1
|
de Almeida TCR, Vilela GF, Garcia J, Bazzana MJF, Thomasi SS, Teófilo CR, Magriotis ZM, Okumura LL, Saczk AA. Development of an electrochemical sensor composed of macaúba biochar for assessing caffeine levels. Food Chem 2025; 480:143925. [PMID: 40121875 DOI: 10.1016/j.foodchem.2025.143925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 03/07/2025] [Accepted: 03/16/2025] [Indexed: 03/25/2025]
Abstract
This study addresses the development of analytical methods to monitor caffeine concentration levels in stimulant beverages and capsules. For this purpose, an electrochemical sensor modified with macaúba biomass biochar was developed to evaluate caffeine by differential pulse voltammetry. The results indicate that the ideal proportions of the carbon paste components are 50 % graphite, 20 % macaúba biochar and 30 % binder. The voltametric analysis revealed a caffeine oxidation peak at 1.45 V, with linearity in the concentration range from 1.35 × 10-5 mol·L-1 to 1.03 × 10-4 mol·L-1. The calculated limits of detection (LOD) for soluble coffee, traditional cola-based soft drinks and central nervous system stimulant capsules were 3.52 μmol·L-1, 0.65 μmol·L-1 and 0.82 μmol·L-1, respectively, and the limits of quantification (LOQ) were 11.7 μmol·L-1, 2.15 μmol·L-1 and 2.74 μmol·L-1, respectively. Thus, this method is effective for evaluating caffeine, representing a promising alternative to traditional methods, being a low-cost method that proposes an environmentally friendly analysis.
Collapse
Affiliation(s)
| | | | - Juliana Garcia
- Department of Chemistry - Federal University of Lavras, Lavras, MG, Brazil
| | | | | | - Carla Rhaira Teófilo
- Department of Engineering and Materials- Federal University of Lavras, Lavras, MG, Brazil
| | - Zuy Maria Magriotis
- Department of Engineering and Materials- Federal University of Lavras, Lavras, MG, Brazil
| | | | | |
Collapse
|
2
|
D’Almeida TZ, Gomes MJ, Engel LE, Giometti IC, Ferreira NZ, Stuani R, Corrêa CR, Castoldi RC, Nunes SG, Aguiar AF, Castilho AC, Okoshi MP, Pacagnelli FL. Effects of high-intensity interval training on physical performance, systolic blood pressure, oxidative stress and inflammatory markers in skeletal muscle of spontaneously hypertensive rats. PLoS One 2025; 20:e0316441. [PMID: 39903719 PMCID: PMC11793773 DOI: 10.1371/journal.pone.0316441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 12/11/2024] [Indexed: 02/06/2025] Open
Abstract
AIM To investigate whether high-intensity interval training (HIIT) improves physical performance, systolic blood pressure, and markers of oxidative stress and inflammation in skeletal muscle of spontaneously hypertensive rats (SHR). METHODS Nineteen male SHR rats were randomly assigned to two groups: sedentary (SHRC) and trained (SHR+T). The SHR+T group trained five times a week for eight weeks on a treadmill, while the SHR group remained without any exercise stimulus throughout the experimental period. Maximum physical performance and systolic blood pressure (SBP) were assessed before and after the training period. The following variables were measured in the tibialis anterior (TA) muscle: gene expression of the NADPH oxidase complex (NOX2, NOX4, p22phox, p47phox) and the NF-kB pathway (NF-kB and Ik-B), lipid peroxidation (malonaldehyde; MDA), protein carbonylation, hydrophilic antioxidant capacity (HAC) and pro-inflammatory cytokines (IL-6 and TNF-α). RESULTS SHR+T rats showed higher physical performance and levels of IL-6, and lower SBP and protein carbonylation (p<0.05), compared with SHRC rats. No significant differences (p>0.05) were observed in the other variables. SIGNIFICANCE Our results indicate that HIIT is an effective non-pharmacologic strategy to improve physical performance, reduce SBP, and modulate the skeletal muscle oxidative damage and inflammation in hypertensive rats.
Collapse
Affiliation(s)
| | - Mariana Janini Gomes
- Department of Kinesiology and Sports Management, Texas A&M University, College Station, TX, United States of America
| | | | | | - Natalia Zamberlan Ferreira
- Department of Kinesiology and Sports Management, Texas A&M University, College Station, TX, United States of America
| | - Rafael Stuani
- Postgraduate Program in Animal Science, UNOESTE, Presidente Prudente, Brazil
- Postgraduate Program in Health Science, UNOESTE, Presidente Prudente, Brazil
| | | | - Robson Chacon Castoldi
- Department of Pharmacology, Institute of Biosciences, Júlio de Mesquita Filho Paulista State University, Botucatu, São Paulo, Brazil
| | - Sarah Gomes Nunes
- Postgraduate Program in Animal Science, UNOESTE, Presidente Prudente, Brazil
| | - Andreo Fernando Aguiar
- Postgraduate Program in Physical Exercise in Health Promotion, Northern University of Paraná, Londrina, PR, Brazil
| | | | | | | |
Collapse
|
3
|
Kaneguchi A, Sakitani N, Umehara T. Histological changes in skeletal muscle induced by heart failure in human patients and animal models: A scoping review. Acta Histochem 2024; 126:152210. [PMID: 39442432 DOI: 10.1016/j.acthis.2024.152210] [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: 07/27/2024] [Revised: 10/15/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024]
Abstract
OBJECTIVE This scoping review aimed to characterize the histological changes in skeletal muscle after heart failure (HF) and to identify gaps in knowledge. METHODS On April 03, 2024, systematic searches were performed for papers in which histological analyses were conducted on skeletal muscle sampled from patients with HF or animal models of HF. Screening and data extraction were conducted by two independent authors. RESULTS AND CONCLUSION A total of 118 papers were selected, including 33 human and 85 animal studies. Despite some disagreements among studies, some trends were observed. These trends included a slow-to-fast transition, a decrease in muscle fiber size, capillary to muscle fiber ratio, and mitochondrial activity and content, and an increase in apoptosis. These changes may contribute to the fatigability and decrease in muscle strength observed after HF. Although there were some disagreements between the results of human and animal studies, the results were generally similar. Animal models of HF will therefore be useful in elucidating the histological changes in skeletal muscle that occur in human patients with HF. Because the muscles subjected to histological analysis were mostly thigh muscles in humans and mostly lower leg muscles in animals, it remains uncertain whether changes similar to those seen in lower limb (hindlimb) muscles after HF also occur in upper limb (forelimb) muscles. The results of this review will consolidate the current knowledge on HF-induced histological changes in skeletal muscle and consequently aid in the rehabilitation of patients with HF and future studies.
Collapse
Affiliation(s)
- Akinori Kaneguchi
- Department of Rehabilitation, Faculty of Rehabilitation, Hiroshima International University, Kurose-Gakuendai 555-36, Higashi-Hiroshima, Hiroshima, 739-2695, Japan.
| | - Naoyoshi Sakitani
- Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology, Hayashi-cho 2217-4, Takamatsu, Kagawa, 761-0395, Japan
| | - Takuya Umehara
- Department of Rehabilitation, Faculty of Rehabilitation, Hiroshima International University, Kurose-Gakuendai 555-36, Higashi-Hiroshima, Hiroshima, 739-2695, Japan
| |
Collapse
|
4
|
Chaudhari M, Zelko I, Lorkiewicz P, Hoetker D, Nong Y, Doelling B, Brittian K, Bhatnagar A, Srivastava S, Baba SP. Metabolic pathways for removing reactive aldehydes are diminished in the skeletal muscle during heart failure. Skelet Muscle 2024; 14:24. [PMID: 39425168 PMCID: PMC11488087 DOI: 10.1186/s13395-024-00354-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 09/25/2024] [Indexed: 10/21/2024] Open
Abstract
Muscle wasting is a serious complication in heart failure patients. Oxidative stress and inflammation are implicated in the pathogenesis of muscle wasting. Oxidative stress leads to the formation of toxic lipid peroxidation products, such as 4-hydroxy-2-nonenal (HNE), which covalently bind with proteins and DNA and activate atrophic pathways. Whether the formation of lipid peroxidation products and metabolic pathways that remove these toxic products are affected during heart failure-associated skeletal muscle wasting has never been studied. Male C57BL/6J mice were subjected to sham and transverse aortic constriction (TAC) surgeries for 4, 8 or 14 weeks. Different skeletal muscle beds were weighed, and the total cross-sectional area of the gastrocnemius muscle was measured via immunohistochemistry. Muscle function and muscle stiffness were measured by a grip strength meter and atomic force microscope, respectively. Atrophic and inflammatory marker levels were measured via qRT‒PCR. The levels of acrolein and HNE-protein adducts, aldehyde-removing enzymes, the histidyl dipeptide-synthesizing enzyme carnosine synthase (CARNS), and amino acid transporters in the gastrocnemius muscle were measured via Western blotting and qRT‒PCR. Histidyl dipeptides and histidyl dipeptide aldehyde conjugates in the Gastrocnemius and soleus muscles were analyzed by LC/MS-MS. Body weight, gastrocnemius muscle and soleus muscle weights and the total cross-sectional area of the gastrocnemius muscle were decreased after 14 weeks of TAC. Heart weight, cardiac function, grip strength and muscle stiffness were decreased in the TAC-operated mice. Expression of the atrophic and inflammatory markers Atrogin1 and TNF-α, respectively, was increased ~ 1.5-2fold in the gastrocnemius muscle after 14 weeks of TAC (p < 0.05 and p = 0.004 vs sham). The formation of HNE and acrolein protein adducts was increased, and the expression of the aldehyde-removing enzyme aldehyde dehydrogenase (ALDH2) was decreased in the gastrocnemius muscle of TAC mice. Carnosine (sham: 5.76 ± 1.3 vs TAC: 4.72 ± 0.7 nmol/mg tissue, p = 0.04) and total histidyl dipeptide levels (carnosine and anserine; sham: 11.97 ± 1.5 vs TAC: 10.13 ± 1.4 nmol/mg tissue, p < 0.05) were decreased in the gastrocnemius muscle of TAC mice. Depletion of histidyl dipeptides diminished the aldehyde removal capacity of the atrophic gastrocnemius muscle. Furthermore, CARNS and TAUT protein expression were decreased in the atrophic gastrocnemius muscle. Our data reveals that reduced expression of ALDH2 and depletion of histidyl dipeptides in the gastrocnemius muscle during heart failure leads to the accumulation of toxic aldehydes and might contribute to muscle wasting.
Collapse
Affiliation(s)
- Mamata Chaudhari
- Center for Cardiometabolic Science, Louisville, KY, USA
- Department of Medicine, Christina Lee Brown Envirome Institute, University of Louisville, 580 South Preston Street, Delia Baxter Building, Room 304A, Louisville, KY, 40202, USA
| | - Igor Zelko
- Center for Cardiometabolic Science, Louisville, KY, USA
- Department of Medicine, Christina Lee Brown Envirome Institute, University of Louisville, 580 South Preston Street, Delia Baxter Building, Room 304A, Louisville, KY, 40202, USA
| | - Pawel Lorkiewicz
- Center for Cardiometabolic Science, Louisville, KY, USA
- Department of Medicine, Christina Lee Brown Envirome Institute, University of Louisville, 580 South Preston Street, Delia Baxter Building, Room 304A, Louisville, KY, 40202, USA
| | - David Hoetker
- Center for Cardiometabolic Science, Louisville, KY, USA
- Department of Medicine, Christina Lee Brown Envirome Institute, University of Louisville, 580 South Preston Street, Delia Baxter Building, Room 304A, Louisville, KY, 40202, USA
| | - Yibing Nong
- Center for Cardiometabolic Science, Louisville, KY, USA
- Department of Medicine, Christina Lee Brown Envirome Institute, University of Louisville, 580 South Preston Street, Delia Baxter Building, Room 304A, Louisville, KY, 40202, USA
| | - Benjamin Doelling
- Center for Cardiometabolic Science, Louisville, KY, USA
- Department of Medicine, Christina Lee Brown Envirome Institute, University of Louisville, 580 South Preston Street, Delia Baxter Building, Room 304A, Louisville, KY, 40202, USA
| | - Kenneth Brittian
- Center for Cardiometabolic Science, Louisville, KY, USA
- Department of Medicine, Christina Lee Brown Envirome Institute, University of Louisville, 580 South Preston Street, Delia Baxter Building, Room 304A, Louisville, KY, 40202, USA
| | - Aruni Bhatnagar
- Center for Cardiometabolic Science, Louisville, KY, USA
- Department of Medicine, Christina Lee Brown Envirome Institute, University of Louisville, 580 South Preston Street, Delia Baxter Building, Room 304A, Louisville, KY, 40202, USA
| | | | - Shahid P Baba
- Center for Cardiometabolic Science, Louisville, KY, USA.
- Department of Medicine, Christina Lee Brown Envirome Institute, University of Louisville, 580 South Preston Street, Delia Baxter Building, Room 304A, Louisville, KY, 40202, USA.
| |
Collapse
|
5
|
Gouveia M, Schmidt C, Basilio PG, Aveiro SS, Domingues P, Xia K, Colón W, Vitorino R, Ferreira R, Santos M, Vieira SI, Ribeiro F. Exercise training decreases the load and changes the content of circulating SDS-resistant protein aggregates in patients with heart failure with reduced ejection fraction. Mol Cell Biochem 2024; 479:2711-2722. [PMID: 37902886 PMCID: PMC11455743 DOI: 10.1007/s11010-023-04884-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 10/15/2023] [Indexed: 11/01/2023]
Abstract
BACKGROUND Heart failure (HF) often disrupts the protein quality control (PQC) system leading to protein aggregate accumulation. Evidence from tissue biopsies showed that exercise restores PQC system in HF; however, little is known about its effects on plasma proteostasis. AIM To determine the effects of exercise training on the load and composition of plasma SDS-resistant protein aggregates (SRA) in patients with HF with reduced ejection fraction (HFrEF). METHODS Eighteen patients with HFrEF (age: 63.4 ± 6.5 years; LVEF: 33.4 ± 11.6%) participated in a 12-week combined (aerobic plus resistance) exercise program (60 min/session, twice per week). The load and content of circulating SRA were assessed using D2D SDS-PAGE and mass spectrometry. Cardiorespiratory fitness, quality of life, and circulating levels of high-sensitive C-reactive protein, N-terminal pro-B-type natriuretic peptide (NT-proBNP), haptoglobin and ficolin-3, were also evaluated at baseline and after the exercise program. RESULTS The exercise program decreased the plasma SRA load (% SRA/total protein: 38.0 ± 8.9 to 36.1 ± 9.7%, p = 0.018; % SRA/soluble fraction: 64.3 ± 27.1 to 59.8 ± 27.7%, p = 0.003). Plasma SRA of HFrEF patients comprised 31 proteins, with α-2-macroglobulin and haptoglobin as the most abundant ones. The exercise training significantly increased haptoglobin plasma levels (1.03 ± 0.40 to 1.11 ± 0.46, p = 0.031), while decreasing its abundance in SRA (1.83 ± 0.54 × 1011 to 1.51 ± 0.59 × 1011, p = 0.049). Cardiorespiratory fitness [16.4(5.9) to 19.0(5.2) ml/kg/min, p = 0.002], quality of life, and circulating NT-proBNP [720.0(850.0) to 587.0(847.3) pg/mL, p = 0.048] levels, also improved after the exercise program. CONCLUSION Exercise training reduced the plasma SRA load and enhanced PQC, potentially via haptoglobin-mediated action, while improving cardiorespiratory fitness and quality of life of patients with HFrEF.
Collapse
Affiliation(s)
- Marisol Gouveia
- Department of Medical Sciences, iBiMED - Institute of Biomedicine, University of Aveiro, Building 30, Agras do Crasto - Campus Universitário de Santiago, Aveiro, 3810-193, Portugal.
| | - Cristine Schmidt
- Surgery and Physiology Department, Faculty of Medicine, University of Porto, Porto, Portugal
- Research Centre in Physical Activity, Health and Leisure, Faculty of Sport, University of Porto, Porto, Portugal
- Laboratory for Integrative and Translational Research in Population Health (ITR), Porto, Portugal
| | - Priscilla Gois Basilio
- Research Centre in Physical Activity, Health and Leisure, Faculty of Sport, University of Porto, Porto, Portugal
| | - Susana S Aveiro
- Mass Spectrometry Centre, Department of Chemistry, LAQV REQUIMTE, University of Aveiro, Aveiro, Portugal
- GreenCoLab - Green Ocean Association, University of Algarve, Faro, Portugal
| | - Pedro Domingues
- Mass Spectrometry Centre, Department of Chemistry, LAQV REQUIMTE, University of Aveiro, Aveiro, Portugal
| | - Ke Xia
- Department of Chemistry and Chemical Biology, Rensselaer Polytechnic Institute, Troy, NY, USA
- Centre for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Wilfredo Colón
- Department of Chemistry and Chemical Biology, Rensselaer Polytechnic Institute, Troy, NY, USA
- Centre for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Rui Vitorino
- Department of Medical Sciences, iBiMED - Institute of Biomedicine, University of Aveiro, Building 30, Agras do Crasto - Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
- Surgery and Physiology Department, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Rita Ferreira
- Department of Chemistry, QOPNA & LAQV-REQUIMTE, University of Aveiro, Aveiro, Portugal
| | - Mário Santos
- Laboratory for Integrative and Translational Research in Population Health (ITR), Porto, Portugal
- Serviço de Cardiologia, Hospital Santo António, Centro Hospitalar Universitário do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, UMIB, University of Porto, Porto, Portugal
| | - Sandra I Vieira
- Department of Medical Sciences, iBiMED - Institute of Biomedicine, University of Aveiro, Building 30, Agras do Crasto - Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
| | - Fernando Ribeiro
- School of Health Sciences, iBiMED - Institute of Biomedicine, University of Aveiro, Aveiro, Portugal
| |
Collapse
|
6
|
Packer M, Anker SD, Butler J, Cleland JGF, Kalra PR, Mentz RJ, Ponikowski P. Identification of three mechanistic pathways for iron-deficient heart failure. Eur Heart J 2024; 45:2281-2293. [PMID: 38733250 PMCID: PMC11231948 DOI: 10.1093/eurheartj/ehae284] [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: 11/13/2023] [Revised: 02/29/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
Current understanding of iron-deficient heart failure is based on blood tests that are thought to reflect systemic iron stores, but the available evidence suggests greater complexity. The entry and egress of circulating iron is controlled by erythroblasts, which (in severe iron deficiency) will sacrifice erythropoiesis to supply iron to other organs, e.g. the heart. Marked hypoferraemia (typically with anaemia) can drive the depletion of cardiomyocyte iron, impairing contractile performance and explaining why a transferrin saturation < ≈15%-16% predicts the ability of intravenous iron to reduce the risk of major heart failure events in long-term trials (Type 1 iron-deficient heart failure). However, heart failure may be accompanied by intracellular iron depletion within skeletal muscle and cardiomyocytes, which is disproportionate to the findings of systemic iron biomarkers. Inflammation- and deconditioning-mediated skeletal muscle dysfunction-a primary cause of dyspnoea and exercise intolerance in patients with heart failure-is accompanied by intracellular skeletal myocyte iron depletion, which can be exacerbated by even mild hypoferraemia, explaining why symptoms and functional capacity improve following intravenous iron, regardless of baseline haemoglobin or changes in haemoglobin (Type 2 iron-deficient heart failure). Additionally, patients with advanced heart failure show myocardial iron depletion due to both diminished entry into and enhanced egress of iron from the myocardium; the changes in iron proteins in the cardiomyocytes of these patients are opposite to those expected from systemic iron deficiency. Nevertheless, iron supplementation can prevent ventricular remodelling and cardiomyopathy produced by experimental injury in the absence of systemic iron deficiency (Type 3 iron-deficient heart failure). These observations, taken collectively, support the possibility of three different mechanistic pathways for the development of iron-deficient heart failure: one that is driven through systemic iron depletion and impaired erythropoiesis and two that are characterized by disproportionate depletion of intracellular iron in skeletal and cardiac muscle. These mechanisms are not mutually exclusive, and all pathways may be operative at the same time or may occur sequentially in the same patients.
Collapse
Affiliation(s)
- Milton Packer
- Baylor Heart and Vascular Institute, Baylor University Medical Center, 621 North Hall Street, Dallas, TX 75226, USA
- Imperial College, London, UK
| | - Stefan D Anker
- Department of Cardiology of German Heart Center Charité, Institute of Health Center for Regenerative Therapies, German Centre for Cardiovascular Research, partner site Berlin, Charité Universitätsmedizin, Berlin, Germany
| | - Javed Butler
- Baylor Scott and White Research Institute, Baylor University Medical Center, Dallas, TX, USA
- University of Mississippi Medical Center, Jackson, MS, USA
| | - John G F Cleland
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Paul R Kalra
- Department of Cardiology, Portsmouth Hospitals University NHS Trust, Portsmouth, UK
- College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK
- Faculty of Science and Health, University of Portsmouth, Portsmouth, UK
| | - Robert J Mentz
- Division of Cardiology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
- Duke Clinical Research Institute, Durham, NC, USA
| | - Piotr Ponikowski
- Institute of Heart Diseases, Wroclaw Medical University, Wroclaw, Poland
- Institute of Heart Diseases, University Hospital, Wroclaw, Poland
| |
Collapse
|
7
|
Yan J, Wu B, Lu B, Zhu Z, Di N, Yang C, Xu Q, Fan L, Hu Y. Association between baseline office blood pressure level and the incidence and development of long-term frailty in the community-dwelling very elderly with hypertension. Hypertens Res 2024; 47:1523-1532. [PMID: 38459173 DOI: 10.1038/s41440-024-01614-w] [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: 09/25/2023] [Revised: 01/15/2024] [Accepted: 01/20/2024] [Indexed: 03/10/2024]
Abstract
Frailty is the most important risk factor causing disability in the elderly. Hypertension is one of the most common chronic diseases in the elderly and is closely related to frailty, but there is still controversy about the association between blood pressure and frailty. To explore the association between baseline blood pressure level and the incident and development of long-term frailty in the community-dwelling very elderly (i.e., over 80 years old [1]) with hypertension, in order to provide a basis for scientific blood pressure management of very elderly hypertension. In this study, very elderly hypertensive patients who received comprehensive geriatric assessment from January to June 2019 and with complete data were included, and follow-up was conducted from January 1 to February 14, 2023. A total of 330 very elderly individuals with hypertension were enrolled in this study. FRAIL scale was used to evaluate frailty. Binomial logistic regression analysis was used to calculate the OR and 95%CI between baseline systolic blood pressure (SBP), diastolic blood pressure (DBP), pulse pressure (PP) levels and long-term incident and development of frailty. The dose-response relationship between baseline office SBP, DBP or PP levels and incident frailty and its development was analyzed by Generalized Additive Model (GAM) using smooth curve fitting and threshold effect analysis. Smooth curve fitting and threshold effect analysis showed that the relationship between baseline office SBP level and incident frailty was U-shaped, with the nadir of the U-shaped curve at 135 mmHg after adjustment. Baseline office SBP, PP level and development frailty was U-shaped and the nadir was 140 mmHg and 77 mmHg. In the community-dwelling very elderly with hypertension, baseline office SBP level had a relationship with long-term incident frailty and its development and PP level had a relationship with long-term development of frailty.
Collapse
Affiliation(s)
- Jin Yan
- Graduate School of Chinese PLA General Hospital & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, 100853, Beijing, China
| | - Bing Wu
- Graduate School of Chinese PLA General Hospital & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, 100853, Beijing, China
| | - Bingjie Lu
- Graduate School of Jilin Sport University, 130022, Changchun, China
| | - Zhihui Zhu
- Ningxia University, 750021, Yinchuan, China
| | - Ning Di
- Ningxia University, 750021, Yinchuan, China
| | - Cunmei Yang
- Geriatric Health Care Department 4th of The Second Medical Center, Chinese PLA General Hospital, 100853, Beijing, China
| | - Qiuli Xu
- Geriatric Health Care Department 4th of The Second Medical Center, Chinese PLA General Hospital, 100853, Beijing, China
| | - Li Fan
- Cardiovascular Department of The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, 100853, Beijing, China.
| | - Yixin Hu
- Geriatric Health Care Department 4th of The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, 100853, Beijing, China.
| |
Collapse
|
8
|
Lisi V, Senesi G, Balbi C. Converging protective pathways: Exploring the linkage between physical exercise, extracellular vesicles and oxidative stress. Free Radic Biol Med 2023; 208:718-727. [PMID: 37739138 DOI: 10.1016/j.freeradbiomed.2023.09.021] [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] [Received: 07/25/2023] [Revised: 08/27/2023] [Accepted: 09/18/2023] [Indexed: 09/24/2023]
Abstract
Physical Exercise (EXR) has been shown to have numerous beneficial effects on various systems in the human body. It leads to a decrease in the risk of mortality from chronic diseases, such as cardiovascular disease, cancer, metabolic and central nervous system disorders. EXR results in improving cardiovascular fitness, cognitive function, immune activity, endocrine action, and musculoskeletal health. These positive effects make EXR a valuable intervention for promoting overall health and well-being in individuals of all ages. These beneficial effects are partially mediated by the role of the regular EXR in the adaptation to redox homeostasis counteracting the sudden increase of ROS, the hallmark of many chronic diseases. EXR can trigger the release of numerous humoral factors, e.g. protein, microRNA (miRs), and DNA, that can be shuttled as cargo of Extracellular vesicles (EVs). EVs show different cargo modification after oxidative stress stimuli as well as after EXR. In this review, we aim to highlight the main studies on the role of EVs released during EXR and oxidative stress conditions in enhancing the antioxidant enzymes pathway and in the decrease of oxidative stress environment mediated by their cargo.
Collapse
Affiliation(s)
- Veronica Lisi
- Unit of Biology and Genetics of Movement, Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis 15, 00135 Rome, Italy.
| | - Giorgia Senesi
- Cellular and Molecular Cardiology, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Carolina Balbi
- Cellular and Molecular Cardiology, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland; Center for Molecular Cardiology, Zurich, Switzerland
| |
Collapse
|
9
|
Mangner N, Winzer EB, Linke A, Adams V. Locomotor and respiratory muscle abnormalities in HFrEF and HFpEF. Front Cardiovasc Med 2023; 10:1149065. [PMID: 37965088 PMCID: PMC10641491 DOI: 10.3389/fcvm.2023.1149065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 10/02/2023] [Indexed: 11/16/2023] Open
Abstract
Heart failure (HF) is a chronic and progressive syndrome affecting worldwide billions of patients. Exercise intolerance and early fatigue are hallmarks of HF patients either with a reduced (HFrEF) or a preserved (HFpEF) ejection fraction. Alterations of the skeletal muscle contribute to exercise intolerance in HF. This review will provide a contemporary summary of the clinical and molecular alterations currently known to occur in the skeletal muscles of both HFrEF and HFpEF, and thereby differentiate the effects on locomotor and respiratory muscles, in particular the diaphragm. Moreover, current and future therapeutic options to address skeletal muscle weakness will be discussed focusing mainly on the effects of exercise training.
Collapse
Affiliation(s)
- Norman Mangner
- Department of Internal Medicine and Cardiology, Heart Center Dresden, Technische Universität Dresden, Dresden, Germany
| | - Ephraim B. Winzer
- Department of Internal Medicine and Cardiology, Heart Center Dresden, Technische Universität Dresden, Dresden, Germany
| | - Axel Linke
- Department of Internal Medicine and Cardiology, Heart Center Dresden, Technische Universität Dresden, Dresden, Germany
| | - Volker Adams
- Laboratory of Molecular and Experimental Cardiology, Heart Center Dresden, Technische Universität Dresden, Dresden, Germany
- Dresden Cardiovascular Research Institute and Core Laboratories GmbH, Dresden, Germany
| |
Collapse
|
10
|
Ren W, Xu Z, Pan S, Ma Y, Li H, Wu F, Bo W, Cai M, Tian Z. Irisin and ALCAT1 mediated aerobic exercise-alleviated oxidative stress and apoptosis in skeletal muscle of mice with myocardial infarction. Free Radic Biol Med 2022; 193:526-537. [PMID: 36336228 DOI: 10.1016/j.freeradbiomed.2022.10.321] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 10/26/2022] [Accepted: 10/30/2022] [Indexed: 11/06/2022]
Abstract
Skeletal muscle in patients with heart failure (HF) exhibits altered structure, function and metabolism. Myocardial infarction (MI) is the most common cause of HF. Oxidative stress and cell apoptosis are involved in the pathophysiology of MI/HF-induced skeletal muscle atrophy. It is well recognized that aerobic exercise (AE) could prevent skeletal muscle atrophy after MI, but the underlying mechanism and molecular targets are still not fully clarified. In this study, Fndc5-/- and Alcat1-/- mice were used to establish the MI model and subjected to six weeks of moderate-intensity AE. C2C12 cells were treated with H2O2 and recombinant human Irisin (rhIrisin), or transduced with a lentiviral vector to mediate the overexpression of ALCAT1 (LV-Alcat1). Results showed that MI reduced Irisin expression and antioxidant capacity of skeletal muscle, increased ALCAT1 expression, induced protein degradation and cell apoptosis, which were partly reversed by AE; Knockout of Fndc5 further aggravated MI-induced oxidative stress and cell apoptosis in skeletal muscle, and partly weakened the beneficial effects of AE. In contrast, knockout of Alcat1 reduced MI-induced oxidative stress and cell apoptosis and strengthened the beneficial effects of AE. rhIrisin and AICAR intervention inhibited ALCAT1 expression, oxidative stress and cell apoptosis, which induced by H2O2 or LV-Alcat1 in C2C12 cells. These findings reveal that AE could alleviate the levels of oxidative stress and apoptosis in skeletal muscle following MI, partly via up-regulating Irisin and inhibiting ALCAT1 expression.
Collapse
Affiliation(s)
- Wujing Ren
- Institute of Sports Biology, College of Physical Education, Shaanxi Normal University, Xi'an, 710119, China
| | - Zujie Xu
- Division of Sports Science and Physical Education, Tsinghua University, Beijing, 100081, China
| | - Shou Pan
- Institute of Sports Biology, College of Physical Education, Shaanxi Normal University, Xi'an, 710119, China
| | - Yixuan Ma
- Institute of Sports Biology, College of Physical Education, Shaanxi Normal University, Xi'an, 710119, China
| | - Hangzhuo Li
- Institute of Sports Biology, College of Physical Education, Shaanxi Normal University, Xi'an, 710119, China; School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Fangnan Wu
- Institute of Sports Biology, College of Physical Education, Shaanxi Normal University, Xi'an, 710119, China; School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Wenyan Bo
- Institute of Sports Biology, College of Physical Education, Shaanxi Normal University, Xi'an, 710119, China
| | - Mengxin Cai
- Institute of Sports Biology, College of Physical Education, Shaanxi Normal University, Xi'an, 710119, China.
| | - Zhenjun Tian
- Institute of Sports Biology, College of Physical Education, Shaanxi Normal University, Xi'an, 710119, China.
| |
Collapse
|
11
|
Winzer EB, Augstein A, Schauer A, Mueller S, Fischer-Schaepmann T, Goto K, Hommel J, van Craenenbroeck EM, Wisløff U, Pieske B, Halle M, Linke A, Adams V. Impact of Different Training Modalities on Molecular Alterations in Skeletal Muscle of Patients With Heart Failure With Preserved Ejection Fraction: A Substudy of the OptimEx Trial. Circ Heart Fail 2022; 15:e009124. [DOI: 10.1161/circheartfailure.121.009124] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Background:
Exercise intolerance is a cardinal feature of heart failure with preserved ejection fraction and so far exercise training (ET) is the most effective treatment. Since the improvement in exercise capacity is only weakly associated with changes in diastolic function other mechanisms, like changes in the skeletal muscle, contribute to improvement in peak oxygen consumption. The aim of the present study was to analyze molecular changes in skeletal muscle of patients with heart failure with preserved ejection fraction performing different ET modalities.
Methods:
Skeletal muscle biopsies were taken at study begin and after 3 and 12 months from patients with heart failure with preserved ejection fraction randomized either into a control group (guideline based advice for ET), a high-intensity interval training group (HIIT) or a moderate continuous training group. The first 3 months of ET were supervised in-hospital followed by 9 months home-based ET. Protein and mRNA expression of atrophy-related proteins, enzyme activities of enzymes linked to energy metabolism and satellite cells (SCs) were quantified.
Results:
Exercise capacity improved 3 months after moderate continuous exercise training and HIIT. This beneficial effect was lost after 12 months. HIIT mainly improved markers of energy metabolism and the amount and function of SC, with minor changes in markers for muscle atrophy. Only slight changes were observed after moderate continuous exercise training. The molecular changes were no longer detectable after 12 months.
Conclusions:
Despite similar improvements in exercise capacity by HIIT and moderate continuous exercise training after 3 months, only HIIT altered proteins related to energy metabolism and amount/function of SC. These effects were lost after switching from in-hospital to at-home-based ET.
Registration:
URL:
https://www.clinicaltrials.gov
; Unique identifier: NCT02078947.
Collapse
Affiliation(s)
- Ephraim B. Winzer
- Laboratory of Molecular and Experimental Cardiology, Technische Universität Dresden, Heart Center Dresden, Germany (E.B.W., A.A., A.S., K.G., J.H., A.L., V.A.)
| | - Antje Augstein
- Laboratory of Molecular and Experimental Cardiology, Technische Universität Dresden, Heart Center Dresden, Germany (E.B.W., A.A., A.S., K.G., J.H., A.L., V.A.)
| | - Antje Schauer
- Laboratory of Molecular and Experimental Cardiology, Technische Universität Dresden, Heart Center Dresden, Germany (E.B.W., A.A., A.S., K.G., J.H., A.L., V.A.)
| | - Stephan Mueller
- Department of Prevention and Sports Medicine, University Hospital Klinikum rechts der Isar, Technical University of Munich, Germany (S.M., M.H.)
- DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (S.M., M.H.)
| | - Tina Fischer-Schaepmann
- Department of Internal Medicine/Cardiology, Heart Center Leipzig – University Hospital, Helios Stiftungsprofessur, Germany (T.F.-S.)
| | - Keita Goto
- Laboratory of Molecular and Experimental Cardiology, Technische Universität Dresden, Heart Center Dresden, Germany (E.B.W., A.A., A.S., K.G., J.H., A.L., V.A.)
| | - Jennifer Hommel
- Laboratory of Molecular and Experimental Cardiology, Technische Universität Dresden, Heart Center Dresden, Germany (E.B.W., A.A., A.S., K.G., J.H., A.L., V.A.)
| | - Emeline M. van Craenenbroeck
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Belgium (E.M.v.C.)
- Department of Cardiology, Antwerp University Hospital, Belgium (E.M.v.C.)
| | - Ulrik Wisløff
- Cardiac Exercise Research Group at Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway (U.W.)
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology, Campus Virchow Klinikum, Charité Universitätsmedizin Berlin, Germany (B.P.)
| | - Martin Halle
- Department of Prevention and Sports Medicine, University Hospital Klinikum rechts der Isar, Technical University of Munich, Germany (S.M., M.H.)
- DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (S.M., M.H.)
| | - Axel Linke
- Laboratory of Molecular and Experimental Cardiology, Technische Universität Dresden, Heart Center Dresden, Germany (E.B.W., A.A., A.S., K.G., J.H., A.L., V.A.)
| | - Volker Adams
- Laboratory of Molecular and Experimental Cardiology, Technische Universität Dresden, Heart Center Dresden, Germany (E.B.W., A.A., A.S., K.G., J.H., A.L., V.A.)
- Dresden Cardiovascular Research Institute and Core Laboratories GmbH, Germany (V.A.)
| |
Collapse
|
12
|
Hu S, Wan X, Li X, Wang X. Aerobic exercise alleviates pyroptosis-related diseases by regulating NLRP3 inflammasome. Front Physiol 2022; 13:965366. [PMID: 36187801 PMCID: PMC9520335 DOI: 10.3389/fphys.2022.965366] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 07/25/2022] [Indexed: 11/29/2022] Open
Abstract
Pyroptosis plays a crucial role in a variety of human diseases, including atherosclerosis, obesity, diabetes, depression, and Alzheimer’s disease, which usually release pyroptosis-related cytokines due to inflammation. Many studies have demonstrated that aerobic exercise is a good option for decreasing the release of pyroptosis-related cytokines. However, the molecular mechanisms of aerobic exercise on pyroptosis-related diseases remain unknown. In this review, the effects of aerobic exercise on pyroptosis in endothelial cells, adipocytes and hippocampal cells, and their potential mechanisms are summarized. In endothelial cells, aerobic exercise could inhibit NOD-like receptor protein 3 (NLRP3) inflammasome-mediated pyroptosis by improving the endothelial function, while reducing vascular inflammation and oxidative stress. In adipocytes, aerobic exercise has been shown to inhibit pyroptosis by ameliorating inflammation and insulin resistance. Moreover, aerobic exercise could restrict pyroptosis by attenuating microglial activation, neuroinflammation, and amyloid-beta deposition in hippocampal cells. In summary, aerobic exercise alleviates the pyroptosis-related diseases by regulating the NLRP3 inflammation si0067naling.
Collapse
Affiliation(s)
- Shujuan Hu
- School of Education and Physical Education, Yangtze University, Jingzhou, China
- School of Physical Education and Science, Jishou University, Jishou, China
| | - Xingxia Wan
- Department of Biochemistry and Molecular Biology, Health Science Center, Yangtze University, Jingzhou, China
| | - Xianhui Li
- College of Pharmacy, Jishou University, Jishou, China
| | - Xianwang Wang
- Department of Biochemistry and Molecular Biology, Health Science Center, Yangtze University, Jingzhou, China
- *Correspondence: Xianwang Wang,
| |
Collapse
|
13
|
Ağaşcıoğlu E, Çolak R, Çakatay U. Redox status biomarkers in the fast-twitch extensor digitorum longus resulting from the hypoxic exercise. NAGOYA JOURNAL OF MEDICAL SCIENCE 2022; 84:433-447. [PMID: 35967949 PMCID: PMC9350571 DOI: 10.18999/nagjms.84.2.433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 11/01/2021] [Indexed: 11/29/2022]
Abstract
The fast-twitch muscle may be affected from over-produced reactive oxygen species (ROS) during hypoxia/hypoxic exercise. The study aims to investigate redox status biomarkers in the fast-twitch extensor digitorum longus (EDL) muscle after hypoxic exercise. Male Sprague Dawley rats (eight-week-old) were randomly divided into six groups of the experimental "live high train high (LHTH), live high train low (LHTL) and live low train low (LLTL)" and their respective controls. Before the EDLs' extraction, the animals exercised for a 4-week familiarization period, then they exercised for four-weeks at different altitudes. The LHTH group had higher ratios of lipid hydroperoxides (LHPs) than the experimental groups of LHTL (p=0.004) and LLTL (p=0.002), while having no difference than its control 'LH'. Similarly, a higher percentage of advanced oxidation protein products (AOPP) was determined in the LHTH than the LHTL (p=0.041) and LLTL (p=0.048). Furthermore, oxidation of thiol fractions was the lowest in the LHTH and LH. However, redox biomarkers and thiol fractions illustrated no significant change in the LHTL and LLTL that might ensure redox homeostasis due to higher oxygen consumption. The study shows that not hypoxic exercise/exercise, but hypoxia might itself lead to a redox imbalance in the fast-twitch EDL muscle.
Collapse
Affiliation(s)
- Eda Ağaşcıoğlu
- Department of Recreation, Faculty of Sports Sciences, Lokman Hekim University, Ankara, Turkey
| | - Rıdvan Çolak
- Department of Physical Education and Sports, Ardahan University, Ardahan, Turkey
| | - Ufuk Çakatay
- Department of Medical Biochemistry, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| |
Collapse
|
14
|
Rodrigues LF, Pelozin BRA, da Silva Junior ND, Soci UPR, do Carmo EC, da Mota GDFA, Cachofeiro V, Lahera V, Oliveira EM, Fernandes T. Angiotensin II Promotes Skeletal Muscle Angiogenesis Induced by Volume-Dependent Aerobic Exercise Training: Effects on miRNAs-27a/b and Oxidant-Antioxidant Balance. Antioxidants (Basel) 2022; 11:651. [PMID: 35453336 PMCID: PMC9026451 DOI: 10.3390/antiox11040651] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/08/2022] [Accepted: 03/14/2022] [Indexed: 12/28/2022] Open
Abstract
Aerobic exercise training (ET) produces beneficial adaptations in skeletal muscles, including angiogenesis. The renin-angiotensin system (RAS) is highly involved in angiogenesis stimuli. However, the molecular mechanisms underlying capillary growth in skeletal muscle induced by aerobic ET are not completely understood. This study aimed to investigate the effects of volume-dependent aerobic ET on skeletal muscle angiogenesis involving the expression of miRNAs-27a and 27b on RAS and oxidant-antioxidant balance. Eight-week-old female Wistar rats were divided into three groups: sedentary control (SC), trained protocol 1 (P1), and trained protocol 2 (P2). P1 consisted of 60 min/day of swimming, 5×/week, for 10 weeks. P2 consisted of the same protocol as P1 until the 8th week, but in the 9th week, rats trained 2×/day, and in the 10th week, trained 3×/day. Angiogenesis and molecular analyses were performed in soleus muscle samples. Furthermore, to establish ET-induced angiogenesis through RAS, animals were treated with an AT1 receptor blocker (losartan). Aerobic ET promoted higher VO2 peak and exercise tolerance values. In contrast, miRNA-27a and -27b levels were reduced in both trained groups, compared with the SC group. This was in parallel with an increase in the ACE1/Ang II/VEGF axis, which led to a higher capillary-to-fiber ratio. Moreover, aerobic ET induced an antioxidant profile increasing skeletal muscle SOD2 and catalase gene expression, which was accompanied by high nitrite levels and reduced nitrotyrosine concentrations in the circulation. Additionally, losartan treatment partially re-established the miRNAs expression and the capillary-to-fiber ratio in the trained groups. In summary, aerobic ET promoted angiogenesis through the miRNA-27a/b-ACE1/Ang II/VEGF axis and improved the redox balance. Losartan treatment demonstrates the participation of RAS in ET-induced vascular growth. miRNAs and RAS components are promising potential targets to modulate angiogenesis for combating vascular diseases, as well as potential biomarkers to monitor training interventions and physical performance.
Collapse
Affiliation(s)
- Luis Felipe Rodrigues
- Laboratory of Biochemistry and Molecular Biology of Exercise, School of Physical Education and Sport, University of Sao Paulo, Sao Paulo 05508-030, Brazil; (L.F.R.); (B.R.A.P.); (N.D.d.S.J.); (U.P.R.S.); (G.d.F.A.d.M.)
| | - Bruno Rocha Avila Pelozin
- Laboratory of Biochemistry and Molecular Biology of Exercise, School of Physical Education and Sport, University of Sao Paulo, Sao Paulo 05508-030, Brazil; (L.F.R.); (B.R.A.P.); (N.D.d.S.J.); (U.P.R.S.); (G.d.F.A.d.M.)
| | - Natan Daniel da Silva Junior
- Laboratory of Biochemistry and Molecular Biology of Exercise, School of Physical Education and Sport, University of Sao Paulo, Sao Paulo 05508-030, Brazil; (L.F.R.); (B.R.A.P.); (N.D.d.S.J.); (U.P.R.S.); (G.d.F.A.d.M.)
- Physiotherapy Program, Ibirapuera University, Sao Paulo 04661-100, Brazil
| | - Ursula Paula Renó Soci
- Laboratory of Biochemistry and Molecular Biology of Exercise, School of Physical Education and Sport, University of Sao Paulo, Sao Paulo 05508-030, Brazil; (L.F.R.); (B.R.A.P.); (N.D.d.S.J.); (U.P.R.S.); (G.d.F.A.d.M.)
| | | | - Glória de Fatima Alves da Mota
- Laboratory of Biochemistry and Molecular Biology of Exercise, School of Physical Education and Sport, University of Sao Paulo, Sao Paulo 05508-030, Brazil; (L.F.R.); (B.R.A.P.); (N.D.d.S.J.); (U.P.R.S.); (G.d.F.A.d.M.)
| | - Victoria Cachofeiro
- Department of Physiology, School of Medicine, Universidad Complutense de Madrid, 28040 Madrid, Spain; (V.C.); (V.L.)
| | - Vicente Lahera
- Department of Physiology, School of Medicine, Universidad Complutense de Madrid, 28040 Madrid, Spain; (V.C.); (V.L.)
| | - Edilamar Menezes Oliveira
- Laboratory of Biochemistry and Molecular Biology of Exercise, School of Physical Education and Sport, University of Sao Paulo, Sao Paulo 05508-030, Brazil; (L.F.R.); (B.R.A.P.); (N.D.d.S.J.); (U.P.R.S.); (G.d.F.A.d.M.)
| | - Tiago Fernandes
- Laboratory of Biochemistry and Molecular Biology of Exercise, School of Physical Education and Sport, University of Sao Paulo, Sao Paulo 05508-030, Brazil; (L.F.R.); (B.R.A.P.); (N.D.d.S.J.); (U.P.R.S.); (G.d.F.A.d.M.)
| |
Collapse
|
15
|
Sun L, Li FH, Han C, Wang ZZ, Gao KK, Qiao YB, Ma S, Xie T, Wang J. Alterations in mitochondrial biogenesis and respiratory activity, inflammation of the senescence-associated secretory phenotype, and lipolysis in the perirenal fat and liver of rats following lifelong exercise and detraining. FASEB J 2021; 35:e21890. [PMID: 34460990 DOI: 10.1096/fj.202100868r] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 08/04/2021] [Accepted: 08/16/2021] [Indexed: 12/25/2022]
Abstract
The primary aims of this study were to determine the effects of lifelong exercise and detraining on age-related alterations in mitochondrial function, inflammation associated with senescence-associated secretory phenotype (SASP), and lipolysis in the perirenal fat and liver of rats. Female Sprague-Dawley rats were randomly assigned to four groups: young control (n = 12), old control (n = 12), detraining (n = 12), and lifelong exercise (n = 12). We then investigated mitochondrial function, SASP-associated inflammation, and lipolysis in the perirenal fat and liver using qRT-PCR and western blotting to assess the expression of AKT, hypoxia-inducible factor 1α (HIF-1α), nuclear factor-kappa B (NF-κB), c-jun kinase (JNK), and p38 mitogen-activated protein kinase (p38MAPK). In the tissues of both the perirenal fat and liver, lifelong exercise significantly improved mitochondrial function, SASP-associated inflammation, and lipolysis. Meanwhile, pathways associated with inflammatory regulation were inhibited, predominantly via the activation of phosphorylated-AKT (p-AKT) and suppression of HIF-1α in both tissues, and via JNK in the perirenal fat and p38MAPK in the liver. Furthermore, detraining activated NF-κB expression in both tissues and induced the upregulation of serum high-sensitivity C-reactive protein (hsCRP) levels. Collectively, lifelong exercise was found to exert beneficial effects by ameliorating age-related alterations in mitochondrial function, SASP-associated inflammation, and lipolysis in perirenal fat and liver tissues, potentially inhibiting inflammation via the JNK and p38 MAPK pathways, respectively, as well as the HIF-1α and AKT pathways in both tissues. In contrast, detraining induced high levels of circulating hsCRP by activating the NF-κB signaling pathway in both tissues.
Collapse
Affiliation(s)
- Lei Sun
- School of Sport Sciences, Nanjing Normal University, Nanjing, P.R. China
| | - Fang-Hui Li
- School of Sport Sciences, Nanjing Normal University, Nanjing, P.R. China
| | - Chong Han
- School of Sport Sciences, Nanjing Normal University, Nanjing, P.R. China
| | - Zhuang-Zhi Wang
- School of Sport Sciences, Nanjing Normal University, Nanjing, P.R. China
| | - Ke-Ke Gao
- School of Sport Sciences, Nanjing Normal University, Nanjing, P.R. China
| | - Yi-Bo Qiao
- School of Sport Sciences, Nanjing Normal University, Nanjing, P.R. China
| | - Song Ma
- School of Sport Sciences, Nanjing Normal University, Nanjing, P.R. China
| | - Tian Xie
- School of Sport Sciences, Nanjing Normal University, Nanjing, P.R. China
| | - Jing Wang
- School of Sport Sciences, Nanjing Normal University, Nanjing, P.R. China
| |
Collapse
|
16
|
Alves CRR, Eichelberger EJ, das Neves W, Ribeiro MAC, Bechara LRG, Voltarelli VA, de Almeida NR, Hagen L, Sharma A, Ferreira JCB, Swoboda KJ, Slupphaug G, Brum PC. Cancer-induced muscle atrophy is determined by intrinsic muscle oxidative capacity. FASEB J 2021; 35:e21714. [PMID: 34118107 DOI: 10.1096/fj.202100263r] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 05/07/2021] [Accepted: 05/18/2021] [Indexed: 01/15/2023]
Abstract
We tested the hypothesis that cancer cachexia progression would induce oxidative post-translational modifications (Ox-PTMs) associated with skeletal muscle wasting, with different responses in muscles with the prevalence of glycolytic and oxidative fibers. We used cysteine-specific isotopic coded affinity tags (OxICAT) and gel-free mass spectrometry analysis to investigate the cysteine Ox-PTMs profile in the proteome of both plantaris (glycolytic) and soleus (oxidative) muscles in tumor-bearing and control rats. Histological analysis revealed muscle atrophy in type II fibers in plantaris muscle, with no changes in plantaris type I fibers and no differences in both soleus type I and II fibers in tumor-bearing rats when compared to healthy controls. Tumor progression altered the Ox-PTMs profile in both plantaris and soleus. However, pathway analysis including the differentially oxidized proteins revealed tricarboxylic acid cycle and oxidative phosphorylation as main affected pathways in plantaris muscle from tumor-bearing rats, while the same analysis did not show main metabolic pathways affected in the soleus muscle. In addition, cancer progression affected several metabolic parameters such as ATP levels and markers of oxidative stress associated with muscle atrophy in plantaris muscle, but not in soleus. However, isolated soleus from tumor-bearing rats had a reduced force production capacity when compared to controls. These novel findings demonstrate that tumor-bearing rats have severe muscle atrophy exclusively in glycolytic fibers. Cancer progression is associated with cysteine Ox-PTMs in the skeletal muscle, but these modifications affect different pathways in a glycolytic muscle compared to an oxidative muscle, indicating that intrinsic muscle oxidative capacity determines the response to cancer cachectic effects.
Collapse
Affiliation(s)
- Christiano R R Alves
- School of Physical Education and Sport, University of Sao Paulo, Sao Paulo, Brazil.,Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Eric J Eichelberger
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Willian das Neves
- School of Physical Education and Sport, University of Sao Paulo, Sao Paulo, Brazil
| | - Márcio A C Ribeiro
- Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Luiz R G Bechara
- Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Vanessa A Voltarelli
- School of Physical Education and Sport, University of Sao Paulo, Sao Paulo, Brazil
| | - Ney R de Almeida
- School of Physical Education and Sport, University of Sao Paulo, Sao Paulo, Brazil
| | - Lars Hagen
- Department of Clinical and Molecular Medicine, NTNU Norwegian University of Science and Technology, Trondheim, Norway.,PROMEC Proteomics and Modomics Experimental Core, NTNU and the Central Norway Regional Health Authority, Trondheim, Norway
| | - Animesh Sharma
- Department of Clinical and Molecular Medicine, NTNU Norwegian University of Science and Technology, Trondheim, Norway.,PROMEC Proteomics and Modomics Experimental Core, NTNU and the Central Norway Regional Health Authority, Trondheim, Norway
| | - Julio C B Ferreira
- Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Kathryn J Swoboda
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Geir Slupphaug
- Department of Clinical and Molecular Medicine, NTNU Norwegian University of Science and Technology, Trondheim, Norway.,PROMEC Proteomics and Modomics Experimental Core, NTNU and the Central Norway Regional Health Authority, Trondheim, Norway
| | - Patricia C Brum
- School of Physical Education and Sport, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
17
|
Physical Activity and Redox Balance in the Elderly: Signal Transduction Mechanisms. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11052228] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Reactive Oxygen Species (ROS) are molecules naturally produced by cells. If their levels are too high, the cellular antioxidant machinery intervenes to bring back their quantity to physiological conditions. Since aging often induces malfunctioning in this machinery, ROS are considered an effective cause of age-associated diseases. Exercise stimulates ROS production on one side, and the antioxidant systems on the other side. The effects of exercise on oxidative stress markers have been shown in blood, vascular tissue, brain, cardiac and skeletal muscle, both in young and aged people. However, the intensity and volume of exercise and the individual subject characteristics are important to envisage future strategies to adequately personalize the balance of the oxidant/antioxidant environment. Here, we reviewed the literature that deals with the effects of physical activity on redox balance in young and aged people, with insights into the molecular mechanisms involved. Although many molecular pathways are involved, we are still far from a comprehensive view of the mechanisms that stand behind the effects of physical activity during aging. Although we believe that future precision medicine will be able to transform exercise administration from wellness to targeted prevention, as yet we admit that the topic is still in its infancy.
Collapse
|
18
|
Neuroprotective Benefits of Exercise and MitoQ on Memory Function, Mitochondrial Dynamics, Oxidative Stress, and Neuroinflammation in D-Galactose-Induced Aging Rats. Brain Sci 2021; 11:brainsci11020164. [PMID: 33514020 PMCID: PMC7910851 DOI: 10.3390/brainsci11020164] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 01/19/2021] [Accepted: 01/23/2021] [Indexed: 01/06/2023] Open
Abstract
Exercise and antioxidants have health benefits that improve cognitive impairment and may act synergistically. In this study, we examined the effects of treadmill exercise (TE) and mitochondria-targeted antioxidant mitoquinone (MitoQ), individually or combined, on learning and memory, mitochondrial dynamics, NADPH oxidase activity, and neuroinflammation and antioxidant activity in the hippocampus of D-galactose-induced aging rats. TE alone and TE combined with MitoQ in aging rats reduced mitochondrial fission factors (Drp1, Fis1) and increased mitochondrial fusion factors (Mfn1, Mfn2, Opa1). These groups also exhibited improved NADPH oxidase activity and antioxidant activity (SOD-2, catalase). TE or MitoQ alone decreased neuroinflammatory response (COX-2, TNF-α), but the suppression was greater with their combination. In addition, aging-increased neuroinflammation in the dentate gyrus was decreased in TE but not MitoQ treatment. Learning and memory tests showed that, contrarily, MitoQ alone demonstrated some similar effects to TE but not a definitive improvement. In conclusion, this study demonstrated that MitoQ exerted some positive effects on aging when used as an isolated treatment, but TE had a more effective role on cognitive impairment, oxidative stress, inflammation, and mitochondria dysfunction. Our findings suggest that the combination of TE and MitoQ exerted no synergistic effects and indicated regular exercise should be the first priority in neuroprotection of age-related cognitive decline.
Collapse
|
19
|
Frailty is not associated with hypertension, blood pressure or antihypertensive medication in community-dwelling older adults: A cross-sectional comparison across 3 frailty instruments. Exp Gerontol 2021; 146:111245. [PMID: 33476700 DOI: 10.1016/j.exger.2021.111245] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 01/04/2021] [Accepted: 01/13/2021] [Indexed: 12/29/2022]
Abstract
AIM The present study investigated whether hypertension, blood pressure, and antihypertensive therapy were associated with frailty status in community-dwelling older adults. In addition, we tested whether such associations were consistent across different frailty instruments. MATERIAL AND METHODS Two-hundred older adults were enrolled in the study. Participant frailty status was determined according to a modified physical frailty phenotype (mFP), the FRAIL scale, and the Study of Osteoporotic Fracture (SOF) index. Blood pressure was assessed three times, in three different days, and mean values were used in the final analysis. Information pertaining to disease conditions and antihypertensive therapy were collected by two researchers through self-report and careful review of medical charts. RESULTS No significant differences in hemodynamic parameters, hypertension diagnosis, and antihypertensive therapy were observed across frailty statuses, regardless of the frailty assessment tool used. CONCLUSION Findings of the present study indicate that hypertension, blood pressure levels and antihypertensive medication were not cross-sectionally associated with frailty status in cognitively preserved community-dwelling older adults with low prevalence of comorbidities, regardless of the tool used for frailty identification.
Collapse
|
20
|
Carvalho MR, Mendonça MLM, Oliveira JML, Romanenghi RB, Morais CS, Ota GE, Lima ARR, Oliveira RJ, Filiú WFO, Okoshi K, Okoshi MP, Oliveira-Junior SA, Martinez PF. Influence of high-intensity interval training and intermittent fasting on myocardium apoptosis pathway and cardiac morphology of healthy rats. Life Sci 2021; 264:118697. [PMID: 33130084 DOI: 10.1016/j.lfs.2020.118697] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 10/22/2020] [Accepted: 10/28/2020] [Indexed: 10/23/2022]
Abstract
AIM To evaluate the influence of intermittent fasting and high-intensity interval training (HIIT) on myocardial apoptosis signaling and cardiac morphological characteristics in healthy rats. METHODS Male Wistar rats (n = 60) were divided into four groups: sedentary control (SED-C), intermittent fasting (SED-IF), high-intensity interval training (HIIT-C), and high-intensity interval training plus intermittent fasting (HIIT-IF). SED-C and HIIT-C groups were treated daily with ad libitum chow; SED-IF and HIIT-IF received the same standard chow every other day. HIIT-C and HIIT-IF rats were submitted to an HIIT protocol five times a week for 12 weeks. At the end of the experiment, functional capacity, cardiac morphology, and expression of apoptosis signaling pathways-related proteins were analyzed. KEY FINDINGS HIIT increased cardiomyocyte cross-sectional area, collagen interstitial fraction, and the pro-apoptotic proteins AIF and caspase-3 expression, and reduced pro-apoptotic protein CYTC expression and the cleaved-to-non-cleaved PARP-1 ratio in myocardium. Intermittent fasting reduced cardiomyocyte cross-sectional area, collagen interstitial fraction, and expression of Bax, CYTC and cleaved PARP-1, and increased expression of the anti-apoptotic protein BCL-2. SMAC, ARC, and caspase-8 expression was not changed by HIIT or intermittent fasting. SIGNIFICANCE HIIT promotes cardiomyocyte hypertrophy and interstitial fibrosis, and modulates the apoptosis signaling pathway in healthy rat myocardium. Intermittent fasting reduces pro-apoptotic and increases antiapoptotic signaling, besides attenuating HIIT-induced cardiomyocyte hypertrophy and myocardial interstitial fibrosis.
Collapse
Affiliation(s)
- Marianna R Carvalho
- Striated Muscle Study Laboratory, Federal University of Mato Grosso do Sul (UFMS), Campo Grande, Brazil
| | - Maria Lua M Mendonça
- Striated Muscle Study Laboratory, Federal University of Mato Grosso do Sul (UFMS), Campo Grande, Brazil
| | - Jéssica M L Oliveira
- Striated Muscle Study Laboratory, Federal University of Mato Grosso do Sul (UFMS), Campo Grande, Brazil
| | - Rodrigo B Romanenghi
- Striated Muscle Study Laboratory, Federal University of Mato Grosso do Sul (UFMS), Campo Grande, Brazil
| | - Camila S Morais
- Striated Muscle Study Laboratory, Federal University of Mato Grosso do Sul (UFMS), Campo Grande, Brazil
| | - Gabriel E Ota
- Striated Muscle Study Laboratory, Federal University of Mato Grosso do Sul (UFMS), Campo Grande, Brazil
| | - Aline R R Lima
- Internal Medicine Department, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu, Brazil
| | - Rodrigo J Oliveira
- Striated Muscle Study Laboratory, Federal University of Mato Grosso do Sul (UFMS), Campo Grande, Brazil
| | - Wander F O Filiú
- Striated Muscle Study Laboratory, Federal University of Mato Grosso do Sul (UFMS), Campo Grande, Brazil
| | - Katashi Okoshi
- Internal Medicine Department, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu, Brazil
| | - Marina P Okoshi
- Internal Medicine Department, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu, Brazil
| | - Silvio A Oliveira-Junior
- Striated Muscle Study Laboratory, Federal University of Mato Grosso do Sul (UFMS), Campo Grande, Brazil
| | - Paula F Martinez
- Striated Muscle Study Laboratory, Federal University of Mato Grosso do Sul (UFMS), Campo Grande, Brazil.
| |
Collapse
|
21
|
Romanello V, Sandri M. The connection between the dynamic remodeling of the mitochondrial network and the regulation of muscle mass. Cell Mol Life Sci 2020; 78:1305-1328. [PMID: 33078210 PMCID: PMC7904552 DOI: 10.1007/s00018-020-03662-0] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 09/02/2020] [Accepted: 09/28/2020] [Indexed: 12/11/2022]
Abstract
The dynamic coordination of processes controlling the quality of the mitochondrial network is crucial to maintain the function of mitochondria in skeletal muscle. Changes of mitochondrial proteolytic system, dynamics (fusion/fission), and mitophagy induce pathways that affect muscle mass and performance. When muscle mass is lost, the risk of disease onset and premature death is dramatically increased. For instance, poor quality of muscles correlates with the onset progression of several age-related disorders such as diabetes, obesity, cancer, and aging sarcopenia. To date, there are no drug therapies to reverse muscle loss, and exercise remains the best approach to improve mitochondrial health and to slow atrophy in several diseases. This review will describe the principal mechanisms that control mitochondrial quality and the pathways that link mitochondrial dysfunction to muscle mass regulation.
Collapse
Affiliation(s)
- Vanina Romanello
- Venetian Institute of Molecular Medicine, via Orus 2, 35129, Padova, Italy.
- Department of Biomedical Science, University of Padova, via G. Colombo 3, 35100, Padova, Italy.
| | - Marco Sandri
- Venetian Institute of Molecular Medicine, via Orus 2, 35129, Padova, Italy.
- Department of Biomedical Science, University of Padova, via G. Colombo 3, 35100, Padova, Italy.
- Department of Medicine, McGill University, Montreal, Canada.
| |
Collapse
|
22
|
Exercise as a Therapeutic Strategy for Sarcopenia in Heart Failure: Insights into Underlying Mechanisms. Cells 2020; 9:cells9102284. [PMID: 33066240 PMCID: PMC7602002 DOI: 10.3390/cells9102284] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/06/2020] [Accepted: 10/10/2020] [Indexed: 12/16/2022] Open
Abstract
Sarcopenia, a syndrome commonly seen in elderly populations, is often characterized by a gradual loss of skeletal muscle, leading to the decline of muscle strength and physical performance. Growing evidence suggests that the prevalence of sarcopenia increases in patients with heart failure (HF), which is a dominant pathogenesis in the aging heart. HF causes diverse metabolic complications that may result in sarcopenia. Therefore, sarcopenia may act as a strong predictor of frailty, disability, and mortality associated with HF. Currently, standard treatments for slowing muscle loss in patients with HF are not available. Therefore, here, we review the pathophysiological mechanisms underlying sarcopenia in HF as well as current knowledge regarding the beneficial effects of exercise on sarcopenia in HF and related mechanisms, including hormonal changes, myostatin, oxidative stress, inflammation, apoptosis, autophagy, the ubiquitin-proteasome system, and insulin resistance.
Collapse
|
23
|
Powers SK, Ozdemir M, Hyatt H. Redox Control of Proteolysis During Inactivity-Induced Skeletal Muscle Atrophy. Antioxid Redox Signal 2020; 33:559-569. [PMID: 31941357 PMCID: PMC7454189 DOI: 10.1089/ars.2019.8000] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Significance: Skeletal muscles play essential roles in key body functions including breathing, locomotion, and glucose homeostasis; therefore, maintaining healthy skeletal muscles is important. Prolonged periods of muscle inactivity (e.g., bed rest, mechanical ventilation, or limb immobilization) result in skeletal muscle atrophy and weakness. Recent Advances: Disuse skeletal muscle atrophy occurs due to both accelerated proteolysis and decreased protein synthesis with proteolysis playing a leading role in some types of inactivity-induced atrophy. Although all major proteolytic systems are involved in inactivity-induced proteolysis in skeletal muscles, growing evidence indicates that both calpain and autophagy play an important role. Regulation of proteolysis in skeletal muscle is under complex control, but it is established that activation of both calpain and autophagy is directly linked to oxidative stress. Critical Issues: In this review, we highlight the experimental evidence that supports a cause and effect link between reactive oxygen species (ROS) and activation of both calpain and autophagy in skeletal muscle fibers during prolonged inactivity. We also review the sources of oxidant production in muscle fibers during inactivity-induced atrophy, and provide a detailed discussion on how ROS activates both calpain and autophagy during disuse muscle wasting. Future Directions: Future studies are required to delineate the specific mechanisms by which ROS activates both calpain and autophagy in skeletal muscles during prolonged periods of contractile inactivity. This knowledge is essential to develop the most effective strategies to protect against disuse muscle atrophy. Antioxid. Redox Signal. 33, 559-569.
Collapse
Affiliation(s)
- Scott K Powers
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, USA
| | - Mustafa Ozdemir
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, USA
| | - Hayden Hyatt
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
24
|
Alves CRR, Neves WD, de Almeida NR, Eichelberger EJ, Jannig PR, Voltarelli VA, Tobias GC, Bechara LRG, de Paula Faria D, Alves MJN, Hagen L, Sharma A, Slupphaug G, Moreira JBN, Wisloff U, Hirshman MF, Negrão CE, de Castro G, Chammas R, Swoboda KJ, Ruas JL, Goodyear LJ, Brum PC. Exercise training reverses cancer-induced oxidative stress and decrease in muscle COPS2/TRIP15/ALIEN. Mol Metab 2020; 39:101012. [PMID: 32408015 PMCID: PMC7283151 DOI: 10.1016/j.molmet.2020.101012] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 05/04/2020] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE We tested the hypothesis that exercise training would attenuate metabolic impairment in a model of severe cancer cachexia. METHODS We used multiple in vivo and in vitro methods to explore the mechanisms underlying the beneficial effects induced by exercise training in tumor-bearing rats. RESULTS Exercise training improved running capacity, prolonged lifespan, reduced oxidative stress, and normalized muscle mass and contractile function in tumor-bearing rats. An unbiased proteomic screening revealed COP9 signalosome complex subunit 2 (COPS2) as one of the most downregulated proteins in skeletal muscle at the early stage of cancer cachexia. Exercise training normalized muscle COPS2 protein expression in tumor-bearing rats and mice. Lung cancer patients with low endurance capacity had low muscle COPS2 protein expression as compared to age-matched control subjects. To test whether decrease in COPS2 protein levels could aggravate or be an intrinsic compensatory mechanism to protect myotubes from cancer effects, we performed experiments in vitro using primary myotubes. COPS2 knockdown in human myotubes affected multiple cellular pathways, including regulation of actin cytoskeleton. Incubation of cancer-conditioned media in mouse myotubes decreased F-actin expression, which was partially restored by COPS2 knockdown. Direct repeat 4 (DR4) response elements have been shown to positively regulate gene expression. COPS2 overexpression decreased the DR4 activity in mouse myoblasts, and COPS2 knockdown inhibited the effects of cancer-conditioned media on DR4 activity. CONCLUSIONS These studies demonstrated that exercise training may be an important adjuvant therapy to counteract cancer cachexia and uncovered novel mechanisms involving COPS2 to regulate myotube homeostasis in cancer cachexia.
Collapse
Affiliation(s)
- Christiano R R Alves
- School of Physical Education and Sport, University of Sao Paulo, Sao Paulo, Brazil; Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA; Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA.
| | - Willian das Neves
- School of Physical Education and Sport, University of Sao Paulo, Sao Paulo, Brazil; Instituto do Cancer do Estado de Sao Paulo ICESP, Hospital das Clinicas HC FMUSP, Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Ney R de Almeida
- School of Physical Education and Sport, University of Sao Paulo, Sao Paulo, Brazil
| | - Eric J Eichelberger
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Paulo R Jannig
- School of Physical Education and Sport, University of Sao Paulo, Sao Paulo, Brazil; Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Vanessa A Voltarelli
- School of Physical Education and Sport, University of Sao Paulo, Sao Paulo, Brazil
| | - Gabriel C Tobias
- School of Physical Education and Sport, University of Sao Paulo, Sao Paulo, Brazil
| | - Luiz R G Bechara
- School of Physical Education and Sport, University of Sao Paulo, Sao Paulo, Brazil
| | - Daniele de Paula Faria
- Department of Radiology and Oncology, Faculdade de Medicina da Universidade de São Paulo, Sao Paulo, Brazil
| | - Maria J N Alves
- Heart Institute, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Lars Hagen
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway; Proteomics and Modomics Experimental Core, PROMEC, at NTNU and the Central Norway Regional Health Authority, Stjørdal, Norway
| | - Animesh Sharma
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway; Proteomics and Modomics Experimental Core, PROMEC, at NTNU and the Central Norway Regional Health Authority, Stjørdal, Norway
| | - Geir Slupphaug
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway; Proteomics and Modomics Experimental Core, PROMEC, at NTNU and the Central Norway Regional Health Authority, Stjørdal, Norway
| | - José B N Moreira
- K.G. Jebsen Center of Exercise in Medicine at Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - Ulrik Wisloff
- K.G. Jebsen Center of Exercise in Medicine at Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - Michael F Hirshman
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Carlos E Negrão
- School of Physical Education and Sport, University of Sao Paulo, Sao Paulo, Brazil; Heart Institute, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Gilberto de Castro
- Instituto do Cancer do Estado de Sao Paulo ICESP, Hospital das Clinicas HC FMUSP, Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Roger Chammas
- Department of Radiology and Oncology, Faculdade de Medicina da Universidade de São Paulo, Sao Paulo, Brazil
| | - Kathryn J Swoboda
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Jorge L Ruas
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Laurie J Goodyear
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Patricia C Brum
- School of Physical Education and Sport, University of Sao Paulo, Sao Paulo, Brazil.
| |
Collapse
|
25
|
Campos JC, Baehr LM, Ferreira ND, Bozi LHM, Andres AM, Ribeiro MAC, Gottlieb RA, Bodine SC, Ferreira JCB. β 2 -adrenoceptor activation improves skeletal muscle autophagy in neurogenic myopathy. FASEB J 2020; 34:5628-5641. [PMID: 32112488 DOI: 10.1096/fj.201902305r] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 02/03/2020] [Accepted: 02/14/2020] [Indexed: 01/21/2023]
Abstract
β2 -adrenoceptor agonists improve autophagy and re-establish proteostasis in cardiac cells; therefore, suggesting autophagy as a downstream effector of β2 -adrenoceptor signaling pathway. Here, we used the pharmacological and genetic tools to determine the autophagy effect of sustained β2 -adrenoceptor activation in rodents with neurogenic myopathy, which display impaired skeletal muscle autophagic flux. Sustained β2 -adrenoceptor activation using Formoterol (10 μg kg-1 day-1 ), starting at the onset of neurogenic myopathy, prevents disruption of autophagic flux in skeletal muscle 14 days after sciatic nerve constriction. These changes are followed by reduction of the cytotoxic protein levels and increased skeletal muscle cross-sectional area and contractility properties. Of interest, sustained administration of Formoterol at lower concentration (1 μg kg-1 day-1 ) induces similar improvements in skeletal muscle autophagic flux and contractility properties in neurogenic myopathy, without affecting the cross-sectional area. Sustained pharmacological inhibition of autophagy using Chloroquine (50 mg kg-1 day-1 ) abolishes the beneficial effects of β2 -adrenoceptor activation on the skeletal muscle proteostasis and contractility properties in neurogenic myopathy. Further supporting an autophagy mechanism for β2 -adrenoceptor activation, skeletal muscle-specific deletion of ATG7 blunts the beneficial effects of β2 -adrenoceptor on skeletal muscle proteostasis and contractility properties in neurogenic myopathy in mice. These findings suggest autophagy as a critical downstream effector of β2 -adrenoceptor signaling pathway in skeletal muscle.
Collapse
Affiliation(s)
- Juliane C Campos
- Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Leslie M Baehr
- Department of Internal Medicine, Endocrinology and Metabolism Division, University of Iowa, Iowa City, IA, USA
| | - Nikolas D Ferreira
- Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Luiz H M Bozi
- Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Allen M Andres
- The Cedars-Sinai Heart Institute and the Barbra Streisand Women's Heart Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Márcio A C Ribeiro
- Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Roberta A Gottlieb
- The Cedars-Sinai Heart Institute and the Barbra Streisand Women's Heart Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Sue C Bodine
- Department of Internal Medicine, Endocrinology and Metabolism Division, University of Iowa, Iowa City, IA, USA
| | - Julio C B Ferreira
- Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
- Department of Chemical & Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
26
|
Henríquez-Olguín C, Boronat S, Cabello-Verrugio C, Jaimovich E, Hidalgo E, Jensen TE. The Emerging Roles of Nicotinamide Adenine Dinucleotide Phosphate Oxidase 2 in Skeletal Muscle Redox Signaling and Metabolism. Antioxid Redox Signal 2019; 31:1371-1410. [PMID: 31588777 PMCID: PMC6859696 DOI: 10.1089/ars.2018.7678] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Significance: Skeletal muscle is a crucial tissue to whole-body locomotion and metabolic health. Reactive oxygen species (ROS) have emerged as intracellular messengers participating in both physiological and pathological adaptations in skeletal muscle. A complex interplay between ROS-producing enzymes and antioxidant networks exists in different subcellular compartments of mature skeletal muscle. Recent evidence suggests that nicotinamide adenine dinucleotide phosphate (NADPH) oxidases (NOXs) are a major source of contraction- and insulin-stimulated oxidants production, but they may paradoxically also contribute to muscle insulin resistance and atrophy. Recent Advances: Pharmacological and molecular biological tools, including redox-sensitive probes and transgenic mouse models, have generated novel insights into compartmentalized redox signaling and suggested that NOX2 contributes to redox control of skeletal muscle metabolism. Critical Issues: Major outstanding questions in skeletal muscle include where NOX2 activation occurs under different conditions in health and disease, how NOX2 activation is regulated, how superoxide/hydrogen peroxide generated by NOX2 reaches the cytosol, what the signaling mediators are downstream of NOX2, and the role of NOX2 for different physiological and pathophysiological processes. Future Directions: Future research should utilize and expand the current redox-signaling toolbox to clarify the NOX2-dependent mechanisms in skeletal muscle and determine whether the proposed functions of NOX2 in cells and animal models are conserved into humans.
Collapse
Affiliation(s)
- Carlos Henríquez-Olguín
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports (NEXS), Faculty of Science, University of Copenhagen, Copenhagen, Denmark.,Muscle Cell Physiology Laboratory, Center for Exercise, Metabolism, and Cancer, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - Susanna Boronat
- Oxidative Stress and Cell Cycle Group, Universitat Pompeu Fabra, Barcelona, Spain
| | - Claudio Cabello-Verrugio
- Laboratory of Muscle Pathology, Fragility and Aging, Department of Biological Sciences, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Santiago, Chile.,Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago, Chile
| | - Enrique Jaimovich
- Muscle Cell Physiology Laboratory, Center for Exercise, Metabolism, and Cancer, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - Elena Hidalgo
- Oxidative Stress and Cell Cycle Group, Universitat Pompeu Fabra, Barcelona, Spain
| | - Thomas E Jensen
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports (NEXS), Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
27
|
Himori K, Tatebayashi D, Ashida Y, Yamada T. Eccentric training enhances the αB-crystallin binding to the myofibrils and prevents skeletal muscle weakness in adjuvant-induced arthritis rat. J Appl Physiol (1985) 2019; 127:71-80. [DOI: 10.1152/japplphysiol.00102.2019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Patients with rheumatoid arthritis (RA) frequently suffer from muscle weakness. We examined whether eccentric training prevents skeletal muscle weakness in adjuvant-induced arthritis (AIA) rat, a widely used animal model for RA. AIA was induced in the knees of Wistar rats by injection of complete Freund’s adjuvant. To induce eccentric contractions (ECCs), neuromuscular electrical stimulation (45 V) was applied to the plantar flexor muscles simultaneously with forced dorsiflexion of the ankle joint (0–40°) and was given every 6 s. ECC exercise was applied every other day for a total of 11 sessions and consisted of 4 sets of 5 contractions. There was a significant reduction in in vitro maximum Ca2+-activated force in skinned fibers in gastrocnemius muscle from AIA rats. These changes were associated with reduced expression levels of contractile proteins (i.e., myosin and actin), increased levels of inflammation redox stress-related biomarkers (i.e., TNF-α, malondialdehyde-protein adducts, NADPH oxidase 2, and neuronal nitric oxide synthase), and autolyzed active calpain-1 in AIA muscles. ECC training markedly enhanced the steady-state levels of αB-crystallin, a small heat shock protein, and its binding to the myofibrils and prevented the AIA-induced myofibrillar dysfunction, reduction in contractile proteins, and inflammation-oxidative stress insults. Our findings demonstrate that ECC training preserves myofibrillar function without muscle damage in AIA rats, which is at least partially attributable to the protective effect of αB-crystallin on the myofibrils against oxidative stress-mediated protein degeneration. Thus ECC training can be a safe and effective intervention, counteracting the loss of muscle strength in RA patients. NEW & NOTEWORTHY Eccentric contractions (ECCs) are regarded as an effective way to increase muscle strength. No studies, however, assess safety and effectiveness of ECC training on muscle weakness associated with rheumatoid arthritis. Here, we used adjuvant-induced arthritis (AIA) rats to demonstrate that ECC training prevents intrinsic contractile dysfunction without muscle damage in AIA rats, which may be attributed to the protective effect of αB-crystallin on the myofibrils against inflammation-oxidative stress insults.
Collapse
Affiliation(s)
- Koichi Himori
- Graduate School of Health Sciences, Sapporo Medical University, Sapporo, Japan
| | - Daisuke Tatebayashi
- Graduate School of Health Sciences, Sapporo Medical University, Sapporo, Japan
| | - Yuki Ashida
- Graduate School of Health Sciences, Sapporo Medical University, Sapporo, Japan
| | - Takashi Yamada
- Graduate School of Health Sciences, Sapporo Medical University, Sapporo, Japan
| |
Collapse
|
28
|
Di Meo S, Napolitano G, Venditti P. Mediators of Physical Activity Protection against ROS-Linked Skeletal Muscle Damage. Int J Mol Sci 2019; 20:E3024. [PMID: 31226872 PMCID: PMC6627449 DOI: 10.3390/ijms20123024] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 06/12/2019] [Accepted: 06/17/2019] [Indexed: 12/24/2022] Open
Abstract
Unaccustomed and/or exhaustive exercise generates excessive free radicals and reactive oxygen and nitrogen species leading to muscle oxidative stress-related damage and impaired contractility. Conversely, a moderate level of free radicals induces the body's adaptive responses. Thus, a low oxidant level in resting muscle is essential for normal force production, and the production of oxidants during each session of physical training increases the body's antioxidant defenses. Mitochondria, NADPH oxidases and xanthine oxidases have been identified as sources of free radicals during muscle contraction, but the exact mechanisms underlying exercise-induced harmful or beneficial effects yet remain elusive. However, it is clear that redox signaling influences numerous transcriptional activators, which regulate the expression of genes involved in changes in muscle phenotype. The mitogen-activated protein kinase family is one of the main links between cellular oxidant levels and skeletal muscle adaptation. The family components phosphorylate and modulate the activities of hundreds of substrates, including transcription factors involved in cell response to oxidative stress elicited by exercise in skeletal muscle. To elucidate the complex role of ROS in exercise, here we reviewed the literature dealing on sources of ROS production and concerning the most important redox signaling pathways, including MAPKs that are involved in the responses to acute and chronic exercise in the muscle, particularly those involved in the induction of antioxidant enzymes.
Collapse
Affiliation(s)
- Sergio Di Meo
- Dipartimento di Biologia, Università di Napoli Federico II, Complesso Universitario Monte Sant'Angelo, Via Cinthia, I-80126 Napoli, Italy.
| | - Gaetana Napolitano
- Dipartimento di Scienze e Tecnologie, Università degli Studi di Napoli Parthenope, via Acton n. 38-I-80133 Napoli, Italy.
| | - Paola Venditti
- Dipartimento di Biologia, Università di Napoli Federico II, Complesso Universitario Monte Sant'Angelo, Via Cinthia, I-80126 Napoli, Italy.
| |
Collapse
|
29
|
Taghi Beigi H, Esfarjani F, Marandi SM, Karami H. The Effect of 8-Week Aerobic Exercise on the Expression of Regulatory Subunits of NADPH Oxidase 2 (p47phox and p67phox) in the Cardiac Tissue of Diabetic Rats. INTERNATIONAL JOURNAL OF BASIC SCIENCE IN MEDICINE 2019. [DOI: 10.15171/ijbsm.2019.05] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Introduction: Oxidative stress seems to play a major role in diabetes-induced cardiac dysfunction, known as diabetic cardiomyopathy (DCM). Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 2 is considered as one of the main enzymatic systems which primarily contributes to the production of reactive oxygen species in various organs including the heart. The present study aimed to investigate the effects of 8 weeks of aerobic exercise (AE) on the expression of p47phox and p67phox, which are regarded as the regulatory subunits of NADPH oxidase 2 in the cardiac tissue in diabetic rats. Methods: A total of 36 male Wistar rats with a mean weight of 231±25 g were randomly divided into non-diabetic, control diabetic, and trained diabetic groups (each containing 12 rats). Nicotinamide and streptozotocin were used to induce diabetes in the rats. The cardiac muscle was removed under sterile conditions 48 hours following the last training session. Finally, the mRNA levels of p47phox and p67phox were evaluated using the real-time polymerase chain reaction. Results: The results showed that diabetes induction significantly increased the gene expressions of p47phox and p67phox in the cardiac tissue of diabetic rats. The expression of these genes was significantly attenuated after 8 weeks of AE. Conclusion: In general, AE was found to prevent the negative effects of diabetes by suppressing p47phox and p67phox in the cardiac tissue of diabetic rats. Therefore, this can improve cardiac function and may be a potential preventive or therapeutic modality for DCM.
Collapse
Affiliation(s)
- Hamid Taghi Beigi
- Department of Exercise Physiology, Faculty of Sport Sciences, University of Isfahan, Isfahan, Iran
| | - Fahimeh Esfarjani
- Department of Exercise Physiology, Faculty of Sport Sciences, University of Isfahan, Isfahan, Iran
| | - Seyed Mohammad Marandi
- Department of Exercise Physiology, Faculty of Sport Sciences, University of Isfahan, Isfahan, Iran
| | - Hadi Karami
- Department of Molecular Medicine and Biotechnology, Faculty of Medical Sciences, Arak University of Medical Sciences, Arak, Iran
| |
Collapse
|
30
|
Kiyuna LA, Albuquerque RPE, Chen CH, Mochly-Rosen D, Ferreira JCB. Targeting mitochondrial dysfunction and oxidative stress in heart failure: Challenges and opportunities. Free Radic Biol Med 2018; 129:155-168. [PMID: 30227272 PMCID: PMC6309415 DOI: 10.1016/j.freeradbiomed.2018.09.019] [Citation(s) in RCA: 137] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 08/28/2018] [Accepted: 09/14/2018] [Indexed: 02/06/2023]
Abstract
Mitochondrial dysfunction characterized by impaired bioenergetics, oxidative stress and aldehydic load is a hallmark of heart failure. Recently, different research groups have provided evidence that selective activation of mitochondrial detoxifying systems that counteract excessive accumulation of ROS, RNS and reactive aldehydes is sufficient to stop cardiac degeneration upon chronic stress, such as heart failure. Therefore, pharmacological and non-pharmacological approaches targeting mitochondria detoxification may play a critical role in the prevention or treatment of heart failure. In this review we discuss the most recent findings on the central role of mitochondrial dysfunction, oxidative stress and aldehydic load in heart failure, highlighting the most recent preclinical and clinical studies using mitochondria-targeted molecules and exercise training as effective tools against heart failure.
Collapse
Affiliation(s)
- Ligia Akemi Kiyuna
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, Brazil
| | | | - Che-Hong Chen
- Department of Chemical and Systems Biology, Stanford University School of Medicine, USA
| | - Daria Mochly-Rosen
- Department of Chemical and Systems Biology, Stanford University School of Medicine, USA
| | | |
Collapse
|
31
|
Exercise prevents impaired autophagy and proteostasis in a model of neurogenic myopathy. Sci Rep 2018; 8:11818. [PMID: 30087400 PMCID: PMC6081439 DOI: 10.1038/s41598-018-30365-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 07/26/2018] [Indexed: 01/07/2023] Open
Abstract
Increased proteolytic activity has been widely associated with skeletal muscle atrophy. However, elevated proteolysis is also critical for the maintenance of cellular homeostasis by disposing cytotoxic proteins and non-functioning organelles. We recently demonstrated that exercise activates autophagy and re-establishes proteostasis in cardiac diseases. Here, we characterized the impact of exercise on skeletal muscle autophagy and proteostasis in a model of neurogenic myopathy induced by sciatic nerve constriction in rats. Neurogenic myopathy, characterized by progressive atrophy and impaired contractility, was paralleled by accumulation of autophagy-related markers and loss of acute responsiveness to both colchicine and chloroquine. These changes were correlated with elevated levels of damaged proteins, chaperones and pro-apoptotic markers compared to control animals. Sustained autophagy inhibition using chloroquine in rats (50 mg.kg-1.day-1) or muscle-specific deletion of Atg7 in mice was sufficient to impair muscle contractility in control but not in neurogenic myopathy, suggesting that dysfunctional autophagy is critical in skeletal muscle pathophysiology. Finally, 4 weeks of aerobic exercise training (moderate treadmill running, 5x/week, 1 h/day) prior to neurogenic myopathy improved skeletal muscle autophagic flux and proteostasis. These changes were followed by spared muscle mass and better contractility properties. Taken together, our findings suggest the potential value of exercise in maintaining skeletal muscle proteostasis and slowing down the progression of neurogenic myopathy.
Collapse
|
32
|
Cangemi R, Loffredo L, Battaglia S, Perri L, Santulli M, Carnevale R, Nocella C, Muscolo M, Masala D, Violi F. Does Regular Physical Exercise Preserve Artery Dilation by Lowering Nox2-Related Oxidative Stress? Antioxid Redox Signal 2018; 28:1576-1581. [PMID: 28990416 DOI: 10.1089/ars.2017.7296] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Habitual physical activity has beneficial effects on cardiovascular risk reduction by improving vascular function but the underlying mechanism is still unclear. To address this issue, we performed a cross-sectional study comparing 50 physically active (PA) adults with 50 sedentary controls matched for age, sex, and cardiovascular risk factors. PA subjects had significantly higher flow-mediated dilation (FMD) than controls and higher serum levels of nitrite/nitrate, a marker of nitric oxide generation. In addition, PA subjects showed lower levels of urinary isoprostanes, a marker of reactive oxygen species (ROS) production, and lower serum levels of sNox2-dp, a validated assay to measure Nox2 activity, one of the most important enzymes producing ROS in the blood cells. FMD was independently correlated with sNox2-dp, after adjusting for possible confounding factors. Our observation leads to the hypothesis that, in adults, regular exercise preserves artery dilation through Nox2 decreased activity. Antioxid. Redox Signal. 28, 1576-1581.
Collapse
Affiliation(s)
- Roberto Cangemi
- 1 Department of Internal Medicine and Medical Specialties, Sapienza University of Rome , Rome, Italy
| | - Lorenzo Loffredo
- 1 Department of Internal Medicine and Medical Specialties, Sapienza University of Rome , Rome, Italy
| | - Simona Battaglia
- 1 Department of Internal Medicine and Medical Specialties, Sapienza University of Rome , Rome, Italy
| | - Ludovica Perri
- 1 Department of Internal Medicine and Medical Specialties, Sapienza University of Rome , Rome, Italy
| | - Maria Santulli
- 2 Department of Experimental Medicine, Sapienza University of Rome , Rome, Italy
| | - Roberto Carnevale
- 3 Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome , Rome, Italy
| | - Cristina Nocella
- 3 Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome , Rome, Italy
| | - Miryam Muscolo
- 1 Department of Internal Medicine and Medical Specialties, Sapienza University of Rome , Rome, Italy
| | - Daniele Masala
- 4 Department of Human, Social and Health Sciences, University of Cassino and Southern Latium , Cassino, Italy
| | - Francesco Violi
- 1 Department of Internal Medicine and Medical Specialties, Sapienza University of Rome , Rome, Italy
| |
Collapse
|
33
|
Deletion of NAD(P)H Oxidase 2 Prevents Angiotensin II-Induced Skeletal Muscle Atrophy. BIOMED RESEARCH INTERNATIONAL 2018; 2018:3194917. [PMID: 29487866 PMCID: PMC5816890 DOI: 10.1155/2018/3194917] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 12/01/2017] [Accepted: 12/12/2017] [Indexed: 12/20/2022]
Abstract
Skeletal muscle atrophy is induced by an imbalance between protein synthesis and degradation. Our previous studies reported that angiotensin II (AII) directly induced muscle atrophy in mice. This study investigated the role of NAD(P)H oxidase 2 (Nox2) activation by AII in the induction of skeletal muscle atrophy. For 4 weeks, either saline (vehicle: V) or AII (1000 ng kg−1 min−1) was infused into male wild-type (WT) and Nox2 knockout (KO) mice via osmotic minipumps. Experiments were performed in the following 4 groups: WT + V, KO + V, WT + AII, and KO + AII. Body weight, muscle weight, and myocyte cross-sectional area were significantly decreased in WT + AII compared to WT + V mice, and these changes were not observed in KO + AII mice. Akt phosphorylation of Ser473 and p70S6K of Thr389 was decreased, gene expression levels of MuRF-1 and atrogin-1 were increased in WT + AII compared to WT + V, and these changes were significantly attenuated in KO + AII mice. The deletion of Nox2 prevented AII-induced skeletal muscle atrophy via improving the balance between protein synthesis and degradation. Therefore, Nox2 may be a therapeutic target for AII-induced skeletal muscle atrophy.
Collapse
|
34
|
Ábrigo J, Elorza AA, Riedel CA, Vilos C, Simon F, Cabrera D, Estrada L, Cabello-Verrugio C. Role of Oxidative Stress as Key Regulator of Muscle Wasting during Cachexia. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:2063179. [PMID: 29785242 PMCID: PMC5896211 DOI: 10.1155/2018/2063179] [Citation(s) in RCA: 146] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 02/07/2018] [Indexed: 12/11/2022]
Abstract
Skeletal muscle atrophy is a pathological condition mainly characterized by a loss of muscular mass and the contractile capacity of the skeletal muscle as a consequence of muscular weakness and decreased force generation. Cachexia is defined as a pathological condition secondary to illness characterized by the progressive loss of muscle mass with or without loss of fat mass and with concomitant diminution of muscle strength. The molecular mechanisms involved in cachexia include oxidative stress, protein synthesis/degradation imbalance, autophagy deregulation, increased myonuclear apoptosis, and mitochondrial dysfunction. Oxidative stress is one of the most common mechanisms of cachexia caused by different factors. It results in increased ROS levels, increased oxidation-dependent protein modification, and decreased antioxidant system functions. In this review, we will describe the importance of oxidative stress in skeletal muscles, its sources, and how it can regulate protein synthesis/degradation imbalance, autophagy deregulation, increased myonuclear apoptosis, and mitochondrial dysfunction involved in cachexia.
Collapse
Affiliation(s)
- Johanna Ábrigo
- 1Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas, Universidad Andres Bello, Santiago, Chile
- 2Millennium Institute of Immunology and Immunotherapy, Santiago, Chile
| | - Alvaro A. Elorza
- 2Millennium Institute of Immunology and Immunotherapy, Santiago, Chile
- 3Centro de Investigaciones Biomédicas, Facultad de Ciencias Biológicas & Facultad de Medicina, Universidad Andres Bello, Santiago, Chile
| | - Claudia A. Riedel
- 1Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas, Universidad Andres Bello, Santiago, Chile
- 2Millennium Institute of Immunology and Immunotherapy, Santiago, Chile
| | - Cristian Vilos
- 4Laboratory of Nanomedicine and Targeted Delivery, Center for Integrative Medicine and Innovative Science, Faculty of Medicine, and Center for Bioinformatics and Integrative Biology, Faculty of Biological Sciences, Universidad Andres Bello, Santiago, Chile
- 5Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago, Chile
| | - Felipe Simon
- 1Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas, Universidad Andres Bello, Santiago, Chile
- 2Millennium Institute of Immunology and Immunotherapy, Santiago, Chile
| | - Daniel Cabrera
- 6Departamento de Gastroenterología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
- 7Departamento de Ciencias Químicas y Biológicas, Facultad de Salud, Universidad Bernardo O'Higgins, Santiago, Chile
| | - Lisbell Estrada
- 8Centro Integrativo de Biología y Química Aplicada, Universidad Bernardo O'Higgins, Santiago, Chile
| | - Claudio Cabello-Verrugio
- 1Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas, Universidad Andres Bello, Santiago, Chile
- 2Millennium Institute of Immunology and Immunotherapy, Santiago, Chile
| |
Collapse
|
35
|
Krylatov AV, Maslov LN, Voronkov NS, Boshchenko AA, Popov SV, Gomez L, Wang H, Jaggi AS, Downey JM. Reactive Oxygen Species as Intracellular Signaling Molecules in the Cardiovascular System. Curr Cardiol Rev 2018; 14:290-300. [PMID: 29962348 PMCID: PMC6300799 DOI: 10.2174/1573403x14666180702152436] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 06/13/2018] [Accepted: 06/20/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Redox signaling plays an important role in the lives of cells. This signaling not only becomes apparent in pathologies but is also thought to be involved in maintaining physiological homeostasis. Reactive Oxygen Species (ROS) can activate protein kinases: CaMKII, PKG, PKA, ERK, PI3K, Akt, PKC, PDK, JNK, p38. It is unclear whether it is a direct interaction of ROS with these kinases or whether their activation is a consequence of inhibition of phosphatases. ROS have a biphasic effect on the transport of Ca2+ in the cell: on one hand, they activate the sarcoplasmic reticulum Ca2+-ATPase, which can reduce the level of Ca2+ in the cell, and on the other hand, they can inactivate Ca2+-ATPase of the plasma membrane and open the cation channels TRPM2, which promote Ca2+-loading and subsequent apoptosis. ROS inhibit the enzyme PHD2, which leads to the stabilization of HIF-α and the formation of the active transcription factor HIF. CONCLUSION Activation of STAT3 and STAT5, induced by cytokines or growth factors, may include activation of NADPH oxidase and enhancement of ROS production. Normal physiological production of ROS under the action of cytokines activates the JAK/STAT while excessive ROS production leads to their inhibition. ROS cause the activation of the transcription factor NF-κB. Physiological levels of ROS control cell proliferation and angiogenesis. ROS signaling is also involved in beneficial adaptations to survive ischemia and hypoxia, while further increases in ROS can trigger programmed cell death by the mechanism of apoptosis or autophagy. ROS formation in the myocardium can be reduced by moderate exercise.
Collapse
Affiliation(s)
| | - Leonid N. Maslov
- Address correspondence to this author at the Cardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of
Science, Tomsk, Russia; Tel: 3822 262174; Fax: 3822 555057;
E-mail:
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Biolo G, Di Girolamo FG, Fiotti N, Mearelli F, Sarto P. Exercise-mediated reactive oxygen species generation in athletes and in patients with chronic disease. Intern Emerg Med 2017; 12:741-744. [PMID: 28567558 DOI: 10.1007/s11739-017-1689-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 05/24/2017] [Indexed: 10/19/2022]
Affiliation(s)
- Gianni Biolo
- Department of Medical, Surgical and Health Sciences, Clinica Medica, ASUITs, University of Trieste, Cattinara University Hospital, Strada di Fiume 447, 34149, Trieste, Italy.
| | - Filippo Giorgio Di Girolamo
- Department of Medical, Surgical and Health Sciences, Clinica Medica, ASUITs, University of Trieste, Cattinara University Hospital, Strada di Fiume 447, 34149, Trieste, Italy
| | - Nicola Fiotti
- Department of Medical, Surgical and Health Sciences, Clinica Medica, ASUITs, University of Trieste, Cattinara University Hospital, Strada di Fiume 447, 34149, Trieste, Italy
| | - Filippo Mearelli
- Department of Medical, Surgical and Health Sciences, Clinica Medica, ASUITs, University of Trieste, Cattinara University Hospital, Strada di Fiume 447, 34149, Trieste, Italy
| | | |
Collapse
|
37
|
Arginine and aerobic training prevent endothelial and metabolic alterations in rats at high risk for the development of the metabolic syndrome. Br J Nutr 2017; 118:1-10. [DOI: 10.1017/s0007114517001702] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
AbstractEndothelial function is a key mechanism in the development of CVD. Arginine and exercise are important non-pharmacological strategies for mitigating the impact of metabolic changes in the metabolic syndrome, but the effect of their combined administration is unknown. Thus, the aim of this study was to investigate the isolated and combined effects of aerobic training and arginine supplementation on metabolic variables and vascular reactivity in rats at high risk for developing the metabolic syndrome. Wistar rats were divided into two groups: control and fructose (F – water with 10 % fructose). After 2 weeks, the F group was divided into four groups: F, fructose+arginine (FA, 880 mg/kg per d of l-arginine), fructose+training (FT) and fructose+arginine+training (FTA); treatments lasted for 8 weeks, and no difference was observed in body mass gain. Arginine did not improve the body protein content, and both the FA and FT groups show a reversal of the increase in adipose tissue. Insulin increase was prevented by training and arginine, without additive effect, and the increase in serum TAG was prevented only by training. The F group showed impaired endothelium-dependent vasodilation and hyperreactivity to phenylephrine, but arginine and training were capable of preventing these effects, even separately. Higher nitric oxide level was observed in the FA and FT groups, and no potentiating effect was detected. Thus, only training was able to prevent the increase in TAG and improve the protein mass, and training and arginine exert similar effects on fat content, insulin and endothelial function, but these effects are not additive.
Collapse
|