1
|
K Soman S, Swain M, Dagda RK. BDNF-TrkB Signaling in Mitochondria: Implications for Neurodegenerative Diseases. Mol Neurobiol 2025; 62:1756-1769. [PMID: 39030441 PMCID: PMC11909598 DOI: 10.1007/s12035-024-04357-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 07/09/2024] [Indexed: 07/21/2024]
Abstract
Brain-derived neurotrophic factor (BDNF) plays a pivotal role in neuronal development, synaptic plasticity, and overall neuronal health by binding to its receptor, tyrosine receptor kinase B (TrkB). This review delves into the intricate mechanisms through which BDNF-TrkB signaling influences mitochondrial function and potentially influences pathology in neurodegenerative diseases. This review highlights the BDNF-TrkB signaling pathway which regulates mitochondrial bioenergetics, biogenesis, and dynamics, mitochondrial processes vital for synaptic transmission and plasticity. Furthermore, we explore how the BDNF-TrkB-PKA signaling in the cytosol and in mitochondria affects mitochondrial transport and distribution and mitochondrial content, which is crucial for supporting the energy demands of synapses. The dysregulation of this signaling pathway is linked to various neurodegenerative diseases, including Alzheimer's and Parkinson's disease, which are characterized by mitochondrial dysfunction and reduced BDNF expression. By examining seminal studies that have characterized this signaling pathway in health and disease, the present review underscores the potential of enhancing BDNF-TrkB signaling to mitigate mitochondrial dysfunction in neurodegenerative diseases, offering insights into therapeutic strategies to enhance neuronal resilience and function.
Collapse
Affiliation(s)
- Smijin K Soman
- Department of Pharmacology, University of Nevada, Reno School of Medicine, 1664 North Virginia Street, Reno, NV, 89557, USA
| | - Maryann Swain
- Department of Pharmacology, University of Nevada, Reno School of Medicine, 1664 North Virginia Street, Reno, NV, 89557, USA
| | - Ruben K Dagda
- Department of Pharmacology, University of Nevada, Reno School of Medicine, 1664 North Virginia Street, Reno, NV, 89557, USA.
| |
Collapse
|
2
|
Brown AD, Marko AD, Marko DM, Baranowski BJ, Silvera S, Finch MS, Yang AJ, Dhaliwal R, Ryan CR, Roy BD, Fajardo VA, MacPherson REK. Brain-derived neurotrophic factor drives muscle adaptation similar to aerobic training in mice. FASEB J 2025; 39:e70321. [PMID: 39853792 PMCID: PMC11760663 DOI: 10.1096/fj.202402421r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 12/13/2024] [Accepted: 01/06/2025] [Indexed: 01/26/2025]
Abstract
This study, in vivo and in vitro, investigated the role of brain-derived neurotrophic factor (BDNF) in skeletal muscle adaptations to aerobic exercise. BDNF is a contraction-induced protein that may play a role in muscle adaptations to aerobic exercise. BDNF is involved in muscle repair, increased fat oxidation, and mitochondrial biogenesis, all of which are adaptations observed with aerobic training. The purpose of this study was two-pronged and investigated the skeletal muscle BDNF response to (1) acute and (2) chronic exercise in male C57BL/6J mice. It also examined if chronic BDNF treatment resulted in similar adaptations to chronic exercise. In aim 1, mice underwent a 2 hr. treadmill exercise bout. In aim 2, mice were assigned to one of four groups: (1) control (CON); (2) endurance training (ET; treadmill running 1 h/day, 5 days/wk); (3) BDNF (BDNF; 0.5 mg/kg·bw, 5 days/wk); (4) endurance training and BDNF (ET + BDNF) for 8 weeks. Results demonstrated that the soleus (SOL) had higher BDNF content compared with the extensor digitorum longus (EDL) and that SOL BDNF increased with acute exercise. After chronic exercise and BDNF treatment, treadmill testing to exhaustion demonstrated a main effect of BDNF and ET on increasing exercise capacity. In vitro contractile assessment of the EDL revealed BDNF treatment resulted in similar increases in the max rate of relaxation as ET. EDL force-frequency analysis showed ET + BDNF produced higher force than CON and BDNF, indicating an additive effect. BDNF increased EDL mitochondrial proteins, COXIV, and CS. These results demonstrate that BDNF contributes to muscle adaptations observed with ET.
Collapse
Affiliation(s)
- Alexander D. Brown
- Department of Health Sciences, Faculty of Applied Health SciencesBrock UniversitySt. CatharinesOntarioCanada
| | - Alexander D. Marko
- Department of Health Sciences, Faculty of Applied Health SciencesBrock UniversitySt. CatharinesOntarioCanada
| | - Daniel M. Marko
- Department of Health Sciences, Faculty of Applied Health SciencesBrock UniversitySt. CatharinesOntarioCanada
| | - Bradley J. Baranowski
- Department of Health Sciences, Faculty of Applied Health SciencesBrock UniversitySt. CatharinesOntarioCanada
| | - Sebastian Silvera
- Department of Health Sciences, Faculty of Applied Health SciencesBrock UniversitySt. CatharinesOntarioCanada
| | - Michael S. Finch
- Department of Health Sciences, Faculty of Applied Health SciencesBrock UniversitySt. CatharinesOntarioCanada
| | - Alex J. Yang
- Department of Health Sciences, Faculty of Applied Health SciencesBrock UniversitySt. CatharinesOntarioCanada
| | - Roopan Dhaliwal
- Department of Health Sciences, Faculty of Applied Health SciencesBrock UniversitySt. CatharinesOntarioCanada
| | - Chantal R. Ryan
- Department of Health Sciences, Faculty of Applied Health SciencesBrock UniversitySt. CatharinesOntarioCanada
| | - Brian D. Roy
- Department of KinesiologyFaculty of Applied Health Sciences, Brock UniversitySt. CatharinesOntarioCanada
| | - Val A. Fajardo
- Department of KinesiologyFaculty of Applied Health Sciences, Brock UniversitySt. CatharinesOntarioCanada
| | - Rebecca E. K. MacPherson
- Department of Health Sciences, Faculty of Applied Health SciencesBrock UniversitySt. CatharinesOntarioCanada
- Centre for NeuroscienceBrock UniversitySt. CatharinesOntarioCanada
| |
Collapse
|
3
|
Li X, Xie E, Sun S, Shen J, Ding Y, Wang J, Peng X, Zheng R, Farag MA, Xiao J. Flavonoids for gastrointestinal tract local and associated systemic effects: A review of clinical trials and future perspectives. J Adv Res 2025:S2090-1232(25)00033-5. [PMID: 39798849 DOI: 10.1016/j.jare.2025.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 01/06/2025] [Accepted: 01/06/2025] [Indexed: 01/15/2025] Open
Abstract
BACKGROUND Flavonoids are naturally occurring dietary phytochemicals with significant antioxidant effects aside from several health benefits. People often consume them in combination with other food components. Compiling data establishes a link between bioactive flavonoids and prevention of several diseases in animal models, including cardiovascular diseases, diabetes, gut dysbiosis, and metabolic dysfunction-associated steatotic liver disease (MASLD). However, numerous clinical studies have demonstrated the ineffectiveness of flavonoids contradicting rodent models, thereby challenging the validity of using flavonoids as dietary supplements. AIM OF REVIEW This review provides a clinical perspective to emphasize the effective roles of dietary flavonoids as well as to summarize their specific mechanisms in animals briefly. KEY SCIENTIFIC CONCEPTS OF REVIEW First, this review offers an in-depth elucidation of the metabolic processes of flavonoids within human, encompassing the small, large intestine, and the liver. Furthermore, the review provides a comprehensive overview of the various functions of flavonoids in the gastrointestinal tract, including hindering the breakdown and assimilation of macronutrients, such as polysaccharides and lipids, regulating gut hormone secretion as well as inhibition of mineral iron absorption. In the large intestine, an unabsorbed major portion of flavonoids interact with the gut flora leading to their biotransformation. Once absorbed and circulated in the bloodstream, bioactive flavonoids or their metabolites exert numerous beneficial systemic effects. Lastly, we examine the protective effects of flavonoids in several metabolic disorders, including endothelial dysfunction, MASLD, cardiovascular disease, obesity, hyperlipidemia, and insulin resistance. In conclusion, this review outlines the safety and future prospects of flavonoids in the field of health, especially in the prevention of metabolic syndrome (MetS).
Collapse
Affiliation(s)
- Xiaopeng Li
- Center of Nutrition and Food Sciences Hunan Agricultural Products Processing Institute Hunan Academy of Agricultural Sciences Changsha China.
| | - Enjun Xie
- School of Public Health Zhejiang University School of Medicine Hangzhou China
| | - Shumin Sun
- School of Public Health Zhejiang University School of Medicine Hangzhou China
| | - Jie Shen
- School of Public Health Zhejiang University School of Medicine Hangzhou China
| | - Yujin Ding
- National Clinical Research Center for Metabolic Diseases Metabolic Syndrome Research Center Department of Metabolism and Endocrinology The Second Xiangya Hospital of Central South University Changsha China
| | - Jiaqi Wang
- Ausnutria Dairy Co., Ltd., Changsha 410200 China
| | - Xiaoyu Peng
- Ausnutria Dairy Co., Ltd., Changsha 410200 China
| | - Ruting Zheng
- Ausnutria Dairy Co., Ltd., Changsha 410200 China
| | - Mohamed A Farag
- Pharmacognosy Department, Faculty of Pharmacy, Cairo University, Cairo 11562 Egypt
| | - Jianbo Xiao
- Universidade de Vigo, Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Instituto de Agroecoloxía e Alimentación (IAA) - CITEXVI 36310 Vigo, Spain; Research Group on Food, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Isabel Torres 21 39011 Santander, Spain.
| |
Collapse
|
4
|
Pang BPS, Iu ECY, Hang M, Chan WS, Tse MCL, Yeung CTY, Wang M, Siu PMF, Lee CW, Ye K, So H, Chan CB. Deficiency of muscle-generated brain-derived neurotrophic factor causes inflammatory myopathy through reactive oxygen species-mediated necroptosis and pyroptosis. Redox Biol 2024; 78:103418. [PMID: 39531828 PMCID: PMC11602578 DOI: 10.1016/j.redox.2024.103418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 10/21/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024] Open
Abstract
Idiopathic inflammatory myopathy (commonly known as myositis) is a group of immune-related diseases characterized by muscle damage, weakness, and fatigue with unknown causes. Although overactivated innate immunity is a widely believed cause of myositis onset, the mechanism that provokes and maintains a high immune response in myositis patients is still unclear. This study aims to test if brain-derived neurotrophic factor (BDNF) deficiency per se is sufficient to cause myositis and determine its underlying mechanism. We found that ablating BDNF production in skeletal muscle is sufficient to trigger myositis development in mice. Muscle-specific Bdnf knockout (MBKO) mice displayed extensive myocyte necrosis, mononuclear cell infiltration, and myophagocytosis. In association with these damages, elevated production of pro-inflammatory cytokines such as interleukin (IL) 23, IL-1β, IL-18, and tumor necrosis factor α (TNFα) was found in the muscle of MBKO mice. Disruption of sarcolemma integrity was also detected in MBKO mice, which is a result of necroptosis executioner Mixed lineage kinase domain-like protein (MLKL) and pyroptosis executioner Gasdermin D (GSDMD) activation. Mechanistically, diminishing BDNF synthesis in myotubes enhances the accumulation of mitochondrial reactive oxygen species (mtROS), which sensitizes the cells towards TNFα-induced receptor-interacting protein kinase (RIPs) activation and promotes the formation of NLR family pyrin domain containing 3 (NLRP3)-containing inflammasome. BDNF deficiency-induced cell death could be alleviated by scavenging mtROS, suppressing the activity of GSDMD, or inhibiting receptor-interacting kinase 3 (RIP3). Similarly, supplementation of BDNF mimetics, suppression of RIP3 activity, increasing the intramyocellular antioxidant, or enhancing mitophagy ameliorated the myopathies of MBKO mice and improved their muscle strength. Together, our study demonstrates that insufficient BDNF production in mouse muscle causes the development of pathological features of myositis via enhancing oxidative stress, necroptosis, and pyroptosis in myofibers.
Collapse
Affiliation(s)
- Brian Pak Shing Pang
- School of Biological Sciences, Faculty of Science, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Elsie Chit Yu Iu
- School of Biological Sciences, Faculty of Science, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Miaojia Hang
- School of Biological Sciences, Faculty of Science, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Wing Suen Chan
- School of Biological Sciences, Faculty of Science, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Margaret Chui Ling Tse
- School of Biological Sciences, Faculty of Science, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Connie Tsz Ying Yeung
- School of Biological Sciences, Faculty of Science, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Mingfu Wang
- Shenzhen Key Laboratory of Food Nutrition and Health, Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, Hong Kong Special Administrative Region
| | - Parco Ming Fai Siu
- Division of Kinesiology, School of Public Health, The University of Hong Kong, Pok Fu Lam, Hong Kong Special Administrative Region
| | - Chi Wai Lee
- Department of Biology, Hong Kong Baptist University, Hong Kong Special Administrative Region
| | - Keqiang Ye
- Faculty of Life and Health Sciences, and Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Hong Kong Special Administrative Region
| | - Ho So
- Department of Medicine & Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Chi Bun Chan
- School of Biological Sciences, Faculty of Science, The University of Hong Kong, Hong Kong Special Administrative Region; State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong Special Administrative Region.
| |
Collapse
|
5
|
Martins LA, Schiavo A, Paz LV, Xavier LL, Mestriner RG. Neural underpinnings of fine motor skills under stress and anxiety: A review. Physiol Behav 2024; 282:114593. [PMID: 38782244 DOI: 10.1016/j.physbeh.2024.114593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 05/17/2024] [Accepted: 05/21/2024] [Indexed: 05/25/2024]
Abstract
This review offers a comprehensive examination of how stress and anxiety affect motor behavior, particularly focusing on fine motor skills and gait adaptability. We explore the role of several neurochemicals, including brain-derived neurotrophic factor (BDNF) and dopamine, in modulating neural plasticity and motor control under these affective states. The review highlights the importance of developing therapeutic strategies that enhance motor performance by leveraging the interactions between key neurochemicals. Additionally, we investigate the complex interplay between emotional-cognitive states and sensorimotor behaviors, showing how stress and anxiety disrupt neural integration, leading to impairments in skilled movements and negatively impacting quality of life. Synthesizing evidence from human and rodent studies, we provide a detailed understanding of the relationships among stress, anxiety, and motor behavior. Our findings reveal neurophysiological pathways, behavioral outcomes, and potential therapeutic targets, emphasizing the intricate connections between neurobiological mechanisms, environmental factors, and motor performance.
Collapse
Affiliation(s)
- Lucas Athaydes Martins
- Pontifical Catholic University of Rio Grande do Sul (PUCRS). Graduate Program in Biomedical Gerontology, Av. Ipiranga, 6681, Porto Alegre, Brazil; Pontifical Catholic University of Rio Grande do Sul (PUCRS). Neuroscience, Motor Behavior, and Rehabilitation Research Group (NECORE-CNPq), Av. Ipiranga, 6681, Porto Alegre, Brazil
| | - Aniuska Schiavo
- Pontifical Catholic University of Rio Grande do Sul (PUCRS). Graduate Program in Biomedical Gerontology, Av. Ipiranga, 6681, Porto Alegre, Brazil; Pontifical Catholic University of Rio Grande do Sul (PUCRS). Neuroscience, Motor Behavior, and Rehabilitation Research Group (NECORE-CNPq), Av. Ipiranga, 6681, Porto Alegre, Brazil
| | - Lisiê Valéria Paz
- Pontifical Catholic University of Rio Grande do Sul (PUCRS). Graduate Program in Cellular and Molecular Biology, Av. Ipiranga, 6681, Porto Alegre, Brazil
| | - Léder Leal Xavier
- Pontifical Catholic University of Rio Grande do Sul (PUCRS). Neuroscience, Motor Behavior, and Rehabilitation Research Group (NECORE-CNPq), Av. Ipiranga, 6681, Porto Alegre, Brazil; Pontifical Catholic University of Rio Grande do Sul (PUCRS). Graduate Program in Cellular and Molecular Biology, Av. Ipiranga, 6681, Porto Alegre, Brazil
| | - Régis Gemerasca Mestriner
- Pontifical Catholic University of Rio Grande do Sul (PUCRS). Graduate Program in Biomedical Gerontology, Av. Ipiranga, 6681, Porto Alegre, Brazil; Pontifical Catholic University of Rio Grande do Sul (PUCRS). Neuroscience, Motor Behavior, and Rehabilitation Research Group (NECORE-CNPq), Av. Ipiranga, 6681, Porto Alegre, Brazil; Pontifical Catholic University of Rio Grande do Sul (PUCRS). Graduate Program in Cellular and Molecular Biology, Av. Ipiranga, 6681, Porto Alegre, Brazil.
| |
Collapse
|
6
|
Hemasree GNS, Satish KS, Rajalekshmi SG, Burri RR, Murthy TPK. Exploration of interaction interface of TRKβ/BDNF through fingerprint analysis to disinter potential agonists. Mol Divers 2024; 28:1531-1549. [PMID: 37389778 DOI: 10.1007/s11030-023-10673-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 06/12/2023] [Indexed: 07/01/2023]
Abstract
Tyrosine Kinase beta (TRKβ), is a type I membrane receptor which plays a major role in various signalling pathways. TRKβ was found to be upregulated in various cancers and contrastingly downregulated in various neurodegenerative disorders. Hitherto, contemporary drug research is oriented towards discovery of TRKβ inhibitors, thus neglecting the development of TRKβ agonists. This research is aimed at identifying FDA approved drugs exhibiting repurposable potential as TRKβ agonists by mapping them with fingerprints of the BDNF/TRKβ interaction interface. Initially, crucial interacting residues were retrieved and a receptor grid was generated around it. TRKβ agonists were retrieved from literature search and a drug library was created for each agonist based on its structural and side effect similarities. Subsequently, molecular docking and dynamics were performed for each library to identify the drugs possessing affinity towards the binding pocket of TRKβ. The study revealed molecular interactions of Perospirone, Droperidol, Urapidil, and Clobenzorex with the crucial amino acids lining the active binding pocket of TRKβ. Subsequent network pharmacological analysis of the above drugs revealed their interactions with key proteins involved in neurotransmitter signalling pathways. Clobenzorex displayed high stability in dynamics simulation and therefore this drug is recommended for further experimental evaluations to attain better mechanistic insights and predict its implications in correcting neuropathological aberrations. This study's focus on the interaction interface between TRKβ and BDNF, combined with the utilization of fingerprint analysis for drug repurposing, contributes to our understanding of neurotrophic signalling and holds potential for identifying new therapeutic options for neurological disorders.
Collapse
Affiliation(s)
- G N S Hemasree
- Faculty of Pharmacy, M.S.Ramaiah University of Applied Sciences, Bangalore, Karnataka, 560054, India
| | - Kshreeraja S Satish
- Department of Pharmacy Practice, Faculty of Pharmacy, M.S.Ramaiah University of Applied Sciences, Bangalore, Karnataka, 560054, India
| | - Saraswathy Ganesan Rajalekshmi
- Department of Pharmacy Practice, Faculty of Pharmacy, M.S.Ramaiah University of Applied Sciences, Bangalore, Karnataka, 560054, India.
- Pharmacological Modelling and Simulation Centre, Faculty of Pharmacy, M.S.Ramaiah University of Applied Sciences, Bangalore, Karnataka, 560054, India.
| | | | - T P Krishna Murthy
- Department of Biotechnology, M S Ramaiah Institute of Technology, Bangalore, Karnataka, 560054, India
| |
Collapse
|
7
|
Zhang MR, Zuo BY, Song YC, Guo DD, Li QL, Lyu JX, Zhu H, Zhao J, Hang PZ. BDNF mimetics recover palmitic acid-induced injury in cardiomyocytes by ameliorating Akt-dependent mitochondrial impairments. Toxicol Appl Pharmacol 2024; 486:116951. [PMID: 38705401 DOI: 10.1016/j.taap.2024.116951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 04/21/2024] [Accepted: 05/02/2024] [Indexed: 05/07/2024]
Abstract
Cardiac lipotoxicity is a prevalent consequence of lipid metabolism disorders occurring in cardiomyocytes, which in turn precipitates the onset of heart failure. Mimetics of brain-derived neurotrophic factor (BDNF), such as 7,8-dihydroxyflavone (DHF) and 7,8,3'-trihydroxyflavone (THF), have demonstrated significant cardioprotective effects. However, it remains unclear whether these mimetics can protect cardiomyocytes against lipotoxicity. The aim of this study was to examine the impact of DHF and THF on the lipotoxic effects induced by palmitic acid (PA), as well as the concurrent mitochondrial dysfunction. H9c2 cells were subjected to treatment with PA alone or in conjunction with DHF or THF. Various factors such as cell viability, lactate dehydrogenase (LDH) release, death ratio, and mitochondrial function including mitochondrial membrane potential (MMP), mitochondrial-derived reactive oxygen species (mito-SOX) production, and mitochondrial respiration were assessed. PA dose-dependently reduced cell viability, which was restored by DHF or THF. Additionally, both DHF and THF decreased LDH content, death ratio, and mito-SOX production, while increasing MMP and regulating mitochondrial oxidative phosphorylation in cardiomyocytes. Moreover, DHF and THF specifically activated Akt signaling. The protective effects of DHF and THF were abolished when an Akt inhibitor was used. In conclusion, BDNF mimetics attenuate PA-induced injury in cardiomyocytes by alleviating mitochondrial impairments through the activation of Akt signaling.
Collapse
Affiliation(s)
- Man-Ru Zhang
- Department of Pharmacy, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China; College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Bang-Yun Zuo
- Medical College, Yangzhou University, Yangzhou 225009, China
| | - Yu-Chen Song
- Department of Pharmacy, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China; Medical College, Yangzhou University, Yangzhou 225009, China
| | - Dan-Dan Guo
- Department of Pharmacy, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China; Medical College, Yangzhou University, Yangzhou 225009, China
| | - Qing-Liu Li
- Department of Pharmacy, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China; Medical College, Yangzhou University, Yangzhou 225009, China
| | - Jin-Xiu Lyu
- Department of Pharmacy, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Hua Zhu
- Department of Pharmacy, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China.
| | - Jing Zhao
- Department of Pharmacy, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China.
| | - Peng-Zhou Hang
- Department of Pharmacy, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China.
| |
Collapse
|
8
|
Zhou N, Gong L, Zhang E, Wang X. Exploring exercise-driven exerkines: unraveling the regulation of metabolism and inflammation. PeerJ 2024; 12:e17267. [PMID: 38699186 PMCID: PMC11064867 DOI: 10.7717/peerj.17267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 03/28/2024] [Indexed: 05/05/2024] Open
Abstract
Exercise has many beneficial effects that provide health and metabolic benefits. Signaling molecules are released from organs and tissues in response to exercise stimuli and are widely termed exerkines, which exert influence on a multitude of intricate multi-tissue processes, such as muscle, adipose tissue, pancreas, liver, cardiovascular tissue, kidney, and bone. For the metabolic effect, exerkines regulate the metabolic homeostasis of organisms by increasing glucose uptake and improving fat synthesis. For the anti-inflammatory effect, exerkines positively influence various chronic inflammation-related diseases, such as type 2 diabetes and atherosclerosis. This review highlights the prospective contribution of exerkines in regulating metabolism, augmenting the anti-inflammatory effects, and providing additional advantages associated with exercise. Moreover, a comprehensive overview and analysis of recent advancements are provided in this review, in addition to predicting future applications used as a potential biomarker or therapeutic target to benefit patients with chronic diseases.
Collapse
Affiliation(s)
- Nihong Zhou
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing, China
- School of Sport Science, Beijing Sport University, Beijing, China
| | - Lijing Gong
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing, China
- Key Laboratory for Performance Training & Recovery of General Administration of Sport, Beijing Sport University, Beijing, China
| | - Enming Zhang
- Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Lund University, Malmö, Sweden
- NanoLund Center for NanoScience, Lund University, Lund, Sweden
| | - Xintang Wang
- Key Laboratory for Performance Training & Recovery of General Administration of Sport, Beijing Sport University, Beijing, China
- China Institute of Sport and Health Science, Beijing Sport University, Beijing, China
| |
Collapse
|
9
|
Chan WS, Ng CF, Pang BPS, Hang M, Tse MCL, Iu ECY, Ooi XC, Yang X, Kim JK, Lee CW, Chan CB. Exercise-induced BDNF promotes PPARδ-dependent reprogramming of lipid metabolism in skeletal muscle during exercise recovery. Sci Signal 2024; 17:eadh2783. [PMID: 38502732 PMCID: PMC11022078 DOI: 10.1126/scisignal.adh2783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 02/29/2024] [Indexed: 03/21/2024]
Abstract
Post-exercise recovery is essential to resolve metabolic perturbations and promote long-term cellular remodeling in response to exercise. Here, we report that muscle-generated brain-derived neurotrophic factor (BDNF) elicits post-exercise recovery and metabolic reprogramming in skeletal muscle. BDNF increased the post-exercise expression of the gene encoding PPARδ (peroxisome proliferator-activated receptor δ), a transcription factor that is a master regulator of lipid metabolism. After exercise, mice with muscle-specific Bdnf knockout (MBKO) exhibited impairments in PPARδ-regulated metabolic gene expression, decreased intramuscular lipid content, reduced β-oxidation, and dysregulated mitochondrial dynamics. Moreover, MBKO mice required a longer period to recover from a bout of exercise and did not show increases in exercise-induced endurance capacity. Feeding naïve mice with the bioavailable BDNF mimetic 7,8-dihydroxyflavone resulted in effects that mimicked exercise-induced adaptations, including improved exercise capacity. Together, our findings reveal that BDNF is an essential myokine for exercise-induced metabolic recovery and remodeling in skeletal muscle.
Collapse
Affiliation(s)
- Wing Suen Chan
- School of Biological Sciences, the University of Hong Kong, 5N10 Kadoorie Biological Sciences Building, Pokfulam Road, Hong Kong, China
| | - Chun Fai Ng
- School of Biological Sciences, the University of Hong Kong, 5N10 Kadoorie Biological Sciences Building, Pokfulam Road, Hong Kong, China
| | - Brian Pak Shing Pang
- School of Biological Sciences, the University of Hong Kong, 5N10 Kadoorie Biological Sciences Building, Pokfulam Road, Hong Kong, China
| | - Miaojia Hang
- School of Biological Sciences, the University of Hong Kong, 5N10 Kadoorie Biological Sciences Building, Pokfulam Road, Hong Kong, China
| | - Margaret Chui Ling Tse
- School of Biological Sciences, the University of Hong Kong, 5N10 Kadoorie Biological Sciences Building, Pokfulam Road, Hong Kong, China
| | - Elsie Chit Yu Iu
- School of Biological Sciences, the University of Hong Kong, 5N10 Kadoorie Biological Sciences Building, Pokfulam Road, Hong Kong, China
| | - Xin Ci Ooi
- School of Biological Sciences, the University of Hong Kong, 5N10 Kadoorie Biological Sciences Building, Pokfulam Road, Hong Kong, China
| | - Xiuying Yang
- Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica of Peking Union Medical College, Beijing 101399, China
| | - Jason K. Kim
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | - Chi Wai Lee
- Department of Biology, Hong Kong Baptist University, Hong Kong, China
| | - Chi Bun Chan
- School of Biological Sciences, the University of Hong Kong, 5N10 Kadoorie Biological Sciences Building, Pokfulam Road, Hong Kong, China
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
10
|
Wang L, Yan Y, Wu L, Peng J. Natural products in non-alcoholic fatty liver disease (NAFLD): Novel lead discovery for drug development. Pharmacol Res 2023; 196:106925. [PMID: 37714392 DOI: 10.1016/j.phrs.2023.106925] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/06/2023] [Accepted: 09/12/2023] [Indexed: 09/17/2023]
Abstract
With changing lifestyles, non-alcoholic fatty liver disease (NAFLD) has become the most prevalent liver disease worldwide. A substantial increase in the incidence, mortality, and associated burden of NAFLD-related advanced liver disease is expected. Currently, the initial diagnosis of NAFLD is still based on ultrasound and there is no approved treatment method. Lipid-lowering drugs, vitamin supplementation, and lifestyle improvement treatments are commonly used in clinical practice. However, most lipid-lowering drugs can produce poor patient compliance and specific adverse effects. Therefore, the exploration of bio-diagnostic markers and active lead compounds for the development of innovative drugs is urgently needed. More and more studies have reported the anti-NAFLD effects and mechanisms of natural products (NPs), which have become an important source for new drug development to treat NAFLD due to their high activity and low side effects. At present, berberine and silymarin have been approved by the US FDA to enter clinical phase IV studies, demonstrating the potential of NPs against NAFLD. Studies have found that the regulation of lipid metabolism, insulin resistance, oxidative stress, and inflammation-related pathways may play important roles in the process. With the continuous updating of technical means and scientific theories, in-depth research on the targets and mechanisms of NPs against NAFLD can provide new possibilities to find bio-diagnostic markers and innovative drugs. As we know, FXR agonists, PPARα agonists, and dual CCR2/5 inhibitors are gradually coming on stage for the treatment of NAFLD. Whether NPs can exert anti-NAFLD effects by regulating these targets or some unknown targets remains to be further studied. Therefore, the study reviewed the potential anti-NAFLD NPs and their targets. Some works on the discovery of new targets and the docking of active lead compounds were also discussed. It is hoped that this review can provide some reference values for the development of non-invasive diagnostic markers and new drugs against NAFLD in the clinic.
Collapse
Affiliation(s)
- Lu Wang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Yonghuan Yan
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Linfang Wu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Jinyong Peng
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China.
| |
Collapse
|
11
|
Swain M, Soman SK, Tapia K, Dagda RY, Dagda RK. Brain-derived neurotrophic factor protects neurons by stimulating mitochondrial function through protein kinase A. J Neurochem 2023; 167:104-125. [PMID: 37688457 PMCID: PMC10543477 DOI: 10.1111/jnc.15945] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 07/28/2023] [Accepted: 08/01/2023] [Indexed: 09/10/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) stimulates dendrite outgrowth and synaptic plasticity by activating downstream protein kinase A (PKA) signaling. Recently, BDNF has been shown to modulate mitochondrial respiration in isolated brain mitochondria, suggesting that BDNF can modulate mitochondrial physiology. However, the molecular mechanisms by which BDNF stimulates mitochondrial function in neurons remain to be elucidated. In this study, we surmised that BDNF binds to the TrkB receptor and translocates to mitochondria to govern mitochondrial physiology in a PKA-dependent manner. Confocal microscopy and biochemical subcellular fractionation assays confirm the localization of the TrkB receptor in mitochondria. The translocation of the TrkB receptor to mitochondria was significantly enhanced upon treating primary cortical neurons with exogenous BDNF, leading to rapid PKA activation. Showing a direct role of BDNF in regulating mitochondrial structure/function, time-lapse confocal microscopy in primary cortical neurons showed that exogenous BDNF enhances mitochondrial fusion, anterograde mitochondrial trafficking, and mitochondrial content within dendrites, which led to increased basal and ATP-linked mitochondrial respiration and glycolysis as assessed by an XF24e metabolic analyzer. BDNF-mediated regulation of mitochondrial structure/function requires PKA activity as treating primary cortical neurons with a pharmacological inhibitor of PKA or transiently expressing constructs that target an inhibitor peptide of PKA (PKI) to the mitochondrion abrogated BDNF-mediated mitochondrial fusion and trafficking. Mechanistically, western/Phos-tag blots show that BDNF stimulates PKA-mediated phosphorylation of Drp1 and Miro-2 to promote mitochondrial fusion and elevate mitochondrial content in dendrites, respectively. Effects of BDNF on mitochondrial function were associated with increased resistance of neurons to oxidative stress and dendrite retraction induced by rotenone. Overall, this study revealed new mechanisms of BDNF-mediated neuroprotection, which entails enhancing mitochondrial health and function of neurons.
Collapse
Affiliation(s)
- Maryann Swain
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, 1664 North Virginia Street, Nevada, 89557, USA
| | - Smijin K. Soman
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, 1664 North Virginia Street, Nevada, 89557, USA
| | - Kylea Tapia
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, 1664 North Virginia Street, Nevada, 89557, USA
| | - Raul Y. Dagda
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, 1664 North Virginia Street, Nevada, 89557, USA
| | - Ruben K. Dagda
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, 1664 North Virginia Street, Nevada, 89557, USA
| |
Collapse
|
12
|
KIM SUJIN, PARK DONGHO, LEE SANGHYUN, KWAK HYOBUM, KANG JUHEE. Contribution of High-Intensity Interval Exercise in the Fasted State to Fat Browning: Potential Roles of Lactate and β-Hydroxybutyrate. Med Sci Sports Exerc 2023; 55:1160-1171. [PMID: 36790381 PMCID: PMC10242519 DOI: 10.1249/mss.0000000000003136] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
PURPOSE Fat browning contributes to energy consumption and may have metabolic benefits against obesity; however, the potential roles of lactate and β-hydroxybutyrate (β-HB) in fat browning remain unclear. We investigated the roles of a single bout of aerobic exercise that increases lactate and β-HB levels in the fasted state on the regulation of fat browning in rats and humans. METHODS Male Sprague-Dawley rats were exposed to 24-h fasting and/or a single bout moderate-intensity aerobic exercise (40 min): sedentary (CON), exercise (ND-EX), fasting (FAST), and exercise + fasting (F-EX). Adult men ( n = 13) were randomly assigned into control with food intake (CON), exercise with intensity at onset of blood lactate accumulation in the fasted state (F-OBLA), and high-intensity interval exercise in the fasted state (F-HIIE) until each participant expended 350 kcal of energy. For evaluating the effects of exercise intensity in rats, we conducted another set of animal experiment, including groups of sedentary fed control, fasting control, and exercise with moderate-intensity or HIIE for 40 min after a 24-h fasting. RESULTS Regardless of fasting, single bout of exercise increases the concentration of lactate and β-HB in rats, but the exercise in the fasted state increases the β-HB level more significantly in rats and humans. F-EX-activated fat browning (AMPK-SirT1-PGC1α pathway and PRDM16) and thermogenic factor (UCP1) in white fat of rats. In rats and humans, exercise in the fasted state increased the blood levels of fat browning-related adipomyokines. In particular, compared with F-OBLA, F-HIIE more efficiently increases free fatty acid as well as blood levels of fat browning adipomyokines in humans, which was correlated with blood levels of lactate and β-HB. In rats that performed exercise with different intensity, the higher plasma lactate and β-HB levels, and higher expression of p-AMPK, UCP1, and PRDM16 in white adipose tissue of HIIE group than those of moderate-intensity group, were observed. CONCLUSIONS A single bout of aerobic exercise in the fasted state significantly induced fat browning-related pathways, free fatty acid, and adipomyokines, particularly F-HIIE in human. Although further evidence for supporting our results is required in humans, aerobic exercise in the fasted state with high intensity that increase lactate and β-HB may be a modality of fat browning.
Collapse
Affiliation(s)
- SUJIN KIM
- Department of Pharmacology and Research Center for Controlling Intercellular Communication, College of Medicine, Inha University, Incheon, REPUBLIC OF KOREA
| | - DONG-HO PARK
- Department of Kinesiology, Inha University, Incheon, REPUBLIC OF KOREA
- Program in Biomedical Science and Engineering, Inha University, Incheon, REPUBLIC OF KOREA
| | - SANG-HYUN LEE
- Department of Kinesiology, Inha University, Incheon, REPUBLIC OF KOREA
| | - HYO-BUM KWAK
- Department of Kinesiology, Inha University, Incheon, REPUBLIC OF KOREA
- Program in Biomedical Science and Engineering, Inha University, Incheon, REPUBLIC OF KOREA
| | - JU-HEE KANG
- Department of Pharmacology and Research Center for Controlling Intercellular Communication, College of Medicine, Inha University, Incheon, REPUBLIC OF KOREA
- Program in Biomedical Science and Engineering, Inha University, Incheon, REPUBLIC OF KOREA
| |
Collapse
|
13
|
Hang PZ, Ge FQ, Zhang MR, Li QH, Yu HQ, Song YC, Guo DD, Zhao J, Zhu H. BDNF mimetic 7,8-dihydroxyflavone rescues rotenone-induced cytotoxicity in cardiomyocytes by ameliorating mitochondrial dysfunction. Free Radic Biol Med 2023; 198:83-91. [PMID: 36764626 DOI: 10.1016/j.freeradbiomed.2023.02.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/25/2023] [Accepted: 02/07/2023] [Indexed: 02/11/2023]
Abstract
The relationship between mitochondrial dysfunction and cardiovascular disease pathogenesis is well recognized. 7,8-Dihydroxyflavone (7,8-DHF), a mimetic of brain-derived neurotrophic factor, inhibits mitochondrial impairments and improves cardiac function. However, the regulatory role of 7,8-DHF in the mitochondrial function of cardiomyocytes is not fully understood. To investigate the potential mito-protective effects of 7,8-DHF in cardiomyocytes, we treated H9c2 or HL-1 cells with the mitochondrial respiratory complex I inhibitor rotenone (Rot) as an in vitro model of mitochondrial dysfunction. We found that 7,8-DHF effectively eliminated various concentrations of Rot-induced cell death and reduced lactate dehydrogenase release. 7,8-DHF significantly improved mitochondrial membrane potential and inhibited mitochondrial reactive oxygen species. Moreover, 7,8-DHF decreased routine and leak respiration, restored protein levels of mitochondrial complex I-IV, and increased ATP production in Rot-treated H9c2 cells. The protective role of 7,8-DHF in Rot-induced damage was validated in HL-1 cells. Nuclear phosphorylation protein expression of signal transducer and activator of transcription 3 (STAT3) was significantly increased by 7,8-DHF. The present study suggests that 7,8-DHF rescues Rot-induced cytotoxicity by inhibiting mitochondrial dysfunction and promoting nuclear translocation of p-STAT3 in cardiomyocytes, thus nominating 7,8-DHF as a new pharmacological candidate agent against mitochondrial dysfunction in cardiac diseases.
Collapse
Affiliation(s)
- Peng-Zhou Hang
- Department of Pharmacy, Clinical Medical College, Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, 225001, China
| | - Feng-Qin Ge
- Department of Pharmacy, Clinical Medical College, Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, 225001, China; Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Man-Ru Zhang
- Department of Pharmacy, Clinical Medical College, Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, 225001, China; College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Qi-Hang Li
- Department of Pharmacy, Clinical Medical College, Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, 225001, China; Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Hua-Qing Yu
- Department of Pharmacy, Clinical Medical College, Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, 225001, China; College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Yu-Chen Song
- Department of Pharmacy, Clinical Medical College, Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, 225001, China; Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Dan-Dan Guo
- Department of Pharmacy, Clinical Medical College, Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, 225001, China; Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Jing Zhao
- Department of Pharmacy, Clinical Medical College, Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, 225001, China.
| | - Hua Zhu
- Department of Pharmacy, Clinical Medical College, Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, 225001, China.
| |
Collapse
|
14
|
7,8-Dihydroxyflavone Attenuates Inflammatory Response and Insulin Resistance Induced by the Paracrine Interaction between Adipocytes and Macrophages. Int J Mol Sci 2023; 24:ijms24043520. [PMID: 36834930 PMCID: PMC9961847 DOI: 10.3390/ijms24043520] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023] Open
Abstract
Obesity-induced inflammation and insulin resistance are mediated by macrophage infiltration into adipose tissue. We investigated the effects of 7,8-dihydroxyflavone (7,8-DHF), a flavone found in plants, on the inflammatory response and insulin resistance induced by the interaction between adipocytes and macrophages. Hypertrophied 3T3-L1 adipocytes were cocultured with RAW 264.7 macrophages and treated with 7,8-DHF (3.12, 12.5, and 50 μM). The inflammatory cytokines and free fatty acid (FFA) release were evaluated by assay kits, and signaling pathways were determined by immunoblotting. Coculture of adipocytes and macrophages increased inflammatory mediators, such as nitric oxide (NO), monocyte chemoattractant protein-1 (MCP-1), tumor necrosis factor-alpha (TNF-α), and interleukin-6 (IL-6) and FFA secretion but suppressed the production of anti-inflammatory adiponectin. 7,8-DHF counteracted the coculture-induced changes (p < 0.001). 7,8-DHF also inhibited c-Jun N-terminal kinase (JNK) activation and blocked nuclear factor kappa B (NF-κB) nuclear translocation in the coculture system (p < 0.01). In addition, adipocytes cocultured with macrophages did not increase glucose uptake and Akt phosphorylation in response to insulin. However, 7,8-DHF treatment recovered the impaired responsiveness to insulin (p < 0.01). These findings show that 7,8-DHF alleviates inflammation and adipocyte dysfunction in the coculture of hypertrophied 3T3-L1 adipocytes and RAW 264.7 macrophages, indicating its potential as a therapeutic agent for obesity-induced insulin resistance.
Collapse
|
15
|
Yang HM, Kim J, Shin D, Kim JY, You J, Lee HC, Jang HD, Kim HS. Resistin impairs mitochondrial homeostasis via cyclase-associated protein 1-mediated fission, leading to obesity-induced metabolic diseases. Metabolism 2023; 138:155343. [PMID: 36356648 DOI: 10.1016/j.metabol.2022.155343] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 10/15/2022] [Accepted: 10/29/2022] [Indexed: 11/09/2022]
Abstract
OBJECTIVE One of the suggested mechanisms of obesity-induced insulin resistance is mitochondrial dysfunction in target tissues such as skeletal muscle. In our study, we examined whether resistin, an adipokine associated with obesity-mediated insulin resistance, induced metabolic disorders by impairing mitochondrial homeostasis. METHODS The morphology and function of mitochondria of skeletal muscle were examined in resistin-knockout and humanized resistin mice that were subjected to high-fat diet for 3 months. Morphology was examined by transmission electron microscopy. Mitochondria bioenergetics of skeletal muscle were evaluated using a Seahorse XF96 analyzer. Human skeletal myoblasts were used for in vitro studies on signaling mechanisms in responses to resistin. RESULTS A high-fat diet in humanized resistin mice increased fragmented and shorter mitochondria in the skeletal muscle, whereas resistin-knockout mice had healthy normal mitochondria. In vitro studies showed that human resistin treatment impaired mitochondrial homeostasis by inducing mitochondrial fission, leading to a decrease in ATP production and mitochondrial dysfunction. Induction of mitochondrial fission by resistin was accompanied by increased formation of mitochondria-associated ER membranes (MAM). At the same time, resistin induced up-regulation of the protein kinase A (PKA) pathway. This activation of PKA induced phosphorylation of Drp1 at serine 616, leading to Drp1 activation and subsequent induction of mitochondrial fission. The key molecule that mediated human resistin-induced mitochondrial fission was adenylyl cyclase-associated protein 1 (CAP1), which was reported as a bona fide receptor for human resistin. Moreover, our newly developed biomimetic selective blocking peptide could repress human resistin-mediated mitochondrial dysfunction. High-fat diet-fed mice showed lower exercise capacity and higher insulin resistance, which was prevented by a novel peptide to block the binding of resistin to CAP1 or in the CAP1-knockdown mice. CONCLUSIONS Our study demonstrated that human resistin induces mitochondrial dysfunction by inducing abnormal mitochondrial fission. This result suggests that the resistin-CAP1 complex could be a potential therapeutic target for the treatment of obesity-related metabolic diseases such as diabetes and cardiometabolic diseases.
Collapse
Affiliation(s)
- Han-Mo Yang
- Strategic Center of Cell & Bio Therapy, Seoul National University Hospital, Seoul, Republic of Korea; Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Joonoh Kim
- Strategic Center of Cell & Bio Therapy, Seoul National University Hospital, Seoul, Republic of Korea; Department of Molecular Medicine & Biopharmaceutical Science, Graduate School of Convergence Science and Technology and College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Dasom Shin
- Strategic Center of Cell & Bio Therapy, Seoul National University Hospital, Seoul, Republic of Korea; Department of Molecular Medicine & Biopharmaceutical Science, Graduate School of Convergence Science and Technology and College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Ju-Young Kim
- Strategic Center of Cell & Bio Therapy, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jihye You
- Strategic Center of Cell & Bio Therapy, Seoul National University Hospital, Seoul, Republic of Korea; Department of Molecular Medicine & Biopharmaceutical Science, Graduate School of Convergence Science and Technology and College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Hyun-Chae Lee
- Strategic Center of Cell & Bio Therapy, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hyun-Duk Jang
- Strategic Center of Cell & Bio Therapy, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hyo-Soo Kim
- Strategic Center of Cell & Bio Therapy, Seoul National University Hospital, Seoul, Republic of Korea; Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea; Department of Molecular Medicine & Biopharmaceutical Science, Graduate School of Convergence Science and Technology and College of Medicine, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
16
|
Roh E, Hwang SY, Song E, Park MJ, Yoo HJ, Baik SH, Kim M, Won CW, Choi KM. Association of plasma brain-derived neurotrophic factor levels and frailty in community-dwelling older adults. Sci Rep 2022; 12:18605. [PMID: 36329115 PMCID: PMC9633836 DOI: 10.1038/s41598-022-19706-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 09/02/2022] [Indexed: 11/06/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF), an exercise-induced neurotrophin, is an important factor in memory consolidation and cognitive function. This study evaluates the association between plasma BDNF levels and frailty in community-dwelling older adults. Plasma BDNF levels were analyzed in a total of 302 individuals aged 70-84 years from the Korean Frailty and Aging Cohort Study. There were 30 (9.9%) participants with frailty. They were older and had a higher prevalence of dementia and depression than those without frailty. There were no differences in the proportion of male sex between the frail and non-frail groups. Plasma BDNF levels were significantly lower in participants with frailty than in those without frailty. The presence of frailty was significantly associated with plasma BDNF levels (odds ratio 0.508, 95% confidence interval 0.304-0.849) as well as age, hemoglobin, and the presence of dementia, and depression. After adjustment for confounding factors, the significant association between plasma BDNF and frailty was maintained (0.495, 0.281-0.874). This association remained consistent after exclusion of individuals with dementia, depression, stroke, diabetes, and osteoporosis. Plasma BDNF levels were significantly associated with frailty in community-dwelling older adults. Our study may suggest the possible role of BDNF as a novel biomarker of frailty.
Collapse
Affiliation(s)
- Eun Roh
- grid.488421.30000000404154154Division of Endocrinology and Metabolism, Department of Internal Medicine, Hallym University Sacred Heart Hospital, Anyang, 14068 Republic of Korea
| | - Soon Young Hwang
- grid.222754.40000 0001 0840 2678Department of Biostatistics, Korea University College of Medicine, Seoul, 08308 Republic of Korea
| | - Eyun Song
- grid.411134.20000 0004 0474 0479Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University Guro Hospital, Korea University College of Medicine, 148 Gurodong-ro, Guro-gu, Seoul, 08308 Republic of Korea
| | - Min Jeong Park
- grid.411134.20000 0004 0474 0479Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University Guro Hospital, Korea University College of Medicine, 148 Gurodong-ro, Guro-gu, Seoul, 08308 Republic of Korea
| | - Hye Jin Yoo
- grid.411134.20000 0004 0474 0479Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University Guro Hospital, Korea University College of Medicine, 148 Gurodong-ro, Guro-gu, Seoul, 08308 Republic of Korea
| | - Sei Hyun Baik
- grid.411134.20000 0004 0474 0479Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University Guro Hospital, Korea University College of Medicine, 148 Gurodong-ro, Guro-gu, Seoul, 08308 Republic of Korea
| | - Miji Kim
- grid.289247.20000 0001 2171 7818East-West Medical Research Institute, Kyung Hee University, Seoul, 02447 Republic of Korea
| | - Chang Won Won
- grid.289247.20000 0001 2171 7818Department of Family Medicine, College of Medicine, Kyung Hee University, Seoul, 02447 Republic of Korea
| | - Kyung Mook Choi
- grid.411134.20000 0004 0474 0479Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University Guro Hospital, Korea University College of Medicine, 148 Gurodong-ro, Guro-gu, Seoul, 08308 Republic of Korea
| |
Collapse
|
17
|
Association of serum brain-derived neurotrophic factor with hepatic enzymes, AST/ALT ratio, and FIB-4 index in middle-aged and older women. PLoS One 2022; 17:e0273056. [PMID: 35998179 PMCID: PMC9398011 DOI: 10.1371/journal.pone.0273056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 08/01/2022] [Indexed: 11/19/2022] Open
Abstract
Substantial evidence suggests an important role of liver function in brain health. Liver function is clinically assessed by measuring the activity of hepatic enzymes in the peripheral blood. Brain-derived neurotrophic factor (BDNF) is an important regulator of brain function. Therefore, we hypothesized that blood BDNF levels are associated with liver function and fibrosis. To test this hypothesis, in this cross-sectional study, we investigated whether serum BDNF concentration is associated with liver enzyme activity, aspartate aminotransferase (AST)/ alanine aminotransferase (ALT) ratio, and fibrosis-4 (FIB-4) index in middle-aged and older women. We found that serum BDNF level showed a significant positive association with ALT and γ-glutamyltranspeptidase (GGT) activity and negative association with FIB-4 index, and a trend of negative association with the AST/ALT ratio after adjustment for age. Additionally, these associations remained statistically significant even after adjustment for body mass index (BMI) and fasting blood glucose level. These results demonstrate associations of serum BDNF levels with liver enzymes and hepatic fibrosis-related indices, which may underlie liver-brain interactions.
Collapse
|
18
|
Is Brain-Derived Neurotrophic Factor a Metabolic Hormone in Peripheral Tissues? BIOLOGY 2022; 11:biology11071063. [PMID: 36101441 PMCID: PMC9312804 DOI: 10.3390/biology11071063] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/05/2022] [Accepted: 07/15/2022] [Indexed: 12/06/2022]
Abstract
Simple Summary The activity of brain-derived neurotrophic factor (BDF) in the central nervous system has been well-studied, but its physiological role in other organs has not been clearly defined. This review summarizes the current findings on the functionality of BDNF in various peripheral tissues and discusses several unresolved questions in the field. Abstract Brain-derived neurotrophic factor (BDNF) is an important growth factor in the central nervous system. In addition to its well-known activities in promoting neuronal survival, neuron differentiation, and synaptic plasticity, neuronal BDNF also regulates energy homeostasis by modulating the hypothalamus’s hormonal signals. In the past decades, several peripheral tissues, including liver, skeletal muscle, and white adipose tissue, were demonstrated as the active sources of BDNF synthesis in response to different metabolic challenges. Nevertheless, the functions of BDNF in these tissues remain obscure. With the use of tissue-specific Bdnf knockout animals and the availability of non-peptidyl BDNF mimetic, increasing evidence has reported that peripheral tissues-derived BDNF might play a significant role in maintaining systemic metabolism, possibly through the regulation of mitochondrial dynamics in the various tissues. This article reviews the autocrine/paracrine/endocrine functions of BDNF in non-neuronal tissues and discusses the unresolved questions about BDNF’s function.
Collapse
|
19
|
Sahin E, Saglam N, Erdem S, Alvuroglu E, Abidin I, Yulug E, Alver A. 7,8-Dihydroxyflavone alleviates Endoplasmic Reticulum Stress in cafeteria diet-induced metabolic syndrome. Life Sci 2022; 306:120781. [PMID: 35835252 DOI: 10.1016/j.lfs.2022.120781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 06/29/2022] [Accepted: 07/01/2022] [Indexed: 10/17/2022]
Abstract
AIMS Prolonged Endoplasmic Reticulum Stress (ERS) is involved in the pathogenesis of metabolic syndrome, including type-2 diabetes mellitus, cardiovascular diseases, atherosclerosis, obesity, and fatty liver disease. There have been significant efforts to discover molecules to treat ERS and/or to ameliorate associate symptoms. In this study, we investigated the effect of 7,8-Dihydroxyflavone (7,8-DHF) on ERS in liver and pancreas tissues in a cafeteria (CAF) diet induced metabolic syndrome model. MAIN METHODS Male C57BL/6 mice were fed CAF diet for 16 weeks and 7,8-DHF was administered intraperitoneally (5 mg/kg/day) for last four weeks. 78-kDa glucose-regulated protein (GRP78) and C/EBP homologous protein (CHOP) in liver and pancreas tissues, insulin and interleukin-1β (IL-1β) in serum were analyzed by ELISA method and serum biochemistry parameters were analyzed with autoanalyzer. GRP78 and CHOP gene expression levels were determined by qRT-PCR. In addition, histopathological analyzes were performed on liver and pancreas tissues. KEY FINDINGS Findings revealed that CAF diet caused metabolic abnormalities, insulin resistance and inflammation in serum and triggered ERS in pancreas and liver tissues. 7,8-DHF treatment significantly reduced metabolic abnormalities by reducing serum biochemical parameters, HOMO-IR and IL-1β levels. qRT-PCR and ELISA results indicated that 7,8-DHF treatment down-regulated GRP78 and CHOP expression and protein levels in the liver and GRP78 expression in pancreas. Efficiency of 7,8-DHF in these tissues was also demonstrated by histopathological tests. SIGNIFICANCE In conclusion, CAF diet-induced metabolic syndrome model, 7,8-DHF suppressed ERS and ERS-induced metabolic disorders in both liver and pancreas. Therefore, 7,8-DHF may potentially be a novel therapeutic compound to ameliorate ERS and related metabolic symptoms.
Collapse
Affiliation(s)
- Elif Sahin
- Department of Medical Biochemistry, Graduate School of Medical Science, Karadeniz Technical University, Trabzon, Turkiye.
| | - Neslihan Saglam
- Department of Medical Biochemistry, Graduate School of Medical Science, Karadeniz Technical University, Trabzon, Turkiye
| | - Seniz Erdem
- Department of Medical Biochemistry, Graduate School of Medical Science, Karadeniz Technical University, Trabzon, Turkiye
| | - Elif Alvuroglu
- Department of Histology and Embryology, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkiye
| | - Ismail Abidin
- Department of Biophysics, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkiye
| | - Esin Yulug
- Department of Histology and Embryology, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkiye
| | - Ahmet Alver
- Department of Medical Biochemistry, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkiye
| |
Collapse
|
20
|
Yang S, Zhu G. 7,8-Dihydroxyflavone and Neuropsychiatric Disorders: A Translational Perspective from the Mechanism to Drug Development. Curr Neuropharmacol 2022; 20:1479-1497. [PMID: 34525922 PMCID: PMC9881092 DOI: 10.2174/1570159x19666210915122820] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 08/26/2021] [Accepted: 09/12/2021] [Indexed: 11/22/2022] Open
Abstract
7,8-Dihydroxyflavone (7,8-DHF) is a kind of natural flavonoid with the potential to cross the blood-brain barrier. 7,8-DHF effectively mimics the effect of brain-derived neurotrophic factor (BDNF) in the brain to selectively activate tyrosine kinase receptor B (TrkB) and downstream signaling pathways, thus playing a neuroprotective role. The preclinical effects of 7,8-DHF have been widely investigated in neuropsychiatric disorders, including Alzheimer's disease (AD), Parkinson's disease (PD), depression, and memory impairment. Besides the effect on TrkB, 7,8-DHF could also function through fighting against oxidative stress, cooperating with estrogen receptors, or regulating intestinal flora. This review focuses on the recent experimental studies on depression, neurodegenerative diseases, and learning and memory functions. Additionally, the structural modification and preparation of 7,8-DHF were also concluded and proposed, hoping to provide a reference for the follow-up research and clinical drug development of 7,8-DHF in the field of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Shaojie Yang
- Key Laboratory of Xin’an Medicine, the Ministry of Education, Anhui University of Chinese Medicine, Hefei, Anhui, 230038, China
| | - Guoqi Zhu
- Key Laboratory of Xin’an Medicine, the Ministry of Education, Anhui University of Chinese Medicine, Hefei, Anhui, 230038, China,Address correspondence to this author at the Anhui University of Chinese Medicine, Meishan Road 103, Hefei 230038, China; E-mail:
| |
Collapse
|
21
|
Anti-lipogenic and thermogenic potency of Padina tetrastromatica bioactives in hypertrophied 3T3-L1 cells and their efficacy based thermogenic food supplement to mitigate obesity. ALGAL RES 2022. [DOI: 10.1016/j.algal.2022.102695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
22
|
Li T, Li X, Huang X, Yu H, Li S, Zhang Z, Xie Y, Song X, Liu J, Yang X, Liu G. Mitochondriomics reveals the underlying neuroprotective mechanism of TrkB receptor agonist R13 in the 5×FAD mice. Neuropharmacology 2022; 204:108899. [PMID: 34838815 DOI: 10.1016/j.neuropharm.2021.108899] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 11/20/2021] [Accepted: 11/23/2021] [Indexed: 02/07/2023]
Abstract
Decreased energy metabolism and mitochondrial biogenesis defects are implicated in the pathogenesis of Alzheimer's disease (AD). In present study, mitochondriomics analysis revealed significant effects of R13, a prodrug of 7,8-dihydroxyflavone, on mitochondrial protein expression profile, including the proteins related to the biological processes: fatty acid beta-oxidation, fatty acid metabolic process, mitochondrial electron transport, and mitochondrial respiratory chain. Cluster analysis demonstrated that R13 promoted mitochondrial oxidative phosphorylation (OXPHOS). The functional analysis showed that R13 increased ATP levels and enhanced OXPHOS including complex Ⅰ, Ⅱ, Ⅲ and Ⅳ. R13 treatment increased mitochondrial biogenesis by regulating the levels of p-AMPKα, p-CREB, PGC-1α, NRF1 and TFAM as a consequence of activation of TrkB receptor in the 5 × FAD mice. Finally, R13 significantly reduced the levels of tau phosphorylation and Aβ plaque. Our data suggest that R13 may be used for treating AD via enhancing mitochondrial biogenesis and metabolism.
Collapse
Affiliation(s)
- Ting Li
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao Li
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Medical Key Discipline of Health Toxicology (2020-2024), Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, China; Department of Pathology, Wuhan No. 1 Hospital, Wuhan, 430022, China
| | - Xi Huang
- Department of Neurology,The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, 518020, China; The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Hao Yu
- Shenzhen Polytechnic, Shenzhen, 518055, China
| | - Shupeng Li
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Zaijun Zhang
- Institute of New Drug Research and Guangzhou, Key Laboratory of Innovative Chemical Drug Research in Cardio-Cerebrovascular Diseases, Jinan University College of Pharmacy, Guangzhou, 510632, China
| | - Yongmei Xie
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Xiangrong Song
- Department of Critical Care Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jianjun Liu
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Medical Key Discipline of Health Toxicology (2020-2024), Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, China
| | - Xifei Yang
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Medical Key Discipline of Health Toxicology (2020-2024), Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, China.
| | - Gongping Liu
- Department of Pathology, Wuhan No. 1 Hospital, Wuhan, 430022, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, JS, China.
| |
Collapse
|
23
|
Valenti D, Stagni F, Emili M, Guidi S, Bartesaghi R, Vacca RA. Impaired Brain Mitochondrial Bioenergetics in the Ts65Dn Mouse Model of Down Syndrome Is Restored by Neonatal Treatment with the Polyphenol 7,8-Dihydroxyflavone. Antioxidants (Basel) 2021; 11:antiox11010062. [PMID: 35052567 PMCID: PMC8773005 DOI: 10.3390/antiox11010062] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/22/2021] [Accepted: 12/24/2021] [Indexed: 12/18/2022] Open
Abstract
Down syndrome (DS), a major genetic cause of intellectual disability, is characterized by numerous neurodevelopmental defects. Previous in vitro studies highlighted a relationship between bioenergetic dysfunction and reduced neurogenesis in progenitor cells from the Ts65Dn mouse model of DS, suggesting a critical role of mitochondrial dysfunction in neurodevelopmental alterations in DS. Recent in vivo studies in Ts65Dn mice showed that neonatal supplementation (Days P3–P15) with the polyphenol 7,8-dihydroxyflavone (7,8-DHF) fully restored hippocampal neurogenesis. The current study was aimed to establish whether brain mitochondrial bioenergetic defects are already present in Ts65Dn pups and whether early treatment with 7,8-DHF positively impacts on mitochondrial function. In the brain and cerebellum of P3 and P15 Ts65Dn pups we found a strong impairment in the oxidative phosphorylation apparatus, resulting in a deficit in mitochondrial ATP production and ATP content. Administration of 7,8-DHF (dose: 5 mg/kg/day) during Days P3–P15 fully restored bioenergetic dysfunction in Ts65Dn mice, reduced the levels of oxygen radicals and reinstated the hippocampal levels of PGC-1α. No pharmacotherapy is available for DS. From current findings, 7,8-DHF emerges as a treatment with a good translational potential for improving mitochondrial bioenergetics and, thus, mitochondria-linked neurodevelopmental alterations in DS.
Collapse
Affiliation(s)
- Daniela Valenti
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council (CNR), 70126 Bari, Italy;
- Correspondence: (D.V.); (R.B.)
| | - Fiorenza Stagni
- Department for Life Quality Studies, University of Bologna, 47921 Rimini, Italy;
| | - Marco Emili
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy; (M.E.); (S.G.)
| | - Sandra Guidi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy; (M.E.); (S.G.)
| | - Renata Bartesaghi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy; (M.E.); (S.G.)
- Correspondence: (D.V.); (R.B.)
| | - Rosa Anna Vacca
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council (CNR), 70126 Bari, Italy;
| |
Collapse
|
24
|
Li Y, Cheng Y, Zhou Y, Du H, Zhang C, Zhao Z, Chen Y, Zhou Z, Mei J, Wu W, Chen M. High fat diet-induced obesity leads to depressive and anxiety-like behaviors in mice via AMPK/mTOR-mediated autophagy. Exp Neurol 2021; 348:113949. [PMID: 34902357 DOI: 10.1016/j.expneurol.2021.113949] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 12/02/2021] [Accepted: 12/07/2021] [Indexed: 12/28/2022]
Abstract
Depression is one of the most common mental illnesses in modern society. In recent years, several studies show that there are disturbances in lipid metabolism in depressed patients. High-fat diet may lead to anxiety and depression, but the mechanisms involved remain unclear. In our study, we found that 8 weeks of high-fat feeding effectively induced metabolic disorders, including obesity and hyperlipidemia in mice. Interestingly, the mice also showed depressive and anxiety-like behaviors. We further found activated microglia and astrocyte, increased neuroinflammation, decreased autophagy and BDNF levels in mice after high-fat feeding. Besides, high-fat feeding can also inhibit AMPK phosphorylation and induce mTOR phosphorylation. After treating with the mTOR inhibitor rapamycin, autophagy and BDNF levels were elevated. The number of activated microglia and astrocyte, and pro-inflammation levels were reduced. Besides, rapamycin can also reduce the body weight and serum lipid level in high fat feeding mice. Depressive and anxiety-like behaviors were also ameliorated to some extent after rapamycin treatment. In summary, these results suggest that high-fat diet-induced obesity may lead to depressive and anxiety-like behaviors in mice by inhibiting AMPK phosphorylation and promoting mTOR shift to phosphorylation to inhibit autophagy. Therefore, improving lipid metabolism or enhancing autophagy through the AMPK/mTOR pathway could be potential targets for the treatment of obesity depression.
Collapse
Affiliation(s)
- Yong Li
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Yujie Cheng
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Yuan Zhou
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Hongmei Du
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Cui Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Zhentao Zhao
- Department of Otorhinolaryngology, Head and Neck Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China
| | - Yuenan Chen
- Department of Clinical Pharmacy, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Zhongnan Zhou
- Department of Clinical Pharmacy, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Jinyu Mei
- Department of Otorhinolaryngology, Head and Neck Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China.
| | - Wenning Wu
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China.
| | - Ming Chen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
25
|
Ahuja P, Ng CF, Pang BPS, Chan WS, Tse MCL, Bi X, Kwan HLR, Brobst D, Herlea-Pana O, Yang X, Du G, Saengnipanthkul S, Noh HL, Jiao B, Kim JK, Lee CW, Ye K, Chan CB. Muscle-generated BDNF (brain derived neurotrophic factor) maintains mitochondrial quality control in female mice. Autophagy 2021; 18:1367-1384. [PMID: 34689722 DOI: 10.1080/15548627.2021.1985257] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Mitochondrial remodeling is dysregulated in metabolic diseases but the underlying mechanism is not fully understood. We report here that BDNF (brain derived neurotrophic factor) provokes mitochondrial fission and clearance in skeletal muscle via the PRKAA/AMPK-PINK1-PRKN/Parkin and PRKAA-DNM1L/DRP1-MFF pathways. Depleting Bdnf expression in myotubes reduced fatty acid-induced mitofission and mitophagy, which was associated with mitochondrial elongation and impaired lipid handling. Muscle-specific bdnf knockout (MBKO) mice displayed defective mitofission and mitophagy, and accumulation of dysfunctional mitochondria in the muscle when they were fed with a high-fat diet (HFD). These animals also have exacerbated body weight gain, increased intramyocellular lipid deposition, reduced energy expenditure, poor metabolic flexibility, and more insulin resistance. In contrast, consuming a BDNF mimetic (7,8-dihydroxyflavone) increased mitochondrial content, and enhanced mitofission and mitophagy in the skeletal muscles. Hence, BDNF is an essential myokine to maintain mitochondrial quality and function, and its repression in obesity might contribute to impaired metabolism.Abbreviation: 7,8-DHF: 7,8-dihydroxyflavone; ACACA/ACC: acetyl Coenzyme A carboxylase alpha; ACAD: acyl-Coenzyme A dehydrogenase family; ACADVL: acyl-Coenzyme A dehydrogenase, very long chain; ACOT: acyl-CoA thioesterase; CAMKK2: calcium/calmodulin-dependent protein kinase kinase 2, beta; BDNF: brain derived neurotrophic factor; BNIP3: BCL2/adenovirus E1B interacting protein 3; BNIP3L/NIX: BCL2/adenovirus E1B interacting protein 3-like; CCL2/MCP-1: chemokine (C-C motif) ligand 2; CCL5: chemokine (C-C motif) ligand 5; CNS: central nervous system; CPT1B: carnitine palmitoyltransferase 1b, muscle; Cpt2: carnitine palmitoyltransferase 2; CREB: cAMP responsive element binding protein; DNM1L/DRP1: dynamin 1-like; E2: estrogen; EHHADH: enoyl-CoenzymeA hydratase/3-hydroxyacyl CoenzymeA dehydrogenase; ESR1/ER-alpha: estrogen receptor 1 (alpha); FA: fatty acid; FAO: fatty acid oxidation; FCCP: carbonyl cyanide-4-(trifluoromethoxy)phenylhydrazone; FFA: free fatty acids; FGF21: fibroblast growth factor 21; FUNDC1: FUN14 domain containing 1; HADHA: hydroxyacyl-CoA dehydrogenase trifunctional multienzyme complex subunit alpha; HFD: high-fat diet; iWAT: inguinal white adipose tissues; MAP1LC3A/LC3A: microtubule-associated protein 1 light chain 3 alpha; MBKO; muscle-specific bdnf knockout; IL6/IL-6: interleukin 6; MCEE: methylmalonyl CoA epimerase; MFF: mitochondrial fission factor; NTRK2/TRKB: neurotrophic tyrosine kinase, receptor, type 2; OPTN: optineurin; PA: palmitic acid; PARL: presenilin associated, rhomboid-like; PDH: pyruvate dehydrogenase; PINK1: PTEN induced putative kinase 1; PPARGC1A/PGC-1α: peroxisome proliferative activated receptor, gamma, coactivator 1 alpha; PRKAA/AMPK: protein kinase, AMP-activated, alpha 2 catalytic subunit; ROS: reactive oxygen species; TBK1: TANK-binding kinase 1; TG: triacylglycerides; TNF/TNFα: tumor necrosis factor; TOMM20: translocase of outer mitochondrial membrane 20; ULK1: unc-51 like kinase 1.
Collapse
Affiliation(s)
- Palak Ahuja
- School of Biological Sciences, The University of Hong Kong, Hong Kong, China, Hong Kong
| | - Chun Fai Ng
- School of Biological Sciences, The University of Hong Kong, Hong Kong, China, Hong Kong
| | - Brian Pak Shing Pang
- School of Biological Sciences, The University of Hong Kong, Hong Kong, China, Hong Kong
| | - Wing Suen Chan
- School of Biological Sciences, The University of Hong Kong, Hong Kong, China, Hong Kong
| | - Margaret Chui Ling Tse
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, China, Hong Kong
| | - Xinyi Bi
- School of Biological Sciences, The University of Hong Kong, Hong Kong, China, Hong Kong
| | - Hiu-Lam Rachel Kwan
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, China, Hong Kong
| | - Daniel Brobst
- Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Oana Herlea-Pana
- Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Xiuying Yang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines and Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica of Peking Union Medical College, Beijing, China
| | - Guanhua Du
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines and Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica of Peking Union Medical College, Beijing, China
| | - Suchaorn Saengnipanthkul
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Hye Lim Noh
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Baowei Jiao
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Jason K Kim
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA.,Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Chi Wai Lee
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, China, Hong Kong
| | - Keqiang Ye
- Department of Pathology, Emory University School of Medicine, Atlanta, USA
| | - Chi Bun Chan
- School of Biological Sciences, The University of Hong Kong, Hong Kong, China, Hong Kong.,State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong
| |
Collapse
|
26
|
A novel knockout mouse model of the noncoding antisense Brain-Derived Neurotrophic Factor ( Bdnf) gene displays increased endogenous Bdnf protein and improved memory function following exercise. Heliyon 2021; 7:e07570. [PMID: 34377851 PMCID: PMC8327352 DOI: 10.1016/j.heliyon.2021.e07570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/10/2021] [Accepted: 07/10/2021] [Indexed: 11/26/2022] Open
Abstract
Brain-derived neurotrophic factor (Bdnf) expression is tightly controlled at the transcriptional and post-transcriptional levels. Previously, we showed that inhibition of noncoding Bdnf antisense (Bdnf-AS) RNA upregulates Bdnf protein. Here, we generated a Bdnf-antisense knockout (Bdnf-AS KO) mouse model by deleting 6 kilobases upstream of Bdnf-AS. After verifying suppression of Bdnf-AS, baseline behavioral tests indicated no significant difference in knockout and wild type mice, except for enhanced cognitive function in the knockout mice in the Y-maze. Following acute involuntary exercise, Bdnf-AS KO mice were re-assessed and a significant increase in Bdnf mRNA and protein were observed. Following long-term involuntary exercise, we observed a significant increase in nonspatial and spatial memory in novel object recognition and Barnes maze tests in young and aged Bdnf-AS KO mice. Our data provides evidence for the beneficial effects of endogenous Bdnf upregulation and the synergistic effect of Bdnf-AS knockout on exercise and memory retention.
Collapse
|
27
|
Krassovskaia PM, Chaves AB, Houmard JA, Broskey NT. Exercise during Pregnancy: Developmental Programming Effects and Future Directions in Humans. Int J Sports Med 2021; 43:107-118. [PMID: 34344043 DOI: 10.1055/a-1524-2278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Epidemiological studies show that low birth weight is associated with mortality from cardiovascular disease in adulthood, indicating that chronic diseases could be influenced by hormonal or metabolic insults encountered in utero. This concept, now known as the Developmental Origins of Health and Disease hypothesis, postulates that the intrauterine environment may alter the structure and function of the organs of the fetus as well as the expression of genes that impart an increased vulnerability to chronic diseases later in life. Lifestyle interventions initiated during the prenatal period are crucial as there is the potential to attenuate progression towards chronic diseases. However, how lifestyle interventions such as physical activity directly affect human offspring metabolism and the potential mechanisms involved in regulating metabolic balance at the cellular level are not known. The purpose of this review is to highlight the effects of exercise during pregnancy on offspring metabolic health and emphasize gaps in the current human literature and suggestions for future research.
Collapse
Affiliation(s)
- Polina M Krassovskaia
- Human Performance Laboratory, Department of Kinesiology, East Carolina University, Greenville, United States.,East Carolina Diabetes & Obesity Institute, East Carolina University, Greenville, United States
| | - Alec B Chaves
- Human Performance Laboratory, Department of Kinesiology, East Carolina University, Greenville, United States.,East Carolina Diabetes & Obesity Institute, East Carolina University, Greenville, United States
| | - Joseph A Houmard
- Human Performance Laboratory, Department of Kinesiology, East Carolina University, Greenville, United States.,East Carolina Diabetes & Obesity Institute, East Carolina University, Greenville, United States
| | - Nicholas T Broskey
- Human Performance Laboratory, Department of Kinesiology, East Carolina University, Greenville, United States.,East Carolina Diabetes & Obesity Institute, East Carolina University, Greenville, United States
| |
Collapse
|
28
|
Koito Y, Yanishi M, Kimura Y, Tsukaguchi H, Kinoshita H, Matsuda T. Serum Brain-Derived Neurotrophic Factor and Myostatin Levels Are Associated With Skeletal Muscle Mass in Kidney Transplant Recipients. Transplant Proc 2021; 53:1939-1944. [PMID: 34253381 DOI: 10.1016/j.transproceed.2021.04.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 04/01/2021] [Accepted: 04/19/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Sarcopenia, or reduced muscle mass, can be an important complication in kidney transplant recipients. The skeletal muscles were recently reported to secrete various myokines, such as brain-derived neurotrophic factor (BDNF) and myostatin, to regulate their mass, function, or both. The aim of the present study was to analyze the interrelationship between myokines (BDNF and myostatin) and skeletal muscle mass in kidney transplant recipients. METHODS The study population comprised 40 patients who underwent kidney transplantation at Kansai Medical University Hospital. Twenty patients had low skeletal muscle mass index (SMI) values, as measured on dual-energy x-ray absorptiometry, and were categorized into 2 groups (low SMI and normal). RESULTS Mean serum BDNF levels were 15.7 ng/mL in the low SMI group and 17.8 ng/mL in the normal group (P = .013). Mean serum myostatin levels were 362 pg/mL in the low SMI and 267 pg/mL in the normal group (P = .024). There was a significant positive correlation among metabolic equivalents and serum BDNF levels (r = 0.817; P < .001) and a significant negative correlation among metabolic equivalents and serum myostatin levels (r = -0.541; P < .001). Receiver operating characteristic analysis showed that serum BDNF and level of area under curve was 0.712, and serum myostatin level of area under the curve was 0.690. Serum BDNF and myostatin levels showed no significant difference. CONCLUSION These results suggest that BDNF and myostatin are potential biomarkers of reduced muscle mass in kidney transplant recipients.
Collapse
Affiliation(s)
- Yuya Koito
- Department of Urology and Andrology, Kansai Medical University, Osaka, Japan
| | - Masaaki Yanishi
- Department of Urology and Andrology, Kansai Medical University, Osaka, Japan.
| | - Yutaka Kimura
- Health Science Center, Kansai Medical University, Osaka, Japan
| | - Hiroyasu Tsukaguchi
- Second Department of Internal Medicine, Division of Nephrology, Kansai Medical University, Osaka, Japan
| | - Hidefumi Kinoshita
- Department of Urology and Andrology, Kansai Medical University, Osaka, Japan
| | - Tadashi Matsuda
- Department of Urology and Andrology, Kansai Medical University, Osaka, Japan
| |
Collapse
|
29
|
Zhao J, Li P, Zhu H, Ge F, Liu J, Xia J, Hang P. 7,8-Dihydroxyflavone suppresses proliferation and induces apoptosis of human osteosarcoma cells. Acta Biochim Biophys Sin (Shanghai) 2021; 53:903-911. [PMID: 34019097 DOI: 10.1093/abbs/gmab060] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Indexed: 02/07/2023] Open
Abstract
Recent studies suggest that 7,8-dihydroxyflavone (7,8-DHF) inhibits the development of several tumors. However, its role in osteosarcoma (OS) remains unknown. This study was designed to investigate the effects and underlying mechanisms of 7,8-DHF that may influence OS development. Human OS cell lines (U2OS and 143B) were treated with 7,8-DHF; cell viability and cell migration were assessed by methylthiazolyldiphenyl-tetrazolium bromide (MTT) assay and wound-healing assay, respectively; and cell death and apoptosis were evaluated by LIVE/DEAD staining and terminal deoxynucleotidyl transferase (TdT) dUTP nick-end labeling (TUNEL) assay, respectively. Reactive oxygen species production was measured using 2,7-dichlorodihydrofluorescein diacetate probe. Akt, Bcl-xL/Bcl-2 asociated death promoter (Bad), p38 mitogen-activated protein kinase (MAPK), extracellular regulated protein kinase (ERK), and c-Jun N-terminal kinase (JNK) expression and their respective phosphorylation levels were detected by western blot analysis. We found that 7,8-DHF reduced cell viability in a dose-dependent manner and also promoted apoptosis, inhibited migration, and induced oxidative stress in OS cells. Moreover, 7,8-DHF inhibited Akt, Bad, and p38MAPK, but activated ERK and JNK signals. In summary, our results suggest that 7,8-DHF inhibits OS progression, possibly by regulating Akt/Bad and MAPK signaling. These findings provide new evidence for the pharmacological effects of 7,8-DHF that may improve drug therapy for OS patients.
Collapse
Affiliation(s)
- Jing Zhao
- Department of Pharmacy, Clinical Medical College, Yangzhou University, Northern Jiangsu People’s Hospital, Yangzhou 225001, China
| | - Peifeng Li
- Institute of Clinical Pharmacology, The Second Affiliated Hospital of Harbin Medical University (The University Key Laboratory of Drug Research), Harbin 150086, China
| | - Hua Zhu
- Department of Pharmacy, Clinical Medical College, Yangzhou University, Northern Jiangsu People’s Hospital, Yangzhou 225001, China
| | - Fengqin Ge
- Department of Pharmacy, Clinical Medical College, Yangzhou University, Northern Jiangsu People’s Hospital, Yangzhou 225001, China
| | - Jie Liu
- Institute of Clinical Pharmacology, The Second Affiliated Hospital of Harbin Medical University (The University Key Laboratory of Drug Research), Harbin 150086, China
| | - Jingjun Xia
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Pengzhou Hang
- Department of Pharmacy, Clinical Medical College, Yangzhou University, Northern Jiangsu People’s Hospital, Yangzhou 225001, China
| |
Collapse
|
30
|
Podyma B, Parekh K, Güler AD, Deppmann CD. Metabolic homeostasis via BDNF and its receptors. Trends Endocrinol Metab 2021; 32:488-499. [PMID: 33958275 PMCID: PMC8192464 DOI: 10.1016/j.tem.2021.04.005] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/10/2021] [Accepted: 04/12/2021] [Indexed: 12/12/2022]
Abstract
Metabolic disorders result from dysregulation of central nervous system and peripheral metabolic energy homeostatic pathways. To maintain normal energy balance, neural circuits must integrate feedforward and feedback signals from the internal metabolic environment to orchestrate proper food intake and energy expenditure. These signals include conserved meal and adipocyte cues such as glucose and leptin, respectively, in addition to more novel players including brain-derived neurotrophic factor (BDNF). In particular, BDNF's two receptors, tropomyosin related kinase B (TrkB) and p75 neurotrophin receptor (p75NTR), are increasingly appreciated to be involved in whole body energy homeostasis. At times, these two receptors even seem to functionally oppose one another's actions, providing the framework for a potential neurotrophin mediated energy regulatory axis, which we explore further here.
Collapse
Affiliation(s)
- Brandon Podyma
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA; Medical Scientist Training Program, School of Medicine, University of Virginia, Charlottesville, VA 22908-0738, USA.
| | - Kavya Parekh
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA
| | - Ali D Güler
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA
| | | |
Collapse
|
31
|
Longo M, Meroni M, Paolini E, Macchi C, Dongiovanni P. Mitochondrial dynamics and nonalcoholic fatty liver disease (NAFLD): new perspectives for a fairy-tale ending? Metabolism 2021; 117:154708. [PMID: 33444607 DOI: 10.1016/j.metabol.2021.154708] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 01/05/2021] [Accepted: 01/07/2021] [Indexed: 12/12/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) includes a broad spectrum of liver dysfunctions and it is predicted to become the primary cause of liver failure and hepatocellular carcinoma. Mitochondria are highly dynamic organelles involved in multiple metabolic/bioenergetic pathways in the liver. Emerging evidence outlined that hepatic mitochondria adapt in number and functionality in response to external cues, as high caloric intake and obesity, by modulating mitochondrial biogenesis, and maladaptive mitochondrial response has been described from the early stages of NAFLD. Indeed, mitochondrial plasticity is lost in progressive NAFLD and these organelles may assume an aberrant phenotype to drive or contribute to hepatocarcinogenesis. Severe alimentary regimen and physical exercise represent the cornerstone for NAFLD care, although the low patients' compliance is urging towards the discovery of novel pharmacological treatments. Mitochondrial-targeted drugs aimed to recover mitochondrial lifecycle and to modulate oxidative stress are becoming attractive molecules to be potentially introduced for NAFLD management. Although the path guiding the switch from bench to bedside remains tortuous, the study of mitochondrial dynamics is providing intriguing perspectives for future NAFLD healthcare.
Collapse
Affiliation(s)
- Miriam Longo
- General Medicine and Metabolic Diseases, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Pad. Granelli, via F Sforza 35, 20122 Milan, Italy; Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milano, Italy
| | - Marica Meroni
- General Medicine and Metabolic Diseases, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Pad. Granelli, via F Sforza 35, 20122 Milan, Italy; Department of Pathophysiology and Transplantation, Università degli Studi di Milano, 20122 Milano, Italy
| | - Erika Paolini
- General Medicine and Metabolic Diseases, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Pad. Granelli, via F Sforza 35, 20122 Milan, Italy; Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milano, Italy
| | - Chiara Macchi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milano, Italy
| | - Paola Dongiovanni
- General Medicine and Metabolic Diseases, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Pad. Granelli, via F Sforza 35, 20122 Milan, Italy.
| |
Collapse
|
32
|
Zierold S, Buschmann K, Gachkar S, Bochenek ML, Velmeden D, Hobohm L, Vahl CF, Schäfer K. Brain-Derived Neurotrophic Factor Expression and Signaling in Different Perivascular Adipose Tissue Depots of Patients With Coronary Artery Disease. J Am Heart Assoc 2021; 10:e018322. [PMID: 33666096 PMCID: PMC8174206 DOI: 10.1161/jaha.120.018322] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background Brain‐derived neurotrophic factor (BDNF) is expressed in neuronal and nonneuronal cells and may affect vascular functions via its receptor, tropomyosin‐related kinase B (TrkB). In this study, we determined the expression of BDNF in different perivascular adipose tissue (PVAT) depots of patients with established coronary atherosclerosis. Methods and Results Serum, vascular tissue, and PVAT surrounding the proximal aorta (C‐PVAT) or internal mammary artery (IMA‐PVAT) was obtained from 24 patients (79% men; mean age, 71.7±9.7 years; median body mass index, 27.4±4.8 kg/m2) with coronary atherosclerosis undergoing elective coronary artery bypass surgery. BDNF protein levels were significantly higher in C‐PVAT compared with IMA‐PVAT, independent of obesity, metabolic syndrome, or systemic biomarkers of inflammation. mRNA transcripts of TrkB, the BDNF receptor, were significantly reduced in aorta compared with IMA. Vessel wall TrkB immunosignals colocalized with cells expressing smooth muscle cell markers, and confocal microscopy and flow cytometry confirmed BDNF receptor expression in human aortic smooth muscle cells. Significantly elevated levels of protein tyrosine phosphatase 1B, a negative regulator of TrkB signaling in the brain, were also observed in C‐PVAT. In vitro, inhibition of protein tyrosine phosphatase 1B blunted the effects of BDNF on smooth muscle cell proliferation, migration, differentiation, and collagen production, possibly by upregulation of low‐affinity p75 neurotrophin receptors. Expression of nerve growth factor or its receptor tropomyosin‐related kinase A did not differ between C‐PVAT and IMA‐PVAT. Conclusions Elevated expression of BDNF in parallel with local upregulation of negative regulators of neurotrophin signaling in perivascular fat and lower TrkB expression suggest that vascular BDNF signaling is reduced or lost in patients with coronary atherosclerosis.
Collapse
Affiliation(s)
- Sarah Zierold
- Department of Cardiology Cardiology I University Medical Center Mainz Mainz Germany
| | - Katja Buschmann
- Department of Cardiothoracic and Vascular Surgery University Medical Center Mainz Mainz Germany
| | - Sogol Gachkar
- Department of Cardiology Cardiology I University Medical Center Mainz Mainz Germany
| | - Magdalena L Bochenek
- Department of Cardiology Cardiology I University Medical Center Mainz Mainz Germany.,Center for Thrombosis and Hemostasis University Medical Center Mainz Mainz Germany
| | - David Velmeden
- Department of Cardiology Cardiology I University Medical Center Mainz Mainz Germany
| | - Lukas Hobohm
- Department of Cardiology Cardiology I University Medical Center Mainz Mainz Germany.,Center for Thrombosis and Hemostasis University Medical Center Mainz Mainz Germany
| | | | - Katrin Schäfer
- Department of Cardiology Cardiology I University Medical Center Mainz Mainz Germany
| |
Collapse
|
33
|
Sharma P, Wu G, Kumaraswamy D, Burchat N, Ye H, Gong Y, Zhao L, Lam YY, Sampath H. Sex-Dependent Effects of 7,8-Dihydroxyflavone on Metabolic Health Are Associated with Alterations in the Host Gut Microbiome. Nutrients 2021; 13:637. [PMID: 33669347 PMCID: PMC7920311 DOI: 10.3390/nu13020637] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/05/2021] [Accepted: 02/09/2021] [Indexed: 12/12/2022] Open
Abstract
7,8-Dihydroxyflavone (DHF) is a naturally occurring flavonoid that has been reported to protect against a variety of pathologies. Chronic administration of DHF prevents high-fat diet (HFD)-induced obesity in female, but not male, mice. However, the mechanisms underlying this sexual dimorphism have not been elucidated. We have discovered that oral DHF supplementation significantly attenuates fat mass, hepatic lipid accumulation, and adipose tissue inflammation in female mice. In contrast, male mice were not protected from adiposity, and had a paradoxical worsening of hepatic lipid accumulation and adipose tissue inflammation upon DHF supplementation. Consistent with these sexually dimorphic effects on body weight and metabolic health, 7,8-DHF induced early and stable remodeling of the female intestinal microbiome. DHF supplementation significantly increased gut microbial diversity, and suppressed potentially detrimental bacteria, particularly Desulfovibrionaceae, which are pro-inflammatory and positively associated with obesity and inflammation. Changes in the female gut microbiome preceded alterations in body weights, and in silico analyses indicated that these early microbial changes were highly predictive of subsequent weight gain in female mice. While some alterations in the intestinal microbiome were also observed in male DHF-supplemented mice, these changes were distinct from those in females and, importantly, were not predictive of subsequent body weight changes in male animals. The temporality of microbial changes preceding alterations in body weight in female mice suggests a role for the gut microbiome in mediating the sexually dimorphic effects of DHF on body weight. Given the significant clinical interest in this flavonoid across a wide range of pathologies, further elucidation of these sexually dimorphic effects will aid the development of effective clinical therapies.
Collapse
Affiliation(s)
- Priyanka Sharma
- Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, New Brunswick, NJ 08901, USA; (P.S.); (D.K.); (N.B.); (H.Y.)
- Center for Microbiome, Nutrition, and Health, New Jersey Institute for Food, Nutrition, and Health, New Brunswick, NJ 08901, USA; (G.W.); (Y.G.); (L.Z.)
| | - Guojun Wu
- Center for Microbiome, Nutrition, and Health, New Jersey Institute for Food, Nutrition, and Health, New Brunswick, NJ 08901, USA; (G.W.); (Y.G.); (L.Z.)
- Department of Biochemistry and Microbiology, Rutgers University, New Brunswick, NJ 08901, USA
| | - Deeptha Kumaraswamy
- Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, New Brunswick, NJ 08901, USA; (P.S.); (D.K.); (N.B.); (H.Y.)
- Center for Microbiome, Nutrition, and Health, New Jersey Institute for Food, Nutrition, and Health, New Brunswick, NJ 08901, USA; (G.W.); (Y.G.); (L.Z.)
| | - Natalie Burchat
- Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, New Brunswick, NJ 08901, USA; (P.S.); (D.K.); (N.B.); (H.Y.)
- Center for Microbiome, Nutrition, and Health, New Jersey Institute for Food, Nutrition, and Health, New Brunswick, NJ 08901, USA; (G.W.); (Y.G.); (L.Z.)
| | - Hong Ye
- Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, New Brunswick, NJ 08901, USA; (P.S.); (D.K.); (N.B.); (H.Y.)
- Center for Microbiome, Nutrition, and Health, New Jersey Institute for Food, Nutrition, and Health, New Brunswick, NJ 08901, USA; (G.W.); (Y.G.); (L.Z.)
| | - Yongjia Gong
- Center for Microbiome, Nutrition, and Health, New Jersey Institute for Food, Nutrition, and Health, New Brunswick, NJ 08901, USA; (G.W.); (Y.G.); (L.Z.)
- Department of Biochemistry and Microbiology, Rutgers University, New Brunswick, NJ 08901, USA
| | - Liping Zhao
- Center for Microbiome, Nutrition, and Health, New Jersey Institute for Food, Nutrition, and Health, New Brunswick, NJ 08901, USA; (G.W.); (Y.G.); (L.Z.)
- Department of Biochemistry and Microbiology, Rutgers University, New Brunswick, NJ 08901, USA
| | - Yan Y. Lam
- Center for Microbiome, Nutrition, and Health, New Jersey Institute for Food, Nutrition, and Health, New Brunswick, NJ 08901, USA; (G.W.); (Y.G.); (L.Z.)
- Department of Biochemistry and Microbiology, Rutgers University, New Brunswick, NJ 08901, USA
| | - Harini Sampath
- Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, New Brunswick, NJ 08901, USA; (P.S.); (D.K.); (N.B.); (H.Y.)
- Center for Microbiome, Nutrition, and Health, New Jersey Institute for Food, Nutrition, and Health, New Brunswick, NJ 08901, USA; (G.W.); (Y.G.); (L.Z.)
- Department of Nutritional Sciences, Rutgers University, New Brunswick, NJ 08901, USA
| |
Collapse
|
34
|
Pang BPS, Chan WS, Chan CB. Mitochondria Homeostasis and Oxidant/Antioxidant Balance in Skeletal Muscle-Do Myokines Play a Role? Antioxidants (Basel) 2021; 10:antiox10020179. [PMID: 33513795 PMCID: PMC7911667 DOI: 10.3390/antiox10020179] [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: 12/29/2020] [Revised: 01/19/2021] [Accepted: 01/21/2021] [Indexed: 12/19/2022] Open
Abstract
Mitochondria are the cellular powerhouses that generate adenosine triphosphate (ATP) to substantiate various biochemical activities. Instead of being a static intracellular structure, they are dynamic organelles that perform constant structural and functional remodeling in response to different metabolic stresses. In situations that require a high ATP supply, new mitochondria are assembled (mitochondrial biogenesis) or formed by fusing the existing mitochondria (mitochondrial fusion) to maximize the oxidative capacity. On the other hand, nutrient overload may produce detrimental metabolites such as reactive oxidative species (ROS) that wreck the organelle, leading to the split of damaged mitochondria (mitofission) for clearance (mitophagy). These vital processes are tightly regulated by a sophisticated quality control system involving energy sensing, intracellular membrane interaction, autophagy, and proteasomal degradation to optimize the number of healthy mitochondria. The effective mitochondrial surveillance is particularly important to skeletal muscle fitness because of its large tissue mass as well as its high metabolic activities for supporting the intensive myofiber contractility. Indeed, the failure of the mitochondrial quality control system in skeletal muscle is associated with diseases such as insulin resistance, aging, and muscle wasting. While the mitochondrial dynamics in cells are believed to be intrinsically controlled by the energy content and nutrient availability, other upstream regulators such as hormonal signals from distal organs or factors generated by the muscle itself may also play a critical role. It is now clear that skeletal muscle actively participates in systemic energy homeostasis via producing hundreds of myokines. Acting either as autocrine/paracrine or circulating hormones to crosstalk with other organs, these secretory myokines regulate a large number of physiological activities including insulin sensitivity, fuel utilization, cell differentiation, and appetite behavior. In this article, we will review the mechanism of myokines in mitochondrial quality control and ROS balance, and discuss their translational potential.
Collapse
|
35
|
Hang PZ, Zhu H, Li PF, Liu J, Ge FQ, Zhao J, Du ZM. The Emerging Role of BDNF/TrkB Signaling in Cardiovascular Diseases. Life (Basel) 2021; 11:life11010070. [PMID: 33477900 PMCID: PMC7833389 DOI: 10.3390/life11010070] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/08/2021] [Accepted: 01/16/2021] [Indexed: 02/06/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is one of the most abundantneurotrophins in the central nervous system. Numerous studies suggestthat BDNF has extensive roles by binding to its specific receptor, tropomyosin-related kinase receptor B (TrkB), and thereby triggering downstream signaling pathways. Recently, growing evidence highlightsthat the BDNF/TrkB pathway is expressed in the cardiovascular system andclosely associated with the development and outcome of cardiovascular diseases (CVD), including coronary artery disease, heart failure, cardiomyopathy, hypertension, and metabolic diseases. Furthermore, circulating BDNF has also been revealed as a new potential biomarker for both diagnosis and prognosis of CVD. In this review, we discuss the current evidence of the emerging role of BDNF/TrkBsignalingand address the challenges that remain in translating these discoveries to novel therapeutic strategies for CVD.
Collapse
Affiliation(s)
- Peng-Zhou Hang
- Department of Pharmacy, Clinical Medical College, Yangzhou University, Northern Jiangsu People’s Hospital, Yangzhou 225001, China; (P.-Z.H.); (H.Z.); (F.-Q.G.)
- Institute of Clinical Pharmacology, The Second Affiliated Hospital of Harbin Medical University (University Key Laboratory of Drug Research), Harbin 150086, China; (P.-F.L.); (J.L.)
| | - Hua Zhu
- Department of Pharmacy, Clinical Medical College, Yangzhou University, Northern Jiangsu People’s Hospital, Yangzhou 225001, China; (P.-Z.H.); (H.Z.); (F.-Q.G.)
| | - Pei-Feng Li
- Institute of Clinical Pharmacology, The Second Affiliated Hospital of Harbin Medical University (University Key Laboratory of Drug Research), Harbin 150086, China; (P.-F.L.); (J.L.)
| | - Jie Liu
- Institute of Clinical Pharmacology, The Second Affiliated Hospital of Harbin Medical University (University Key Laboratory of Drug Research), Harbin 150086, China; (P.-F.L.); (J.L.)
| | - Feng-Qin Ge
- Department of Pharmacy, Clinical Medical College, Yangzhou University, Northern Jiangsu People’s Hospital, Yangzhou 225001, China; (P.-Z.H.); (H.Z.); (F.-Q.G.)
| | - Jing Zhao
- Medical Research Center, Clinical Medical College, Yangzhou University, Northern Jiangsu People’s Hospital, Yangzhou 225001, China
- Correspondence: or (J.Z.); or (Z.-M.D.); Tel.: +86-514-8737-3691 (J.Z.); +86-451-8660-5353 (Z.-M.D.); Fax: +86-514-8737-3039 (J.Z.); +86-451-8666-5559 (Z.-M.D.)
| | - Zhi-Min Du
- Institute of Clinical Pharmacology, The Second Affiliated Hospital of Harbin Medical University (University Key Laboratory of Drug Research), Harbin 150086, China; (P.-F.L.); (J.L.)
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau 999078, China
- Correspondence: or (J.Z.); or (Z.-M.D.); Tel.: +86-514-8737-3691 (J.Z.); +86-451-8660-5353 (Z.-M.D.); Fax: +86-514-8737-3039 (J.Z.); +86-451-8666-5559 (Z.-M.D.)
| |
Collapse
|
36
|
Zhu W, Yang F, Cai X, Zhang W, Zhang J, Cai M, Li X, Xiang J, Cai D. Role of glucocorticoid receptor phosphorylation-mediated synaptic plasticity in anxiogenic and depressive behaviors induced by monosodium glutamate. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2021; 394:151-164. [PMID: 32444989 DOI: 10.1007/s00210-020-01845-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 02/19/2020] [Indexed: 12/15/2022]
Abstract
Psychiatric diseases and metabolic disorders frequently cooccur, yet the mechanisms underlying this interaction remain unknown. The aim of this study was to determine the role of glucocorticoid receptor (GR) phosphorylation in the comorbidity of metabolic and psychiatric disorders. Neonatal Sprague-Dawley rats were subcutaneously injected with monosodium glutamate (MSG) every 2 days for 10 days after birth. Metabolic and behavioral tests were performed 12 weeks later. Golgi staining and transmission electron microscopy (TEM) were performed to evaluate synaptic structural plasticity. Changes in GR phosphorylation and the BDNF/TrkB pathway were evaluated by western blotting and immunofluorescence. We found that MSG-treated rats displayed significant metabolic abnormalities accompanied by anxiogenic and depressive behaviors, an altered synaptic ultrastructure and the loss of dendritic spines. The expression of phosphorylated GR was reduced in the brain. Furthermore, a specific agonist of BDNF/TrkB significantly reversed the reduction in GR phosphorylation, as well as the metabolic and behavioral outcomes. These findings indicate that a decrease in BDNF/TrkB pathway-dependent GR phosphorylation is a long-term effect of MSG treatment that may contribute to metabolic and behavioral disturbances.
Collapse
Affiliation(s)
- Wen Zhu
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Laboratory of Neurology, Institute of Integrative Medicine, Fudan University, Shanghai, 200032, China
| | - Feng Yang
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Laboratory of Neurology, Institute of Integrative Medicine, Fudan University, Shanghai, 200032, China
| | - Xiaofang Cai
- Department of Stomatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Wen Zhang
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Laboratory of Neurology, Institute of Integrative Medicine, Fudan University, Shanghai, 200032, China
| | - Jingsi Zhang
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Laboratory of Neurology, Institute of Integrative Medicine, Fudan University, Shanghai, 200032, China
| | - Min Cai
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Laboratory of Neurology, Institute of Integrative Medicine, Fudan University, Shanghai, 200032, China
| | - Xiangting Li
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Laboratory of Neurology, Institute of Integrative Medicine, Fudan University, Shanghai, 200032, China
| | - Jun Xiang
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Laboratory of Neurology, Institute of Integrative Medicine, Fudan University, Shanghai, 200032, China.
| | - Dingfang Cai
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Laboratory of Neurology, Institute of Integrative Medicine, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
37
|
Zhao Z, Xue F, Gu Y, Han J, Jia Y, Ye K, Zhang Y. Crosstalk between the muscular estrogen receptor α and BDNF/TrkB signaling alleviates metabolic syndrome via 7,8-dihydroxyflavone in female mice. Mol Metab 2020; 45:101149. [PMID: 33352311 PMCID: PMC7811170 DOI: 10.1016/j.molmet.2020.101149] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 12/15/2020] [Accepted: 12/16/2020] [Indexed: 02/06/2023] Open
Abstract
Objective 7,8-Dihydroxyflavone (7,8-DHF), a small molecular mimetic of brain-derived neurotrophic factor (BDNF), alleviates high-fat diet-induced obesity in female mice in a sex-specific manner by activating muscular tropomyosin-related kinase B (TrkB). However, the underlying molecular mechanism for this sex difference is unknown. Moreover, muscular estrogen receptor α (ERα) plays a critical role in metabolic diseases. Impaired ERα action is often accompanied by metabolic syndrome (MetS) in postmenopausal women. This study investigated whether muscular ERα is involved in the metabolic effects of 7,8-DHF. Methods For the in vivo studies, 72 female C57BL/6J mice were given a low-fat diet or high-fat diet, and both received daily intragastric administration of vehicle or 7,8-DHF for 24 weeks. The hypothalamic-pituitary-ovarian (HPO) axis function was assessed by investigating typical sex-related serum hormones and the ovarian reserve. Indicators of menopausal MetS, including lipid metabolism, insulin sensitivity, bone density, and serum inflammatory cytokines, were also evaluated. The expression levels of ERα and other relevant signaling molecules were also examined. In vitro, the molecular mechanism involved in the interplay of ERα and TrkB receptors was verified in differentiated C2C12 myotubes using several inhibitors and a lentivirus short hairpin RNA-knockdown strategy. Results Long-term oral administration of 7,8-DHF acted as a protective factor for the female HPO axis function, protecting against ovarian failure, earlier menopause, and sex hormone disorders, which was paralleled by the alleviation of MetS coupled with the production of ERα-rich, TrkB-activated, and uncoupling protein 1 (UCP1) high thermogenic skeletal muscle tissues. 7,8-DHF-stimulated transactivation of ERα at serine 118 (S118) and tyrosine 537 (Y537), which was crucial to activate the BDNF/TrkB signaling cascades. In turn, activation of BDNF/TrkB signaling was also required for the ligand-independent activation of ERα, especially at the Y537 phosphorylation site. In addition, Src family kinases played a core role in the interplay of ERα and TrkB, synergistically activating the signaling pathways related to energy metabolism. Conclusions These findings revealed a novel role of 7,8-DHF in protecting the function of the female HPO axis and activating tissue-specific ERα, which improves our understanding of this sex difference in 7,8-DHF-mediated maintenance of metabolic homeostasis and provides new therapeutic strategies for managing MetS in women. 7,8-DHF improves hypothalamic-pituitary-ovarian axis function in mature adult female mice. 7,8-DHF protects against ovarian failure and onset of earlier menopause. 7,8-DHF-induced transactivation of ERα is crucial to activate BDNF/TrkB signaling cascades. 7,8-DHF-induced activations of ERα and BDNF/TrkB signaling are interdependent. Src family kinases play a core role in the crosstalk of ERα and BDNF/TrkB signaling pathways.
Collapse
Affiliation(s)
- Zhenlei Zhao
- Department of Food Science and Nutrition, School of Biosystems Engineering and Food Science, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang Engineering Center for Food Technology and Equipment, Zhejiang University, Hangzhou 310058, China
| | - Fan Xue
- Department of Food Science and Nutrition, School of Biosystems Engineering and Food Science, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang Engineering Center for Food Technology and Equipment, Zhejiang University, Hangzhou 310058, China
| | - Yanpei Gu
- Department of Food Science and Nutrition, School of Biosystems Engineering and Food Science, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang Engineering Center for Food Technology and Equipment, Zhejiang University, Hangzhou 310058, China
| | - Jianxin Han
- Department of Food Science and Nutrition, School of Biosystems Engineering and Food Science, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang Engineering Center for Food Technology and Equipment, Zhejiang University, Hangzhou 310058, China
| | - Yingxian Jia
- Department of Gynecology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Keqiang Ye
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | - Ying Zhang
- Department of Food Science and Nutrition, School of Biosystems Engineering and Food Science, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang Engineering Center for Food Technology and Equipment, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
38
|
Emili M, Guidi S, Uguagliati B, Giacomini A, Bartesaghi R, Stagni F. Treatment with the flavonoid 7,8-Dihydroxyflavone: a promising strategy for a constellation of body and brain disorders. Crit Rev Food Sci Nutr 2020; 62:13-50. [DOI: 10.1080/10408398.2020.1810625] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Marco Emili
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Sandra Guidi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Beatrice Uguagliati
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Andrea Giacomini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Renata Bartesaghi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Fiorenza Stagni
- Department for Life Quality Studies, University of Bologna, Rimini, Italy
| |
Collapse
|
39
|
Zhang D, He S, Wang Q, Pu S, Zhou Z, Wu Q. Impact of Aging on the Characterization of Brown and White Adipose Tissue-Derived Stem Cells in Mice. Cells Tissues Organs 2020; 209:26-36. [PMID: 32526740 DOI: 10.1159/000507434] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 03/23/2020] [Indexed: 12/16/2022] Open
Abstract
Adipose tissue enriched with adipose tissue-derived stem cells (ASCs) is often used for stem cell-based therapies. However, the characteristics of ASCs from different types of adipose tissue have varying biochemical and functional properties. We aimed to investigate how age affected the biological and functional characteristics of ASCs from brown (BAT) and white adipose tissue (WAT). ASCs were obtained and cultured from mouse BAT and WAT at different ages: young (2 months of age) and older mice (22 months of age). Mesenchymal markers were characterized by flow cytometry, and cell proliferation, apoptosis, differentiation potential, senescence, and metabolism were then determined. The percentage of WAT was higher in elderly mice, and the percentage of BAT was higher in young mice. All ASC sample phenotypes were characterized as CD29+/CD44+/CD105+/CD45-; the proliferation rate was not statistically different among all age groups. However, the number of senescent cells and the percentage of apoptosis in elderly mouse ASCs were significantly increased, and the ability of osteogenic and lipogenic differentiation was decreased in these same animals. In addition, ASCs from young mice were more inclined to undergo osteogenic differentiation, especially BAT-ASCs, whose gene expression of fat-consuming components was also significantly higher than of WAT-ASCs. The results indicated that ASCs derived from both WAT and BAT possessed different characteristics of fat metabolism and cell differentiation relative to the osteo- and adipolineages. In particular, because BAT-ASCs from young mice contributed to fat consumption, if used for cell grafting, they may potentially be attractive vehicles for treating obesity.
Collapse
Affiliation(s)
- Daxiu Zhang
- School of Life Sciences, Guangxi Normal University, Guilin, China.,Key Laboratory of Stem Cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, China.,Research Center for Biomedical Sciences, Guangxi Normal University, Guilin, China
| | - Shuangli He
- School of Life Sciences, Guangxi Normal University, Guilin, China.,Key Laboratory of Stem Cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, China.,Research Center for Biomedical Sciences, Guangxi Normal University, Guilin, China
| | - Qian Wang
- School of Life Sciences, Guangxi Normal University, Guilin, China.,Key Laboratory of Stem Cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, China.,Research Center for Biomedical Sciences, Guangxi Normal University, Guilin, China
| | - Shiming Pu
- School of Life Sciences, Guangxi Normal University, Guilin, China.,Key Laboratory of Stem Cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, China.,Research Center for Biomedical Sciences, Guangxi Normal University, Guilin, China
| | - Zuping Zhou
- School of Life Sciences, Guangxi Normal University, Guilin, China.,Key Laboratory of Stem Cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, China.,Research Center for Biomedical Sciences, Guangxi Normal University, Guilin, China
| | - Qiong Wu
- School of Life Sciences, Guangxi Normal University, Guilin, China, .,Key Laboratory of Stem Cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, China, .,Research Center for Biomedical Sciences, Guangxi Normal University, Guilin, China,
| |
Collapse
|
40
|
Nakano I, Kinugawa S, Hori H, Fukushima A, Yokota T, Takada S, Kakutani N, Obata Y, Yamanashi K, Anzai T. Serum Brain-Derived Neurotrophic Factor Levels Are Associated with Skeletal Muscle Function but Not with Muscle Mass in Patients with Heart Failure. Int Heart J 2020; 61:96-102. [DOI: 10.1536/ihj.19-400] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Ippei Nakano
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University
| | - Shintaro Kinugawa
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University
| | - Hiroaki Hori
- Department of Rehabilitation, Hokkaido University Hospital
| | - Arata Fukushima
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University
| | - Takashi Yokota
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University
| | - Shingo Takada
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University
| | - Naoya Kakutani
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University
- Research Fellow of the Japan Society for the Promotion of Science
| | - Yoshikuni Obata
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University
| | - Katsuma Yamanashi
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University
| | - Toshihisa Anzai
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University
| |
Collapse
|
41
|
Chan CB, Ahuja P, Ye K. Developing Insulin and BDNF Mimetics for Diabetes Therapy. Curr Top Med Chem 2019; 19:2188-2204. [PMID: 31660832 DOI: 10.2174/1568026619666191010160643] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 08/29/2019] [Accepted: 09/05/2019] [Indexed: 01/06/2023]
Abstract
Diabetes is a global public health concern nowadays. The majority of diabetes mellitus (DM) patients belong to type 2 diabetes mellitus (T2DM), which is highly associated with obesity. The general principle of current therapeutic strategies for patients with T2DM mainly focuses on restoring cellular insulin response by potentiating the insulin-induced signaling pathway. In late-stage T2DM, impaired insulin production requires the patients to receive insulin replacement therapy for maintaining their glucose homeostasis. T2DM patients also demonstrate a drop of brain-derived neurotrophic factor (BDNF) in their circulation, which suggests that replenishing BDNF or enhancing its downstream signaling pathway may be beneficial. Because of their protein nature, recombinant insulin or BDNF possess several limitations that hinder their clinical application in T2DM treatment. Thus, developing orally active "insulin pill" or "BDNF pill" is essential to provide a more convenient and effective therapy. This article reviews the current development of non-peptidyl chemicals that mimic insulin or BDNF and their potential as anti-diabetic agents.
Collapse
Affiliation(s)
- Chi Bun Chan
- School of Biological Sciences, The University of Hong Kong, Hong Kong
| | - Palak Ahuja
- School of Biological Sciences, The University of Hong Kong, Hong Kong
| | - Keqiang Ye
- Department of Pathology and Laboratory Medicine, Emory University of School of Medicine, Atlanta, GA, United States
| |
Collapse
|
42
|
Máderová D, Krumpolec P, Slobodová L, Schön M, Tirpáková V, Kovaničová Z, Klepochová R, Vajda M, Šutovský S, Cvečka J, Valkovič L, Turčáni P, Krššák M, Sedliak M, Tsai CL, Ukropcová B, Ukropec J. Acute and regular exercise distinctly modulate serum, plasma and skeletal muscle BDNF in the elderly. Neuropeptides 2019; 78:101961. [PMID: 31506171 DOI: 10.1016/j.npep.2019.101961] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 08/26/2019] [Accepted: 08/27/2019] [Indexed: 12/14/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) participates in orchestrating the adaptive response to exercise. However, the importance of transient changes in circulating BDNF for eliciting whole-body and skeletal muscle exercise benefits in humans remains relatively unexplored. Here, we investigated effects of acute aerobic exercise and 3-month aerobic-strength training on serum, plasma and skeletal muscle BDNF in twenty-two sedentary older individuals (69.0 ± 8.0 yrs., 9 M/13F). BDNF response to acute exercise was additionally evaluated in young trained individuals (25.1 ± 2.1 yrs., 3 M/5F). Acute aerobic exercise transiently increased serum BDNF in sedentary (16%, p = .007) but not in trained elderly or young individuals. Resting serum or plasma BDNF was not regulated by exercise training in the elderly. However, subtle training-related changes of serum BDNF positively correlated with improvements in walking speed (R = 0.59, p = .005), muscle mass (R = 0.43, p = .04) and cognitive performance (R = 0.41, p = .05) and negatively with changes in body fat (R = -0.43, p = .04) and triglyceridemia (R = -0.53, p = .01). Individuals who increased muscle BDNF protein in response to 3-month training (responders) displayed stronger acute exercise-induced increase in serum BDNF than non-responders (p = .006). In addition, muscle BDNF protein content positively correlated with type II-to-type I muscle fiber ratio (R = 0.587, p = .008) and with the rate of post-exercise muscle ATP re-synthesis (R = 0.703, p = .005). Contrary to serum, acute aerobic exercise resulted in a decline of plasma BDNF 1 h post-exercise in both elderly-trained (-34%, p = .002) and young-trained individuals (-48%, p = .034). Acute circulating BDNF regulation by exercise was dependent on the level of physical fitness and correlated with training-induced improvements in metabolic and cognitive functions. Our observations provide an indirect evidence that distinct exercise-induced changes in serum and plasma BDNF as well as training-related increase in muscle BDNF protein, paralleled by improvements in muscle and whole-body clinical phenotypes, are involved in the coordinated adaptive response to exercise in humans.
Collapse
Affiliation(s)
- Denisa Máderová
- Institute of Experimental Endocrinology, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Patrik Krumpolec
- Institute of Experimental Endocrinology, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Bratislava, Slovakia; Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Lucia Slobodová
- Institute of Experimental Endocrinology, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Bratislava, Slovakia; Institute of Pathophysiology, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Martin Schön
- Institute of Experimental Endocrinology, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Bratislava, Slovakia; Institute of Pathophysiology, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Veronika Tirpáková
- Institute of Sports Medicine, Faculty of Medicine, Slovak Medical University, Bratislava, Slovakia
| | - Zuzana Kovaničová
- Institute of Pathophysiology, Faculty of Medicine, Comenius University, Bratislava, Slovakia; Institute of Experimental Endocrinology, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Radka Klepochová
- High Field MR Centre, Department of Biomedical Imaging and Imaged-Guided Therapy, Medical University of Vienna, Vienna, Austria; Christian Doppler Laboratory for Clinical Molecular Imaging, MOLIMA, Medical University of Vienna, Vienna, Austria
| | - Matej Vajda
- Faculty of Physical Education and Sports, Comenius University, Bratislava, Slovakia
| | - Stanislav Šutovský
- 1st Department of Neurology, Faculty of Medicine, Comenius University & University Hospital Bratislava, Slovakia
| | - Ján Cvečka
- Faculty of Physical Education and Sports, Comenius University, Bratislava, Slovakia
| | - Ladislav Valkovič
- High Field MR Centre, Department of Biomedical Imaging and Imaged-Guided Therapy, Medical University of Vienna, Vienna, Austria; Oxford Centre for Clinical Magnetic Resonance Research (OCMR), University of Oxford, Oxford, United Kingdom
| | - Peter Turčáni
- 1st Department of Neurology, Faculty of Medicine, Comenius University & University Hospital Bratislava, Slovakia
| | - Martin Krššák
- High Field MR Centre, Department of Biomedical Imaging and Imaged-Guided Therapy, Medical University of Vienna, Vienna, Austria; Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria; Christian Doppler Laboratory for Clinical Molecular Imaging, MOLIMA, Medical University of Vienna, Vienna, Austria
| | - Milan Sedliak
- Faculty of Physical Education and Sports, Comenius University, Bratislava, Slovakia
| | - Chia-Liang Tsai
- Institute of Physical Education, Health and Leisure Studies, National Cheng Kung University, Tainan, Taiwan, ROC
| | - Barbara Ukropcová
- Institute of Experimental Endocrinology, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Bratislava, Slovakia; Institute of Pathophysiology, Faculty of Medicine, Comenius University, Bratislava, Slovakia; Faculty of Physical Education and Sports, Comenius University, Bratislava, Slovakia.
| | - Jozef Ukropec
- Institute of Experimental Endocrinology, Biomedical Research Center, University Science Park for Biomedicine, Slovak Academy of Sciences, Bratislava, Slovakia.
| |
Collapse
|
43
|
Wang Z, Wang SP, Shao Q, Li PF, Sun Y, Luo LZ, Yan XQ, Fan ZY, Hu J, Zhao J, Hang PZ, Du ZM. Brain-derived neurotrophic factor mimetic, 7,8-dihydroxyflavone, protects against myocardial ischemia by rebalancing optic atrophy 1 processing. Free Radic Biol Med 2019; 145:187-197. [PMID: 31574344 DOI: 10.1016/j.freeradbiomed.2019.09.033] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 09/26/2019] [Accepted: 09/27/2019] [Indexed: 01/09/2023]
Abstract
Brain-derived neurotrophic factor (BDNF)/tropomyosin-related kinase B (TrkB) pathway is associated with ischemic heart diseases (IHD). 7,8-dihydroxyflavone (7,8-DHF), BDNF mimetic, is a potent agonist of TrkB. We aimed to investigate the effects and the underlying mechanisms of 7,8-DHF on cardiac ischemia. Myocardial ischemic mouse model was induced by ligation of left anterior descending coronary artery. 7,8-DHF (5 mg/kg) was administered intraperitoneally two days after ischemia for four weeks. Echocardiography, HE staining and transmission electron microscope were used to examine the function, histology and ultrastructure of the heart. H9c2 cells were treated with hydrogen peroxide (H2O2), 7,8-DHF or TrkB inhibitor ANA-12. The effects of 7,8-DHF on cell viability, mitochondrial membrane potential (MMP) and mitochondrial superoxide generation were examined. Furthermore, mitochondrial fission and protein expression of mitochondrial dynamics (Mfn2 [mitofusin 2], OPA1 [optic atrophy 1], Drp1 [dynamin-related protein 1] and Fis-1 [fission 1]) was detected by mitotracker green staining and western blot, respectively. 7,8-DHF attenuated cardiac dysfunction and cardiomyocyte abnormality of myocardial ischemic mice. Moreover, 7,8-DHF increased cell viability and reduced cell death accompanied by improving MMP, inhibiting mitochondrial superoxide and preventing excessive mitochondrial fission of H2O2-treated H9c2 cells. The cytoprotective effects of 7,8-DHF were antagonized by ANA-12. Mechanistically, 7,8-DHF repressed OMA1-dependent conversion of L-OPA1 into S-OPA1, which was abolished by Akt inhibitor. In conclusion, 7,8-DHF protects against cardiac ischemic injury by inhibiting the proteolytic cleavage of OPA1. These findings provide a novel pharmacological effect of 7,8-DHF on mitochondrial dynamics and a new potential target for IHD.
Collapse
Affiliation(s)
- Zhen Wang
- Institute of Clinical Pharmacology, The Second Affiliated Hospital (The University Key Laboratory of Drug Research, Heilongjiang Province), Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150086, China
| | - Shi-Peng Wang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Qun Shao
- Department of Cardiology, The Third Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Pei-Feng Li
- Institute of Clinical Pharmacology, The Second Affiliated Hospital (The University Key Laboratory of Drug Research, Heilongjiang Province), Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150086, China
| | - Yue Sun
- Institute of Clinical Pharmacology, The Second Affiliated Hospital (The University Key Laboratory of Drug Research, Heilongjiang Province), Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150086, China
| | - Lan-Zi Luo
- Institute of Clinical Pharmacology, The Second Affiliated Hospital (The University Key Laboratory of Drug Research, Heilongjiang Province), Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150086, China
| | - Xiu-Qing Yan
- Institute of Clinical Pharmacology, The Second Affiliated Hospital (The University Key Laboratory of Drug Research, Heilongjiang Province), Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150086, China
| | - Zi-Yi Fan
- Institute of Clinical Pharmacology, The Second Affiliated Hospital (The University Key Laboratory of Drug Research, Heilongjiang Province), Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150086, China
| | - Juan Hu
- Institute of Clinical Pharmacology, The Second Affiliated Hospital (The University Key Laboratory of Drug Research, Heilongjiang Province), Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150086, China
| | - Jing Zhao
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University (Key Laboratory of Cardiac Diseases and Heart Failure, Harbin Medical University), Harbin, 150001, China.
| | - Peng-Zhou Hang
- Institute of Clinical Pharmacology, The Second Affiliated Hospital (The University Key Laboratory of Drug Research, Heilongjiang Province), Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150086, China.
| | - Zhi-Min Du
- Institute of Clinical Pharmacology, The Second Affiliated Hospital (The University Key Laboratory of Drug Research, Heilongjiang Province), Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150086, China; State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China.
| |
Collapse
|
44
|
Kumar D, Dwivedi DK, Lahkar M, Jangra A. Hepatoprotective potential of 7,8-Dihydroxyflavone against alcohol and high-fat diet induced liver toxicity via attenuation of oxido-nitrosative stress and NF-κB activation. Pharmacol Rep 2019; 71:1235-1243. [PMID: 31670060 DOI: 10.1016/j.pharep.2019.07.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 05/11/2019] [Accepted: 07/06/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Fatty liver diseases are the most common and major health concern arises from the modern lifestyle and alcohol (ethanol) abuse. The prevalence of non-alcoholic fatty liver diseases (NAFLD) has been observed prominently in obese and diabetic individuals, while alcoholic liver disease is common in alcoholic persons. Fatty liver disease, such as steatohepatitis, leads to fibrosis, cirrhosis and eventually hepatocellular carcinoma. The present study was designed to investigate the effect of 7,8-Dihydroxyflavone (7,8-DHF) against high-fat diet (HFD) and ethanol (EtOH)-induced hepatotoxicity in rats. METHODS Male Wistar rats (150-200 g) were fed HFD (58% calories from fat) and EtOH (3-15% in drinking water) for 12 weeks. 7,8-DHF was administered intraperitoneally at the dose of 5 mg/kg/day for the last four weeks. After 12 weeks, biochemical, ELISA, RT-PCR, and histological studies have been carried out. RESULTS Biochemical analyses revealed the involvement of oxidative stress and inflammation in the liver of HFD and EtOH-fed rats. 7,8-DHF treatment significantly reduced HFD and EtOH-induced oxidative stress as evidenced by the reduction of lipid peroxidation and augmentation of reduced glutathione level. Moreover, IL-1β level was found significantly reduced in 7,8-DHF treated EtOH, HFD and EtOH+HFD groups. The semi-quantitative RT-PCR results indicated down-regulation of Nrf-2 and HO-1 and up-regulation of NF-κB and iNOS mRNA expression level in the liver of HFD and EtOH-fed rats, which was ameliorated by 7,8-DHF treatment. CONCLUSION The present study suggested that 7,8-DHF could be an effective pharmacological intervention in combating HFD and EtOH-induced hepatotoxicity.
Collapse
Affiliation(s)
- Dinesh Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, India
| | - Durgesh Kumar Dwivedi
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, India
| | - Mangala Lahkar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, India; Department of Pharmacology, Gauhati Medical College, Guwahati, Assam, India
| | - Ashok Jangra
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, India; Department of Pharmacology, KIET School of Pharmacy, Krishna Group of Institutions, Ghaziabad, Uttar Pradesh, India.
| |
Collapse
|
45
|
BDNF-mediated mitophagy alleviates high-glucose-induced brain microvascular endothelial cell injury. Apoptosis 2019; 24:511-528. [DOI: 10.1007/s10495-019-01535-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
46
|
Pilitsi E, Farr OM, Polyzos SA, Perakakis N, Nolen-Doerr E, Papathanasiou AE, Mantzoros CS. Pharmacotherapy of obesity: Available medications and drugs under investigation. Metabolism 2019; 92:170-192. [PMID: 30391259 DOI: 10.1016/j.metabol.2018.10.010] [Citation(s) in RCA: 150] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 10/13/2018] [Accepted: 10/23/2018] [Indexed: 02/07/2023]
Abstract
Obesity is a chronic disease with a continuously rising prevalence that currently affects more than half a billion people worldwide. Energy balance and appetite are highly regulated via central and peripheral mechanisms, and weight loss triggers a homeostatic response leading to weight regain. Lifestyle and behavioral modifications are the cornerstones of obesity management; however, they often fail to achieve or sustain long-term weight loss. Pharmacotherapy added onto lifestyle modifications results in an additional, albeit limited, weight reduction. Regardless, this weight reduction of 5-10% conveys multiple cardiovascular and metabolic benefits. In this review, evidence on the food and drug administration (FDA)-approved medications, i.e., orlistat, lorcaserin, phentermine/topiramate, liraglutide and naltrexone/bupropion, is summarized. Furthermore, anti-obesity agents in the pipeline for potential future therapeutic use are presented.
Collapse
Affiliation(s)
- Eleni Pilitsi
- Division of Endocrinology, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, MA 02215
| | - Olivia M Farr
- Division of Endocrinology, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, MA 02215.
| | - Stergios A Polyzos
- First Department of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Nikolaos Perakakis
- Division of Endocrinology, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, MA 02215
| | - Eric Nolen-Doerr
- Department of Medicine, Boston Medical Center, Boston, MA, 02118, United States of America
| | - Aimilia-Eirini Papathanasiou
- Division of Pediatric Newborn Medicine, Brigham and Women's Hospital/Harvard Medical School, Boston, MA 02215, United States of America
| | - Christos S Mantzoros
- Division of Endocrinology, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, MA 02215; Section of Endocrinology, VA Boston Healthcare System, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
47
|
Peradze N, Farr OM, Mantzoros CS. Research developments in metabolism 2018. Metabolism 2019; 91:70-79. [PMID: 30503805 DOI: 10.1016/j.metabol.2018.11.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 11/15/2018] [Indexed: 12/16/2022]
Affiliation(s)
- Natia Peradze
- Section of Endocrinology, Beth-Israel Deaconess Medical Center/Harvard Medical School, Boston, MA, United States of America.
| | - Olivia M Farr
- Section of Endocrinology, Beth-Israel Deaconess Medical Center/Harvard Medical School, Boston, MA, United States of America
| | - Christos S Mantzoros
- Section of Endocrinology, Beth-Israel Deaconess Medical Center/Harvard Medical School, Boston, MA, United States of America
| |
Collapse
|