thorns C, Santos RA, Bader M, Raasch W. Lack of weight gain after angiotensin AT1 receptor blockade in diet-induced obesity is partly mediated by an angiotensin-(1-7)/Mas-dependent pathway. Br J Pharmacol 2015; 172:3764-78. [PMID: 25906670 DOI: 10.1111/bph.13172] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 03/24/2015] [Accepted: 04/04/2015] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND AND PURPOSE Angiotensin AT1 receptor antagonists induce weight loss; however, the mechanism underlying this phenomenon is unknown. The Mas receptor agonist angiotensin-(1-7) is a metabolite of angiotensin I and of angiotensin II . As an agonist of Mas receptors, angiotensin-(1-7) has beneficial cardiovascular and metabolic effects. EXPERIMENTAL APPROACH We investigated the anti-obesity effects of transgenically overexpressed angiotensin-(1-7) in rats. We secondly examined whether weight loss due to telmisartan (8 mg·kg(-1) ·d(-1) ) in diet-induced obese Sprague Dawley (SD) rats can be blocked when the animals were co-treated with the Mas receptor antagonist A779 (24 or 72 μg·kg(-1) ·d(-1) ). KEY RESULTS In contrast to wild-type controls, transgenic rats overexpressing angiotensin-(1-7) had 1.) diminished body weight when they were regularly fed with chow; 2.) were protected from developing obesity although they were fed with cafeteria diet (CD); 3.) showed a reduced energy intake that was mainly related to a lower CD intake; 5.) remained responsive to leptin despite chronic CD feeding; 6.) had a higher, strain-dependent energy expenditure, and 7.) were protected from developing insulin resistance despite CD feeding. Telmisartan-induced weight loss in SD rats was partially antagonized after a high, but not a low dose of A779. CONCLUSIONS AND IMPLICATIONS Angiotensin-(1-7) regulated food intake and body weight and contributed to the weight loss after AT1 receptor blockade. Angiotensin-(1-7)-like agonists may be drug candidates for treating obesity.
Collapse
Affiliation(s)
- Johanna Schuchard
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany
| | - Martina Winkler
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Ines Stölting
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Franziska Schuster
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Florian M Vogt
- Department for Radiology and Nuclear Medicine, University of Lübeck, Lübeck, Germany
| | - Jörg Barkhausen
- Department for Radiology and Nuclear Medicine, University of Lübeck, Lübeck, Germany
| | - Christoph Thorns
- Department of Pathology, University Clinic Schleswig-Holstein, Luebeck, Germany
| | - Robson A Santos
- National Institute of Science and Technology in Nanobiopharmaceutics, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Michael Bader
- National Institute of Science and Technology in Nanobiopharmaceutics, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.,Max-Delbrück-Center for Molecular Medicine (MDC), Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany.,Center for Structural and Cell Biology in Medicine, Institute for Biology, University of Lübeck, Lübeck, Germany.,Charité - University Medicine Berlin, Berlin, Germany
| | - Walter Raasch
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany
| |
Collapse
|
46
|
Favre GA, Esnault VLM, Van Obberghen E. Modulation of glucose metabolism by the renin-angiotensin-aldosterone system. Am J Physiol Endocrinol Metab 2015; 308:E435-49. [PMID: 25564475 DOI: 10.1152/ajpendo.00391.2014] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The renin-angiotensin-aldosterone system (RAAS) is an enzymatic cascade functioning in a paracrine and autocrine fashion. In animals and humans, RAAS intrinsic to tissues modulates food intake, metabolic rate, adiposity, insulin sensitivity, and insulin secretion. A large array of observations shows that dysregulation of RAAS in the metabolic syndrome favors type 2 diabetes. Remarkably, angiotensin-converting enzyme inhibitors, suppressing the synthesis of angiotensin II (ANG II), and angiotensin receptor blockers, targeting the ANG II type 1 receptor, prevent diabetes in patients with hypertensive or ischemic cardiopathy. These drugs interrupt the negative feedback loop of ANG II on the RAAS cascade, which results in increased production of angiotensins. In addition, they change the tissue expression of RAAS components. Therefore, the concept of a dual axis of RAAS regarding glucose homeostasis has emerged. The RAAS deleterious axis increases the production of inflammatory cytokines and raises oxidative stress, exacerbating the insulin resistance and decreasing insulin secretion. The beneficial axis promotes adipogenesis, blocks the production of inflammatory cytokines, and lowers oxidative stress, thereby improving insulin sensitivity and secretion. Currently, drugs targeting RAAS are not given for the purpose of preventing diabetes in humans. However, we anticipate that in the near future the discovery of novel means to modulate the RAAS beneficial axis will result in a decisive therapeutic breakthrough.
Collapse
Affiliation(s)
- Guillaume A Favre
- Institut National de la Sante et de la Recherche Medicale, U 1081, Institute for Research on Cancer and Aging of Nice (IRCAN), "Aging and Diabetes" Team, Nice, France; Centre National de la Recherche Scientifique, UMR7284, IRCAN, Nice, France; University of Nice-Sophia Antipolis, Nice, France; Nephrology Department, University Hospital, Nice, France; and
| | - Vincent L M Esnault
- Institut National de la Sante et de la Recherche Medicale, U 1081, Institute for Research on Cancer and Aging of Nice (IRCAN), "Aging and Diabetes" Team, Nice, France; Centre National de la Recherche Scientifique, UMR7284, IRCAN, Nice, France; University of Nice-Sophia Antipolis, Nice, France; Nephrology Department, University Hospital, Nice, France; and
| | - Emmanuel Van Obberghen
- Institut National de la Sante et de la Recherche Medicale, U 1081, Institute for Research on Cancer and Aging of Nice (IRCAN), "Aging and Diabetes" Team, Nice, France; Centre National de la Recherche Scientifique, UMR7284, IRCAN, Nice, France; University of Nice-Sophia Antipolis, Nice, France; Clinical Chemistry Laboratory, University Hospital, Nice, France
| |
Collapse
|
47
|
Oriowo MA. Perivascular adipose tissue, vascular reactivity and hypertension. Med Princ Pract 2015; 24 Suppl 1:29-37. [PMID: 24503717 PMCID: PMC6489082 DOI: 10.1159/000356380] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2013] [Accepted: 10/09/2013] [Indexed: 12/13/2022] Open
Abstract
Most blood vessels are surrounded by a variable amount of adventitial adipose tissue, perivascular adipose tissue (PVAT), which was originally thought to provide mechanical support for the vessel. It is now known that PVAT secretes a number of bioactive substances including vascular endothelial growth factor, tumor necrosis factor-alpha (TNF-α), leptin, adiponectin, insulin-like growth factor, interleukin-6, plasminogen activator substance, resistin and angiotensinogen. Several studies have shown that PVAT significantly modulated vascular smooth muscle contractions induced by a variety of agonists and electrical stimulation by releasing adipocyte-derived relaxing (ADRF) and contracting factors. The identity of ADRF is not yet known. However, several vasodilators have been suggested including adiponectin, angiotensin 1-7, hydrogen sulfide and methyl palmitate. The anticontractile effect of PVAT is mediated through the activation of potassium channels since it is abrogated by inhibiting potassium channels. Hypertension is characterized by a reduction in the size and amount of PVAT and this is associated with the attenuated anticontractile effect of PVAT in hypertension. However, since a reduction in size and amount of PVAT and the attenuated anticontractile effect of PVAT were already evident in prehypertensive rats with no evidence of impaired release of ADRF, there is the possibility that the anticontractile effect of PVAT was not directly related to an altered function of the adipocytes per se. Hypertension is characterized by low-grade inflammation and infiltration of macrophages. One of the adipokines secreted by macrophages is TNF-α. It has been shown that exogenously administered TNF-α enhanced agonist-induced contraction of a variety of vascular smooth muscle preparations and reduced endothelium-dependent relaxation. Other procontractile factors released by the PVAT include angiotensin II and superoxide. It is therefore possible that the loss could be due to an increased amount of these proinflammatory and procontractile factors. More studies are definitely required to confirm this.
Collapse
Affiliation(s)
- Mabayoje A Oriowo
- Department of Pharmacology and Toxicology, Faculty of Medicine, Health Sciences Centre, Kuwait University, Jabriya, Kuwait
| |
Collapse
|
48
|
Abstract
The renin-angiotensin (Ang) system is involved in maintaining cardiovascular function by regulating blood pressure and electrolyte homeostasis. More recently, alternative pathways within the renin-angiotensin system have been described, such as the ACE-2/Ang-(1-7)/Mas axis, with opposite effects to the ones of the ACE/Ang-II/AT1 axis. Correspondingly, our previous work reported that Ang-(1-7) via its receptor Mas inhibits the mRNA expression of the proinflammatory cytokines interleukin 6 (IL-6) and tumor necrosis factor-α increased by lipopolysaccharide (LPS) in mouse peritoneal macrophages. These data led us to investigate the functional role of the Ang-(1-7)/Mas axis in an in vivo LPS model. In this work, we present evidence that Ang-(1-7) via Mas significantly reduced the LPS-increased production of circulating cytokines, such as IL-6, IL-12, and CXCL-1. This inhibitory effect was mediated by Mas because it was not detectable in Mas-deficient (Mas) mice. Accordingly, IL-6, CXCL-1, and CXCL-2 levels were higher after LPS treatment in the absence of Mas. Mas mice were less resistant to LPS-induced endotoxemia, their survival rate being 50% compared with 95% in wild-type mice. Telemetric analyses showed that Mas mice presented more pronounced LPS-induced hypothermia with a 3°C lower body temperature compared with wild-type mice. Altogether, our findings suggest that Ang-(1-7) and Mas inhibit LPS-induced cytokine production and hypothermia and thereby protect mice from the fatal consequences of endotoxemia.
Collapse
|
49
|
Moreira de Macêdo S, Guimarães TA, Feltenberger JD, Sousa Santos SH. The role of renin-angiotensin system modulation on treatment and prevention of liver diseases. Peptides 2014; 62:189-96. [PMID: 25453980 DOI: 10.1016/j.peptides.2014.10.005] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 10/10/2014] [Accepted: 10/10/2014] [Indexed: 02/07/2023]
Abstract
The renin-angiotensin system (RAS) is now recognized as an important modulator of body metabolic processes. The discovery of angiotensin-converting enzyme 2 (ACE2) has renewed interest in the potential therapeutic role of RAS modulation. Recent studies have pointed out the importance of the local balance between ACE/Ang-II/AT1 and ACE2/Ang-(1-7)/Mas arms to avoid liver metabolic diseases. Furthermore, non-alcoholic fatty liver disease is an increasing health problem that includes a spectrum of hepatic steatosis, steatohepatitis and fibrosis. Some new studies revealed that RAS imbalance appears to promote hepatic fibrogenesis; while the activation of ACE2/Ang-(1-7)/Mas counter-regulatory axis is able to prevent liver injuries. In this context, the aim of the present review is to discuss the importance of RAS in the development and prevention of liver disease. AT1 receptor activation by Ang II induces hepatic stellate cell contraction and proliferation, causes oxidative stress, endothelial dysfunction, cell growth and inflammation. In addition, both AT1 blocker administration and ACE inhibitors lead to a reduction in inflammation and improvement of hepatic fibrosis. Conversely, Ang-(1-7) infusion reduces fibrosis and proliferation mainly by suppression of hepatic stellate cell activation; Mas receptor antagonism aggravates liver fibrosis and severe liver steatosis. In conclusion, the use of ACE/Ang II/AT1 axis inhibitors associated with ACE2/Ang(1-7)/Mas axis activation is a promising new strategy serving as a novel therapeutic regimen to prevent and treat chronic liver diseases as well as acute liver injury.
Collapse
|
50
|
Andrade JMO, Lemos FDO, da Fonseca Pires S, Millán RDS, de Sousa FB, Guimarães ALS, Qureshi M, Feltenberger JD, de Paula AMB, Neto JTM, Lopes MTP, Andrade HMD, Santos RAS, Santos SHS. Proteomic white adipose tissue analysis of obese mice fed with a high-fat diet and treated with oral angiotensin-(1-7). Peptides 2014; 60:56-62. [PMID: 25102447 DOI: 10.1016/j.peptides.2014.07.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 07/25/2014] [Accepted: 07/25/2014] [Indexed: 02/06/2023]
Abstract
Angiotensin-(1-7) has been described as a new potential therapeutic tool for the treatment and prevention of metabolic disorders by regulating several pathways in visceral white adipose tissue (vWAT). The aim of this study was to access the proteins differentially regulated by Ang-(1-7) using proteomic analysis of visceral adipose tissue. Male mice were divided into three groups and fed for 60 days, with each group receiving one of the following diets: standard diet+HPβCD (ST), high fat diet+HPβCD (HFD) and high fat diet+Ang-(1-7)/HPβCD (HFD+Ang-(1-7)). Body weight, fat weight and food intake were measured. At the end of treatment, Ang-(1-7) induced a decrease in body and fat weight. Differential proteomic analysis using two-dimensional electrophoresis (2-DE) combined with mass spectrometry were performed. Results of protein mapping of mesenteric adipose tissue using 2-DE revealed the presence of about 450 spots in each gel (n=3/treatment) with great reproducibility (>70%). Image analysis and further statistical analysis allowed the detection and identification of eight proteins whose expression was modulated in response to HFD when compared to ST. Among these, two proteins showed a sensitive response to Ang-(1-7) treatment (eno1 and aldehyde dehydrogenase). In addition, three proteins were expressed statistically different between HFD+Ang-(1-7) and HFD groups, and four proteins were modulated compared to standard diet. In conclusion, comparative proteomic analysis of a mice model of diet-induced obesity allowed us to outline possible pathways involved in the response to Ang-(1-7), suggesting that Ang-(1-7) may be a useful tool for the treatment of metabolic disorders.
Collapse
Affiliation(s)
- João Marcus Oliveira Andrade
- Laboratory of Health Science, Postgraduate Program in Health Sciences, Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Minas Gerais, Brazil; Nursing Department, Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Minas Gerais, Brazil
| | - Fernanda de Oliveira Lemos
- Pharmacology Department, Biological Sciences Institute (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | - Simone da Fonseca Pires
- Parasitology Department, Biological Sciences Institute (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | | | - Frederico Barros de Sousa
- Department of Chemistry, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | - André Luiz Sena Guimarães
- Laboratory of Health Science, Postgraduate Program in Health Sciences, Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Minas Gerais, Brazil
| | - Mahboob Qureshi
- Touro University Nevada College of Osteopathic Medicine, Las Vegas, NV, United States
| | | | - Alfredo Maurício Batista de Paula
- Laboratory of Health Science, Postgraduate Program in Health Sciences, Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Minas Gerais, Brazil
| | - Jaime Tolentino Miranda Neto
- Physical Training School, Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Minas Gerais, Brazil
| | - Miriam Teresa Paz Lopes
- Pharmacology Department, Biological Sciences Institute (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | - Hélida Monteiro de Andrade
- Parasitology Department, Biological Sciences Institute (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | - Robson Augusto Souza Santos
- National Institute of Science and Technology (INCT-NanoBiofar), Physiology Department, Biological Sciences Institute (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | - Sérgio Henrique Sousa Santos
- Laboratory of Health Science, Postgraduate Program in Health Sciences, Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Minas Gerais, Brazil; Pharmacology Department, Biological Sciences Institute (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|