101
|
Gopar-Cuevas Y, Duarte-Jurado AP, Diaz-Perez RN, Saucedo-Cardenas O, Loera-Arias MJ, Montes-de-Oca-Luna R, Rodriguez-Rocha H, Garcia-Garcia A. Pursuing Multiple Biomarkers for Early Idiopathic Parkinson's Disease Diagnosis. Mol Neurobiol 2021; 58:5517-5532. [PMID: 34350555 DOI: 10.1007/s12035-021-02500-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 07/16/2021] [Indexed: 12/21/2022]
Abstract
Parkinson's disease (PD) ranks first in the world as a neurodegenerative movement disorder and occurs most commonly in an idiopathic form. PD patients may have motor symptoms, non-motor symptoms, including cognitive and behavioral changes, and symptoms related to autonomic nervous system (ANS) failures, such as gastrointestinal, urinary, and cardiovascular symptoms. Unfortunately, the diagnostic accuracy of PD by general neurologists is relatively low. Currently, there is no objective molecular or biochemical test for PD; its diagnosis is based on clinical criteria, mainly by cardinal motor symptoms, which manifest when patients have lost about 60-80% of dopaminergic neurons. Therefore, it is urgent to establish a panel of biomarkers for the early and accurate diagnosis of PD. Once the disease is accurately diagnosed, it may be easier to unravel idiopathic PD's pathogenesis, and ultimately, finding a cure. This review discusses several biomarkers' potential to set a panel for early idiopathic PD diagnosis and future directions.
Collapse
Affiliation(s)
- Yareth Gopar-Cuevas
- Departamento de Histología, Facultad de Medicina, Universidad Autónoma de Nuevo León, 64460, Monterrey, Mexico
| | - Ana P Duarte-Jurado
- Departamento de Histología, Facultad de Medicina, Universidad Autónoma de Nuevo León, 64460, Monterrey, Mexico
| | - Rosa N Diaz-Perez
- Departamento de Histología, Facultad de Medicina, Universidad Autónoma de Nuevo León, 64460, Monterrey, Mexico
| | - Odila Saucedo-Cardenas
- Departamento de Histología, Facultad de Medicina, Universidad Autónoma de Nuevo León, 64460, Monterrey, Mexico.,Departamento de Genética Molecular, Centro de Investigación Biomédica del Noreste, Delegación Nuevo León, Instituto Mexicano del Seguro Social, Monterrey, Mexico
| | - Maria J Loera-Arias
- Departamento de Histología, Facultad de Medicina, Universidad Autónoma de Nuevo León, 64460, Monterrey, Mexico
| | - Roberto Montes-de-Oca-Luna
- Departamento de Histología, Facultad de Medicina, Universidad Autónoma de Nuevo León, 64460, Monterrey, Mexico
| | - Humberto Rodriguez-Rocha
- Departamento de Histología, Facultad de Medicina, Universidad Autónoma de Nuevo León, 64460, Monterrey, Mexico.
| | - Aracely Garcia-Garcia
- Departamento de Histología, Facultad de Medicina, Universidad Autónoma de Nuevo León, 64460, Monterrey, Mexico.
| |
Collapse
|
102
|
Abstract
➤ Oxidative stress has been implicated as a causative factor in many disease states, possibly including the diminished bone mineral density in osteoporosis. ➤ Understanding the effects of oxidative stress on the development of osteoporosis may lead to further research improving preventative and therapeutic measures that can combat this important contributor to morbidity and mortality worldwide. ➤ A diet rich in whole plant foods with high antioxidant content along with antioxidant-preserving lifestyle changes may improve bone mineral density and reduce the risk of fragility-related fractures. While it is not explicitly clear if antioxidant activity is the effector of this change, the current evidence supports this possibility. ➤ Supplementation with isolated antioxidants may also provide some osteoprotective benefits, but whole plant food-derived antioxidants potentially have more overall benefits. Larger-scale clinical trials are needed to give credence to definitive clinical recommendations.
Collapse
Affiliation(s)
- Jeff S Kimball
- Loma Linda University Medical Center, Loma Linda, California
| | | | | |
Collapse
|
103
|
The ketogenic diet as a therapeutic intervention strategy in mitochondrial disease. Int J Biochem Cell Biol 2021; 138:106050. [PMID: 34298163 DOI: 10.1016/j.biocel.2021.106050] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/02/2021] [Accepted: 07/16/2021] [Indexed: 11/21/2022]
Abstract
Classical mitochondrial disease (MD) represents a group of complex metabolic syndromes primarily linked to dysfunction of the mitochondrial ATP-generating oxidative phosphorylation (OXPHOS) system. To date, effective therapies for these diseases are lacking. Here we discuss the ketogenic diet (KD), being a high-fat, moderate protein, and low carbohydrate diet, as a potential intervention strategy. We concisely review the impact of the KD on bioenergetics, ROS/redox metabolism, mitochondrial dynamics and mitophagy. Next, the consequences of the KD in (models of) MD, as well as KD adverse effects, are described. It is concluded that the current experimental evidence suggests that the KD can positively impact on mitochondrial bioenergetics, mitochondrial ROS/redox metabolism and mitochondrial dynamics. However, more information is required on the bioenergetic consequences and mechanistic mode-of-action aspects of the KD at the cellular level and in MD patients.
Collapse
|
104
|
He J, Liu R, Zheng W, Guo H, Yang Y, Zhao R, Yao W. High ambient temperature exposure during late gestation disrupts glycolipid metabolism and hepatic mitochondrial function tightly related to gut microbial dysbiosis in pregnant mice. Microb Biotechnol 2021; 14:2116-2129. [PMID: 34272826 PMCID: PMC8449678 DOI: 10.1111/1751-7915.13893] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 07/03/2021] [Indexed: 12/11/2022] Open
Abstract
As global warming intensifies, emerging evidence has demonstrated high ambient temperature during pregnancy negatively affects maternal physiology with compromised pregnant outcomes; however, little is known about the roles of gut microbiota and its underlying mechanisms in this process. Here, for the first time, we explored the potential mechanisms of gut microbiota involved in the disrupted glycolipid metabolism via hepatic mitochondrial function. Our results indicate heat stress (HS) reduces fat and protein contents and serum levels of insulin and triglyceride (TG), while increases that of non-esterified fatty acid (NEFA), β-hydroxybutyric acid (B-HBA), creatinine and blood urea nitrogen (BUN) (P < 0.05). Additionally, HS downregulates both mitochondrial genes (mtDNA) and nuclear encoding mitochondrial functional genes with increasing serum levels of malondialdehyde (MDA) and 8-hydroxydeoxyguanosine (8-OHdG) (P < 0.05). Regarding microbial response, HS boosts serum levels of lipopolysaccharide (LPS) (P < 0.05) and alters β-diversity (ANOSIM, P < 0.01), increasing the proportions of Escherichia-Shigella, Acinetobacter and Klebsiella (q < 0.05), while reducing that of Ruminiclostridium, Blautia, Lachnospiraceae_NK4A136_group, Clostridium VadinBB60 and Muribaculaceae (q < 0.05). PICRUSt analysis predicts that HS upregulates 11 KEGG pathways, mainly including bile secretion and bacterial invasion of epithelial cells. The collective results suggest that microbial dysbiosis due to late gestational HS has strong associations with damaged hepatic mitochondrial function and disrupted metabolic profiles.
Collapse
Affiliation(s)
- Jianwen He
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.,Affiliated Hospital of Shaanxi University of Chinese Medicine, Shaanxi University of Chinese Medicine, Xianyang, 712000, China
| | - Riliang Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Weijiang Zheng
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Huiduo Guo
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yunnan Yang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Ruqian Zhao
- Key Lab of Animal Physiology and Biochemistry, Nanjing Agricultural University, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Nanjing, 210095, China
| | - Wen Yao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.,Key Lab of Animal Physiology and Biochemistry, Nanjing Agricultural University, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Nanjing, 210095, China
| |
Collapse
|
105
|
Wu X, Xiong F, Fang H, Zhang J, Chang M. Crosstalks between NOD1 and Histone H2A Contribute to Host Defense against Streptococcus agalactiae Infection in Zebrafish. Antibiotics (Basel) 2021; 10:antibiotics10070861. [PMID: 34356784 PMCID: PMC8300774 DOI: 10.3390/antibiotics10070861] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/08/2021] [Accepted: 07/10/2021] [Indexed: 11/30/2022] Open
Abstract
Correlation studies about NOD1 and histones have not been reported. In the present study, we report the functional correlation between NOD1 and the histone H2A variant in response to Streptococcus agalactiae infection. In zebrafish, NOD1 deficiency significantly promoted S. agalactiae proliferation and decreased larval survival. Transcriptome analysis revealed that the significantly enriched pathways in NOD1−/− adult zebrafish were mainly involved in immune and metabolism. Among 719 immunity-associated DEGs at 48 hpi, 74 DEGs regulated by NOD1 deficiency were histone variants. Weighted gene co-expression network analysis identified that H2A, H2B, and H3 had significant associations with NOD1 deficiency. Above all, S. agalactiae infection could induce the expression of intracellular histone H2A, as well as NOD1 colocalized with histone H2A, both in the cytoplasm and cell nucleus in the case of S. agalactiae infection. The overexpression of H2A variants such as zfH2A-6 protected against S. agalactiae infection and could improve cell survival in NOD1-deficient cells. Furthermore, NOD1 could interact with zfH2A-6 and cooperate with zfH2A-6 to inhibit the proliferation of S. agalactiae. NOD1 also showed a synergetic effect in inducing the expression of many antibacterial genes, especially antibacterial pattern recognition receptors PGRP2, PGRP5, and PGRP6. Collectively, these results firstly highlight the roles of NOD1 deficiency in the regulation of immune-related and metabolic pathways, and the correlation between zebrafish NOD1 and histone H2A variant in the defense against S. agalactiae infection.
Collapse
Affiliation(s)
- Xiaoman Wu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Aquaculture Disease Control, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; (X.W.); (F.X.); (H.F.); (J.Z.)
| | - Fan Xiong
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Aquaculture Disease Control, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; (X.W.); (F.X.); (H.F.); (J.Z.)
| | - Hong Fang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Aquaculture Disease Control, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; (X.W.); (F.X.); (H.F.); (J.Z.)
| | - Jie Zhang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Aquaculture Disease Control, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; (X.W.); (F.X.); (H.F.); (J.Z.)
| | - Mingxian Chang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Aquaculture Disease Control, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; (X.W.); (F.X.); (H.F.); (J.Z.)
- Innovation Academy for Seed Design, Chinese Academy of Sciences, Wuhan 430072, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Correspondence:
| |
Collapse
|
106
|
Wu Y, Wang CZ, Wan JY, Yao H, Yuan CS. Dissecting the Interplay Mechanism between Epigenetics and Gut Microbiota: Health Maintenance and Disease Prevention. Int J Mol Sci 2021; 22:6933. [PMID: 34203243 PMCID: PMC8267743 DOI: 10.3390/ijms22136933] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/10/2021] [Accepted: 06/24/2021] [Indexed: 12/12/2022] Open
Abstract
The gut microbiota exists throughout the full life cycle of the human body, and it has been proven to have extensive impacts on health and disease. Accumulating evidence demonstrates that the interplay between gut microbiota and host epigenetics plays a multifaceted role in health maintenance and disease prevention. Intestinal microflora, along with their metabolites, could regulate multiple epigenetic pathways; e.g., DNA methylation, miRNA, or histone modification. Moreover, epigenetic factors can serve as mediators to coordinate gut microbiota within the host. Aiming to dissect this interplay mechanism, the present review summarizes the research profile of gut microbiota and epigenetics in detail, and further interprets the biofunctions of this interplay, especially the regulation of intestinal inflammation, the improvement of metabolic disturbances, and the inhibition of colitis events. This review provides new insights into the interplay of epigenetics and gut microbiota, and attempts to reveal the mysteries of health maintenance and disease prevention from this new perspective.
Collapse
Affiliation(s)
- Yuqi Wu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China;
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Chong-Zhi Wang
- Tang Center for Herbal Medicine Research, The University of Chicago, Chicago, IL 60637, USA; (C.-Z.W.); (C.-S.Y.)
- Department of Anesthesia and Critical Care, The University of Chicago, Chicago, IL 60637, USA
| | - Jin-Yi Wan
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China;
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Haiqiang Yao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China;
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Chun-Su Yuan
- Tang Center for Herbal Medicine Research, The University of Chicago, Chicago, IL 60637, USA; (C.-Z.W.); (C.-S.Y.)
- Department of Anesthesia and Critical Care, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
107
|
Mediterranean Diet and White Blood Cell Count-A Randomized Controlled Trial. Foods 2021; 10:foods10061268. [PMID: 34199545 PMCID: PMC8227102 DOI: 10.3390/foods10061268] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 05/31/2021] [Indexed: 12/26/2022] Open
Abstract
We aimed to assess the effects of the antioxidant-rich Mediterranean diet (MedDiet) on white blood cell count. Our study population included participants in the PREvención con DIeta MEDiterránea study (average age 67 years old, 58% women, high cardiovascular risk). We assessed whether a MedDiet intervention enriched in extra-virgin olive oil or nuts, versus a low-fat control diet, modified the incidence of leukocytosis (>11 × 109 leukocytes/L), mild leukopenia (<4.5 × 109 leukocytes/L), or severe leukopenia (<3.5 × 109 leukocytes/L) in individuals without the condition at baseline (n = 3190, n = 2925, and n = 3190, respectively). We also examined whether MedDiet modified the association between leukocyte count alterations and all-cause mortality. Both MedDiet interventions were associated with a lower risk of developing leukopenia (incidence rates: 5.06% in control diet, 3.29% in MedDiet groups combined; hazard ratio [95% confidence interval]: 0.54 [0.36-0.80]) and severe leukopenia (incidence rates: 1.26% in control diet, 0.46% in MedDiet groups combined; hazard ratio: 0.25 [0.10-0.60]). High cumulative adherence to a MedDiet was linked to lower risk of leukocytosis (incidence rates: 2.08% in quartile 1, 0.65% in quartile 4; HRQ4-Q1: 0.29 [0.085-0.99]) and attenuated the association between leukopenia and all-cause mortality (P-interaction = 0.032). In brief, MedDiet decreased the incidence of white blood cell count-related alterations in high cardiovascular risk individuals.
Collapse
|
108
|
Ribó-Coll M, Lassale C, Sacanella E, Ros E, Toledo E, Sorlí JV, Babio N, Lapetra J, Gómez-Gracia E, Alonso-Gómez ÁM, Fiol M, Serra-Majem L, Pinto X, Castañer O, Díez-Espino J, González JI, Becerra-Tomás N, Cofán M, Díaz-López A, Estruch R, Hernáez Á. Mediterranean diet and antihypertensive drug use: a randomized controlled trial. J Hypertens 2021; 39:1230-1237. [PMID: 33496530 DOI: 10.1097/hjh.0000000000002765] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To examine in older individuals at high cardiovascular risk whether following a Mediterranean diet decreased the necessity of antihypertensive drugs and modulated their associated cardiovascular risk. METHODS In the PREvención con DIeta MEDiterránea study, we assessed whether volunteers randomly allocated to an intervention with a Mediterranean diet enriched with extra-virgin olive oil or nuts (relative to a low-fat control diet) disclosed differences in the risk of: initiating antihypertensive medication in nonusers at baseline (n = 2188); and escalating therapy in participants using one, two, or three drugs at baseline (n = 2361, n = 1579, and n = 554, respectively). We also assessed whether allocation to Mediterranean diet modified the association between antihypertensive drug use and incident cardiovascular events. RESULTS Participants allocated to Mediterranean diet interventions were associated with lower risk of initiating antihypertensive therapy [5-year incidence rates: 47.1% in the control diet, 43.0% in MedDiets; hazard ratio = 0.84, 95% CI (0.74--0.97), in a model adjusted for age, sex, and recruitment site]. Volunteers using two drugs at baseline in the Mediterranean diet intervention enriched with extra-virgin olive oil decreased their risk of therapy escalation [5-year incidence rates: 22.9% in the control diet, 20.1% in the MedDiet; hazard ratio = 0.77, 95% CI (0.60--0.99)]. Allocation to Mediterranean diet interventions attenuated the association between antihypertensive therapy at baseline and incidence of major adverse cardiovascular events (P interaction = 0.003). CONCLUSION In an older population at high cardiovascular risk, following a Mediterranean diet reduced the risk of initiating or escalating antihypertensive medication and attenuated cardiovascular risk in antihypertensive drug users.
Collapse
Affiliation(s)
- Margarita Ribó-Coll
- Cardiovascular Risk, Nutrition, and Aging Research Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS)
- PhD program in Food Science and Nutrition, Faculty of Pharmacy and Food Science, Universitat de Barcelona, Barcelona
| | - Camille Lassale
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid
- Cardiovascular Risk and Nutrition Research Group, Hospital del Mar Medical Research Institute (IMIM)
| | - Emilio Sacanella
- Cardiovascular Risk, Nutrition, and Aging Research Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS)
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid
- Internal Medicine Service, Hospital Clínic
| | - Emilio Ros
- Cardiovascular Risk, Nutrition, and Aging Research Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS)
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid
- Lipid Clinic, Endocrinology and Nutrition Service, Hospital Clínic, Barcelona
| | - Estefanía Toledo
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid
- Department of Preventive Medicine and Public Health, Universidad de Navarra, Pamplona
| | - José V Sorlí
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid
- Department of Preventive Medicine and Public Health, Universidad de Valencia, Valencia
| | - Nancy Babio
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid
- Unitat de Nutrició Humana, Departament de Bioquimica i Biotecnologia, Universitat Rovira i Virgili
- Institut d'Investigació Pere Virgili (IISPV), Hospital Universitari Sant Joan de Reus, Reus
| | - José Lapetra
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid
- Department of Family Medicine, Research Unit, Distrito Sanitario Atención Primaria Sevilla, Sevilla
| | - Enrique Gómez-Gracia
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid
- Department of Preventive Medicine and Public Health, Universidad de Málaga, Málaga
| | - Ángel M Alonso-Gómez
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid
- Bioaraba Health Research Institute, Osakidetza Basque Health Service, Araba University Hospital, University of the Basque Country UPV/EHU, Vitoria-Gasteiz
| | - Miquel Fiol
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid
- Health Research Institute of the Balearic Islands (IdISBa). Hospital Son Espases. Palma de Mallorca
| | - Lluis Serra-Majem
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid
- Instituto de Investigaciones Biomédicas y Sanitarias, Universidad de Las Palmas de Gran Canaria
- Centro Hospitalario Universitario Insular Materno Infantil, Servicio Canario de Salud, Las Palmas
| | - Xavier Pinto
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid
- Lipids and Vascular Risk Unit, Internal Medicine, Hospital Universitario de Bellvitge, L'Hospitalet de Llobregat
| | - Olga Castañer
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid
- Cardiovascular Risk and Nutrition Research Group, Hospital del Mar Medical Research Institute (IMIM)
| | - Javier Díez-Espino
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid
- Department of Preventive Medicine and Public Health, Universidad de Navarra, Pamplona
- Centro de Salud de Tafalla, Gerencia de Atención Primaria, Servicio Navarro de Salud-Osasunbidea, Tafalla
- Instituto de Investigación Sanitaria de Navarra (IDISNA), Pamplona
| | - José I González
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid
- Department of Preventive Medicine and Public Health, Universidad de Valencia, Valencia
| | - Nerea Becerra-Tomás
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid
- Department of Preventive Medicine and Public Health, Universidad de Valencia, Valencia
- Unitat de Nutrició Humana, Departament de Bioquimica i Biotecnologia, Universitat Rovira i Virgili
- Institut d'Investigació Pere Virgili (IISPV), Hospital Universitari Sant Joan de Reus, Reus
| | - Montserrat Cofán
- Cardiovascular Risk, Nutrition, and Aging Research Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS)
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid
- Lipid Clinic, Endocrinology and Nutrition Service, Hospital Clínic, Barcelona
| | - Andrés Díaz-López
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid
- Unitat de Nutrició Humana, Departament de Bioquimica i Biotecnologia, Universitat Rovira i Virgili
- Institut d'Investigació Pere Virgili (IISPV), Hospital Universitari Sant Joan de Reus, Reus
| | - Ramón Estruch
- Cardiovascular Risk, Nutrition, and Aging Research Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS)
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid
- Internal Medicine Service, Hospital Clínic
| | - Álvaro Hernáez
- Cardiovascular Risk, Nutrition, and Aging Research Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS)
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid
- Blanquerna School of Health Sciences, Universitat Ramon Llull, Barcelona, Spain
| |
Collapse
|
109
|
Abstract
To summarize the literature on the influence of exercise on the gut microbiota of healthy adults.
Collapse
|
110
|
Nurrahma BA, Tsao SP, Wu CH, Yeh TH, Hsieh PS, Panunggal B, Huang HY. Probiotic Supplementation Facilitates Recovery of 6-OHDA-Induced Motor Deficit via Improving Mitochondrial Function and Energy Metabolism. Front Aging Neurosci 2021; 13:668775. [PMID: 34025392 PMCID: PMC8137830 DOI: 10.3389/fnagi.2021.668775] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 04/12/2021] [Indexed: 12/25/2022] Open
Abstract
Parkinson’s disease (PD) is a neurodegenerative disease associated with progressive impairment of motor and non-motor functions in aging people. Overwhelming evidence indicate that mitochondrial dysfunction is a central factor in PD pathophysiology, which impairs energy metabolism. While, several other studies have shown probiotic supplementations to improve host energy metabolism, alleviate the disease progression, prevent gut microbiota dysbiosis and alter commensal bacterial metabolites. But, whether probiotic and/or prebiotic supplementation can affect energy metabolism and cause the impediment of PD progression remains poorly characterized. Therefore, we investigated 8-weeks supplementation effects of probiotic [Lactobacillus salivarius subsp. salicinius AP-32 (AP-32)], residual medium (RM) obtained from the AP-32 culture medium, and combination of AP-32 and RM (A-RM) on unilateral 6-hydroxydopamine (6-OHDA)-induced PD rats. We found that AP-32, RM and A-RM supplementation induced neuroprotective effects on dopaminergic neurons along with improved motor functions in PD rats. These effects were accompanied by significant increases in mitochondrial activities in the brain and muscle, antioxidative enzymes level in serum, and altered SCFAs profile in fecal samples. Importantly, the AP-32 supplement restored muscle mass along with improved motor function in PD rats, and produced the best results among the supplements. Our results demonstrate that probiotic AP-32 and A-RM supplementations can recover energy metabolism via increasing SCFAs producing and mitochondria function. This restoring of mitochondrial function in the brain and muscles with improved energy metabolism might additionally be potentiated by ROS suppression by the elevated generation of antioxidants, and which finally leads to facilitated recovery of 6-OHDA-induced motor deficit. Taken together, this work demonstrates that probiotic AP-32 supplementation could be a potential candidate for alternate treatment strategy to avert PD progression.
Collapse
Affiliation(s)
- Bira Arumndari Nurrahma
- Graduate Institute of Metabolism and Obesity Sciences, College of Nutrition, Taipei Medical University, Taipei City, Taiwan
| | - Shu-Ping Tsao
- Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei City, Taiwan
| | - Chieh-Hsi Wu
- Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei City, Taiwan.,School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei City, Taiwan
| | - Tu-Hsueh Yeh
- Department of Neurology, Taipei Medical University Hospital, Taipei City, Taiwan.,Department of Neurology, College of Medicine and Taipei Neuroscience Institute, Taipei Medical University, Taipei City, Taiwan
| | | | - Binar Panunggal
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei City, Taiwan.,Department of Nutrition Science, Faculty of Medicine, Diponegoro University, Central Java, Indonesia
| | - Hui-Yu Huang
- Graduate Institute of Metabolism and Obesity Sciences, College of Nutrition, Taipei Medical University, Taipei City, Taiwan
| |
Collapse
|
111
|
Jaago M, Timmusk US, Timmusk T, Palm K. Drastic Effects on the Microbiome of a Young Rower Engaged in High-Endurance Exercise After a Month Usage of a Dietary Fiber Supplement. Front Nutr 2021; 8:654008. [PMID: 33996876 PMCID: PMC8119783 DOI: 10.3389/fnut.2021.654008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 03/22/2021] [Indexed: 01/12/2023] Open
Abstract
Food supplements are increasingly used worldwide. However, research on the efficacy of such supplements on athlete's well-being and optimal sports performance is very limited. This study performed in junior academic rowing explores the effects of nutritional supplements to aid to the high energy requirements at periods of intense exercise. Herein, the effects of prebiotic fibers on the intestinal microbiome composition of an 18-year-old athlete exercising at high loads during an 8-month period in a “real-life” setting were examined using next-generation sequencing analysis. Results demonstrated that although the alpha diversity of the subject's microbiome drastically decreased [from 2.11 precompetition to 1.67 (p < 0.05)] upon fiber consumption, the Firmicutes/Bacteroidetes ratio increased significantly [from 3.11 to 4.55, as compared with population average (p < 0.05)]. Underlying these macrolevel microbial alterations were demonstrable shifts from acetate- to butyrate-producing bacteria, although with stable effects on the Veillonella species. To our knowledge, this a unique study that shows pronounced changes in the gut microbiome of the young athlete at the competition season and their favorable compensation by the dietary fiber intake. The data here expand the overall understanding of how the high energy needs in high-intensity sports like academic rowing could be supported by dietary fiber supplement consumption.
Collapse
Affiliation(s)
- Mariliis Jaago
- Protobios Llc, Tallinn, Estonia.,Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | | | - Tõnis Timmusk
- Protobios Llc, Tallinn, Estonia.,Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | - Kaia Palm
- Protobios Llc, Tallinn, Estonia.,Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| |
Collapse
|
112
|
Mitochondrial Dysfunction Contributes to Aging-Related Atrial Fibrillation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5530293. [PMID: 34007402 PMCID: PMC8102104 DOI: 10.1155/2021/5530293] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 04/04/2021] [Accepted: 04/15/2021] [Indexed: 11/17/2022]
Abstract
The incidence of atrial fibrillation (AF) increases with age, and telomere length gradually shortens with age. However, whether telomere length is related to AF is still inconclusive, and the exact mechanism by which aging causes the increased incidence of AF is still unclear. We hypothesize that telomere length is correlated with aging-related AF and that mitochondrial dysfunction plays a role in this. This research recruited 96 elderly male patients with AF who were admitted to the Second Medical Center of Chinese PLA General Hospital from April to October 2018. After matching by age and gender, 96 non-AF elderly male patients who were admitted to the hospital for physical examination during the same period were selected as controls. Anthropometric, clinical, and laboratory analyses were performed on all subjects. The mitochondrial membrane potential (MMP) of peripheral blood leukocytes was detected as the indicator of mitochondrial function. Compared with the control group, the leukocyte telomere length (LTL) was significantly shorter (P < 0.001), and the level of PGC-1α in serum was significantly lower in AF patients. Additionally, in subjects without any other diseases, the AF patients had lower MMP when compared with the control. Multivariate logistic regression confirmed that LTL (OR 0.365; 95% CI 0.235-0.568; P < 0.001) and serum PGC-1α (OR 0.993; 95% CI 0.988-0.997; P = 0.002) were inversely associated with the presence of AF. In addition, ROC analysis indicated the potential diagnostic value of LTL and serum PGC-1α with AUC values of 0.734 and 0.633, respectively. This research concludes that LTL and serum PGC-1α are inversely correlated with the occurrence of aging-related AF and that mitochondrial dysfunction plays a role in this.
Collapse
|
113
|
Wang S, Wu K, Xue D, Zhang C, Rajput SA, Qi D. Mechanism of deoxynivalenol mediated gastrointestinal toxicity: Insights from mitochondrial dysfunction. Food Chem Toxicol 2021; 153:112214. [PMID: 33930483 DOI: 10.1016/j.fct.2021.112214] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 03/21/2021] [Accepted: 04/10/2021] [Indexed: 12/12/2022]
Abstract
Deoxynivalenol (DON) is a mycotoxin predominantly produced by Fusarium genus, and widely contaminates cereals and associated products all over the world. The intestinal toxicity of DON is well established. However, intestinal homeostasis involves mitochondria, which has rarely been considered in the context of DON exposure. We summarize the recent knowledge on mitochondria as a key player in maintaining intestinal homeostasis based on their functions in cellular energy metabolism, redox homeostasis, apoptosis, intestinal immune responses, and orchestrated bidirectional cross-talk with gut microbe. In addition, we discuss the pivotal roles of mitochondrial dysfunction in the intestinal toxicity of DON and highlight promising mitochondrial-targeted therapeutics for DON-induced intestinal injury. Recent studies support that the intestinal toxicity of DON is attributed to mitochondrial dysfunction as a critical factor. Mitochondrial dysfunction characterized by failure in respiratory capacities and ROS overproduction has been demonstrated in intestinal cells exposed to DON. Perturbation of mitochondrial respiration leading to ROS accumulation is implicated in the early initiation of apoptosis. DON-induced intestinal inflammatory response is tightly linked to the mitochondrial ROS, whereas immunosuppression is intimately associated with mitophagy inhibition. DON perturbs the orchestrated bidirectional cross-talk between gut microbe and host mitochondria, which may be involved in DON-induced intestinal toxicity.
Collapse
Affiliation(s)
- Shuai Wang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China.
| | - Kuntan Wu
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China.
| | - Dongfang Xue
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China.
| | - Cong Zhang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China.
| | - Shahid Ali Rajput
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China.
| | - Desheng Qi
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China.
| |
Collapse
|
114
|
Papageorgiou M, Biver E. Interactions of the microbiome with pharmacological and non-pharmacological approaches for the management of ageing-related musculoskeletal diseases. Ther Adv Musculoskelet Dis 2021; 13:1759720X211009018. [PMID: 34104230 PMCID: PMC8172340 DOI: 10.1177/1759720x211009018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/18/2021] [Indexed: 12/18/2022] Open
Abstract
Despite major progress in the understanding of the pathophysiology and therapeutic options for common ageing-related musculoskeletal conditions (i.e. osteoporosis and associated fractures, sarcopenia and osteoarthritis), there is still a considerable proportion of patients who respond sub optimally to available treatments or experience adverse effects. Emerging microbiome research suggests that perturbations in microbial composition, functional and metabolic capacity (i.e. dysbiosis) are associated with intestinal and extra-intestinal disorders including musculoskeletal diseases. Besides its contributions to disease pathogenesis, the role of the microbiome is further extended to shaping individuals' responses to disease therapeutics (i.e. pharmacomicrobiomics). In this review, we focus on the reciprocal interactions between the microbiome and therapeutics for osteoporosis, sarcopenia and osteoarthritis. Specifically, we identify the effects of therapeutics on microbiome's configurations, functions and metabolic output, intestinal integrity and immune function, but also the effects of the microbiome on the metabolism of these therapeutics, which in turn, may influence their bioavailability, efficacy and side-effect profile contributing to variable treatment responses in clinical practice. We further discuss emerging strategies for microbiota manipulation as preventive or therapeutic (alone or complementary to available treatments) approaches for improving outcomes of musculoskeletal health and disease.
Collapse
Affiliation(s)
- Maria Papageorgiou
- Division of Bone Diseases, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Emmanuel Biver
- Division of Bone Diseases, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Rue Gabrielle Perret Gentil 4, Geneva 1205, Switzerland
| |
Collapse
|
115
|
Mach N, Moroldo M, Rau A, Lecardonnel J, Le Moyec L, Robert C, Barrey E. Understanding the Holobiont: Crosstalk Between Gut Microbiota and Mitochondria During Long Exercise in Horse. Front Mol Biosci 2021; 8:656204. [PMID: 33898524 PMCID: PMC8063112 DOI: 10.3389/fmolb.2021.656204] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 02/16/2021] [Indexed: 12/17/2022] Open
Abstract
Endurance exercise has a dramatic impact on the functionality of mitochondria and on the composition of the intestinal microbiome, but the mechanisms regulating the crosstalk between these two components are still largely unknown. Here, we sampled 20 elite horses before and after an endurance race and used blood transcriptome, blood metabolome and fecal microbiome to describe the gut-mitochondria crosstalk. A subset of mitochondria-related differentially expressed genes involved in pathways such as energy metabolism, oxidative stress and inflammation was discovered and then shown to be associated with butyrate-producing bacteria of the Lachnospiraceae family, especially Eubacterium. The mechanisms involved were not fully understood, but through the action of their metabolites likely acted on PPARγ, the FRX-CREB axis and their downstream targets to delay the onset of hypoglycemia, inflammation and extend running time. Our results also suggested that circulating free fatty acids may act not merely as fuel but drive mitochondrial inflammatory responses triggered by the translocation of gut bacterial polysaccharides following endurance. Targeting the gut-mitochondria axis therefore appears to be a potential strategy to enhance athletic performance.
Collapse
Affiliation(s)
- Núria Mach
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
| | - Marco Moroldo
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
| | - Andrea Rau
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France.,BioEcoAgro Joint Research Unit, INRAE, Université de Liège, Université de Lille, Université de Picardie Jules Verne, Estrées-Mons, France
| | - Jérôme Lecardonnel
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
| | - Laurence Le Moyec
- Université d'Évry Val d'Essonne, Université Paris-Saclay, Évry, France ABI UMR 1313, INRAE, Université Paris-Saclay, AgroParisTech, Jouy-en-Josas, France.,MCAM UMR7245, CNRS, Muséum National d'Histoire Naturelle, Paris, France
| | - Céline Robert
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France.,École Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| | - Eric Barrey
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
| |
Collapse
|
116
|
Tippairote T, Bjørklund G, Yaovapak A. The continuum of disrupted metabolic tempo, mitochondrial substrate congestion, and metabolic gridlock toward the development of non-communicable diseases. Crit Rev Food Sci Nutr 2021; 62:6837-6853. [PMID: 33797995 DOI: 10.1080/10408398.2021.1907299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Non-communicable diseases (NCD) are the slow-motion disasters with imminent global health care burden. The current dietary management for NCD is dominated by the calorie balance model. Apart from the quantitative balance of calorie, healthy bioenergetics requires temporal eating and fasting rhythms, and the subsequent switching for different metabolic fuels. We herein term these three bioenergetic attributes, i.e., caloric balance, diurnal eating-fasting rhythm, and metabolic flexibility, as the metabolic tempo. These three attributes are intertwined with each other; alteration of one attribute affects one or more other attributes. Lifestyle-induced disrupted metabolic tempo presents a high flux of mixed carbon substrates to mitochondria, with the resulting congestion and indecisiveness of metabolic switches. Such indecisiveness impairs metabolic flexibility, promotes anabolism, and accumulates the energy storage pools. The triggers from hypoxic inducible factor expression could further promote the metabolic gridlock and adipocyte maladaptation. The maladaptive adipocytes lead to ectopic fat deposition, increased circulating lipid levels, insulin resistance, and chronic systemic inflammation. These continuum set stages for clinical NCDs. We propose that the restoration of all tempo attributes through the combined diet-, time-, and calorie-restricted interventions could be the preferred strategy for NCD management.
Collapse
Affiliation(s)
- Torsak Tippairote
- Nutritional and Environmental Section, Thailand Initiatives for Functional Medicine, Bangkok Thailand.,Nutritional and Environmental Medicine, Healing Passion Medical Center, Bangkok Thailand
| | - Geir Bjørklund
- Nutritional and Environmental Medicine, Council for Nutritional and Environmental Medicine, Mo i Rana, Norway
| | - Augchara Yaovapak
- Nutritional and Environmental Section, Thailand Initiatives for Functional Medicine, Bangkok Thailand.,Nutritional and Environmental Medicine, Healing Passion Medical Center, Bangkok Thailand
| |
Collapse
|
117
|
Barber TM, Valsamakis G, Mastorakos G, Hanson P, Kyrou I, Randeva HS, Weickert MO. Dietary Influences on the Microbiota-Gut-Brain Axis. Int J Mol Sci 2021; 22:ijms22073502. [PMID: 33800707 PMCID: PMC8038019 DOI: 10.3390/ijms22073502] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 02/07/2023] Open
Abstract
Over unimaginable expanses of evolutionary time, our gut microbiota have co-evolved with us, creating a symbiotic relationship in which each is utterly dependent upon the other. Far from confined to the recesses of the alimentary tract, our gut microbiota engage in complex and bi-directional communication with their host, which have far-reaching implications for overall health, wellbeing and normal physiological functioning. Amongst such communication streams, the microbiota–gut–brain axis predominates. Numerous complex mechanisms involve direct effects of the microbiota, or indirect effects through the release and absorption of the metabolic by-products of the gut microbiota. Proposed mechanisms implicate mitochondrial function, the hypothalamus–pituitary–adrenal axis, and autonomic, neuro-humeral, entero-endocrine and immunomodulatory pathways. Furthermore, dietary composition influences the relative abundance of gut microbiota species. Recent human-based data reveal that dietary effects on the gut microbiota can occur rapidly, and that our gut microbiota reflect our diet at any given time, although much inter-individual variation pertains. Although most studies on the effects of dietary macronutrients on the gut microbiota report on associations with relative changes in the abundance of particular species of bacteria, in broad terms, our modern-day animal-based Westernized diets are relatively high in fats and proteins and impoverished in fibres. This creates a perfect storm within the gut in which dysbiosis promotes localized inflammation, enhanced gut wall permeability, increased production of lipopolysaccharides, chronic endotoxemia and a resultant low-grade systemic inflammatory milieu, a harbinger of metabolic dysfunction and many modern-day chronic illnesses. Research should further focus on the colony effects of the gut microbiota on health and wellbeing, and dysbiotic effects on pathogenic pathways. Finally, we should revise our view of the gut microbiota from that of a seething mass of microbes to one of organ-status, on which our health and wellbeing utterly depends. Future guidelines on lifestyle strategies for wellbeing should integrate advice on the optimal establishment and maintenance of a healthy gut microbiota through dietary and other means. Although we are what we eat, perhaps more importantly, we are what our gut microbiota thrive on and they thrive on what we eat.
Collapse
Affiliation(s)
- Thomas M. Barber
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism, University Hospitals Coventry and Warwickshire NHS Trust, Clifford Bridge Road, Coventry CV2 2DX, UK; (T.M.B.); (G.V.); (P.H.); (I.K.); (H.S.R.)
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV2 2DX, UK
| | - Georgios Valsamakis
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism, University Hospitals Coventry and Warwickshire NHS Trust, Clifford Bridge Road, Coventry CV2 2DX, UK; (T.M.B.); (G.V.); (P.H.); (I.K.); (H.S.R.)
- Endocrine Unit, 2nd Department of Obstetrics and Gynaecology and Pathology Department, Aretaieion University Hospital, Athens Medical School, 11528 Athens, Greece;
| | - George Mastorakos
- Endocrine Unit, 2nd Department of Obstetrics and Gynaecology and Pathology Department, Aretaieion University Hospital, Athens Medical School, 11528 Athens, Greece;
| | - Petra Hanson
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism, University Hospitals Coventry and Warwickshire NHS Trust, Clifford Bridge Road, Coventry CV2 2DX, UK; (T.M.B.); (G.V.); (P.H.); (I.K.); (H.S.R.)
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV2 2DX, UK
| | - Ioannis Kyrou
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism, University Hospitals Coventry and Warwickshire NHS Trust, Clifford Bridge Road, Coventry CV2 2DX, UK; (T.M.B.); (G.V.); (P.H.); (I.K.); (H.S.R.)
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV2 2DX, UK
- Aston Medical Research Institute, Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK
| | - Harpal S. Randeva
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism, University Hospitals Coventry and Warwickshire NHS Trust, Clifford Bridge Road, Coventry CV2 2DX, UK; (T.M.B.); (G.V.); (P.H.); (I.K.); (H.S.R.)
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV2 2DX, UK
- Aston Medical Research Institute, Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK
| | - Martin O. Weickert
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism, University Hospitals Coventry and Warwickshire NHS Trust, Clifford Bridge Road, Coventry CV2 2DX, UK; (T.M.B.); (G.V.); (P.H.); (I.K.); (H.S.R.)
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV2 2DX, UK
- Centre for Sport, Exercise and Life Sciences, Faculty of Health & Life Sciences, Coventry University, Coventry CV1 5FB, UK
- Correspondence:
| |
Collapse
|
118
|
Aya V, Flórez A, Perez L, Ramírez JD. Association between physical activity and changes in intestinal microbiota composition: A systematic review. PLoS One 2021; 16:e0247039. [PMID: 33630874 PMCID: PMC7906424 DOI: 10.1371/journal.pone.0247039] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 01/31/2021] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION The intestinal microbiota comprises bacteria, fungi, archaea, protists, helminths and viruses that symbiotically inhabit the digestive system. To date, research has provided limited data on the possible association between an active lifestyle and a healthy composition of human microbiota. This review was aimed to summarize the results of human studies comparing the microbiome of healthy individuals with different physical activity amounts. METHODS We searched Medline/Ovid, NIH/PubMed, and Academic Search Complete between August-October 2020. Inclusion criteria comprised: (a) cross-sectional studies focused on comparing gut microbiome among subjects with different physical activity levels; (b) studies describing human gut microbiome responses to any type of exercise stimulus; (c) studies containing healthy adult women and men. We excluded studies containing diet modifications, probiotic or prebiotic consumption, as well as studies focused on diabetes, hypertension, cancer, hormonal dysfunction. Methodological quality and risk of bias for each study were assessed using the Risk Of Bias In Non-randomized Studies-of Interventions tool. The results from cross-sectional and longitudinal studies are shown independently. RESULTS A total of 17 articles were eligible for inclusion: ten cross-sectional and seven longitudinal studies. Main outcomes vary significantly according to physical activity amounts in longitudinal studies. We identified discrete changes in diversity indexes and relative abundance of certain bacteria in active people. CONCLUSION As literature in this field is rapidly growing, it is important that studies incorporate diverse methods to evaluate other aspects related to active lifestyles such as sleep and dietary patterns. Exploration of other groups such as viruses, archaea and parasites may lead to a better understanding of gut microbiota adaptation to physical activity and sports and its potentially beneficial effects on host metabolism and endurance.
Collapse
Affiliation(s)
- Viviana Aya
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Alberto Flórez
- Grupo In-Novum Educatio, Facultad de Educación, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Luis Perez
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Juan David Ramírez
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| |
Collapse
|
119
|
The Effect of Kefir Supplementation on Improving Human Endurance Exercise Performance and Antifatigue. Metabolites 2021; 11:metabo11030136. [PMID: 33669119 PMCID: PMC7996501 DOI: 10.3390/metabo11030136] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 02/21/2021] [Indexed: 02/06/2023] Open
Abstract
Kefir is an acidic, carbonated, and fermented dairy product produced by fermenting milk with kefir grains. The Lactobacillus species constitutes an important part of kefir grains. In a previous animal study, kefir effectively improved exercise performance and had anti-fatigue effects. The purpose of this research was to explore the benefits of applying kefir to improve exercise performance, reduce fatigue, and improve physiological adaptability in humans. The test used a double-blind crossover design and supplementation for 28 days. Sixteen 20–30 year-old subjects were divided into two groups in a balanced order according to each individual’s initial maximal oxygen uptake and were assigned to receive a placebo (equal flavor, equal calories, 20 g/day) or SYNKEFIR™ (20 g/day) every morning. After the intervention, there were 28 days of wash-out, during which time the subjects did not receive further interventions. After supplementation with SYNKEFIR™, the exercise time to exhaustion was significantly greater than that before ingestion (p = 0.0001) and higher than that in the Placebo group by 1.29-fold (p = 0.0004). In addition, compared with the Placebo group, the SYNKEFIR™ administration group had significantly lower lactate levels in the exercise and recovery (p < 0.05). However, no significant difference was observed in the changes in the gut microbiota. Although no significant changes in body composition were found, SYNKEFIR™ did not cause adverse reactions or harm to the participants’ bodies. In summary, 28 days of supplementation with SYNKEFIR™ significantly improved exercise performance, reduced the production of lactic acid after exercise, and accelerated recovery while also not causing any adverse reactions.
Collapse
|
120
|
Watson MD, Cross BL, Grosicki GJ. Evidence for the Contribution of Gut Microbiota to Age-Related Anabolic Resistance. Nutrients 2021; 13:706. [PMID: 33672207 PMCID: PMC7926629 DOI: 10.3390/nu13020706] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/08/2021] [Accepted: 02/19/2021] [Indexed: 12/11/2022] Open
Abstract
Globally, people 65 years of age and older are the fastest growing segment of the population. Physiological manifestations of the aging process include undesirable changes in body composition, declines in cardiorespiratory fitness, and reductions in skeletal muscle size and function (i.e., sarcopenia) that are independently associated with mortality. Decrements in muscle protein synthetic responses to anabolic stimuli (i.e., anabolic resistance), such as protein feeding or physical activity, are highly characteristic of the aging skeletal muscle phenotype and play a fundamental role in the development of sarcopenia. A more definitive understanding of the mechanisms underlying this age-associated reduction in anabolic responsiveness will help to guide promyogenic and function promoting therapies. Recent studies have provided evidence in support of a bidirectional gut-muscle axis with implications for aging muscle health. This review will examine how age-related changes in gut microbiota composition may impact anabolic response to protein feeding through adverse changes in protein digestion and amino acid absorption, circulating amino acid availability, anabolic hormone production and responsiveness, and intramuscular anabolic signaling. We conclude by reviewing literature describing lifestyle habits suspected to contribute to age-related changes in the microbiome with the goal of identifying evidence-informed strategies to preserve microbial homeostasis, anabolic sensitivity, and skeletal muscle with advancing age.
Collapse
Affiliation(s)
| | | | - Gregory J. Grosicki
- Biodynamics and Human Performance Center, Georgia Southern University (Armstrong Campus), Savannah, GA 31419, USA; (M.D.W.); (B.L.C.)
| |
Collapse
|
121
|
Vona R, Pallotta L, Cappelletti M, Severi C, Matarrese P. The Impact of Oxidative Stress in Human Pathology: Focus on Gastrointestinal Disorders. Antioxidants (Basel) 2021; 10:201. [PMID: 33573222 PMCID: PMC7910878 DOI: 10.3390/antiox10020201] [Citation(s) in RCA: 125] [Impact Index Per Article: 41.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/22/2021] [Accepted: 01/26/2021] [Indexed: 12/15/2022] Open
Abstract
Accumulating evidence shows that oxidative stress plays an essential role in the pathogenesis and progression of many diseases. The imbalance between the production of reactive oxygen species (ROS) and the antioxidant systems has been extensively studied in pulmonary, neurodegenerative cardiovascular disorders; however, its contribution is still debated in gastrointestinal disorders. Evidence suggests that oxidative stress affects gastrointestinal motility in obesity, and post-infectious disorders by favoring the smooth muscle phenotypic switch toward a synthetic phenotype. The aim of this review is to gain insight into the role played by oxidative stress in gastrointestinal pathologies (GIT), and the involvement of ROS in the signaling underlying the muscular alterations of the gastrointestinal tract (GIT). In addition, potential therapeutic strategies based on the use of antioxidants for the treatment of inflammatory gastrointestinal diseases are reviewed and discussed. Although substantial progress has been made in identifying new techniques capable of assessing the presence of oxidative stress in humans, the biochemical-molecular mechanisms underlying GIT mucosal disorders are not yet well defined. Therefore, further studies are needed to clarify the mechanisms through which oxidative stress-related signaling can contribute to the alteration of the GIT mucosa in order to devise effective preventive and curative therapeutic strategies.
Collapse
Affiliation(s)
- Rosa Vona
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy;
| | - Lucia Pallotta
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale del Policlinico, 155, 00161 Rome, Italy; (L.P.); (M.C.); (C.S.)
| | - Martina Cappelletti
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale del Policlinico, 155, 00161 Rome, Italy; (L.P.); (M.C.); (C.S.)
| | - Carola Severi
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale del Policlinico, 155, 00161 Rome, Italy; (L.P.); (M.C.); (C.S.)
| | - Paola Matarrese
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy;
| |
Collapse
|
122
|
Yap YA, Mariño E. Dietary SCFAs Immunotherapy: Reshaping the Gut Microbiota in Diabetes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1307:499-519. [PMID: 32193865 DOI: 10.1007/5584_2020_515] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Diet-microbiota related inflammatory conditions such as obesity, autoimmune type 1 diabetes (T1D), type 2 diabetes (T2D), cardiovascular disease (CVD) and gut infections have become a stigma in Western societies and developing nations. This book chapter examines the most relevant pre-clinical and clinical studies about diet-gut microbiota approaches as an alternative therapy for diabetes. We also discuss what we and others have extensively investigated- the power of dietary short-chain fatty acids (SCFAs) technology that naturally targets the gut microbiota as an alternative method to prevent and treat diabetes and its related complications.
Collapse
Affiliation(s)
- Yu Anne Yap
- Infection and Immunity Program, Biomedicine Discovery Institute, Department of Biochemistry, Monash University, Melbourne, VIC, Australia
| | - Eliana Mariño
- Infection and Immunity Program, Biomedicine Discovery Institute, Department of Biochemistry, Monash University, Melbourne, VIC, Australia.
| |
Collapse
|
123
|
Bórquez JC, Hidalgo M, Rodríguez JM, Montaña A, Porras O, Troncoso R, Bravo-Sagua R. Sucralose Stimulates Mitochondrial Bioenergetics in Caco-2 Cells. Front Nutr 2021; 7:585484. [PMID: 33537337 PMCID: PMC7848014 DOI: 10.3389/fnut.2020.585484] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 12/18/2020] [Indexed: 12/20/2022] Open
Abstract
Sucralose is a non-caloric artificial sweetener widely used in processed foods that reportedly affects energy homeostasis through partially understood mechanisms. Mitochondria are organelles fundamental for cellular bioenergetics that are closely related to the development of metabolic diseases. Here, we addressed whether sucralose alters mitochondrial bioenergetics in the enterocyte cell line Caco-2. Sucralose exposure (0.5-50 mM for 3-24 h) increased cellular reductive power assessed through MTT assay, suggesting enhanced bioenergetics. Low doses of sucralose (0.5 and 5 mM) for 3 h stimulated mitochondrial respiration, measured through oxygraphy, and elevated mitochondrial transmembrane potential and cytoplasmic Ca2+, evaluated by fluorescence microscopy. Contrary to other cell types, the increase in mitochondrial respiration was insensitive to inhibition of mitochondrial Ca2+ uptake. These findings suggest that sucralose alters enterocyte energy homeostasis, contributing to its effects on organismal metabolism.
Collapse
Affiliation(s)
- Juan Carlos Bórquez
- Laboratorio de Investigación en Nutrición y Actividad Física, Instituto de Nutrición y Tecnología de los Alimentos, Universidad de Chile, Santiago, Chile
| | - Miltha Hidalgo
- Laboratory for Research in Functional Nutrition, INTA, Universidad de Chile, Santiago, Chile
| | - Juan M. Rodríguez
- Laboratorio de Investigación en Nutrición y Actividad Física, Instituto de Nutrición y Tecnología de los Alimentos, Universidad de Chile, Santiago, Chile
| | - Alejandra Montaña
- Laboratorio de Investigación en Nutrición y Actividad Física, Instituto de Nutrición y Tecnología de los Alimentos, Universidad de Chile, Santiago, Chile
| | - Omar Porras
- Laboratory for Research in Functional Nutrition, INTA, Universidad de Chile, Santiago, Chile
| | - Rodrigo Troncoso
- Laboratorio de Investigación en Nutrición y Actividad Física, Instituto de Nutrición y Tecnología de los Alimentos, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases, Universidad de Chile, Santiago, Chile
| | - Roberto Bravo-Sagua
- Advanced Center for Chronic Diseases, Universidad de Chile, Santiago, Chile
- Laboratory of Obesity and Metabolism in Geriatrics and Adults, INTA, Universidad de Chile, Santiago, Chile
- Chile State Universities Network on Aging, Universidad de Chile, Santiago, Chile
| |
Collapse
|
124
|
Current Evidence on the Role of the Gut Microbiome in ADHD Pathophysiology and Therapeutic Implications. Nutrients 2021; 13:nu13010249. [PMID: 33467150 PMCID: PMC7830868 DOI: 10.3390/nu13010249] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/12/2021] [Accepted: 01/14/2021] [Indexed: 12/14/2022] Open
Abstract
Studies suggest that the bidirectional relationship existent between the gut microbiome (GM) and the central nervous system (CNS), or so-called the microbiome–gut–brain axis (MGBA), is involved in diverse neuropsychiatric diseases in children and adults. In pediatric age, most studies have focused on patients with autism. However, evidence of the role played by the MGBA in attention deficit/hyperactivity disorder (ADHD), the most common neurodevelopmental disorder in childhood, is still scanty and heterogeneous. This review aims to provide the current evidence on the functioning of the MGBA in pediatric patients with ADHD and the specific role of omega-3 polyunsaturated fatty acids (ω-3 PUFAs) in this interaction, as well as the potential of the GM as a therapeutic target for ADHD. We will explore: (1) the diverse communication pathways between the GM and the CNS; (2) changes in the GM composition in children and adolescents with ADHD and association with ADHD pathophysiology; (3) influence of the GM on the ω-3 PUFA imbalance characteristically found in ADHD; (4) interaction between the GM and circadian rhythm regulation, as sleep disorders are frequently comorbid with ADHD; (5) finally, we will evaluate the most recent studies on the use of probiotics in pediatric patients with ADHD.
Collapse
|
125
|
Shini S, Aland RC, Bryden WL. Avian intestinal ultrastructure changes provide insight into the pathogenesis of enteric diseases and probiotic mode of action. Sci Rep 2021; 11:167. [PMID: 33420315 PMCID: PMC7794591 DOI: 10.1038/s41598-020-80714-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 12/24/2020] [Indexed: 01/27/2023] Open
Abstract
Epithelial damage and loss of barrier integrity occur following intestinal infections in humans and animals. Gut health was evaluated by electron microscopy in an avian model that exposed birds to subclinical necrotic enteritis (NE) and fed them a diet supplemented with the probiotic Bacillus amyloliquefaciens strain H57 (H57). Scanning electron microscopy of ileal mucosa revealed significant villus damage, including focal erosions of epithelial cells and villous atrophy, while transmission electron microscopy demonstrated severe enterocyte damage and loss of cellular integrity in NE-exposed birds. In particular, mitochondria were morphologically altered, appearing irregular in shape or swollen, and containing electron-lucent regions of matrix and damaged cristae. Apical junctional complexes between adjacent enterocytes were significantly shorter, and the adherens junction was saccular, suggesting loss of epithelial integrity in NE birds. Segmented filamentous bacteria attached to villi, which play an important role in intestinal immunity, were more numerous in birds exposed to NE. The results suggest that mitochondrial damage may be an important initiator of NE pathogenesis, while H57 maintains epithelium and improves the integrity of intestinal mucosa. Potential actions of H57 are discussed that further define the mechanisms responsible for probiotic bacteria’s role in maintaining gut health.
Collapse
Affiliation(s)
- Shaniko Shini
- School of Agriculture and Food Sciences, University of Queensland, Gatton, QLD, 4343, Australia.
| | - R Claire Aland
- School of Biomedical Sciences, University of Queensland, St Lucia, QLD, 4071, Australia
| | - Wayne L Bryden
- School of Agriculture and Food Sciences, University of Queensland, Gatton, QLD, 4343, Australia
| |
Collapse
|
126
|
Mancin L, Rollo I, Mota JF, Piccini F, Carletti M, Susto GA, Valle G, Paoli A. Optimizing Microbiota Profiles for Athletes. Exerc Sport Sci Rev 2021; 49:42-49. [PMID: 33044333 DOI: 10.1249/jes.0000000000000236] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Gut microbiome influences athletes' physiology, but because of the complexity of sport performance and the great intervariability of microbiome features, it is not reasonable to define a single healthy microbiota profile for athletes. We suggest the use of specific meta-omics analysis coupled with innovative computational systems to uncover the hidden association between microbes and athlete's physiology and predict personalized recommendation.
Collapse
Affiliation(s)
| | | | - Joao Felipe Mota
- Clinical and Sports Nutrition Research Laboratory (LABINCE), Federal University of Goiás, Goiânia, Goiás, Brazil
| | | | | | | | | | | |
Collapse
|
127
|
Alwehaidah MS, Bakhiet M, AlFadhli S. Mitochondrial Haplogroup Reveals the Genetic Basis of Diabetes Mellitus Type 2 Comorbidity in Psoriasis. Med Princ Pract 2021; 30:62-68. [PMID: 32629455 PMCID: PMC7923845 DOI: 10.1159/000509937] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 07/06/2020] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE Published data show a clear link between psoriasis (Ps) and the increasing prevalence of comorbid conditions, such as diabetes mellitus type 2 (DM2). The role of the mitochondrial genomic haplogroup in the potential coexistence of Ps and DM2 comorbidity is the subject of this study. MATERIAL AND METHODS Ninety-eight Kuwaiti individuals were recruited in 4 cohorts (20 healthy controls, 15 with DM2, 34 with Ps, and 29 with Ps and diabetes mellitus). An Ion Torrent S5XL was used to sequence mitochondrial DNA (mtDNA). χ2 test was used to assess differences in the distribution of each haplogroup between cases and controls (p < 0.05). The Bonferroni correction was applied (p < 0.004). The mtDNA haplogroups were analyzed by HaploGrep. RESULTS Haplogroups R0, U, J, T, N, L3, M, H, X, HV, R, and K were detected in the studied population. Haplogroup M had a high risk for Ps (odds ratio (OR) 4.0, p = 0.003). Haplogroup R0 and J had decreased the risk of DM2 (OR 0.28, p = 0.007). CONCLUSION Our results indicated that mtDNA haplogroups have a potential contribution to the pathogenesis of Ps and DM2 comorbidity. We show for the first time that the comorbidity of diabetes in Ps may be related to mitochondrial dysfunction.
Collapse
Affiliation(s)
- Materah Salem Alwehaidah
- Department of Medical Laboratory, Faculty of Allied Health, Kuwait University, Sulaibekhat, Kuwait,
| | - Moiz Bakhiet
- Department of Molecular Medicine, College of Medical and Medicine Sciences, Arabian Gulf University, Manama, Bahrain
| | - Suad AlFadhli
- Department of Medical Laboratory, Faculty of Allied Health, Kuwait University, Sulaibekhat, Kuwait
| |
Collapse
|
128
|
Dewi L, Rosidi A, Noer ER, Ayuningtyas A. The Prospect for Type 2 Diabetes Mellitus Combined with Exercise and Synbiotics: A Perspective. Curr Diabetes Rev 2021; 17:e012821190875. [PMID: 33511948 DOI: 10.2174/1573399817666210129102956] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 12/08/2020] [Accepted: 12/10/2020] [Indexed: 11/22/2022]
Abstract
Change in gut microbiome diversity (the so-called dysbiosis) is correlated with insulin resistance conditions. Exercise is typically the first management for people with type 2 diabetes mellitus (T2DM), which is generally well-known for improving glucose regulation. The new prebiotics and probiotics, like synbiotics, designed to target specific diseases, require additional studies. While the effectiveness of exercise combined with synbiotics seems promising, this review discusses these agents' possibility of increasing the gut microbiota's diversity. Therefore, they could enhance short-chain fatty acids (SCFA). In particular, the synbiotic interaction on gut microbiota, the exercise mechanism in improving gut microbiota, and the prospect of the synergistic effect of the combination of synbiotic and exercise to improve insulin sensitivity are addressed.
Collapse
Affiliation(s)
- Luthfia Dewi
- Nutrition Department, Faculty of Nursing and Health Science, Universitas Muhammadiyah Semarang 50273, Semarang, Indonesia
| | - Ali Rosidi
- Nutrition Department, Faculty of Nursing and Health Science, Universitas Muhammadiyah Semarang 50273, Semarang, Indonesia
| | - Etika Ratna Noer
- Nutrition Department, Faculty of Medicine, Diponegoro University 50275, Semarang, Indonesia
| | - Annisa Ayuningtyas
- Nutrition Department, Faculty of Nursing and Health Science, Universitas Muhammadiyah Semarang 50273, Semarang, Indonesia
| |
Collapse
|
129
|
Lee HJ, Lee SH, Lee JH, Kim Y, Seong KM, Jin YW, Min KJ. Role of Commensal Microbes in the γ-Ray Irradiation-Induced Physiological Changes in Drosophila melanogaster. Microorganisms 2020; 9:microorganisms9010031. [PMID: 33374132 PMCID: PMC7824294 DOI: 10.3390/microorganisms9010031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/19/2020] [Accepted: 12/22/2020] [Indexed: 12/29/2022] Open
Abstract
Ionizing radiation induces biological/physiological changes and affects commensal microbes, but few studies have examined the relationship between the physiological changes induced by irradiation and commensal microbes. This study investigated the role of commensal microbes in the γ-ray irradiation-induced physiological changes in Drosophila melanogaster. The bacterial load was increased in 5 Gy irradiated flies, but irradiation decreased the number of operational taxonomic units. The mean lifespan of conventional flies showed no significant change by irradiation, whereas that of axenic flies was negatively correlated with the radiation dose. γ-Ray irradiation did not change the average number of eggs in both conventional and axenic flies. Locomotion of conventional flies was decreased after 5 Gy radiation exposure, whereas no significant change in locomotion activity was detected in axenic flies after irradiation. γ-Ray irradiation increased the generation of reactive oxygen species in both conventional and axenic flies, but the increase was higher in axenic flies. Similarly, the amounts of mitochondria were increased in irradiated axenic flies but not in conventional flies. These results suggest that axenic flies are more sensitive in their mitochondrial responses to radiation than conventional flies, and increased sensitivity leads to a reduced lifespan and other physiological changes in axenic flies.
Collapse
Affiliation(s)
- Hwa-Jin Lee
- Department of Biological Sciences, Inha University, Incheon 22212, Korea; (H.-J.L.); (S.-H.L.); (J.-H.L.)
| | - Shin-Hae Lee
- Department of Biological Sciences, Inha University, Incheon 22212, Korea; (H.-J.L.); (S.-H.L.); (J.-H.L.)
| | - Ji-Hyeon Lee
- Department of Biological Sciences, Inha University, Incheon 22212, Korea; (H.-J.L.); (S.-H.L.); (J.-H.L.)
| | - Yongjoong Kim
- Laboratory of Low Dose Risk Assessment, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Sciences, Seoul 01812, Korea; (Y.K.); (K.M.S.); (Y.W.J.)
| | - Ki Moon Seong
- Laboratory of Low Dose Risk Assessment, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Sciences, Seoul 01812, Korea; (Y.K.); (K.M.S.); (Y.W.J.)
| | - Young Woo Jin
- Laboratory of Low Dose Risk Assessment, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Sciences, Seoul 01812, Korea; (Y.K.); (K.M.S.); (Y.W.J.)
| | - Kyung-Jin Min
- Department of Biological Sciences, Inha University, Incheon 22212, Korea; (H.-J.L.); (S.-H.L.); (J.-H.L.)
- Correspondence:
| |
Collapse
|
130
|
Effects of 8 Weeks of 2S-Hesperidin Supplementation on Performance in Amateur Cyclists. Nutrients 2020; 12:nu12123911. [PMID: 33371483 PMCID: PMC7767530 DOI: 10.3390/nu12123911] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/10/2020] [Accepted: 12/18/2020] [Indexed: 12/12/2022] Open
Abstract
2S-Hesperidin is a flavanone (flavonoid) found in high concentrations in citrus fruits. It has an antioxidant and anti-inflammatory effects, improving performance in animals. This study investigated the effects of chronic intake of an orange extract (2S-hesperidin) or placebo on non-oxidative/glycolytic and oxidative metabolism markers and performance markers in amateur cyclists. A double-blind, randomized, placebo-controlled trial was carried out between late September and December 2018. Forty amateur cyclists were randomized into two groups: one taking 500 mg/day 2S-hesperidin and the other taking 500 mg/day placebo (microcellulose) for eight weeks. All participants completed the study. An incremental test was used to evaluate performance, and a step test was used to measure oxygen consumption, carbon dioxide, efficiency and oxidation of carbohydrates and fat by indirect calorimetry. The anaerobic power (non-oxidative) was determined using Wingate tests (30 s). After eight weeks supplementation, there was an increase in the incremental test in estimated functional threshold power (FTP) (3.2%; p ≤ 0.05) and maximum power (2.7%; p ≤ 0.05) with 2S-hesperdin compared to placebo. In the step test, there was a decrease in VO2 (L/min) (−8.3%; p ≤ 0.01) and VO2R (mL/kg/min) (−8.9%; p ≤ 0.01) at VT2 in placebo. However, there were no differences between groups. In the Wingate test, there was a significant increase (p ≤ 0.05) in peak and relative power in both groups, but without differences between groups. Supplementation with an orange extract (2S-hesperdin) 500 mg/day improves estimated FTP and maximum power performance in amateur cyclists.
Collapse
|
131
|
Castro-Barquero S, Ribó-Coll M, Lassale C, Tresserra-Rimbau A, Castañer O, Pintó X, Martínez-González MÁ, Sorlí JV, Salas-Salvadó J, Lapetra J, Gómez-Gracia E, Alonso-Gómez ÁM, Fiol M, Serra-Majem L, Sacanella E, Basterra-Gortari FJ, Portolés O, Babio N, Cofán M, Ros E, Estruch R, Hernáez Á. Mediterranean Diet Decreases the Initiation of Use of Vitamin K Epoxide Reductase Inhibitors and Their Associated Cardiovascular Risk: A Randomized Controlled Trial. Nutrients 2020; 12:E3895. [PMID: 33352771 PMCID: PMC7766197 DOI: 10.3390/nu12123895] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/12/2020] [Accepted: 12/16/2020] [Indexed: 12/29/2022] Open
Abstract
Our aim is to assess whether following a Mediterranean Diet (MedDiet) decreases the risk of initiating antithrombotic therapies and the cardiovascular risk associated with its use in older individuals at high cardiovascular risk. We evaluate whether participants of the PREvención con DIeta MEDiterránea (PREDIMED) study allocated to a MedDiet enriched in extra-virgin olive oil or nuts (versus a low-fat control intervention) disclose differences in the risk of initiation of: (1) vitamin K epoxide reductase inhibitors (acenocumarol/warfarin; n = 6772); (2) acetylsalicylic acid as antiplatelet agent (n = 5662); and (3) other antiplatelet drugs (cilostazol/clopidogrel/dipyridamole/ditazol/ticlopidine/triflusal; n = 6768). We also assess whether MedDiet modifies the association between the antithrombotic drug baseline use and incident cardiovascular events. The MedDiet intervention enriched with extra-virgin olive oil decreased the risk of initiating the use of vitamin K epoxide reductase inhibitors relative to control diet (HR: 0.68 [0.46-0.998]). Their use was also more strongly associated with an increased risk of cardiovascular disease in participants not allocated to MedDiet interventions (HRcontrol diet: 4.22 [1.92-9.30], HRMedDiets: 1.71 [0.83-3.52], p-interaction = 0.052). In conclusion, in an older population at high cardiovascular risk, following a MedDiet decreases the initiation of antithrombotic therapies and the risk of suffering major cardiovascular events among users of vitamin K epoxide reductase inhibitors.
Collapse
Affiliation(s)
- Sara Castro-Barquero
- Cardiovascular Risk, Nutrition and Aging Research Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain; (S.C.-B.); (M.R.-C.); (E.S.); (R.E.)
- Department of Medicine, Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain; (C.L.); (A.T.-R.); (O.C.); (X.P.); (M.Á.M.-G.); (J.V.S.); (J.S.-S.); (J.L.); (E.G.-G.); (Á.M.A.-G.); (M.F.); (L.S.-M.); (F.J.B.-G.); (O.P.); (N.B.); (M.C.); (E.R.)
| | - Margarita Ribó-Coll
- Cardiovascular Risk, Nutrition and Aging Research Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain; (S.C.-B.); (M.R.-C.); (E.S.); (R.E.)
- Faculty of Pharmacy and Food Science, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Camille Lassale
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain; (C.L.); (A.T.-R.); (O.C.); (X.P.); (M.Á.M.-G.); (J.V.S.); (J.S.-S.); (J.L.); (E.G.-G.); (Á.M.A.-G.); (M.F.); (L.S.-M.); (F.J.B.-G.); (O.P.); (N.B.); (M.C.); (E.R.)
- Cardiovascular Risk and Nutrition Research Group, Hospital del Mar Medical Research Institute (IMIM), 08003 Barcelona, Spain
| | - Anna Tresserra-Rimbau
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain; (C.L.); (A.T.-R.); (O.C.); (X.P.); (M.Á.M.-G.); (J.V.S.); (J.S.-S.); (J.L.); (E.G.-G.); (Á.M.A.-G.); (M.F.); (L.S.-M.); (F.J.B.-G.); (O.P.); (N.B.); (M.C.); (E.R.)
- Unitat de Nutrició Humana, Departament de Bioquimica i Biotecnologia, Hospital Universitari Sant Joan de Reus, Universitat Rovira i Virgili, 43201 Reus, Spain
- Institut d′Investigació Pere Virgili (IISPV), 43204 Reus, Spain
- Department of Nutrition, Food Science and Gastronomy, XaRTA, INSA, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain
| | - Olga Castañer
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain; (C.L.); (A.T.-R.); (O.C.); (X.P.); (M.Á.M.-G.); (J.V.S.); (J.S.-S.); (J.L.); (E.G.-G.); (Á.M.A.-G.); (M.F.); (L.S.-M.); (F.J.B.-G.); (O.P.); (N.B.); (M.C.); (E.R.)
- Cardiovascular Risk and Nutrition Research Group, Hospital del Mar Medical Research Institute (IMIM), 08003 Barcelona, Spain
| | - Xavier Pintó
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain; (C.L.); (A.T.-R.); (O.C.); (X.P.); (M.Á.M.-G.); (J.V.S.); (J.S.-S.); (J.L.); (E.G.-G.); (Á.M.A.-G.); (M.F.); (L.S.-M.); (F.J.B.-G.); (O.P.); (N.B.); (M.C.); (E.R.)
- Lipids and Vascular Risk Unit, Internal Medicine Service, Hospital Universitario de Bellvitge, 08907 L’Hospitalet de Llobregat, Spain
| | - Miguel Ángel Martínez-González
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain; (C.L.); (A.T.-R.); (O.C.); (X.P.); (M.Á.M.-G.); (J.V.S.); (J.S.-S.); (J.L.); (E.G.-G.); (Á.M.A.-G.); (M.F.); (L.S.-M.); (F.J.B.-G.); (O.P.); (N.B.); (M.C.); (E.R.)
- Department of Preventive Medicine and Public Health, Universidad de Navarra, 31008 Pamplona, Spain
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA 02115, USA
| | - José V. Sorlí
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain; (C.L.); (A.T.-R.); (O.C.); (X.P.); (M.Á.M.-G.); (J.V.S.); (J.S.-S.); (J.L.); (E.G.-G.); (Á.M.A.-G.); (M.F.); (L.S.-M.); (F.J.B.-G.); (O.P.); (N.B.); (M.C.); (E.R.)
- Department of Preventive Medicine, Universidad de Valencia, 46100 Valencia, Spain
| | - Jordi Salas-Salvadó
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain; (C.L.); (A.T.-R.); (O.C.); (X.P.); (M.Á.M.-G.); (J.V.S.); (J.S.-S.); (J.L.); (E.G.-G.); (Á.M.A.-G.); (M.F.); (L.S.-M.); (F.J.B.-G.); (O.P.); (N.B.); (M.C.); (E.R.)
- Unitat de Nutrició Humana, Departament de Bioquimica i Biotecnologia, Hospital Universitari Sant Joan de Reus, Universitat Rovira i Virgili, 43201 Reus, Spain
- Institut d′Investigació Pere Virgili (IISPV), 43204 Reus, Spain
| | - José Lapetra
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain; (C.L.); (A.T.-R.); (O.C.); (X.P.); (M.Á.M.-G.); (J.V.S.); (J.S.-S.); (J.L.); (E.G.-G.); (Á.M.A.-G.); (M.F.); (L.S.-M.); (F.J.B.-G.); (O.P.); (N.B.); (M.C.); (E.R.)
- Department of Family Medicine, Research Unit, Distrito Sanitario Atención Primaria Sevilla, 41013 Sevilla, Spain
| | - Enrique Gómez-Gracia
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain; (C.L.); (A.T.-R.); (O.C.); (X.P.); (M.Á.M.-G.); (J.V.S.); (J.S.-S.); (J.L.); (E.G.-G.); (Á.M.A.-G.); (M.F.); (L.S.-M.); (F.J.B.-G.); (O.P.); (N.B.); (M.C.); (E.R.)
- Department of Preventive Medicine and Public Health, Universidad de Málaga, 29071 Málaga, Spain
| | - Ángel M. Alonso-Gómez
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain; (C.L.); (A.T.-R.); (O.C.); (X.P.); (M.Á.M.-G.); (J.V.S.); (J.S.-S.); (J.L.); (E.G.-G.); (Á.M.A.-G.); (M.F.); (L.S.-M.); (F.J.B.-G.); (O.P.); (N.B.); (M.C.); (E.R.)
- Bioaraba Health Research Institute, Osakidetza Basque Health Service, Araba University Hospital, University of the Basque Country UPV/EHU, 01009 Vitoria-Gasteiz, Spain
| | - Miquel Fiol
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain; (C.L.); (A.T.-R.); (O.C.); (X.P.); (M.Á.M.-G.); (J.V.S.); (J.S.-S.); (J.L.); (E.G.-G.); (Á.M.A.-G.); (M.F.); (L.S.-M.); (F.J.B.-G.); (O.P.); (N.B.); (M.C.); (E.R.)
- Health Research Institute of the Balearic Islands (IdISBa), Hospital Son Espases, 07120 Palma de Mallorca, Spain
| | - Lluis Serra-Majem
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain; (C.L.); (A.T.-R.); (O.C.); (X.P.); (M.Á.M.-G.); (J.V.S.); (J.S.-S.); (J.L.); (E.G.-G.); (Á.M.A.-G.); (M.F.); (L.S.-M.); (F.J.B.-G.); (O.P.); (N.B.); (M.C.); (E.R.)
- Instituto de Investigaciones Biomédicas y Sanitarias, Universidad de Las Palmas de Gran Canaria, 35016 Las Palmas, Spain
- Centro Hospitalario Universitario Insular Materno Infantil (CHUIMI), Servicio Canario de Salud, 35016 Las Palmas, Spain
| | - Emilio Sacanella
- Cardiovascular Risk, Nutrition and Aging Research Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain; (S.C.-B.); (M.R.-C.); (E.S.); (R.E.)
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain; (C.L.); (A.T.-R.); (O.C.); (X.P.); (M.Á.M.-G.); (J.V.S.); (J.S.-S.); (J.L.); (E.G.-G.); (Á.M.A.-G.); (M.F.); (L.S.-M.); (F.J.B.-G.); (O.P.); (N.B.); (M.C.); (E.R.)
- Internal Medicine Service, Hospital Clínic, 08036 Barcelona, Spain
| | - Francisco Javier Basterra-Gortari
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain; (C.L.); (A.T.-R.); (O.C.); (X.P.); (M.Á.M.-G.); (J.V.S.); (J.S.-S.); (J.L.); (E.G.-G.); (Á.M.A.-G.); (M.F.); (L.S.-M.); (F.J.B.-G.); (O.P.); (N.B.); (M.C.); (E.R.)
- Department of Preventive Medicine and Public Health, Universidad de Navarra, 31008 Pamplona, Spain
- Department of Endocrinology and Nutrition, Complejo Hospitalario de Navarra, 31008 Pamplona, Spain
| | - Olga Portolés
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain; (C.L.); (A.T.-R.); (O.C.); (X.P.); (M.Á.M.-G.); (J.V.S.); (J.S.-S.); (J.L.); (E.G.-G.); (Á.M.A.-G.); (M.F.); (L.S.-M.); (F.J.B.-G.); (O.P.); (N.B.); (M.C.); (E.R.)
- Department of Preventive Medicine, Universidad de Valencia, 46100 Valencia, Spain
| | - Nancy Babio
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain; (C.L.); (A.T.-R.); (O.C.); (X.P.); (M.Á.M.-G.); (J.V.S.); (J.S.-S.); (J.L.); (E.G.-G.); (Á.M.A.-G.); (M.F.); (L.S.-M.); (F.J.B.-G.); (O.P.); (N.B.); (M.C.); (E.R.)
- Unitat de Nutrició Humana, Departament de Bioquimica i Biotecnologia, Hospital Universitari Sant Joan de Reus, Universitat Rovira i Virgili, 43201 Reus, Spain
- Institut d′Investigació Pere Virgili (IISPV), 43204 Reus, Spain
| | - Montserrat Cofán
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain; (C.L.); (A.T.-R.); (O.C.); (X.P.); (M.Á.M.-G.); (J.V.S.); (J.S.-S.); (J.L.); (E.G.-G.); (Á.M.A.-G.); (M.F.); (L.S.-M.); (F.J.B.-G.); (O.P.); (N.B.); (M.C.); (E.R.)
- Lipid Clinic, Endocrinology and Nutrition Service, Hospital Clínic, 08036 Barcelona, Spain
| | - Emilio Ros
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain; (C.L.); (A.T.-R.); (O.C.); (X.P.); (M.Á.M.-G.); (J.V.S.); (J.S.-S.); (J.L.); (E.G.-G.); (Á.M.A.-G.); (M.F.); (L.S.-M.); (F.J.B.-G.); (O.P.); (N.B.); (M.C.); (E.R.)
- Lipid Clinic, Endocrinology and Nutrition Service, Hospital Clínic, 08036 Barcelona, Spain
| | - Ramón Estruch
- Cardiovascular Risk, Nutrition and Aging Research Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain; (S.C.-B.); (M.R.-C.); (E.S.); (R.E.)
- Department of Medicine, Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain; (C.L.); (A.T.-R.); (O.C.); (X.P.); (M.Á.M.-G.); (J.V.S.); (J.S.-S.); (J.L.); (E.G.-G.); (Á.M.A.-G.); (M.F.); (L.S.-M.); (F.J.B.-G.); (O.P.); (N.B.); (M.C.); (E.R.)
- Internal Medicine Service, Hospital Clínic, 08036 Barcelona, Spain
| | - Álvaro Hernáez
- Cardiovascular Risk, Nutrition and Aging Research Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain; (S.C.-B.); (M.R.-C.); (E.S.); (R.E.)
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain; (C.L.); (A.T.-R.); (O.C.); (X.P.); (M.Á.M.-G.); (J.V.S.); (J.S.-S.); (J.L.); (E.G.-G.); (Á.M.A.-G.); (M.F.); (L.S.-M.); (F.J.B.-G.); (O.P.); (N.B.); (M.C.); (E.R.)
- Blanquerna School of Health Sciences, Universitat Ramon Llull, 08025 Barcelona, Spain
| |
Collapse
|
132
|
Li L, Li C, Lv M, Hu Q, Guo L, Xiong D. Correlation between alterations of gut microbiota and miR-122-5p expression in patients with type 2 diabetes mellitus. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1481. [PMID: 33313226 PMCID: PMC7729379 DOI: 10.21037/atm-20-6717] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background To investigate the correlation between gut microbiota and circulating microRNAs (miRNAs) in patients with primary diagnosis of type 2 diabetes mellitus (T2DM) and to explore the possible mechanisms of miRNA-gut microbiota crosstalke network in the regulation of the insulin signaling pathway and glucose homeostasis in T2DM. Methods T2DM patients and normal controls were recruited. Fasting plasma and fecal samples were collected from the subjects, and their biochemical indexes including fasting blood glucose (FBG), glycated hemoglobin (HbAlc), cholesterol (TC), total triglycerides (TG), high-density lipoprotein (HDL), low-density lipoprotein (LDL), and insulin were recorded. The variations in intestinal microbiota in the two groups were analyzed using 16S rRNA third-generation sequencing technology, and the differential expression of miRNAs between the groups was screened using miRNA high-throughput sequencing. The correlation and association between specifically changed intestinal microbiota and miRNA expressions were analyzed using a combination of bioinformatics analysis and statistical methods. Finally, 16S functional gene prediction analysis and target gene enrichment pathway analysis were carried out to predict relevant gut microbiota and miRNAs. Results Compared with normal controls, the biochemical indexes of HAlbc, FBG, TG, TC, LDL, HDL, and insulin were significantly different in T2DM patients (P<0.001, P<0.001, P=0.0125, P=0.98, P<0.001 P=0.022, and P=0.0013, respectively). The two groups also showed significantly different intestinal microbiota distribution and miRNA expression characteristics, including in the counts of Bacteriodes. uniformis and Phascolarctobacterium. Faecium (P=0.023, 0.031), which were negatively correlated (P=0.014, FC = -2.36) with the expression levels of serum miR-122-5p (r=−0.68, −0.60, P=0.01, 0.01). Conclusions This study discovered specific gut microbiota and miRNA characteristics in patients with a primary diagnosis of T2DM. A negative correlation between miR-122-5p and the intestinal bacteria Bacteriodes. uniformis and Phascolarctobacterium. Faecium was also revealed, suggesting that the crosstalke between miRNA and gut microbiota may regulate the insulin secretion and signal transduction by controling key genes of glucose metabolism during the development of T2DM.
Collapse
Affiliation(s)
- Lisha Li
- Department of Laboratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.,College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chaomin Li
- Endocrinology Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Meijun Lv
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiongying Hu
- Department of Laboratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lixuan Guo
- Department of Laboratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Daqian Xiong
- Department of Laboratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
133
|
Liao X, Wu M, Hao Y, Deng H. Exploring the Preventive Effect and Mechanism of Senile Sarcopenia Based on "Gut-Muscle Axis". Front Bioeng Biotechnol 2020; 8:590869. [PMID: 33251202 PMCID: PMC7674676 DOI: 10.3389/fbioe.2020.590869] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/09/2020] [Indexed: 12/11/2022] Open
Abstract
Age-related sarcopenia probably leads to chronic systemic inflammation and plays a vital role in the development of the complications of the disease. Gut microbiota, an environmental factor, is the medium of nutritional support to muscle cells, having significant impact on sarcopenia. Consequently, a significant amount of studies explored and showed the presence of gut microbiome–muscle axis (gut–muscle axis for short), which was possibly considered as the disease interventional target of age-related sarcopenia. However, a variety of nutrients probably affect the changes of the gut–muscle axis so as to affect the healthy balance of skeletal muscle. Therefore, it is necessary to study the mechanism of intestinal–muscle axis, and nutrients play a role in the treatment of senile sarcopenia through this mechanism. This review summarizes the available literature on mechanisms and specific pathways of gut–muscle axis and discusses the potential role and therapeutic feasibility of gut microbiota in age-related sarcopenia to understand the development of age-related sarcopenia and figure out the novel perspective of the potential therapeutic interventional targets.
Collapse
Affiliation(s)
- Xiaoshan Liao
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Nutrition and Food Hygiene, School of Public Health, Southern Medical University, Guangzhou, China
| | - Mengting Wu
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Nutrition and Food Hygiene, School of Public Health, Southern Medical University, Guangzhou, China
| | - Yuting Hao
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Nutrition and Food Hygiene, School of Public Health, Southern Medical University, Guangzhou, China
| | - Hong Deng
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Nutrition and Food Hygiene, School of Public Health, Southern Medical University, Guangzhou, China
| |
Collapse
|
134
|
Toohey JC, Townsend JR, Johnson SB, Toy AM, Vantrease WC, Bender D, Crimi CC, Stowers KL, Ruiz MD, VanDusseldorp TA, Feito Y, Mangine GT. Effects of Probiotic (Bacillus subtilis) Supplementation During Offseason Resistance Training in Female Division I Athletes. J Strength Cond Res 2020; 34:3173-3181. [PMID: 33105368 DOI: 10.1519/jsc.0000000000002675] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Toohey, JC, Townsend, JR, Johnson, SB, Toy, AM, Vantrease, WC, Bender, D, Crimi, CC, Stowers, KL, Ruiz, MD, VanDusseldorp, TA, Feito, Y, and Mangine, GT. Effects of probiotic (Bacillus subtilis) supplementation during offseason resistance training in female Division I athletes. J Strength Cond Res 34(11): 3173-3181, 2020-We examined the effects of probiotic (Bacillus subtilis) supplementation during offseason training in collegiate athletes. Twenty-three Division I female athletes (19.6 ± 1.0 years, 67.5 ± 7.4 kg, and 170.6 ± 6.8 cm) participated in this study and were randomized into either a probiotic (n = 11; DE111) or placebo (n = 12; PL) group while counterbalancing groups for sport. Athletes completed a 10-week resistance training program during the offseason, which consisted of 3-4 workouts per week of upper- and lower-body exercises and sport-specific training. Athletes consumed DE111 (DE111; 5 billion CFU/day) or PL supplement daily for the entire 10-week program. Before and after training, all athletes underwent 1 repetition maximum (1RM) strength testing (squat, deadlift, and bench press), performance testing (vertical jump and pro-agility), and isometric midthigh pull testing. Body composition (body fat [BF]%) was completed using BODPOD and bioelectrical impedance analysis, as well as muscle thickness (MT) measurement of the rectus femoris (RF) and vastus lateralis using ultrasonography. Separate repeated-measures analyses of variance were used to analyze all data. Significant (p ≤ 0.05) main effects for time were observed for improved squat 1RM, deadlift 1RM, bench press 1RM, vertical jump, RF MT, and BF%. Of these, a significant group × time interaction was noted for BF% (p = 0.015), where greater reductions were observed in DE111 (-2.05 ± 1.38%) compared with PL (-0.2 ± 1.6%). No other group differences were observed. These data suggest that probiotic consumption in conjunction with post-workout nutrition had no effect on physical performance but may improve body composition in female Division I soccer and volleyball players after offseason training.
Collapse
Affiliation(s)
- Jeremy C Toohey
- Exercise and Nutrition Science Graduate Program, Lipscomb University, Nashville, Tennessee; and
| | - Jeremy R Townsend
- Exercise and Nutrition Science Graduate Program, Lipscomb University, Nashville, Tennessee; and
| | - Sean B Johnson
- Exercise and Nutrition Science Graduate Program, Lipscomb University, Nashville, Tennessee; and
| | - Ann M Toy
- Exercise and Nutrition Science Graduate Program, Lipscomb University, Nashville, Tennessee; and
| | - William C Vantrease
- Exercise and Nutrition Science Graduate Program, Lipscomb University, Nashville, Tennessee; and
| | - David Bender
- Exercise and Nutrition Science Graduate Program, Lipscomb University, Nashville, Tennessee; and
| | - Chelsea C Crimi
- Exercise and Nutrition Science Graduate Program, Lipscomb University, Nashville, Tennessee; and
| | - Kathryn L Stowers
- Exercise and Nutrition Science Graduate Program, Lipscomb University, Nashville, Tennessee; and
| | - Matthew D Ruiz
- Exercise and Nutrition Science Graduate Program, Lipscomb University, Nashville, Tennessee; and
| | | | - Yuri Feito
- Exercise Science and Sport Management, Kennesaw State University, Kennesaw, Georgia
| | - Gerald T Mangine
- Exercise Science and Sport Management, Kennesaw State University, Kennesaw, Georgia
| |
Collapse
|
135
|
Ribeiro MF, Santos AA, Afonso MB, Rodrigues PM, Sá Santos S, Castro RE, Rodrigues CMP, Solá S. Diet-dependent gut microbiota impacts on adult neurogenesis through mitochondrial stress modulation. Brain Commun 2020; 2:fcaa165. [PMID: 33426525 PMCID: PMC7780462 DOI: 10.1093/braincomms/fcaa165] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 07/23/2020] [Accepted: 08/14/2020] [Indexed: 12/19/2022] Open
Abstract
The influence of dietary factors on brain health and mental function is becoming increasingly recognized. Similarly, mounting evidence supports a role for gut microbiota in modulating central nervous system function and behaviour. Still, the molecular mechanisms responsible for the impact of diet and associated microbiome in adult neurodegeneration are still largely unclear. In this study, we aimed to investigate whether and how changes in diet-associated microbiome and its metabolites impact on adult neurogenesis. Mice were fed a high-fat, choline-deficient diet, developing obesity and several features of the metabolic syndrome, including non-alcoholic steatohepatitis. Strikingly, our results showed, for the first time, that animals fed with this specific diet display premature increased neurogenesis, possibly exhausting the available neural stem cell pool for long-term neurogenesis processes. The high-fat, choline-deficient diet further induced neuroinflammation, oxidative stress, synaptic loss and cell death in different regions of the brain. Notably, this diet-favoured gut dysbiosis in the small intestine and cecum, up-regulating metabolic pathways of short-chain fatty acids, such as propionate and butyrate and significantly increasing propionate levels in the liver. By dissecting the effect of these two specific short-chain fatty acids in vitro, we were able to show that propionate and butyrate enhance mitochondrial biogenesis and promote early neurogenic differentiation of neural stem cells through reactive oxygen species- and extracellular signal-regulated kinases 1/2-dependent mechanism. More importantly, neurogenic niches of high-fat, choline-deficient-fed mice showed increased expression of mitochondrial biogenesis markers, and decreased mitochondrial reactive oxygen species scavengers, corroborating the involvement of this mitochondrial stress-dependent pathway in mediating changes of adult neurogenesis by diet. Altogether, our results highlight a mitochondria-dependent pathway as a novel mediator of the gut microbiota–brain axis upon dietary influences.
Collapse
Affiliation(s)
- Maria F Ribeiro
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - André A Santos
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Marta B Afonso
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Pedro M Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Sónia Sá Santos
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Rui E Castro
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Cecília M P Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Susana Solá
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
136
|
Cross-talk between gut and brain elicited by physical exercise. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165877. [PMID: 32544430 DOI: 10.1016/j.bbadis.2020.165877] [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: 12/30/2019] [Revised: 03/26/2020] [Accepted: 06/08/2020] [Indexed: 12/21/2022]
Abstract
In recent years, a paradigm shift in the bidirectional interactions within the gut-brain axis in normal and pathologic conditions has been evidenced. Although the causal relationship is not completely known, the application of new therapeutic tools such as physical exercise has been described in several studies. However, there are caveats to consider when interpreting the effect of exercise training on the axis. Therefore, an integrative perspective of the gut and the brain's communication pathway is discussed and the role of exercise on influencing this communication highway is explained in this review.
Collapse
|
137
|
Sanborn V, Gunstad J. The Potential Mediation of the Effects of Physical Activity on Cognitive Function by the Gut Microbiome. Geriatrics (Basel) 2020; 5:geriatrics5040063. [PMID: 32992812 PMCID: PMC7709629 DOI: 10.3390/geriatrics5040063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 09/21/2020] [Accepted: 09/23/2020] [Indexed: 12/13/2022] Open
Abstract
The population of older adults is growing dramatically worldwide. As older adults are at greater risk of developing disorders associated with cognitive dysfunction (i.e., dementia), healthcare costs are expected to double by 2040. Evidence suggests dementia may be slowed or prevented by lifestyle interventions, including physical activity (PA). PA is associated with improved cognitive function and may reduce risk for dementia by mitigating known risk factors (i.e., cardiovascular diseases) and/or by enhancing neurochemical processes. An emerging area of research suggests the gut microbiome may have similar neuroprotective effects. Altering the gut microbiome has been found to target physiological processes associated with dementia risk, and it influences gut-brain-microbiome axis signaling, impacting cognitive functioning. The gut microbiome can be altered by several means (i.e., disease, diet, prebiotics, probiotics), including PA. As PA and the gut microbiome independently influence cognitive function and PA changes the composition of the gut microbiome, cognitive improvement due to PA may be partially mediated by the gut microbiome. The present article provides an overview of the literature regarding the complex associations among PA, cognitive function, and the gut microbiome, as well as their underlying biological mechanisms. A comprehensive, theoretical model integrating evidence for the potential mediation is proposed.
Collapse
Affiliation(s)
- Victoria Sanborn
- Department of Psychological Sciences, Kent State University, Kent, OH 44240, USA;
- Correspondence:
| | - John Gunstad
- Department of Psychological Sciences, Kent State University, Kent, OH 44240, USA;
- Brain Health Research Institute, Kent State University, Kent, OH 44240, USA
| |
Collapse
|
138
|
Kirichenko TV, Markina YV, Sukhorukov VN, Khotina VA, Wu WK, Orekhov AN. A Novel Insight at Atherogenesis: The Role of Microbiome. Front Cell Dev Biol 2020; 8:586189. [PMID: 33072766 PMCID: PMC7536348 DOI: 10.3389/fcell.2020.586189] [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: 07/22/2020] [Accepted: 08/31/2020] [Indexed: 12/27/2022] Open
Abstract
There is an important task of current medicine to identify mechanisms and new markers of subclinical atherosclerosis in order to develop early targets for the diagnosis and treatment of this disease, since it causes such widespread diseases as myocardial infarction, stroke, sudden death, and other common reasons of disability and mortality in developed countries. In recent years, studies of the human microbiome in different fields of medicine have become increasingly popular; there is evidence from numerous studies of the significant contribution of microbiome in different steps of atherogenesis. This review attempted to determine the current status of the databases PubMed and Scopus (until May, 2020) to highlight current ideas on the potential role of microbiome and its metabolites in atherosclerosis development, its mechanisms of action in lipids metabolism, endothelial dysfunction, inflammatory pathways, and mitochondrial dysfunction. Results of clinical studies elucidating the relationship of microbiome with subclinical atherosclerosis and cardiovascular disease considered in this article demonstrate strong association of microbiome composition and its metabolites with atherosclerosis and cardiovascular disease. Data on microbiome impact in atherogenesis open a wide perspective to develop new diagnostic and therapeutic approaches, but further comprehensive studies are necessary.
Collapse
Affiliation(s)
- Tatiana V Kirichenko
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Research Institute of Human Morphology, Moscow, Russia
| | - Yuliya V Markina
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Research Institute of Human Morphology, Moscow, Russia
| | - Vasily N Sukhorukov
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Research Institute of Human Morphology, Moscow, Russia
| | - Victoria A Khotina
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Wei-Kai Wu
- Department of Internal Medicine, National Taiwan University Hospital, Bei-Hu Branch, Taipei, Taiwan
| | - Alexander N Orekhov
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Research Institute of Human Morphology, Moscow, Russia.,Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow, Russia
| |
Collapse
|
139
|
Food as medicine: targeting the uraemic phenotype in chronic kidney disease. Nat Rev Nephrol 2020; 17:153-171. [PMID: 32963366 DOI: 10.1038/s41581-020-00345-8] [Citation(s) in RCA: 128] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/10/2020] [Indexed: 02/07/2023]
Abstract
The observation that unhealthy diets (those that are low in whole grains, fruits and vegetables, and high in sugar, salt, saturated fat and ultra-processed foods) are a major risk factor for poor health outcomes has boosted interest in the concept of 'food as medicine'. This concept is especially relevant to metabolic diseases, such as chronic kidney disease (CKD), in which dietary approaches are already used to ameliorate metabolic and nutritional complications. Increased awareness that toxic uraemic metabolites originate not only from intermediary metabolism but also from gut microbial metabolism, which is directly influenced by diet, has fuelled interest in the potential of 'food as medicine' approaches in CKD beyond the current strategies of protein, sodium and phosphate restriction. Bioactive nutrients can alter the composition and metabolism of the microbiota, act as modulators of transcription factors involved in inflammation and oxidative stress, mitigate mitochondrial dysfunction, act as senolytics and impact the epigenome by altering one-carbon metabolism. As gut dysbiosis, inflammation, oxidative stress, mitochondrial dysfunction, premature ageing and epigenetic changes are common features of CKD, these findings suggest that tailored, healthy diets that include bioactive nutrients as part of the foodome could potentially be used to prevent and treat CKD and its complications.
Collapse
|
140
|
Turroni S, Magnani M, Kc P, Lesnik P, Vidal H, Heer M. Gut Microbiome and Space Travelers' Health: State of the Art and Possible Pro/Prebiotic Strategies for Long-Term Space Missions. Front Physiol 2020; 11:553929. [PMID: 33013480 PMCID: PMC7505921 DOI: 10.3389/fphys.2020.553929] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 08/14/2020] [Indexed: 12/20/2022] Open
Abstract
The upcoming exploration missions will imply a much longer duration than any of the missions flown so far. In these missions, physiological adaptation to the new environment leads to changes in different body systems, such as the cardiovascular and musculoskeletal systems, metabolic and neurobehavioral health and immune function. To keep space travelers healthy on their trip to Moon, Mars and beyond and their return to Earth, a variety of countermeasures need to be provided to maintain body functionality. From research on the International Space Station (ISS) we know today, that for instance prescribing an adequate training regime for each individual with the devices available in the respective spacecraft is still a challenge. Nutrient supply is not yet optimal and must be optimized in exploration missions. Food intake is intrinsically linked to changes in the gut microbiome composition. Most of the microbes that inhabit our body supply ecosystem benefit to the host-microbe system, including production of important resources, bioconversion of nutrients, and protection against pathogenic microbes. The gut microbiome has also the ability to signal the host, regulating the processes of energy storage and appetite perception, and influencing immune and neurobehavioral function. The composition and functionality of the microbiome most likely changes during spaceflight. Supporting a healthy microbiome by respective measures in space travelers might maintain their health during the mission but also support rehabilitation when being back on Earth. In this review we are summarizing the changes in the gut microbiome observed in spaceflight and analog models, focusing particularly on the effects on metabolism, the musculoskeletal and immune systems and neurobehavioral disorders. Since space travelers are healthy volunteers, we focus on the potential of countermeasures based on pre- and probiotics supplements.
Collapse
Affiliation(s)
- Silvia Turroni
- Unit of Microbial Ecology of Health, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Marciane Magnani
- Laboratory of Microbial Processes in Foods, Department of Food Engineering, Federal University of Paraíba, João Pessoa, Brazil
| | - Pukar Kc
- Institut National de la Santé et de la Recherche Médicale (Inserm, UMR_S 1166), Hôpital de la Pitié-Salpêtrière, Sorbonne Université, Paris, France
| | - Philippe Lesnik
- Institut National de la Santé et de la Recherche Médicale (Inserm, UMR_S 1166), Hôpital de la Pitié-Salpêtrière, Sorbonne Université, Paris, France.,Institute of Cardiometabolism and Nutrition, Hôpital Pitié-Salpêtrière, Paris, France
| | - Hubert Vidal
- CarMeN Laboratory, INSERM, INRA, Université Claude Bernard Lyon 1, Pierre-Benite, France
| | - Martina Heer
- International University of Applied Sciences, Bad Reichenhall, Germany.,Institute of Nutritional and Food Sciences, University of Bonn, Bonn, Germany
| |
Collapse
|
141
|
Oduro-Donkor D, Turner MC, Farnaud S, Renshaw D, Kyrou I, Hanson P, Hattersley J, Weickert MO, Menon V, Randeva HS, Barber TM. Modification of fecal microbiota as a mediator of effective weight loss and metabolic benefits following bariatric surgery. Expert Rev Endocrinol Metab 2020; 15:363-373. [PMID: 32840125 DOI: 10.1080/17446651.2020.1801412] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 07/22/2020] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Bariatric surgery (primarily Laparoscopic Sleeve Gastrectomy [LSG] and Roux-en-Y Gastric Bypass [RYGB]) is an efficacious and durable therapeutic option for weight loss in obesity. The mechanisms that mediate weight loss following bariatric surgery remain incompletely understood. AREAS COVERED Pubmed search of published data on fecal microbiota, metabolic health, LSG, and RYGB. The fecal microbiome plays a key role in the establishment and maintenance of metabolic wellbeing, and may also contribute (through fecal dysbiosis) to metabolic dysfunction. LSG and RYGB both result in characteristic, procedure-specific changes to the fecal microbiota that may mediate at least some of the resultant weight-loss and metabolically beneficial effects, when applied to the management of obesity. EXPERT OPINION The human fecal microbiome, containing around 100 trillion microbes, evolved over millions of years and interacts symbiotically with its human host. Rodent-based studies have provided insights into the complexities of the gut-microbiome-brain axis. This includes the important role of the gut microbiome in the mediation of normal immunological development, inflammatory pathways, metabolic functioning, hypothalamic appetite regulation, and the absorption of essential nutrients as by-products of bacterial metabolism. Fecal transformation is likely to provide an important therapeutic target for future prevention and management of obesity and metabolic dysfunction.
Collapse
Affiliation(s)
- Dominic Oduro-Donkor
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick , Coventry, UK
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism, University Hospitals Coventry and Warwickshire , Coventry, UK
| | - Mark C Turner
- Centre for Sport, Exercise and Life Sciences, Faculty of Health & Life Sciences, Coventry University , Coventry, UK
| | - Sebastien Farnaud
- Centre for Sport, Exercise and Life Sciences, Faculty of Health & Life Sciences, Coventry University , Coventry, UK
| | - Derek Renshaw
- Centre for Sport, Exercise and Life Sciences, Faculty of Health & Life Sciences, Coventry University , Coventry, UK
| | - Ioannis Kyrou
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick , Coventry, UK
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism, University Hospitals Coventry and Warwickshire , Coventry, UK
- Aston Medical Research Institute, Aston Medical School, Aston University , Birmingham, UK
| | - Petra Hanson
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick , Coventry, UK
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism, University Hospitals Coventry and Warwickshire , Coventry, UK
- NIHR CRF Human Metabolism Research Unit, University Hospitals Coventry and Warwickshire , Coventry, UK
| | - John Hattersley
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism, University Hospitals Coventry and Warwickshire , Coventry, UK
- NIHR CRF Human Metabolism Research Unit, University Hospitals Coventry and Warwickshire , Coventry, UK
| | - Martin O Weickert
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick , Coventry, UK
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism, University Hospitals Coventry and Warwickshire , Coventry, UK
- Centre for Sport, Exercise and Life Sciences, Faculty of Health & Life Sciences, Coventry University , Coventry, UK
- NIHR CRF Human Metabolism Research Unit, University Hospitals Coventry and Warwickshire , Coventry, UK
| | - Vinod Menon
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick , Coventry, UK
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism, University Hospitals Coventry and Warwickshire , Coventry, UK
| | - Harpal S Randeva
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick , Coventry, UK
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism, University Hospitals Coventry and Warwickshire , Coventry, UK
- Aston Medical Research Institute, Aston Medical School, Aston University , Birmingham, UK
- NIHR CRF Human Metabolism Research Unit, University Hospitals Coventry and Warwickshire , Coventry, UK
| | - Thomas M Barber
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick , Coventry, UK
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism, University Hospitals Coventry and Warwickshire , Coventry, UK
- NIHR CRF Human Metabolism Research Unit, University Hospitals Coventry and Warwickshire , Coventry, UK
| |
Collapse
|
142
|
Sato FT, Yap YA, Crisma AR, Portovedo M, Murata GM, Hirabara SM, Ribeiro WR, Marcantonio Ferreira C, Cruz MM, Pereira JNB, Payolla TB, Guima SES, Thomas AM, Setubal JC, Alonso-Vale MIC, Santos MF, Curi R, Marino E, Vinolo MAR. Tributyrin Attenuates Metabolic and Inflammatory Changes Associated with Obesity through a GPR109A-Dependent Mechanism. Cells 2020; 9:E2007. [PMID: 32882837 PMCID: PMC7563536 DOI: 10.3390/cells9092007] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/21/2020] [Accepted: 08/24/2020] [Indexed: 12/21/2022] Open
Abstract
Obesity is linked with altered microbial short-chain fatty acids (SCFAs), which are a signature of gut dysbiosis and inflammation. In the present study, we investigated whether tributyrin, a prodrug of the SCFA butyrate, could improve metabolic and inflammatory profiles in diet-induced obese mice. Mice fed a high-fat diet for eight weeks were treated with tributyrin or placebo for another six weeks. We show that obese mice treated with tributyrin had lower body weight gain and an improved insulin responsiveness and glucose metabolism, partly via reduced hepatic triglycerides content. Additionally, tributyrin induced an anti-inflammatory state in the adipose tissue by reduction of Il-1β and Tnf-a and increased Il-10, Tregs cells and M2-macrophages. Moreover, improvement in glucose metabolism and reduction of fat inflammatory states associated with tributyrin treatment were dependent on GPR109A activation. Our results indicate that exogenous targeting of SCFA butyrate attenuates metabolic and inflammatory dysfunction, highlighting a potentially novel approach to tackle obesity.
Collapse
Affiliation(s)
- Fabio Takeo Sato
- Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas 13083007, Brazil; (F.T.S.); (M.P.)
| | - Yu Anne Yap
- Department of Biochemistry, Biomedicine Discovery Institute, Monash University, Melbourne 3800, Australia;
| | - Amanda Rabello Crisma
- Department of Clinical Analyses, Federal University of Paraná, Curitiba 80060000, Brazil;
| | - Mariana Portovedo
- Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas 13083007, Brazil; (F.T.S.); (M.P.)
| | - Gilson Masahiro Murata
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508000, Brazil; (G.M.M.); (T.B.P.); (R.C.)
| | - Sandro Massao Hirabara
- Interdisciplinary Postgraduate Program in Health Science, Cruzeiro do Sul University, São Paulo 01506000, Brazil; (S.M.H.); (J.N.B.P.)
| | - Willian Rodrigues Ribeiro
- Department of Pharmaceutics Sciences, Institute of Environmental Chemistry and Pharmaceutical Sciences, Federal University of São Paulo, Diadema 09972270, Brazil; (W.R.R.); (C.M.F.)
| | - Caroline Marcantonio Ferreira
- Department of Pharmaceutics Sciences, Institute of Environmental Chemistry and Pharmaceutical Sciences, Federal University of São Paulo, Diadema 09972270, Brazil; (W.R.R.); (C.M.F.)
| | - Maysa Mariana Cruz
- Department of Biological Sciences, Institute of Environmental, Chemical and Pharmaceutical Sciences, Federal University of São Paulo, Diadema 09972270, Brazil; (M.M.C.); (M.I.C.A.-V.)
| | - Joice Naiara Bertaglia Pereira
- Interdisciplinary Postgraduate Program in Health Science, Cruzeiro do Sul University, São Paulo 01506000, Brazil; (S.M.H.); (J.N.B.P.)
| | - Tanyara Baliani Payolla
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508000, Brazil; (G.M.M.); (T.B.P.); (R.C.)
| | - Suzana Eiko Sato Guima
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508000, Brazil; (S.E.S.G.); (A.M.T.); (J.C.S.)
| | - Andrew Maltez Thomas
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508000, Brazil; (S.E.S.G.); (A.M.T.); (J.C.S.)
| | - João Carlos Setubal
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508000, Brazil; (S.E.S.G.); (A.M.T.); (J.C.S.)
| | - Maria Isabel Cardoso Alonso-Vale
- Department of Biological Sciences, Institute of Environmental, Chemical and Pharmaceutical Sciences, Federal University of São Paulo, Diadema 09972270, Brazil; (M.M.C.); (M.I.C.A.-V.)
| | - Marinilce Fagundes Santos
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508000, Brazil;
| | - Rui Curi
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508000, Brazil; (G.M.M.); (T.B.P.); (R.C.)
- Interdisciplinary Postgraduate Program in Health Science, Cruzeiro do Sul University, São Paulo 01506000, Brazil; (S.M.H.); (J.N.B.P.)
- Butantan Institute, São Paulo 05503900, Brazil
| | - Eliana Marino
- Department of Biochemistry, Biomedicine Discovery Institute, Monash University, Melbourne 3800, Australia;
| | - Marco A. R. Vinolo
- Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas 13083007, Brazil; (F.T.S.); (M.P.)
- Experimental Medicine Research Cluster (EMRC), University of Campinas (UNICAMP), Campinas 13083007, Brazil
- Obesity and Comorbidities Research Center (OCRC), University of Campinas (UNICAMP), Campinas 13083864, Brazil
| |
Collapse
|
143
|
Li H, He Y, Zhang C, Ba T, Guo Z, Zhuo Y, He L, Dai H. NOX1 down-regulation attenuated the autophagy and oxidative damage in pig intestinal epithelial cell following transcriptome analysis of transport stress. Gene 2020; 763:145071. [PMID: 32827682 DOI: 10.1016/j.gene.2020.145071] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/31/2020] [Accepted: 08/17/2020] [Indexed: 02/06/2023]
Abstract
The previous study indicated that transport stress resulted in oxidative damage and autophagy/mitophagy elevation, companied by NOX1 over- expression in the jejunal tissues of pigs. However, the transportation-related gene expression profile and NOX1 function in intestine remain to be explicated. In the current study, differentially expressed genes involved in PI3K-Akt and NF-κB pathways, oxidative stress and autophagy process have been identified in pig jejunal tissues after transcriptome analysis following transportation. The physiological functions of NOX1 down-regulation were explored against oxidative damage and excessive autophagy in porcine intestinal epithelial cells (IPEC-1) following NOX1 inhibitor ML171 and H2O2 treatments. NOX1 down-regulation could decrease the content of Malondialdehyde (MDA), Lactic dehydrogenase (LDH) activity and reactive oxygen species (ROS) level, and up-regulate superoxide dismutase (SOD) activity. Furthermore, mitochondrial membrane potential and content were restored, and the expressions of tight junction proteins (Claudin-1 and ZO-1) were also increased. Additionally, NOX1 inhibitior could down-regulate the expression of autophagy-associated proteins (ATG5, LC3, p62), accompanied by activating SIRT1/PGC-1α pathway. NOX1 down-regulation might alleviate oxidative stress-induced mitochondria damage and intestinal mucosal injury via modulating excessive autophagy and SIRT1/PGC-1α signaling pathway. The data will shed light on the molecular mechanism of NOX1 on intestine oxidative damage following pig transportation.
Collapse
Affiliation(s)
- Huari Li
- College of Veterinary Medicine, Huazhong Agricultural University, No.1 Shizishan Street, Wuhan 430070, Hubei, China
| | - Yulong He
- College of Veterinary Medicine, Huazhong Agricultural University, No.1 Shizishan Street, Wuhan 430070, Hubei, China
| | - Cheng Zhang
- College of Veterinary Medicine, Huazhong Agricultural University, No.1 Shizishan Street, Wuhan 430070, Hubei, China
| | - Tongtong Ba
- College of Veterinary Medicine, Huazhong Agricultural University, No.1 Shizishan Street, Wuhan 430070, Hubei, China
| | - Zeheng Guo
- College of Veterinary Medicine, Huazhong Agricultural University, No.1 Shizishan Street, Wuhan 430070, Hubei, China
| | - Yisha Zhuo
- College of Veterinary Medicine, Huazhong Agricultural University, No.1 Shizishan Street, Wuhan 430070, Hubei, China
| | - Lihua He
- College of Veterinary Medicine, Huazhong Agricultural University, No.1 Shizishan Street, Wuhan 430070, Hubei, China
| | - Hanchuan Dai
- College of Veterinary Medicine, Huazhong Agricultural University, No.1 Shizishan Street, Wuhan 430070, Hubei, China.
| |
Collapse
|
144
|
The Interaction between Mitochondrial Oxidative Stress and Gut Microbiota in the Cardiometabolic Consequences in Diet-Induced Obese Rats. Antioxidants (Basel) 2020; 9:antiox9070640. [PMID: 32708095 PMCID: PMC7402124 DOI: 10.3390/antiox9070640] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/14/2020] [Accepted: 07/18/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The objective of this study is to determine the role of mitochondrial oxidative stress in the dysbiosis associated with a high fat diet in rats. In addition, the impact of gut microbiota (GM) in the cardiometabolic consequences of diet-induced obesity in rats has been evaluated. METHODS Male Wistar rats were fed either a high fat diet (HFD) or a control (CT) one for 6 weeks. At the third week, one-half of the animals of each group were treated with the mitochondrial antioxidant MitoTempo (MT; 0.7 mgKg-1day-1 i.p). RESULTS Animals fed an HFD showed a lower microbiota evenness and diversity in comparison to CT rats. This dysbiosis is characterized by a decrease in Firmicutes/Bacteroidetes ratio and relevant changes at family and genera compared with the CT group. This was accompanied by a reduction in colonic mucin-secreting goblet cells. These changes were reversed by MT treatment. The abundance of certain genera could also be relevant in the metabolic consequences of obesity, as well as in the occurrence of cardiac fibrosis associated with obesity. CONCLUSIONS These results support an interaction between GM and mitochondrial oxidative stress and its relation with development of cardiac fibrosis, suggesting new approaches in the management of obesity-related cardiometabolic consequences.
Collapse
|
145
|
Tai YK, Ng C, Purnamawati K, Yap JLY, Yin JN, Wong C, Patel BK, Soong PL, Pelczar P, Fröhlich J, Beyer C, Fong CHH, Ramanan S, Casarosa M, Cerrato CP, Foo ZL, Pannir Selvan RM, Grishina E, Degirmenci U, Toh SJ, Richards PJ, Mirsaidi A, Wuertz‐Kozak K, Chong SY, Ferguson SJ, Aguzzi A, Monici M, Sun L, Drum CL, Wang J, Franco‐Obregón A. Magnetic fields modulate metabolism and gut microbiome in correlation with
Pgc‐1α
expression: Follow‐up to an in vitro magnetic mitohormetic study. FASEB J 2020; 34:11143-11167. [DOI: 10.1096/fj.201903005rr] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 06/07/2020] [Accepted: 06/15/2020] [Indexed: 01/07/2023]
Affiliation(s)
- Yee Kit Tai
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Charmaine Ng
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
| | - Kristy Purnamawati
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Jasmine Lye Yee Yap
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Jocelyn Naixin Yin
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Craig Wong
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Bharati Kadamb Patel
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
| | - Poh Loong Soong
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Pawel Pelczar
- Centre for Transgenic Models University of Basel Basel Switzerland
- Institute of Laboratory Animal Science University of Zürich Zürich Switzerland
| | | | - Christian Beyer
- Centre Suisse d'électronique et de microtechnique, CSEM SA Neuchatel Switzerland
| | - Charlene Hui Hua Fong
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Sharanya Ramanan
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Marco Casarosa
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio” University of Florence Florence Italy
- Institute for Biomechanics ETH Zürich Zürich Switzerland
| | | | - Zi Ling Foo
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Rina Malathi Pannir Selvan
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Elina Grishina
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Ufuk Degirmenci
- Institute of Molecular and Cell Biology, A*STAR Singapore Singapore
| | - Shi Jie Toh
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
| | - Pete J. Richards
- Competence Center for Applied Biotechnology and Molecular Medicine University of Zürich Zürich Switzerland
| | - Ali Mirsaidi
- Competence Center for Applied Biotechnology and Molecular Medicine University of Zürich Zürich Switzerland
| | - Karin Wuertz‐Kozak
- Competence Center for Applied Biotechnology and Molecular Medicine University of Zürich Zürich Switzerland
- Department of Biomedical Engineering Rochester Institute of Technology (RIT) Rochester NY USA
- Cardiovascular Research Institute (CVRI), National University Heart Centre Singapore (NUHCS) Singapore Singapore
| | - Suet Yen Chong
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Cardiovascular Research Institute (CVRI), National University Heart Centre Singapore (NUHCS) Singapore Singapore
| | - Stephen J. Ferguson
- Institute of Molecular and Cell Biology, A*STAR Singapore Singapore
- Competence Center for Applied Biotechnology and Molecular Medicine University of Zürich Zürich Switzerland
| | - Adriano Aguzzi
- Institut für Neuropathologie Universitätsspital Zürich Zürich Switzerland
| | - Monica Monici
- ASAcampus JL, ASA Res. Div. ‐ Dept. of Experimental and Clinical Biomedical Sciences “Mario Serio” University of Florence Florence Italy
| | - Lei Sun
- DUKE‐NUS Graduate Medical School Singapore Singapore Singapore
| | - Chester L. Drum
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Cardiovascular Research Institute (CVRI), National University Heart Centre Singapore (NUHCS) Singapore Singapore
| | - Jiong‐Wei Wang
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Cardiovascular Research Institute (CVRI), National University Heart Centre Singapore (NUHCS) Singapore Singapore
- Department of Physiology Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
| | - Alfredo Franco‐Obregón
- Department of Surgery Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Biolonic Currents Electromagnetic Pulsing Systems Laboratory BICEPS, National University of Singapore Singapore Singapore
- Institute of Molecular and Cell Biology, A*STAR Singapore Singapore
- Department of Physiology Yong Loo Lin School of Medicine, National University of Singapore Singapore Singapore
- Institute for Health Innovation & Technology, iHealthtech National University of Singapore Singapore Singapore
| |
Collapse
|
146
|
Areco VA, Kohan R, Talamoni G, Tolosa de Talamoni NG, Peralta López ME. Intestinal Ca 2+ absorption revisited: A molecular and clinical approach. World J Gastroenterol 2020; 26:3344-3364. [PMID: 32655262 PMCID: PMC7327788 DOI: 10.3748/wjg.v26.i24.3344] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 05/11/2020] [Accepted: 06/10/2020] [Indexed: 02/06/2023] Open
Abstract
Ca2+ has an important role in the maintenance of the skeleton and is involved in the main physiological processes. Its homeostasis is controlled by the intestine, kidney, bone and parathyroid glands. The intestinal Ca2+ absorption occurs mainly via the paracellular and the transcellular pathways. The proteins involved in both ways are regulated by calcitriol and other hormones as well as dietary factors. Fibroblast growth factor 23 (FGF-23) is a strong antagonist of vitamin D action. Part of the intestinal Ca2+ movement seems to be vitamin D independent. Intestinal Ca2+ absorption changes according to different physiological conditions. It is promoted under high Ca2+ demands such as growth, pregnancy, lactation, dietary Ca2+ deficiency and high physical activity. In contrast, the intestinal Ca2+ transport decreases with aging. Oxidative stress inhibits the intestinal Ca2+ absorption whereas the antioxidants counteract the effects of prooxidants leading to the normalization of this physiological process. Several pathologies such as celiac disease, inflammatory bowel diseases, Turner syndrome and others occur with inhibition of intestinal Ca2+ absorption, some hypercalciurias show Ca2+ hyperabsorption, most of these alterations are related to the vitamin D endocrine system. Further research work should be accomplished in order not only to know more molecular details but also to detect possible therapeutic targets to ameliorate or avoid the consequences of altered intestinal Ca2+ absorption.
Collapse
Affiliation(s)
- Vanessa A Areco
- Laboratorio “Dr. Fernando Cañas”, Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, INICSA (CONICET-Universidad Nacional de Córdoba), Córdoba 5000, Argentina
| | - Romina Kohan
- Laboratorio “Dr. Fernando Cañas”, Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, INICSA (CONICET-Universidad Nacional de Córdoba), Córdoba 5000, Argentina
| | - Germán Talamoni
- Laboratorio “Dr. Fernando Cañas”, Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, INICSA (CONICET-Universidad Nacional de Córdoba), Córdoba 5000, Argentina
| | - Nori G Tolosa de Talamoni
- Laboratorio “Dr. Fernando Cañas”, Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, INICSA (CONICET-Universidad Nacional de Córdoba), Córdoba 5000, Argentina
| | - María E Peralta López
- Laboratorio “Dr. Fernando Cañas”, Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, INICSA (CONICET-Universidad Nacional de Córdoba), Córdoba 5000, Argentina
| |
Collapse
|
147
|
Tzemah Shahar R, Koren O, Matarasso S, Shochat T, Magzal F, Agmon M. Attributes of Physical Activity and Gut Microbiome in Adults: A Systematic Review. Int J Sports Med 2020; 41:801-814. [PMID: 32455454 DOI: 10.1055/a-1157-9257] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Growing evidence shows the contribution of physical activity interventions to the gut microbiome. However, specific physical activity characteristics that can modify the gut microbiome are unknown. This review's aim was to explore the contribution of physical activity intervention characteristics on human gut microbiome composition, in terms of diversity, specific bacterial groups, and associated gut microbiome metabolites. A literature search in PubMed; Cochrane Library; CINAHL-EBSCO; SCOPUS; Web of Science; ClinicalTrials.gov; PROSPERO; and ProQuest. Five studies met the inclusion criteria of a physical activity intervention duration of at least five weeks, with any description of the type or dose used. All included studies reported an endurance training; two studies used endurance and an additional muscle-strengthening training regimen. All studies reported using a dietary intervention control. Reported gut microbiome α-diversity changes were non-significant, β-diversity changes were mixed (three studies reported an increase, two reported non-significant changes). All studies reported significant changes in the abundances of specific bacterial/archaea groups and bacteria-related metabolites following interventions. In conclusion, physical activity (regardless of specific characteristics) has significant contribution to gut microbiome composition and associated metabolites. There are no current recommendations for physical activity to promote gut microbiome composition. Future studies should focus on the contribution of current recommended physical activity dose to gut microbiome composition.
Collapse
Affiliation(s)
- Roy Tzemah Shahar
- The Cheryl Spencer Institute of Nursing Research, University of Haifa, Haifa, Israel
| | - Omry Koren
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Sarah Matarasso
- Internal Medicine C, Bnei Zion Medical Centre, Haifa, Israel
| | - Tamar Shochat
- The Cheryl Spencer Institute of Nursing Research, University of Haifa, Haifa, Israel
| | - Faiga Magzal
- Laboratory of Human Health and Nutrition Sciences, MIGAL-Galilee Research Institute, Kiryat Shmona, Israel
| | - Maayan Agmon
- The Cheryl Spencer Institute of Nursing Research, University of Haifa, Haifa, Israel
| |
Collapse
|
148
|
Abstract
The gastrointestinal microbiome plays a pivotal role in physiological homeostasis of the intestine as well as in the pathophysiology of diseases including inflammatory bowel diseases (IBD) and colorectal cancer (CRC). Emerging evidence suggests that gut microbiota signal to the mitochondria of mucosal cells, including epithelial cells and immune cells. Gut microbiota signaling to mitochondria has been shown to alter mitochondrial metabolism, activate immune cells, induce inflammasome signaling, and alter epithelial barrier function. Both dysbiosis of the gut microbiota and mitochondrial dysfunction are associated with chronic intestinal inflammation and CRC. This review discusses mitochondrial metabolism of gut mucosal cells, mitochondrial dysfunction, and known gut microbiota-mediated mitochondrial alterations during IBD and CRC.
Collapse
Affiliation(s)
- Dakota N. Jackson
- Department of Internal Medicine, Division of Gastroenterology, Baylor Scott & White Research Institute, Baylor University Medical Center, Dallas, TX, USA
| | - Arianne L. Theiss
- Department of Internal Medicine, Division of Gastroenterology, Baylor Scott & White Research Institute, Baylor University Medical Center, Dallas, TX, USA,CONTACT Arianne L. Theiss Division of Gastroenterology, Baylor Research Institute, Baylor University Medical Center, 250 Hoblitzelle, 3500 Gaston Avenue, Dallas, TX75246, USA
| |
Collapse
|
149
|
Ruiz E, Penrose HM, Heller S, Nakhoul H, Baddoo M, Flemington EF, Kandil E, Savkovic SD. Bacterial TLR4 and NOD2 signaling linked to reduced mitochondrial energy function in active inflammatory bowel disease. Gut Microbes 2020; 11:350-363. [PMID: 31063017 PMCID: PMC7524318 DOI: 10.1080/19490976.2019.1611152] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 04/12/2019] [Accepted: 04/18/2019] [Indexed: 02/03/2023] Open
Abstract
Inflammatory bowel disease (IBD) has been linked to active signaling with bacterial components and reduced mitochondrial ATP production; however, synergism between both of these disease characteristics remains unclear. We aimed to determine in human IBD transcriptomes the link between a transcriptional signature unique to intestinal cells (ICs) with reduced mitochondrial ATP production (Mito-0) and bacteria triggered signaling using a bioinformatics approach. We generated an IC Mito-0 panel comprised of 199 differentially expressed (DE) transcripts mediated by reduced mitochondrial ATP function (DEGseq, log2 fold-change > |2|, p < .001). Transcripts from this panel were involved in diverse biological functions including regulation of mitochondrial energy (lower ATP), extracellular matrix, cell-cell contact, cytoskeleton, growth, metabolism, and inflammation. Next, unsupervised hierarchical clustering showed that the Mito-0 panel distinctly separated inflamed IBD from non-inflamed transcriptomes, which was also supported by principal component analysis (PCA) revealing distinct variation between sample types based on presence of the Mito-0 signature (PCA, p = 8.77e-09). Utilizing three independent IBD cohorts, we validated that 60 novel transcripts from the Mito-0 panel were significantly increased in inflamed tissue. Subsequently, KEGG generated bacterial TLR4 and NOD2 transcriptional signatures strongly associated with inflamed IBD transcriptomes and with the Mito-0 signature as determined by Spearman's analysis (coefficient of correlation, r = 0.92, p < .05). Herein, using a comprehensive analysis we demonstrated existence of an axis between bacteria triggered signaling and reduced mitochondrial energy function. Furthermore, we identified and validated novel transcripts within this axis as potential drivers and therapeutic targets for human IBD.
Collapse
Affiliation(s)
- Emmanuelle Ruiz
- Division of Endocrine and Oncologic Surgery, Department of Surgery, Tulane University, New Orleans, LA, USA
| | - Harrison M. Penrose
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Sandra Heller
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Hani Nakhoul
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Melody Baddoo
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Erik F. Flemington
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Emad Kandil
- Division of Endocrine and Oncologic Surgery, Department of Surgery, Tulane University, New Orleans, LA, USA
| | - Suzana D. Savkovic
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| |
Collapse
|
150
|
Affiliation(s)
- Suzana D. Savkovic
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA,
| |
Collapse
|