1
|
Xu X, Wen Z. The mediating role of inflammaging between mitochondrial dysfunction and sarcopenia in aging: a review. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL IMMUNOLOGY 2023; 12:109-126. [PMID: 38187366 PMCID: PMC10767199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 11/26/2023] [Indexed: 01/09/2024]
Abstract
Sarcopenia, characterized by the insidious reduction of skeletal muscle mass and strength, detrimentally affects the quality of life in elderly cohorts. Present therapeutic strategies are confined to physiotherapeutic interventions, signaling a critical need for elucidation of the etiological underpinnings to facilitate the development of innovative pharmacotherapies. Recent scientific inquiries have associated mitochondrial dysfunction and inflammation with the etiology of sarcopenia. Mitochondria are integral to numerous fundamental cellular processes within muscle tissue, including but not limited to apoptosis, autophagy, signaling via reactive oxygen species, and the maintenance of protein equilibrium. Deviations in mitochondrial dynamics, coupled with compromised oxidative capabilities, autophagic processes, and protein equilibrium, result in disturbances to muscular architecture and functionality. Mitochondrial dysfunction is particularly detrimental as it diminishes oxidative phosphorylation, escalates apoptotic activity, and hinders calcium homeostasis within muscle cells. Additionally, deleterious feedback loops of deteriorated respiration, exacerbated oxidative injury, and diminished quality control mechanisms precipitate the acceleration of muscular senescence. Notably, mitochondria exhibiting deficient energetic metabolism are pivotal in precipitating the shift from normative muscle aging to a pathogenic state. This analytical review meticulously examines the complex interplay between mitochondrial dysfunction, persistent inflammation, and the pathogenesis of sarcopenia. It underscores the imperative to alleviate inflammation and amend mitochondrial anomalies within geriatric populations as a strategy to forestall and manage sarcopenia. An initial overview provides a succinct exposition of sarcopenia and its clinical repercussions. The discourse then progresses to an examination of the direct correlation between mitochondrial dysfunction and the genesis of sarcopenia. Concomitantly, it accentuates potential synergistic effects between inflammatory responses and mitochondrial insufficiencies during the aging of skeletal muscle, thereby casting light upon emergent therapeutic objectives. In culmination, this review distills the prevailing comprehension of the mitochondrial and inflammatory pathways implicated in sarcopenia and delineates extant lacunae in knowledge to orient subsequent scientific inquiry.
Collapse
Affiliation(s)
- Xin Xu
- Department of Rehabilitation Therapy, School of Health, Shanghai Normal University Tianhua CollegeShanghai, China
| | - Zixing Wen
- Department of Rehabilitation, School of International Medical Technology, Shanghai Sanda UniversityShanghai, China
| |
Collapse
|
2
|
Scrimieri R, Locatelli L, Cazzaniga A, Cazzola R, Malucelli E, Sorrentino A, Iotti S, Maier JA. Ultrastructural features mirror metabolic derangement in human endothelial cells exposed to high glucose. Sci Rep 2023; 13:15133. [PMID: 37704683 PMCID: PMC10499809 DOI: 10.1038/s41598-023-42333-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 09/08/2023] [Indexed: 09/15/2023] Open
Abstract
High glucose-induced endothelial dysfunction is the early event that initiates diabetes-induced vascular disease. Here we employed Cryo Soft X-ray Tomography to obtain three-dimensional maps of high D-glucose-treated endothelial cells and their controls at nanometric spatial resolution. We then correlated ultrastructural differences with metabolic rewiring. While the total mitochondrial mass does not change, high D-glucose promotes mitochondrial fragmentation, as confirmed by the modulation of fission-fusion markers, and dysfunction, as demonstrated by the drop of membrane potential, the decreased oxygen consumption and the increased production of reactive oxygen species. The 3D ultrastructural analysis also indicates the accumulation of lipid droplets in cells cultured in high D-glucose. Indeed, because of the decrease of fatty acid β-oxidation induced by high D-glucose concentration, triglycerides are esterified into fatty acids and then stored into lipid droplets. We propose that the increase of lipid droplets represents an adaptive mechanism to cope with the overload of glucose and associated oxidative stress and metabolic dysregulation.
Collapse
Affiliation(s)
- Roberta Scrimieri
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157, Milan, Italy.
| | - Laura Locatelli
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157, Milan, Italy
| | - Alessandra Cazzaniga
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157, Milan, Italy
| | - Roberta Cazzola
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157, Milan, Italy
| | - Emil Malucelli
- Department of Pharmacy and Biotechnology, Università di Bologna, 40127, Bologna, Italy
| | - Andrea Sorrentino
- Mistral Beamline, ALBA Synchrotron Light Source, Cerdanyola del Valles, 08290, Barcelona, Spain
| | - Stefano Iotti
- Department of Pharmacy and Biotechnology, Università di Bologna, 40127, Bologna, Italy
- National Institute of Biostructures and Biosystems, Viale Delle Medaglie d'Oro 305, 00136, Rome, Italy
| | - Jeanette A Maier
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157, Milan, Italy.
| |
Collapse
|
3
|
Belosludtseva NV, Serov DA, Starinets VS, Penkov NV, Belosludtsev KN. Alterations in Mitochondrial Morphology and Quality Control in Primary Mouse Lung Microvascular Endothelial Cells and Human Dermal Fibroblasts under Hyperglycemic Conditions. Int J Mol Sci 2023; 24:12485. [PMID: 37569860 PMCID: PMC10419261 DOI: 10.3390/ijms241512485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/03/2023] [Accepted: 08/05/2023] [Indexed: 08/13/2023] Open
Abstract
The effect of hyperglycemia on the morphology of individual mitochondria and the state of the mitochondrial network in primary mouse lung microvascular endotheliocytes and human dermal fibroblasts has been investigated. The cells were exposed to high (30 mM) and low (5.5 mM) glucose concentrations for 36 h. In primary endotheliocytes, hyperglycemic stress induced a significant increase in the number of mitochondria and a decrease in the interconnectivity value of the mitochondrial network, which was associated with a decrease in the mean size of the mitochondria. Analysis of the mRNA level of the genes of proteins responsible for mitochondrial biogenesis and mitophagy revealed an increase in the expression level of the Ppargc1a, Pink1, and Parkin genes, indicating stimulated mitochondrial turnover in endotheliocytes under high glucose conditions. In primary fibroblasts, hyperglycemia caused a decrease in the number of mitochondria and an increase in their size. As a result, the mitochondria exhibited higher values for elongation. In parallel, the mRNA level of the Ppargc1a and Mfn2 genes in fibroblasts exposed to hyperglycemia was reduced. These findings indicate that high glucose concentrations induced cell-specific morphological rearrangements of individual mitochondria and the mitochondrial network, which may be relevant during mitochondria-targeted drug testing and therapy for hyperglycemic and diabetic conditions.
Collapse
Affiliation(s)
- Natalia V. Belosludtseva
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia;
| | - Dmitriy A. Serov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, Vavilov St. 38, 119991 Moscow, Russia;
- Institute of Cell Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia;
| | - Vlada S. Starinets
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia;
| | - Nikita V. Penkov
- Institute of Cell Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia;
| | - Konstantin N. Belosludtsev
- Department of Biochemistry, Cell Biology and Microbiology, Mari State University, pl. Lenina 1, 424001 Yoshkar-Ola, Russia
| |
Collapse
|
4
|
Kadam A, Jadiya P, Tomar D. Post-translational modifications and protein quality control of mitochondrial channels and transporters. Front Cell Dev Biol 2023; 11:1196466. [PMID: 37601094 PMCID: PMC10434574 DOI: 10.3389/fcell.2023.1196466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 07/24/2023] [Indexed: 08/22/2023] Open
Abstract
Mitochondria play a critical role in energy metabolism and signal transduction, which is tightly regulated by proteins, metabolites, and ion fluxes. Metabolites and ion homeostasis are mainly mediated by channels and transporters present on mitochondrial membranes. Mitochondria comprise two distinct compartments, the outer mitochondrial membrane (OMM) and the inner mitochondrial membrane (IMM), which have differing permeabilities to ions and metabolites. The OMM is semipermeable due to the presence of non-selective molecular pores, while the IMM is highly selective and impermeable due to the presence of specialized channels and transporters which regulate ion and metabolite fluxes. These channels and transporters are modulated by various post-translational modifications (PTMs), including phosphorylation, oxidative modifications, ions, and metabolites binding, glycosylation, acetylation, and others. Additionally, the mitochondrial protein quality control (MPQC) system plays a crucial role in ensuring efficient molecular flux through the mitochondrial membranes by selectively removing mistargeted or defective proteins. Inefficient functioning of the transporters and channels in mitochondria can disrupt cellular homeostasis, leading to the onset of various pathological conditions. In this review, we provide a comprehensive overview of the current understanding of mitochondrial channels and transporters in terms of their functions, PTMs, and quality control mechanisms.
Collapse
Affiliation(s)
- Ashlesha Kadam
- Department of Internal Medicine, Section of Cardiovascular Medicine, Section of Molecular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Pooja Jadiya
- Department of Internal Medicine, Section of Gerontology and Geriatric Medicine, Section of Molecular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Dhanendra Tomar
- Department of Internal Medicine, Section of Cardiovascular Medicine, Section of Molecular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
5
|
Pilotto AM, Adami A, Mazzolari R, Brocca L, Crea E, Zuccarelli L, Pellegrino MA, Bottinelli R, Grassi B, Rossiter HB, Porcelli S. Near-infrared spectroscopy estimation of combined skeletal muscle oxidative capacity and O 2 diffusion capacity in humans. J Physiol 2022; 600:4153-4168. [PMID: 35930524 PMCID: PMC9481735 DOI: 10.1113/jp283267] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 07/27/2022] [Indexed: 01/05/2023] Open
Abstract
The final steps of the O2 cascade during exercise depend on the product of the microvascular-to-intramyocyteP O 2 ${P}_{{{\rm{O}}}_{\rm{2}}}$ difference and muscle O2 diffusing capacity (D m O 2 $D{{\rm{m}}}_{{{\rm{O}}}_2}$ ). Non-invasive methods to determineD m O 2 $D{{\rm{m}}}_{{{\rm{O}}}_2}$ in humans are currently unavailable. Muscle oxygen uptake (mV ̇ O 2 ${\dot{V}}_{{{\rm{O}}}_{\rm{2}}}$ ) recovery rate constant (k), measured by near-infrared spectroscopy (NIRS) using intermittent arterial occlusions, is associated with muscle oxidative capacity in vivo. We reasoned that k would be limited byD m O 2 $D{{\rm{m}}}_{{{\rm{O}}}_2}$ when muscle oxygenation is low (kLOW ), and hypothesized that: (i) k in well oxygenated muscle (kHIGH ) is associated with maximal O2 flux in fibre bundles; and (ii) ∆k (kHIGH - kLOW ) is associated with capillary density (CD). Vastus lateralis k was measured in 12 participants using NIRS after moderate exercise. The timing and duration of arterial occlusions were manipulated to maintain tissue saturation index within a 10% range either below (LOW) or above (HIGH) half-maximal desaturation, assessed during sustained arterial occlusion. Maximal O2 flux in phosphorylating state was 37.7 ± 10.6 pmol s-1 mg-1 (∼5.8 ml min-1 100 g-1 ). CD ranged 348 to 586 mm-2 . kHIGH was greater than kLOW (3.15 ± 0.45 vs. 1.56 ± 0.79 min-1 , P < 0.001). Maximal O2 flux was correlated with kHIGH (r = 0.80, P = 0.002) but not kLOW (r = -0.10, P = 0.755). Δk ranged -0.26 to -2.55 min-1 , and correlated with CD (r = -0.68, P = 0.015). mV ̇ O 2 ${\dot{V}}_{{{\rm{O}}}_{\rm{2}}}$ k reflects muscle oxidative capacity only in well oxygenated muscle. ∆k, the difference in k between well and poorly oxygenated muscle, was associated with CD, a mediator ofD m O 2 $D{{\rm{m}}}_{{{\rm{O}}}_2}$ . Assessment of muscle k and ∆k using NIRS provides a non-invasive window on muscle oxidative and O2 diffusing capacity. KEY POINTS: We determined post-exercise recovery kinetics of quadriceps muscle oxygen uptake (mV ̇ O 2 ${\dot{V}}_{{{\rm{O}}}_{\rm{2}}}$ ) measured by near-infrared spectroscopy (NIRS) in humans under conditions of both non-limiting (HIGH) and limiting (LOW) O2 availability, for comparison with biopsy variables. The mV ̇ O 2 ${\dot{V}}_{{{\rm{O}}}_{\rm{2}}}$ recovery rate constant in HIGH O2 availability was hypothesized to reflect muscle oxidative capacity (kHIGH ) and the difference in k between HIGH and LOW O2 availability (∆k) was hypothesized to reflect muscle O2 diffusing capacity. kHIGH was correlated with phosphorylating oxidative capacity of permeabilized muscle fibre bundles (r = 0.80). ∆k was negatively correlated with capillary density (r = -0.68) of biopsy samples. NIRS provides non-invasive means of assessing both muscle oxidative and oxygen diffusing capacity in vivo.
Collapse
Affiliation(s)
- Andrea M. Pilotto
- Department of MedicineUniversity of UdineUdineItaly
- Department of Molecular MedicineInstitute of PhysiologyUniversity of PaviaPaviaItaly
| | - Alessandra Adami
- Department of KinesiologyUniversity of Rhode IslandKingstonRIUSA
| | - Raffaele Mazzolari
- Department of Molecular MedicineInstitute of PhysiologyUniversity of PaviaPaviaItaly
- Department of Physical Education and SportUniversity of the Basque Country (UPV/EHU)Vitoria‐GasteizSpain
| | - Lorenza Brocca
- Department of Molecular MedicineInstitute of PhysiologyUniversity of PaviaPaviaItaly
| | - Emanuela Crea
- Department of Molecular MedicineInstitute of PhysiologyUniversity of PaviaPaviaItaly
| | | | - Maria A. Pellegrino
- Department of Molecular MedicineInstitute of PhysiologyUniversity of PaviaPaviaItaly
- Interdipartimental Centre for Biology and Sport MedicineUniversity of PaviaPaviaItaly
| | - Roberto Bottinelli
- Department of Molecular MedicineInstitute of PhysiologyUniversity of PaviaPaviaItaly
- Interdipartimental Centre for Biology and Sport MedicineUniversity of PaviaPaviaItaly
| | - Bruno Grassi
- Department of MedicineUniversity of UdineUdineItaly
| | - Harry B. Rossiter
- Division of Respiratory and Critical Care Physiology and MedicineThe Lundquist Institute for Biomedical Innovation at Harbor–UCLA Medical CenterTorranceCAUSA
| | - Simone Porcelli
- Department of Molecular MedicineInstitute of PhysiologyUniversity of PaviaPaviaItaly
- Institute of Biomedical TechnologiesNational Research CouncilMilanItaly
| |
Collapse
|
6
|
Li H, Chen X, Chen D, Yu B, He J, Zheng P, Luo Y, Yan H, Chen H, Huang Z. Ellagic Acid Alters Muscle Fiber-Type Composition and Promotes Mitochondrial Biogenesis through the AMPK Signaling Pathway in Healthy Pigs. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:9779-9789. [PMID: 35916165 DOI: 10.1021/acs.jafc.2c04108] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Ellagic acid (EA), because of its remarkable health-promoting ability, has aroused widespread interest in the fields of nutrition and medicine. However, no reports showed that EA regulates mitochondrial biogenesis as well as muscle fiber-type composition in pigs. Our study found that dietary 75 and 150 mg/kg EA obviously augmented the slow myosin heavy chain (MyHC) protein level, the number of slow-twitch muscle fibers, and the activity of malate dehydrogenase (MDH) in the longissimus thoracis (LT) muscle of growing-finishing pigs. In contrast, dietary 75 and 150 mg/kg EA decreased the fast MyHC level, the number of fast-twitch muscle fibers, and the activity of lactate dehydrogenase (LDH) in the LT muscle. In addition, our further study found that dietary 75 and 150 mg/kg EA promoted the mitochondrial DNA (mtDNA) content, the mRNA expressions of ATP synthase (ATP5G), mtDNA transcription factor A (TFAM), AMP-activated protein kinase α1 (AMPKα1), peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) and sirtuin 1 (Sirt1), and the level of phospho-LKB1 (P-LKB1), phospho-AMPK (P-AMPK), Sirt1, and PGC-1α in the LT muscle. In vitro, 5, 10, and 20 μmol/L EA treatment upregulated the level of slow MyHC, but only 10 μmol/L EA treatment decreased fast MyHC protein expression in porcine skeletal muscle satellite cells (PSCs). In addition, our data again found that 10 μmol/L EA treatment promoted the mtDNA content, the mRNA levels of ATP5G, mitochondrial transcription factor b1 (TFB1M), citrate synthase (Cs), AMPKα1, PGC-1α, and Sirt1, and the protein expressions of P-AMPK, P-LKB1, PGC-1α, and Sirt1 in PSCs. What is more, inhibition of the AMPK signaling pathway by AMPKα1 siRNA significantly eliminated the improvement of EA on muscle fiber-type composition as well as the mtDNA content in PSCs. In conclusion, EA altered muscle fiber-type composition and promoted mitochondrial biogenesis through the AMPK signaling pathway.
Collapse
Affiliation(s)
- Huawei Li
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan 611130, P. R. China
| | - Xiaoling Chen
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan 611130, P. R. China
| | - Daiwen Chen
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan 611130, P. R. China
| | - Bing Yu
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan 611130, P. R. China
| | - Jun He
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan 611130, P. R. China
| | - Ping Zheng
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan 611130, P. R. China
| | - Yuheng Luo
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan 611130, P. R. China
| | - Hui Yan
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan 611130, P. R. China
| | - Hong Chen
- College of Food Science, Sichuan Agricultural University, Yaan, Sichuan 625014, P. R. China
| | - Zhiqing Huang
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan 611130, P. R. China
| |
Collapse
|
7
|
Wang Z, Li Q, Hao Y, Wang Z, Yang H, Liu J, Wang J. Protective effect of 5-heptadecylresorcinol against obesity-associated skeletal muscle dysfunction by modulating mitochondrial biogenesis via the activation of SIRT3/PGC-1α signaling pathway. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105178] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
|
8
|
Kundu S, Hossain KS, Moni A, Zahan MS, Rahman MM, Uddin MJ. Potentials of ketogenic diet against chronic kidney diseases: pharmacological insights and therapeutic prospects. Mol Biol Rep 2022; 49:9749-9758. [PMID: 35441940 DOI: 10.1007/s11033-022-07460-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 04/06/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Chronic kidney disease (CKD) is a worldwide public health concern. Nutritional interventions become a primary concern in managing various diseases, including CKD. Ketogenic diets (KD) are a popular diet and an increasingly used diet for weight loss. MAIN BODY With the increasing cases of CKD, KD has been proposed as a treatment by many scientists. Several studies have shown that KD can slow down the progression rate of renal abnormalities. Also, this diet is regarded as a safe route for managing CKD. CKD is generally associated with increased inflammation, oxidative stress, fibrosis, autophagy dysfunction, and mitochondrial dysfunction, while all of these can be attenuated by KD. The protective effect of KD is mainly mediated through inhibition of ROS, NF-κB, and p62 signaling. CONCLUSIONS It is suggested that KD could be considered a new strategy for managing and treating CKD more carefully. This review explores the potential of KD on CKD and the mechanism involved in KD-mediated kidney protection.
Collapse
Affiliation(s)
- Sushmita Kundu
- ABEx Bio-Research Center, East Azampur, Dhaka, 1230, Bangladesh
| | | | - Akhi Moni
- ABEx Bio-Research Center, East Azampur, Dhaka, 1230, Bangladesh
| | - Md Sarwar Zahan
- ABEx Bio-Research Center, East Azampur, Dhaka, 1230, Bangladesh
| | - Md Masudur Rahman
- Department of Pathology, Faculty of Veterinary, Animal and Biomedical Sciences, Sylhet Agricultural University, Sylhet, 3100, Bangladesh
| | - Md Jamal Uddin
- ABEx Bio-Research Center, East Azampur, Dhaka, 1230, Bangladesh. .,Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, 03760, Republic of Korea.
| |
Collapse
|
9
|
Lee JK, Suh HN, Yoon SH, Lee KH, Ahn SY, Kim HJ, Kim SH. Non-Destructive Monitoring via Electrochemical NADH Detection in Murine Cells. BIOSENSORS 2022; 12:107. [PMID: 35200367 PMCID: PMC8869533 DOI: 10.3390/bios12020107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 01/25/2022] [Accepted: 02/08/2022] [Indexed: 06/14/2023]
Abstract
Nicotinamide adenine dinucleotide (NADH) is an important cofactor involved in metabolic redox reactions in living cells. The detection of NADH in living animal cells is a challenge. We developed a one-step monitoring method for NADH via an electrocatalytic reaction that uses a surface-modified, screen-printed electrode (SPE) having a redox active monolayer 4'-mercapto-N-phenlyquinone diamine (NPQD) formed by a self-assembled monolayer (SAM) of an aromatic thiol, 4-aminothiophenol (4-ATP). This electrode has a limit of detection (LOD) of 0.49 μM and a sensitivity of 0.0076 ± 0.0006 μM/μA in cell culture media, which indicates that it retains its selectivity. The applicability of this NADH sensor was demonstrated for the first time by cell viability monitoring via NADH-sensing in cell culture supernatants.
Collapse
Affiliation(s)
- Ju Kyung Lee
- Department of Medical IT Convergence, Kumoh National Institute of Technology, Gumi 39177, Korea;
| | - Han Na Suh
- Korea Institute of Toxicology, Jeongeup 56212, Korea; (H.N.S.); (S.H.Y.); (K.H.L.)
| | - Sung Hoon Yoon
- Korea Institute of Toxicology, Jeongeup 56212, Korea; (H.N.S.); (S.H.Y.); (K.H.L.)
- Department of Human and Environmental Toxicology, University of Science & Technology, Daejeon 34113, Korea
| | - Kyu Hong Lee
- Korea Institute of Toxicology, Jeongeup 56212, Korea; (H.N.S.); (S.H.Y.); (K.H.L.)
| | - Sae Young Ahn
- NDD Inc., Gumi 39253, Korea;
- Fuzbien Technology Institute, Rockville, MD 20850, USA
| | - Hyung Jin Kim
- Digital Health Care Research Center, Gumi Electronics and Information Technology Research Institute (GERI), Gumi 39253, Korea;
| | - Sang Hee Kim
- Department of Medical IT Convergence, Kumoh National Institute of Technology, Gumi 39177, Korea;
| |
Collapse
|
10
|
Zhao M, Lian A, Zhong L, Guo R. The regulatory mechanism between lysosomes and mitochondria in the aetiology of cardiovascular diseases. Acta Physiol (Oxf) 2022; 234:e13757. [PMID: 34978753 DOI: 10.1111/apha.13757] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 11/16/2021] [Accepted: 01/01/2022] [Indexed: 11/28/2022]
Abstract
Coordinated action among various organelles maintains cellular functions. For instance, mitochondria and lysosomes are the main organelles contributing to cellular metabolism and provide energy for cardiomyocyte contraction. They also provide essential signalling platforms in the cell that regulate many key processes such as autophagy, apoptosis, oxidative stress, inflammation and cell death. Often, abnormalities in mitochondrial or lysosomal structures and functions bring about cardiovascular diseases (CVDs). Although the communication between mitochondria and lysosomes throughout the cardiovascular system is intensely studied, the regulatory mechanisms have not been completely understood. Thus, we summarize the most recent studies related to mitochondria and lysosomes' role in CVDs and their potential connections and communications under cardiac pathophysiological conditions. Further, we discuss limitations and future perspectives regarding diagnosis, therapeutic strategies and drug discovery in CVDs.
Collapse
Affiliation(s)
- Mengxue Zhao
- College of Life Sciences Institute of Life Science and Green Development Hebei University Baoding China
| | - Andrew Lian
- College of Osteopathic Medicine of the Pacific Western University of Health Sciences Pomona California USA
| | - Li Zhong
- College of Life Sciences Institute of Life Science and Green Development Hebei University Baoding China
- College of Osteopathic Medicine of the Pacific Western University of Health Sciences Pomona California USA
| | - Rui Guo
- College of Life Sciences Institute of Life Science and Green Development Hebei University Baoding China
- The Key Laboratory of Zoological Systematics and Application College of Life Sciences Hebei University Baoding China
| |
Collapse
|
11
|
Differential remodelling of mitochondrial subpopulations and mitochondrial dysfunction are a feature of early stage diabetes. Sci Rep 2022; 12:978. [PMID: 35046471 PMCID: PMC8770458 DOI: 10.1038/s41598-022-04929-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 12/22/2021] [Indexed: 12/28/2022] Open
Abstract
Mitochondrial dysfunction is a feature of type I and type II diabetes, but there is a lack of consistency between reports and links to disease development. We aimed to investigate if mitochondrial structure–function remodelling occurs in the early stages of diabetes by employing a mouse model (GENA348) of Maturity Onset Diabetes in the Young, exhibiting hyperglycemia, but not hyperinsulinemia, with mild left ventricular dysfunction. Employing 3-D electron microscopy (SBF-SEM) we determined that compared to wild-type, WT, the GENA348 subsarcolemma mitochondria (SSM) are ~ 2-fold larger, consistent with up-regulation of fusion proteins Mfn1, Mfn2 and Opa1. Further, in comparison, GENA348 mitochondria are more irregular in shape, have more tubular projections with SSM projections being longer and wider. Mitochondrial density is also increased in the GENA348 myocardium consistent with up-regulation of PGC1-α and stalled mitophagy (down-regulation of PINK1, Parkin and Miro1). GENA348 mitochondria have more irregular cristae arrangements but cristae dimensions and density are similar to WT. GENA348 Complex activity (I, II, IV, V) activity is decreased but the OCR is increased, potentially linked to a shift towards fatty acid oxidation due to impaired glycolysis. These novel data reveal that dysregulated mitochondrial morphology, dynamics and function develop in the early stages of diabetes.
Collapse
|
12
|
Hogan HRH, Hutzenbiler BDE, Robbins CT, Jansen HT. Changing lanes: seasonal differences in cellular metabolism of adipocytes in grizzly bears (Ursus arctos horribilis). J Comp Physiol B 2022; 192:397-410. [PMID: 35024905 DOI: 10.1007/s00360-021-01428-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 12/07/2021] [Accepted: 12/20/2021] [Indexed: 12/21/2022]
Abstract
Obesity is among the most prevalent of health conditions in humans leading to a multitude of metabolic pathologies such as type 2 diabetes and hyperglycemia. However, there are many wild animals that have large seasonal cycles of fat accumulation and loss that do not result in the health consequences observed in obese humans. One example is the grizzly bear (Ursus arctos horribilis) that can have body fat content > 40% that is then used as the energy source for hibernation. Previous in vitro studies found that hibernation season adipocytes exhibit insulin resistance and increased lipolysis. Yet, other aspects of cellular metabolism were not addressed, leaving this in vitro model incomplete. Thus, the current studies were performed to determine if the cellular energetic phenotype-measured via metabolic flux-of hibernating bears was retained in cultured adipocytes and to what extent that was due to serum or intrinsic cellular factors. Extracellular acidification rate and oxygen consumption rate were used to calculate proton efflux rate and total ATP defined as both ATP from glycolysis and from mitochondrial respiration. Hibernation adipocytes treated with hibernation serum produced less ATP and exhibited lower maximal respiration and glycolysis rates than active season adipocytes. These effects were reversed with serum from the opposite season. Insulin had little influence on total ATP production and lipolysis in both hibernation and active serum-treated adipocytes. Together, these results suggest that the metabolic suppression occurring in hibernation adipocytes are downstream of insulin signaling and likely due to a combined reduction in mitochondria number and/or function and glycolytic processes. Future elucidation of the serum components and the cellular mechanisms that enable alterations in mitochondrial function could provide a novel avenue for the development of treatments for human metabolic diseases.
Collapse
Affiliation(s)
- Hannah R Hapner Hogan
- School of Biological Sciences, College of Arts and Sciences, Washington State University, Pullman, WA, 99164, USA.
| | - Brandon D E Hutzenbiler
- Department Integrative Physiology and Neuroscience, College of Veterinary Medicine, Washington State University, Pullman, WA, 99164, USA.,School of the Environment, College of Agricultural, Human and Natural Resource Sciences, Washington State University, Pullman, WA, 99164, USA
| | - Charles T Robbins
- School of Biological Sciences, College of Arts and Sciences, Washington State University, Pullman, WA, 99164, USA.,School of the Environment, College of Agricultural, Human and Natural Resource Sciences, Washington State University, Pullman, WA, 99164, USA
| | - Heiko T Jansen
- Department Integrative Physiology and Neuroscience, College of Veterinary Medicine, Washington State University, Pullman, WA, 99164, USA.
| |
Collapse
|
13
|
Energy transfer between the mitochondrial network and lipid droplets in insulin resistant skeletal muscle. CURRENT OPINION IN PHYSIOLOGY 2021; 24:100487. [PMID: 35274067 PMCID: PMC8903156 DOI: 10.1016/j.cophys.2022.100487] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Mitochondria and lipid droplets in the insulin resistant skeletal muscle of type 2 diabetic individuals have both been heavily investigated independently and are characterized by more fragmented, dysfunctional mitochondrial networks and larger lipid droplets compared to skeletal muscle of healthy individuals. Specialized contacts between mitochondrial and lipid droplet membranes are known to decrease in diabetic muscle, though it remains unclear how energy transfer at the remaining mitochondria-lipid droplet contact sites may be altered by type 2 diabetes. The purpose of this review is to highlight recent data on mitochondrial structure and function and lipid droplet dynamics in type 2 diabetic skeletal muscle and to underscore the need for more detailed investigations into the functional nature of mitochondria-lipid droplet interactions in type 2 diabetes.
Collapse
|
14
|
Laurindo CP, Rego Gregorio KC, Rippi Moreno AC, Viudes Agostinho JM, Campos EC, Nai GA, Nunes MT, Seraphim PM. Resistance training mitigates hepato-cardiac changes and muscle mitochondrial protein reductions in rats with diet-induced obesity. Heliyon 2021; 7:e08374. [PMID: 34841103 PMCID: PMC8605435 DOI: 10.1016/j.heliyon.2021.e08374] [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: 07/31/2021] [Revised: 09/25/2021] [Accepted: 11/10/2021] [Indexed: 11/05/2022] Open
Abstract
Aim To investigate the effect of resistance training (RT) on hepatocardiovascular and muscle mitochondrial parameters in rats that were fed a high-calorie diet for 12 weeks. Main methods The animals were divided into four groups: control (C), exercise (E), obese (O), and obese plus exercise (OE). Group E and OE rats performed resistance training by climbing on a vertical ladder with load attached to the end of the tail (1×/day, 3×/week, for 12 weeks). Group O and OE rats were fed a high-calorie diet containing chow and a cafeteria diet for 12 weeks. Under anesthesia, the heart and liver were removed for histopathological analysis, and the gastrocnemius muscle was removed for Western blotting. Key findings Group O rats were heavier, with increased fat mass, elevated fasting glycemia, and total triglycerides, and exhibited a significant number of Kupffer cells and diffuse steatosis in the liver. Group O rats also showed increased thickness of the right ventricle, septum, and pulmonary artery. All of these parameters were attenuated by RT. PGC1-α protein levels were increased in both exercise groups. The protein levels of OXPHOS complexes III, IV, and V were reduced in Group O, while RT prevented this alteration. Significance RT exerts a protective effect against hepato-cardiac alterations and prevents changes in the muscle mitochondrial protein profile induced by a high-calorie diet.
Collapse
Affiliation(s)
- Caroline Pancera Laurindo
- Department of Physiotherapy - School of Sciences and Technology - Sao Paulo, State University - UNESP, Campus Presidente Prudente, Brazil
| | - Karen C Rego Gregorio
- Department of Physiotherapy - School of Sciences and Technology - Sao Paulo, State University - UNESP, Campus Presidente Prudente, Brazil
| | - Ana Caroline Rippi Moreno
- Department of Physiotherapy - School of Sciences and Technology - Sao Paulo, State University - UNESP, Campus Presidente Prudente, Brazil.,Department of Physiology and Biophysics - Institute of Biomedical Sciences I, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Julia Maia Viudes Agostinho
- Department of Physiotherapy - School of Sciences and Technology - Sao Paulo, State University - UNESP, Campus Presidente Prudente, Brazil
| | - Evelyn Carvalho Campos
- Department of Physiotherapy - School of Sciences and Technology - Sao Paulo, State University - UNESP, Campus Presidente Prudente, Brazil
| | - Gisele Alborghetti Nai
- Department of Pathology - University of Western Sao Paulo, Presidente Prudente, SP, Brazil
| | - Maria Tereza Nunes
- Department of Physiology and Biophysics - Institute of Biomedical Sciences I, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Patrícia Monteiro Seraphim
- Department of Physiotherapy - School of Sciences and Technology - Sao Paulo, State University - UNESP, Campus Presidente Prudente, Brazil
| |
Collapse
|
15
|
Audzeyenka I, Rachubik P, Typiak M, Kulesza T, Topolewska A, Rogacka D, Angielski S, Saleem MA, Piwkowska A. Hyperglycemia alters mitochondrial respiration efficiency and mitophagy in human podocytes. Exp Cell Res 2021; 407:112758. [PMID: 34437881 DOI: 10.1016/j.yexcr.2021.112758] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 07/15/2021] [Accepted: 07/26/2021] [Indexed: 12/19/2022]
Abstract
Podocytes constitute the outer layer of the renal glomerular filtration barrier. Their energy requirements strongly depend on efficient oxidative respiration, which is tightly connected with mitochondrial dynamics. We hypothesized that hyperglycemia modulates energy metabolism in glomeruli and podocytes and contributes to the development of diabetic kidney disease. We found that oxygen consumption rates were severely reduced in glomeruli from diabetic rats and in human podocytes that were cultured in high glucose concentration (30 mM; HG). In these models, all of the mitochondrial respiratory parameters, including basal and maximal respiration, ATP production, and spare respiratory capacity, were significantly decreased. Podocytes that were treated with HG showed a fragmented mitochondrial network, together with a decrease in expression of the mitochondrial fusion markers MFN1, MFN2, and OPA1, and an increase in the activity of the fission marker DRP1. We showed that markers of mitochondrial biogenesis, such as PGC-1α and TFAM, decreased in HG-treated podocytes. Moreover, PINK1/parkin-dependent mitophagy was inhibited in these cells. These results provide evidence that hyperglycemia impairs mitochondrial dynamics and turnover, which may underlie the remarkable deterioration of mitochondrial respiration parameters in glomeruli and podocytes.
Collapse
Affiliation(s)
- Irena Audzeyenka
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Wita Stwosza St. 63, 80-308, Gdansk, Poland; Faculty of Chemistry, University of Gdansk, Wita Stwosza St. 63, 80-308, Gdansk, Poland.
| | - Patrycja Rachubik
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Wita Stwosza St. 63, 80-308, Gdansk, Poland
| | - Marlena Typiak
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Wita Stwosza St. 63, 80-308, Gdansk, Poland
| | - Tomasz Kulesza
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Wita Stwosza St. 63, 80-308, Gdansk, Poland
| | - Anna Topolewska
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Wita Stwosza St. 63, 80-308, Gdansk, Poland; Faculty of Chemistry, University of Gdansk, Wita Stwosza St. 63, 80-308, Gdansk, Poland
| | - Dorota Rogacka
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Wita Stwosza St. 63, 80-308, Gdansk, Poland; Faculty of Chemistry, University of Gdansk, Wita Stwosza St. 63, 80-308, Gdansk, Poland
| | - Stefan Angielski
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Wita Stwosza St. 63, 80-308, Gdansk, Poland
| | - Moin A Saleem
- Bristol Renal, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, United Kingdom
| | - Agnieszka Piwkowska
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Wita Stwosza St. 63, 80-308, Gdansk, Poland; Faculty of Chemistry, University of Gdansk, Wita Stwosza St. 63, 80-308, Gdansk, Poland
| |
Collapse
|
16
|
Nan J, Li J, Lin Y, Saif Ur Rahman M, Li Z, Zhu L. The interplay between mitochondria and store-operated Ca 2+ entry: Emerging insights into cardiac diseases. J Cell Mol Med 2021; 25:9496-9512. [PMID: 34564947 PMCID: PMC8505841 DOI: 10.1111/jcmm.16941] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 08/20/2021] [Accepted: 09/08/2021] [Indexed: 12/14/2022] Open
Abstract
Store‐operated Ca2+ entry (SOCE) machinery, including Orai channels, TRPCs, and STIM1, is key to cellular calcium homeostasis. The following characteristics of mitochondria are involved in the physiological and pathological regulation of cells: mitochondria mediate calcium uptake through calcium uniporters; mitochondria are regulated by mitochondrial dynamic related proteins (OPA1, MFN1/2, and DRP1) and form mitochondrial networks through continuous fission and fusion; mitochondria supply NADH to the electron transport chain through the Krebs cycle to produce ATP; under stress, mitochondria will produce excessive reactive oxygen species to regulate mitochondria‐endoplasmic reticulum interactions and the related signalling pathways. Both SOCE and mitochondria play critical roles in mediating cardiac hypertrophy, diabetic cardiomyopathy, and cardiac ischaemia‐reperfusion injury. All the mitochondrial characteristics mentioned above are determinants of SOCE activity, and vice versa. Ca2+ signalling dictates the reciprocal regulation between mitochondria and SOCE under the specific pathological conditions of cardiomyocytes. The coupling of mitochondria and SOCE is essential for various pathophysiological processes in the heart. Herein, we review the research focussing on the reciprocal regulation between mitochondria and SOCE and provide potential interplay patterns in cardiac diseases.
Collapse
Affiliation(s)
- Jinliang Nan
- Provincial Key Cardiovascular Research Laboratory, Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang Province, Hangzhou, China
| | - Jiamin Li
- Provincial Key Cardiovascular Research Laboratory, Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang Province, Hangzhou, China
| | - Yinuo Lin
- Wenzhou Municipal Key Cardiovascular Research Laboratory, Department of Cardiology, The First Affiliated Hospital, Wenzhou Medical University, Zhejiang Province, Wenzhou, China
| | - Muhammad Saif Ur Rahman
- Zhejiang University-University of Edinburgh Biomedical Institute, Haining, Zhejiang, China.,Clinical Research Center, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
| | - Zhengzheng Li
- Department of Neurology, Research Institute of Experimental Neurobiology, The First Affiliated Hospital, Wenzhou Medical University, Zhejiang Province, Wenzhou, China
| | - Lingjun Zhu
- Provincial Key Cardiovascular Research Laboratory, Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang Province, Hangzhou, China
| |
Collapse
|
17
|
Belosludtsev KN, Starinets VS, Talanov EY, Mikheeva IB, Dubinin MV, Belosludtseva NV. Alisporivir Treatment Alleviates Mitochondrial Dysfunction in the Skeletal Muscles of C57BL/6NCrl Mice with High-Fat Diet/Streptozotocin-Induced Diabetes Mellitus. Int J Mol Sci 2021; 22:9524. [PMID: 34502433 PMCID: PMC8430760 DOI: 10.3390/ijms22179524] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 08/26/2021] [Accepted: 08/30/2021] [Indexed: 01/20/2023] Open
Abstract
Diabetes mellitus is a systemic metabolic disorder associated with mitochondrial dysfunction, with mitochondrial permeability transition (MPT) pore opening being recognized as one of its pathogenic mechanisms. Alisporivir has been recently identified as a non-immunosuppressive analogue of the MPT pore blocker cyclosporin A and has broad therapeutic potential. The purpose of the present work was to study the effect of alisporivir (2.5 mg/kg/day i.p.) on the ultrastructure and functions of the skeletal muscle mitochondria of mice with diabetes mellitus induced by a high-fat diet combined with streptozotocin injections. The glucose tolerance tests indicated that alisporivir increased the rate of glucose utilization in diabetic mice. An electron microscopy analysis showed that alisporivir prevented diabetes-induced changes in the ultrastructure and content of the mitochondria in myocytes. In diabetes, the ADP-stimulated respiration, respiratory control, and ADP/O ratios and the level of ATP synthase in the mitochondria decreased, whereas alisporivir treatment restored these indicators. Alisporivir eliminated diabetes-induced increases in mitochondrial lipid peroxidation products. Diabetic mice showed decreased mRNA levels of Atp5f1a, Ant1, and Ppif and increased levels of Ant2 in the skeletal muscles. The skeletal muscle mitochondria of diabetic animals were sensitized to the MPT pore opening. Alisporivir normalized the expression level of Ant2 and mitochondrial susceptibility to the MPT pore opening. In parallel, the levels of Mfn2 and Drp1 also returned to control values, suggesting a normalization of mitochondrial dynamics. These findings suggest that the targeting of the MPT pore opening by alisporivir is a therapeutic approach to prevent the development of mitochondrial dysfunction and associated oxidative stress in the skeletal muscles in diabetes.
Collapse
Affiliation(s)
- Konstantin N. Belosludtsev
- Department of Biochemistry, Cell Biology and Microbiology, Mari State University, pl. Lenina 1, 424001 Yoshkar-Ola, Russia; (V.S.S.); (M.V.D.)
| | - Vlada S. Starinets
- Department of Biochemistry, Cell Biology and Microbiology, Mari State University, pl. Lenina 1, 424001 Yoshkar-Ola, Russia; (V.S.S.); (M.V.D.)
- Laboratory of Mitochondrial Transport, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia; (E.Y.T.); (I.B.M.); (N.V.B.)
| | - Eugeny Yu. Talanov
- Laboratory of Mitochondrial Transport, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia; (E.Y.T.); (I.B.M.); (N.V.B.)
| | - Irina B. Mikheeva
- Laboratory of Mitochondrial Transport, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia; (E.Y.T.); (I.B.M.); (N.V.B.)
| | - Mikhail V. Dubinin
- Department of Biochemistry, Cell Biology and Microbiology, Mari State University, pl. Lenina 1, 424001 Yoshkar-Ola, Russia; (V.S.S.); (M.V.D.)
| | - Natalia V. Belosludtseva
- Laboratory of Mitochondrial Transport, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia; (E.Y.T.); (I.B.M.); (N.V.B.)
| |
Collapse
|
18
|
Qu Z, Zhou S, Li P, Liu C, Yuan B, Zhang S, Liu A. Natural products and skeletal muscle health. J Nutr Biochem 2021; 93:108619. [DOI: 10.1016/j.jnutbio.2021.108619] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 11/26/2020] [Accepted: 02/01/2021] [Indexed: 12/17/2022]
|
19
|
Bourebaba N, Kornicka-Garbowska K, Marycz K, Bourebaba L, Kowalczuk A. Laurus nobilis ethanolic extract attenuates hyperglycemia and hyperinsulinemia-induced insulin resistance in HepG2 cell line through the reduction of oxidative stress and improvement of mitochondrial biogenesis - Possible implication in pharmacotherapy. Mitochondrion 2021; 59:190-213. [PMID: 34091077 DOI: 10.1016/j.mito.2021.06.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 05/23/2021] [Accepted: 06/01/2021] [Indexed: 02/03/2023]
Abstract
The aim of this study was to establish the potential effect of Laurus nobilis ethanolic extract on improving insulin sensitivity and protecting liver cells from apoptosis, mitochondrial dysfunction, oxidative stress (OS), and inflammation; all of which considered as major alterations occurring during insulin resistance (IR) as well as diabetes onset, in hyperinsulinemic and hyperglycemic-induced HepG2 cell line. Thereby, L. nobilis ethanolic extract has been first chemically characterized using LC-MS/MS technique. Subsequently, HepG2 cells were pre-treated with an optimal concentration of L. nobilis ethanolic extract for 24 h, and then, subjected to 30 mM D-glucose and 500 nM insulin mixture for another 24 h in order to induce hyperinsulinemia and hyperglycaemia (HI/HG) status. Several parameters such as biocompatibility, hepatotoxicity, reactive oxygen species (ROS), mitochondrial transmembrane potential, dynamics, and metabolism, multicaspase activity, glucose uptake, in addition to genes and proteins expression levels were investigated. The obtained results showed that the bioactive extract of Laurus nobilis increased the number of living cells and their proliferation rate, significantly attenuated apoptosis by modulating pro-apoptotic pathways (p21, p53 and Bax genes), allowed a relative normalization of caspases-activity, and decreased the expression of inflammatory markers including c-Jun, NF-κB and Tlr4 transcripts. L. Nobilis ethanolic extract reduced considerably total intracellular ROS levels in challenged HepG2 cells, and regulated the mitochondrial OXPHOS pathway, demonstrating the potential antioxidant effect of the plant. Ethanolic plant extract increased insulin sensitivity, since an elevated expression of master transcripts responsible for insulin sensitivity including IRS1, IRS2, INSR was found. Taken together, obtained data suggest that L. nobilis ethanolic extract offers new insights in the development of potential antioxidant, insulin sensitizing as well as hepatoprotective drugs.
Collapse
Affiliation(s)
- Nabila Bourebaba
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375 Wrocław, Poland; International Institute of Translational Medicine, Jesionowa 11, Malin 55-114 Wisznia Mała, Poland
| | - Katarzyna Kornicka-Garbowska
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375 Wrocław, Poland; International Institute of Translational Medicine, Jesionowa 11, Malin 55-114 Wisznia Mała, Poland
| | - Krzysztof Marycz
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375 Wrocław, Poland; International Institute of Translational Medicine, Jesionowa 11, Malin 55-114 Wisznia Mała, Poland
| | - Lynda Bourebaba
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375 Wrocław, Poland; International Institute of Translational Medicine, Jesionowa 11, Malin 55-114 Wisznia Mała, Poland.
| | - Anna Kowalczuk
- National Medicines Institute, Chełmska 30/34, 00-725 Warsaw, Poland
| |
Collapse
|
20
|
Adipocyte-Mineralocorticoid Receptor Alters Mitochondrial Quality Control Leading to Mitochondrial Dysfunction and Senescence of Visceral Adipose Tissue. Int J Mol Sci 2021; 22:ijms22062881. [PMID: 33809055 PMCID: PMC8001019 DOI: 10.3390/ijms22062881] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 03/03/2021] [Accepted: 03/08/2021] [Indexed: 12/22/2022] Open
Abstract
Mineralocorticoid receptor (MR) expression is increased in the adipose tissue (AT) of obese patients and animals. We previously demonstrated that adipocyte-MR overexpression in mice (Adipo-MROE mice) is associated with metabolic alterations. Moreover, we showed that MR regulates mitochondrial dysfunction and cellular senescence in the visceral AT of obese db/db mice. Our hypothesis is that adipocyte-MR overactivation triggers mitochondrial dysfunction and cellular senescence, through increased mitochondrial oxidative stress (OS). Using the Adipo-MROE mice with conditional adipocyte-MR expression, we evaluated the specific effects of adipocyte-MR on global and mitochondrial OS, as well as on OS-induced damage. Mitochondrial function was assessed by high throughput respirometry. Molecular mechanisms were probed in AT focusing on mitochondrial quality control and senescence markers. Adipo-MROE mice exhibited increased mitochondrial OS and altered mitochondrial respiration, associated with reduced biogenesis and increased fission. This was associated with OS-induced DNA-damage and AT premature senescence. In conclusion, targeted adipocyte-MR overexpression leads to an imbalance in mitochondrial dynamics and regeneration, to mitochondrial dysfunction and to ageing in visceral AT. These data bring new insights into the MR-dependent AT dysfunction in obesity.
Collapse
|
21
|
Curcumin induces mitochondrial biogenesis by increasing cyclic AMP levels via phosphodiesterase 4A inhibition in skeletal muscle. Br J Nutr 2021; 126:1642-1650. [PMID: 33551001 DOI: 10.1017/s0007114521000490] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Previous research has suggested that curcumin potentially induces mitochondrial biogenesis in skeletal muscle via increasing cyclic AMP (cAMP) levels. However, the regulatory mechanisms for this phenomenon remain unknown. The purpose of the present study was to clarify the mechanism by which curcumin activates cAMP-related signalling pathways that upregulate mitochondrial biogenesis and respiration in skeletal muscle. METHODS The effect of curcumin treatment (i.p., 100 mg/kg-BW/d for 28 d) on mitochondrial biogenesis was determined in rats. The effects of curcumin and exercise (swimming for 2 h/d for 3 d) on the cAMP signalling pathway were determined in the absence and presence of phosphodiesterase (PDE) or protein kinase A (PKA) inhibitors. Mitochondrial respiration, citrate synthase (CS) activity, cAMP content and protein expression of cAMP/PKA signalling molecules were analysed. RESULTS Curcumin administration increased cytochrome c oxidase subunit (COX-IV) protein expression, and CS and complex I activity, consistent with the induction of mitochondrial biogenesis by curcumin. Mitochondrial respiration was not altered by curcumin treatment. Curcumin and PDE inhibition tended to increase cAMP levels with or without exercise. In addition, exercise increased the phosphorylation of phosphodiesterase 4A (PDE4A), whereas curcumin treatment strongly inhibited PDE4A phosphorylation regardless of exercise. Furthermore, curcumin promoted AMP-activated protein kinase (AMPK) phosphorylation and PPAR gamma coactivator (PGC-1α) deacetylation. Inhibition of PKA abolished the phosphorylation of AMPK. CONCLUSION The present results suggest that curcumin increases cAMP levels via inhibition of PDE4A phosphorylation, which induces mitochondrial biogenesis through a cAMP/PKA/AMPK signalling pathway. Our data also suggest the possibility that curcumin utilises a regulatory mechanism for mitochondrial biogenesis that is distinct from the exercise-induced mechanism in skeletal muscle.
Collapse
|
22
|
Houzelle A, Jörgensen JA, Schaart G, Daemen S, van Polanen N, Fealy CE, Hesselink MKC, Schrauwen P, Hoeks J. Human skeletal muscle mitochondrial dynamics in relation to oxidative capacity and insulin sensitivity. Diabetologia 2021; 64:424-436. [PMID: 33258025 PMCID: PMC7801361 DOI: 10.1007/s00125-020-05335-w] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 10/02/2020] [Indexed: 12/19/2022]
Abstract
AIMS/HYPOTHESIS Mitochondria operate in networks, adapting to external stresses and changes in cellular metabolic demand and are subject to various quality control mechanisms. On the basis of these traits, we here hypothesise that the regulation of mitochondrial networks in skeletal muscle is hampered in humans with compromised oxidative capacity and insulin sensitivity. METHODS In a cross-sectional design, we compared four groups of participants (selected from previous studies) ranging in aerobic capacity and insulin sensitivity, i.e. participants with type 2 diabetes (n = 11), obese participants without diabetes (n = 12), lean individuals (n = 10) and endurance-trained athletes (n = 12); basal, overnight fasted muscle biopsies were newly analysed for the current study and we compared the levels of essential mitochondrial dynamics and quality control regulatory proteins in skeletal muscle tissue. RESULTS Type 2 diabetes patients and obese participants were older than lean participants and athletes (58.6 ± 4.0 and 56.7 ± 7.2 vs 21.8 ± 2.5 and 25.1 ± 4.3 years, p < 0.001, respectively) and displayed a higher BMI (32.4 ± 3.7 and 31.0 ± 3.7 vs 22.1 ± 1.8 and 21.0 ± 1.5 kg/m2, p < 0.001, respectively) than lean individuals and endurance-trained athletes. Fission protein 1 (FIS1) and optic atrophy protein 1 (OPA1) protein content was highest in muscle from athletes and lowest in participants with type 2 diabetes and obesity, respectively (FIS1: 1.86 ± 0.79 vs 0.79 ± 0.51 AU, p = 0.002; and OPA1: 1.55 ± 0.64 vs 0.76 ± 0.52 AU, p = 0.014), which coincided with mitochondrial network fragmentation in individuals with type 2 diabetes, as assessed by confocal microscopy in a subset of type 2 diabetes patients vs endurance-trained athletes (n = 6). Furthermore, lean individuals and athletes displayed a mitonuclear protein balance that was different from obese participants and those with type 2 diabetes. Mitonuclear protein balance also associated with heat shock protein 60 (HSP60) protein levels, which were higher in athletes when compared with participants with obesity (p = 0.048) and type 2 diabetes (p = 0.002), indicative for activation of the mitochondrial unfolded protein response. Finally, OPA1, FIS1 and HSP60 correlated positively with aerobic capacity (r = 0.48, p = 0.0001; r = 0.55, p < 0.001 and r = 0.61, p < 0.0001, respectively) and insulin sensitivity (r = 0.40, p = 0.008; r = 0.44, p = 0.003 and r = 0.48, p = 0.001, respectively). CONCLUSIONS/INTERPRETATION Collectively, our data suggest that mitochondrial dynamics and quality control in skeletal muscle are linked to oxidative capacity in humans, which may play a role in the maintenance of muscle insulin sensitivity. CLINICAL TRIAL REGISTRY: numbers NCT00943059, NCT01298375 and NL1888 Graphical abstract.
Collapse
Affiliation(s)
- Alexandre Houzelle
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Johanna A Jörgensen
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Gert Schaart
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Sabine Daemen
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Nynke van Polanen
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Ciarán E Fealy
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Matthijs K C Hesselink
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Patrick Schrauwen
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Joris Hoeks
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands.
| |
Collapse
|
23
|
Han WM, Chen XC, Li GR, Wang Y. Acacetin Protects Against High Glucose-Induced Endothelial Cells Injury by Preserving Mitochondrial Function via Activating Sirt1/Sirt3/AMPK Signals. Front Pharmacol 2020; 11:607796. [PMID: 33519472 PMCID: PMC7844858 DOI: 10.3389/fphar.2020.607796] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 11/25/2020] [Indexed: 12/16/2022] Open
Abstract
The strategy of decreasing atherosclerotic cardiovascular disorder is imperative for reducing premature death and improving quality of life in patients with diabetes mellitus. The aim of this study was to investigate whether the natural flavone acacetin could protect against endothelial injury induced by high glucose and attenuate diabetes-accelerated atherosclerosis in streptozotocin-(STZ) induced diabetic ApoE−/− mice model. It was found that in human umbilical vein endothelial cells (HUVECs) cultured with normal 5.5 mM or high 33 mM glucose, acacetin (0.3–3 μM) exerted strong cytoprotective effects by reversing high glucose-induced viability reduction and reducing apoptosis and excess production of intracellular reactive oxygen species (ROS) and malondialdehyde in a concentration-dependent manner. Acacetin countered high glucose-induced depolarization of mitochondrial membrane potential and reduction of ATP product and mitoBcl-2/mitoBax ratio. Silencing Sirt3 abolished the beneficial effects of acacetin. Further analysis revealed that these effects of acacetin rely on Sirt1 activation by increasing NAD+ followed by increasing Sirt3, pAMPK and PGC-1α. In STZ-diabetic mice, acacetin significantly upregulated the decreased signaling molecules (i.e. SOD, Bcl-2, PGC-1α, pAMPK, Sirt3 and Sirt1) in aorta tissue and attenuated atherosclerosis. These results indicate that vascular endothelial protection of acacetin by activating Sirt1/Sirt3/AMPK signals is likely involved in alleviating diabetes-accelerated atherosclerosis by preserving mitochondrial function, which suggests that acacetin may be a drug candidate for treating cardiovascular disorder in patients with diabetes.
Collapse
Affiliation(s)
- Wei-Min Han
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Xu-Chang Chen
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Gui-Rong Li
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen, China.,Nanjing Amazigh Pharma Limited, Nanjing, China
| | - Yan Wang
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
24
|
Oladimeji O, Akinyelu J, Singh M. Nanomedicines for Subcellular Targeting: The Mitochondrial Perspective. Curr Med Chem 2020; 27:5480-5509. [PMID: 31763965 DOI: 10.2174/0929867326666191125092111] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 10/28/2019] [Accepted: 10/30/2019] [Indexed: 01/01/2023]
Abstract
BACKGROUND Over the past decade, there has been a surge in the number of mitochondrialactive therapeutics for conditions ranging from cancer to aging. Subcellular targeting interventions can modulate adverse intracellular processes unique to the compartments within the cell. However, there is a dearth of reviews focusing on mitochondrial nano-delivery, and this review seeks to fill this gap with regards to nanotherapeutics of the mitochondria. METHODS Besides its potential for a higher therapeutic index than targeting at the tissue and cell levels, subcellular targeting takes into account the limitations of systemic drug administration and significantly improves pharmacokinetics. Hence, an extensive literature review was undertaken and salient information was compiled in this review. RESULTS From literature, it was evident that nanoparticles with their tunable physicochemical properties have shown potential for efficient therapeutic delivery, with several nanomedicines already approved by the FDA and others in clinical trials. However, strategies for the development of nanomedicines for subcellular targeting are still emerging, with an increased understanding of dysfunctional molecular processes advancing the development of treatment modules. For optimal delivery, the design of an ideal carrier for subcellular delivery must consider the features of the diseased microenvironment. The functional and structural features of the mitochondria in the diseased state are highlighted and potential nano-delivery interventions for treatment and diagnosis are discussed. CONCLUSION This review provides an insight into recent advances in subcellular targeting, with a focus on en route barriers to subcellular targeting. The impact of mitochondrial dysfunction in the aetiology of certain diseases is highlighted, and potential therapeutic sites are identified.
Collapse
Affiliation(s)
- Olakunle Oladimeji
- Nano-Gene and Drug Delivery Group, Discipline of Biochemistry, School of Life Sciences, University of Kwa-Zulu Natal, Private Bag X54001, Durban, South Africa
| | - Jude Akinyelu
- Nano-Gene and Drug Delivery Group, Discipline of Biochemistry, School of Life Sciences, University of Kwa-Zulu Natal, Private Bag X54001, Durban, South Africa
| | - Moganavelli Singh
- Nano-Gene and Drug Delivery Group, Discipline of Biochemistry, School of Life Sciences, University of Kwa-Zulu Natal, Private Bag X54001, Durban, South Africa
| |
Collapse
|
25
|
Moore TM, Zhou Z, Strumwasser AR, Cohn W, Lin AJ, Cory K, Whitney K, Ho T, Ho T, Lee JL, Rucker DH, Hoang AN, Widjaja K, Abrishami AD, Charugundla S, Stiles L, Whitelegge JP, Turcotte LP, Wanagat J, Hevener AL. Age-induced mitochondrial DNA point mutations are inadequate to alter metabolic homeostasis in response to nutrient challenge. Aging Cell 2020; 19:e13166. [PMID: 33049094 PMCID: PMC7681042 DOI: 10.1111/acel.13166] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 04/10/2020] [Accepted: 04/27/2020] [Indexed: 12/14/2022] Open
Abstract
Mitochondrial dysfunction is frequently associated with impairment in metabolic homeostasis and insulin action, and is thought to underlie cellular aging. However, it is unclear whether mitochondrial dysfunction is a cause or consequence of insulin resistance in humans. To determine the impact of intrinsic mitochondrial dysfunction on metabolism and insulin action, we performed comprehensive metabolic phenotyping of the polymerase gamma (PolG) D257A "mutator" mouse, a model known to accumulate supraphysiological mitochondrial DNA (mtDNA) point mutations. We utilized the heterozygous PolG mutator mouse (PolG+/mut ) because it accumulates mtDNA point mutations ~ 500-fold > wild-type mice (WT), but fails to develop an overt progeria phenotype, unlike PolGmut/mut animals. To determine whether mtDNA point mutations induce metabolic dysfunction, we examined male PolG+/mut mice at 6 and 12 months of age during normal chow feeding, after 24-hr starvation, and following high-fat diet (HFD) feeding. No marked differences were observed in glucose homeostasis, adiposity, protein/gene markers of metabolism, or oxygen consumption in muscle between WT and PolG+/mut mice during any of the conditions or ages studied. However, proteomic analyses performed on isolated mitochondria from 12-month-old PolG+/mut mouse muscle revealed alterations in the expression of mitochondrial ribosomal proteins, electron transport chain components, and oxidative stress-related factors compared with WT. These findings suggest that mtDNA point mutations at levels observed in mammalian aging are insufficient to disrupt metabolic homeostasis and insulin action in male mice.
Collapse
Affiliation(s)
- Timothy M. Moore
- Department of Biological SciencesDana & David Dornsife College of Letters, Arts, and SciencesUniversity of Southern CaliforniaLos AngelesCAUSA
- Division of Endocrinology, Diabetes, and HypertensionDepartment of MedicineDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Zhenqi Zhou
- Division of Endocrinology, Diabetes, and HypertensionDepartment of MedicineDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Alexander R. Strumwasser
- Division of Endocrinology, Diabetes, and HypertensionDepartment of MedicineDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Whitaker Cohn
- Department of Psychiatry and Biobehavioral Sciences & The Semel Institute for Neuroscience and Human BehaviorUniversity of CaliforniaLos AngelesCAUSA
| | - Amanda J. Lin
- Division of Endocrinology, Diabetes, and HypertensionDepartment of MedicineDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Kevin Cory
- Division of Endocrinology, Diabetes, and HypertensionDepartment of MedicineDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Kate Whitney
- Division of Endocrinology, Diabetes, and HypertensionDepartment of MedicineDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Theodore Ho
- Division of Endocrinology, Diabetes, and HypertensionDepartment of MedicineDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Timothy Ho
- Division of Endocrinology, Diabetes, and HypertensionDepartment of MedicineDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Joseph L. Lee
- Division of Endocrinology, Diabetes, and HypertensionDepartment of MedicineDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Daniel H. Rucker
- Division of Endocrinology, Diabetes, and HypertensionDepartment of MedicineDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Austin N. Hoang
- Division of Endocrinology, Diabetes, and HypertensionDepartment of MedicineDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Kevin Widjaja
- Division of Endocrinology, Diabetes, and HypertensionDepartment of MedicineDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Aaron D. Abrishami
- Division of CardiologyDepartment of MedicineDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Sarada Charugundla
- Division of CardiologyDepartment of MedicineDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Linsey Stiles
- Division of Endocrinology, Diabetes, and HypertensionDepartment of MedicineDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Julian P. Whitelegge
- Department of Psychiatry and Biobehavioral Sciences & The Semel Institute for Neuroscience and Human BehaviorUniversity of CaliforniaLos AngelesCAUSA
| | - Lorraine P. Turcotte
- Department of Biological SciencesDana & David Dornsife College of Letters, Arts, and SciencesUniversity of Southern CaliforniaLos AngelesCAUSA
| | - Jonathan Wanagat
- Division of Endocrinology, Diabetes, and HypertensionDepartment of MedicineDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Andrea L. Hevener
- Division of Endocrinology, Diabetes, and HypertensionDepartment of MedicineDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Iris Cantor‐UCLA Women's Health CenterUniversity of CaliforniaLos AngelesCAUSA
| |
Collapse
|
26
|
Abu Bakar MH, Shariff KA, Tan JS, Lee LK. Celastrol attenuates inflammatory responses in adipose tissues and improves skeletal muscle mitochondrial functions in high fat diet-induced obese rats via upregulation of AMPK/SIRT1 signaling pathways. Eur J Pharmacol 2020; 883:173371. [DOI: 10.1016/j.ejphar.2020.173371] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 07/09/2020] [Accepted: 07/13/2020] [Indexed: 12/20/2022]
|
27
|
Díaz-Castro F, Monsalves-Álvarez M, Rojo LE, Del Campo A, Troncoso R. Mifepristone for Treatment of Metabolic Syndrome: Beyond Cushing's Syndrome. Front Pharmacol 2020; 11:429. [PMID: 32390830 PMCID: PMC7193078 DOI: 10.3389/fphar.2020.00429] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 03/19/2020] [Indexed: 12/19/2022] Open
Abstract
A growing body of research indicates that cortisol, the glucocorticoid product of the activation of the hypothalamic-pituitary-adrenal axis, plays a role in the pathophysiology of metabolic syndrome. In this regard, chronic exposure to cortisol is associated with risk factors related to metabolic syndrome like weight gain, type 2 diabetes, hypertension, among others. Mifepristone is the only FDA-approved drug with antiglucocorticoids properties for improved the glycemic control in patients with type 2 patients secondary to endogenous Cushing’s syndrome. Mifepristone also have been shown positive effects in rodents models of diabetes and patients with obesity due to antipsychotic treatment. However, the underlying molecular mechanisms are not fully understood. In this perspective, we summarized the literature regarding the beneficial effects of mifepristone in metabolic syndrome from animal studies to clinical research. Also, we propose a potential mechanism for the beneficial effects in insulin sensitivity which involved the regulation of mitochondrial function in muscle cells.
Collapse
Affiliation(s)
- Francisco Díaz-Castro
- Laboratorio de Investigación en Nutrición y Actividad Física (LABINAF), Instituto de Nutrición y Tecnología en Alimentos (INTA), Universidad de Chile, Santiago, Chile
| | - Matías Monsalves-Álvarez
- Laboratorio de Investigación en Nutrición y Actividad Física (LABINAF), Instituto de Nutrición y Tecnología en Alimentos (INTA), Universidad de Chile, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDIS), Universidad de Chile, Santiago, Chile
| | - Leonel E Rojo
- Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile.,Centro de Biotecnología Acuícola, Universidad de Santiago de Chile, Santiago, Chile
| | - Andrea Del Campo
- Departamento de Farmacia, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Rodrigo Troncoso
- Laboratorio de Investigación en Nutrición y Actividad Física (LABINAF), Instituto de Nutrición y Tecnología en Alimentos (INTA), Universidad de Chile, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDIS), Universidad de Chile, Santiago, Chile
| |
Collapse
|
28
|
Ponnalagu D, Singh H. Insights Into the Role of Mitochondrial Ion Channels in Inflammatory Response. Front Physiol 2020; 11:258. [PMID: 32327997 PMCID: PMC7160495 DOI: 10.3389/fphys.2020.00258] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 03/05/2020] [Indexed: 12/14/2022] Open
Abstract
Mitochondria are the source of many pro-inflammatory signals that cause the activation of the immune system and generate inflammatory responses. They are also potential targets of pro-inflammatory mediators, thus triggering a severe inflammatory response cycle. As mitochondria are a central hub for immune system activation, their dysfunction leads to many inflammatory disorders. Thus, strategies aiming at regulating mitochondrial dysfunction can be utilized as a therapeutic tool to cure inflammatory disorders. Two key factors that determine the structural and functional integrity of mitochondria are mitochondrial ion channels and transporters. They are not only important for maintaining the ionic homeostasis of the cell, but also play a role in regulating reactive oxygen species generation, ATP production, calcium homeostasis and apoptosis, which are common pro-inflammatory signals. The significance of the mitochondrial ion channels in inflammatory response is still not clearly understood and will need further investigation. In this article, we review the different mechanisms by which mitochondria can generate the inflammatory response as well as highlight how mitochondrial ion channels modulate these mechanisms and impact the inflammatory processes.
Collapse
Affiliation(s)
- Devasena Ponnalagu
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Wexner Medical Center, Columbus, OH, United States
| | - Harpreet Singh
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|
29
|
The Role of Reactive Oxygen Species in the Life Cycle of the Mitochondrion. Int J Mol Sci 2020; 21:ijms21062173. [PMID: 32245255 PMCID: PMC7139706 DOI: 10.3390/ijms21062173] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 03/12/2020] [Accepted: 03/19/2020] [Indexed: 02/07/2023] Open
Abstract
Currently, it is known that, in living systems, free radicals and other reactive oxygen and nitrogen species play a double role, because they can cause oxidative damage and tissue dysfunction and serve as molecular signals activating stress responses that are beneficial to the organism. It is also known that mitochondria, because of their capacity to produce free radicals, play a major role in tissue oxidative damage and dysfunction and provide protection against excessive tissue dysfunction through several mechanisms, including the stimulation of permeability transition pore opening. This process leads to mitoptosis and mitophagy, two sequential processes that are a universal route of elimination of dysfunctional mitochondria and is essential to protect cells from the harm due to mitochondrial disordered metabolism. To date, there is significant evidence not only that the above processes are induced by enhanced reactive oxygen species (ROS) production, but also that such production is involved in the other phases of the mitochondrial life cycle. Accumulating evidence also suggests that these effects are mediated through the regulation of the expression and the activity of proteins that are engaged in processes such as genesis, fission, fusion, and removal of mitochondria. This review provides an account of the developments of the knowledge on the dynamics of the mitochondrial population, examining the mechanisms governing their genesis, life, and death, and elucidating the role played by free radicals in such processes.
Collapse
|
30
|
Zhang Z, Cui D, Zhang T, Sun Y, Ding S. Swimming Differentially Affects T2DM-Induced Skeletal Muscle ER Stress and Mitochondrial Dysfunction Related to MAM. Diabetes Metab Syndr Obes 2020; 13:1417-1428. [PMID: 32431525 PMCID: PMC7203063 DOI: 10.2147/dmso.s243024] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 04/02/2020] [Indexed: 12/14/2022] Open
Abstract
PURPOSE Mitochondrial dysfunction and endoplasmic reticulum stress (ERS) are associated with metabolic diseases such as obesity and Type 2 diabetes mellitus (T2DM). Mitochondria and ER are connected via mitochondria-associated membranes (MAM) that are involved in glucose homeostasis and insulin resistance. We postulated that exercise might positively benefit T2DM-induced ER and mitochondrial dysfunction that might be associated with MAM. MATERIALS AND METHODS Mice were fed a high-fat diet and injected with streptozotocin (STZ) to create T2DM models. Glucose tolerance, mitochondrial quality, MAM quality, and ERS were investigated in diabetic mice after six weeks of swimming. RESULTS Type 2 DM induced decreased MAM quantity, impaired mitochondrial quality, and deteriorated ERS in skeletal muscle that led to endoplasmic reticulum-associated degradation (ERAD). Swimming alleviated strong ERS caused by T2DM. Importantly, MAM quantity was positively associated with mitochondrial function and tended to negatively correlate with the ERS branch, ATF6. Moreover, both ATF6 branches of ERS and ERAD were positively associated with the pIRE1α branch of ERS. CONCLUSION Type 2 DM induced glucose intolerance, powerful ERS, and mitochondrial dysfunction associated with decreased amounts of MAM. Swimming improved glucose intolerance and selectively mitigated the ERS in skeletal muscle. Therefore, MAM quality and ATF6 might be novel and important targets for T2DM treatment. Endoplasmic reticulum stress might be an effective target of swimming to improve diabetes.
Collapse
Affiliation(s)
- Zhe Zhang
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai200241, People’s Republic of China
- College of Physical Education and Health, East China Normal University, Shanghai200241, People’s Republic of China
| | - Di Cui
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai200241, People’s Republic of China
- College of Physical Education and Health, East China Normal University, Shanghai200241, People’s Republic of China
| | - Tan Zhang
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai200241, People’s Republic of China
- College of Physical Education and Health, East China Normal University, Shanghai200241, People’s Republic of China
| | - Yi Sun
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai200241, People’s Republic of China
- College of Physical Education and Health, East China Normal University, Shanghai200241, People’s Republic of China
| | - Shuzhe Ding
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai200241, People’s Republic of China
- College of Physical Education and Health, East China Normal University, Shanghai200241, People’s Republic of China
- Correspondence: Shuzhe Ding Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai200241, People’s Republic of ChinaTel +86 13601798505 Email
| |
Collapse
|
31
|
Zhang Y, Yu B, Yu J, Zheng P, Huang Z, Luo Y, Luo J, Mao X, Yan H, He J, Chen D. Butyrate promotes slow-twitch myofiber formation and mitochondrial biogenesis in finishing pigs via inducing specific microRNAs and PGC-1α expression1. J Anim Sci 2019; 97:3180-3192. [PMID: 31228349 DOI: 10.1093/jas/skz187] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 05/17/2019] [Indexed: 12/11/2022] Open
Abstract
The present study aimed to investigate the influence of dietary butyrate supplementation on muscle fiber-type composition and mitochondrial biogenesis of finishing pigs, and the underlying mechanisms. Thirty-two LY (Landrace × Yorkshire) growing pigs with BW of 64.9 ± 5.7 kg were randomly allotted to either control (basal diet) or butyrate diets (0.3% butyrate sodium). Compared with the control group, diet supplemented with butyrate tended to increase average daily gain (P < 0.10). Pigs fed butyrate diet had higher intramuscular fat content, marbling score and pH24 h, and lower shear force and L*24 h in longissimus thoracis (LT) muscle than that fed control diet (P < 0.05). Interestingly, supplemented with butyrate increased (P < 0.05) the mRNA level of myosin heavy chain I (MyHC-I) and the percentage of slow-fibers, and decreased (P < 0.05) the mRNA level of MyHC-IIb in LT muscle. Meanwhile, pigs in butyrate group had an increase in mitochondrial DNA (mtDNA) copy number and the mRNA levels of mtDNA-encoded genes (P < 0.05). Moreover, feeding butyrate diet increased PGC-1α (PPAR γ coactivator 1α) level, decreased miR-133a-3p level and increased its target gene level (TEAD1, TEA domain transcription factor 1), increased miR-208b and miR-499-5p levels and decreased their target genes levels (Sp3 and Sox6, specificity protein 3 and SRY-box containing gene 6; P < 0.05) in the LT muscle. Collectively, these findings suggested that butyrate promoted slow-twitch myofiber formation and mitochondrial biogenesis, and the molecular mechanism may be via upgrading specific microRNAs and PGC-1α expression, finally improving meat quality.
Collapse
Affiliation(s)
- Yong Zhang
- Institute of Animal Nutrition, Sichuan Agricultural University, and Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, People's Republic of China.,School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan, People's Republic of China
| | - Bing Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, and Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, People's Republic of China
| | - Jie Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, and Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, People's Republic of China
| | - Ping Zheng
- Institute of Animal Nutrition, Sichuan Agricultural University, and Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, People's Republic of China
| | - Zhiqing Huang
- Institute of Animal Nutrition, Sichuan Agricultural University, and Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, People's Republic of China
| | - Yuheng Luo
- Institute of Animal Nutrition, Sichuan Agricultural University, and Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, People's Republic of China
| | - Junqiu Luo
- Institute of Animal Nutrition, Sichuan Agricultural University, and Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, People's Republic of China
| | - Xiangbing Mao
- Institute of Animal Nutrition, Sichuan Agricultural University, and Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, People's Republic of China
| | - Honglin Yan
- Institute of Animal Nutrition, Sichuan Agricultural University, and Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, People's Republic of China.,School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan, People's Republic of China
| | - Jun He
- Institute of Animal Nutrition, Sichuan Agricultural University, and Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, People's Republic of China
| | - Daiwen Chen
- Institute of Animal Nutrition, Sichuan Agricultural University, and Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Chengdu, People's Republic of China
| |
Collapse
|
32
|
Malińska D, Więckowski MR, Michalska B, Drabik K, Prill M, Patalas-Krawczyk P, Walczak J, Szymański J, Mathis C, Van der Toorn M, Luettich K, Hoeng J, Peitsch MC, Duszyński J, Szczepanowska J. Mitochondria as a possible target for nicotine action. J Bioenerg Biomembr 2019; 51:259-276. [PMID: 31197632 PMCID: PMC6679833 DOI: 10.1007/s10863-019-09800-z] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 05/19/2019] [Indexed: 12/26/2022]
Abstract
Mitochondria are multifunctional and dynamic organelles deeply integrated into cellular physiology and metabolism. Disturbances in mitochondrial function are involved in several disorders such as neurodegeneration, cardiovascular diseases, metabolic diseases, and also in the aging process. Nicotine is a natural alkaloid present in the tobacco plant which has been well studied as a constituent of cigarette smoke. It has also been reported to influence mitochondrial function both in vitro and in vivo. This review presents a comprehensive overview of the present knowledge of nicotine action on mitochondrial function. Observed effects of nicotine exposure on the mitochondrial respiratory chain, oxidative stress, calcium homeostasis, mitochondrial dynamics, biogenesis, and mitophagy are discussed, considering the context of the experimental design. The potential action of nicotine on cellular adaptation and cell survival is also examined through its interaction with mitochondria. Although a large number of studies have demonstrated the impact of nicotine on various mitochondrial activities, elucidating its mechanism of action requires further investigation.
Collapse
Affiliation(s)
- Dominika Malińska
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warsaw, Poland
| | - Mariusz R Więckowski
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warsaw, Poland
| | - Bernadeta Michalska
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warsaw, Poland
| | - Karolina Drabik
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warsaw, Poland
| | - Monika Prill
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warsaw, Poland
| | - Paulina Patalas-Krawczyk
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warsaw, Poland
| | - Jarosław Walczak
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warsaw, Poland
| | - Jędrzej Szymański
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warsaw, Poland
| | - Carole Mathis
- PMI R&D, Philip Morris Products S.A. (part of Philip Morris International group of companies), Quai Jeanrenaud 5, 2000, Neuchâtel, Switzerland
| | - Marco Van der Toorn
- PMI R&D, Philip Morris Products S.A. (part of Philip Morris International group of companies), Quai Jeanrenaud 5, 2000, Neuchâtel, Switzerland
| | - Karsta Luettich
- PMI R&D, Philip Morris Products S.A. (part of Philip Morris International group of companies), Quai Jeanrenaud 5, 2000, Neuchâtel, Switzerland
| | - Julia Hoeng
- PMI R&D, Philip Morris Products S.A. (part of Philip Morris International group of companies), Quai Jeanrenaud 5, 2000, Neuchâtel, Switzerland
| | - Manuel C Peitsch
- PMI R&D, Philip Morris Products S.A. (part of Philip Morris International group of companies), Quai Jeanrenaud 5, 2000, Neuchâtel, Switzerland
| | - Jerzy Duszyński
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warsaw, Poland.
| | - Joanna Szczepanowska
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warsaw, Poland.
| |
Collapse
|
33
|
Fealy CE, Mulya A, Axelrod CL, Kirwan JP. Mitochondrial dynamics in skeletal muscle insulin resistance and type 2 diabetes. Transl Res 2018; 202:69-82. [PMID: 30153426 DOI: 10.1016/j.trsl.2018.07.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Revised: 07/08/2018] [Accepted: 07/23/2018] [Indexed: 01/09/2023]
Abstract
The traditional view of mitochondria as isolated, spherical, energy producing organelles, is undergoing a revolutionary change. Emerging data show that mitochondria form a dynamic reticulum that is regulated by cycles of fission and fusion. The discovery of proteins that modulate these activities has led to important advances in understanding human disease. Here, we review the latest evidence that connects the emerging field of mitochondrial dynamics to skeletal muscle insulin resistance and propose some potential mechanisms that may explain the long debated link between mitochondria and the development of type 2 diabetes.
Collapse
Affiliation(s)
- CiarÁn E Fealy
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Anny Mulya
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Christopher L Axelrod
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio; Integrated Physiology and Molecular Metabolism Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana
| | - John P Kirwan
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio; Integrated Physiology and Molecular Metabolism Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana.
| |
Collapse
|
34
|
Kou G, Li Z, Wu C, Liu Y, Hu Y, Guo L, Xu X, Zhou Z. Citrus Tangeretin Improves Skeletal Muscle Mitochondrial Biogenesis via Activating the AMPK-PGC1-α Pathway In Vitro and In Vivo: A Possible Mechanism for Its Beneficial Effect on Physical Performance. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:11917-11925. [PMID: 30369237 DOI: 10.1021/acs.jafc.8b04124] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Mitochondrial biogenesis is a key factor, which influences the function of skeletal muscle. Increasingly, flavonoids are reported to have the potential ability of regulating mitochondrial biogenesis. In this study, we investigated the effects of tangeretin, a polymethoxylated flavonoid isolated from mandarin fruits, on mitochondrial biogenesis and its underlying mechanisms. The tangeretin was obtained from the peel of "Dahongpao" tangerine by macroporous adsorptive resins combined with preparative-high performance liquid chromatography. The activity of mitochondrial biogenesis was explored by using mouse-derived C2C12 myoblasts and Kunming mice. Results showed that the purity of tangeretin obtained was 98.64%, and it could effectively activate mitochondrial biogenesis signaling pathway both at gene and at protein levels in C2C12 myoblasts. Animal experiments showed that tangeretin pretreatment could markedly improve exercise performance (the time of hanging wire and run to fatigue was obviously increased 1.6-fold and 2.1-fold in the high-dose tangeretin group, respectively), and the transmission electron microscopy, Western blotting, and immunohistochemistry further indicated that tangeretin increased mitochondria number and activated mitochondrial biogenesis signaling axis. Our findings suggest that tangeretin enhanced mitochondrial biogenesis via activating the AMPK-PGC1-α pathway, resulting in the improvement of exercise performance, and tangeretin may be a potentially novel mitochondria regulator in foods.
Collapse
Affiliation(s)
| | | | | | | | | | - Liya Guo
- Key Lab of Physical Fitness Evaluation and Motor Functional Monitoring , General Administration of Sport of China-Southwest University , Chongqing 400715 , China
| | | | | |
Collapse
|
35
|
Balampanis K, Chasapi A, Kourea E, Tanoglidi A, Hatziagelaki E, Lambadiari V, Dimitriadis G, Lambrou GI, Kalfarentzos F, Melachrinou M, Sotiropoulou-Bonikou G. Inter-tissue expression patterns of the key metabolic biomarker PGC-1α in severely obese individuals: Implication in obesity-induced disease. Hellenic J Cardiol 2018; 60:282-293. [PMID: 30138744 DOI: 10.1016/j.hjc.2018.08.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 07/29/2018] [Accepted: 08/03/2018] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE PGC-1α is already known as a significant regulator of mitochondrial biogenesis, oxidative phosphorylation and fatty acid metabolism. Our study focuses on the role of PGC1α in morbid obesity, in five different tissues, collected from 50 severely obese patients during planned bariatric surgery. METHODS The investigated tissues included subcutaneous adipose tissue (SAT), visceral adipose tissue (VAT), skeletal muscle (SM), extramyocellular adipose tissue (EMAT) and liver. PGC1α expression was investigated with immunohistochemistry and evaluated with microscopy. RESULTS Our findings highlighted significant positive inter-tissue correlations regarding PGC-1α expression between several tissue pairs (VAT-SAT, VAT-SM, VAT-EMAT, SAT-SM, SAT-EMAT, SM-EMAT). Moreover, we found significant negative correlations between PGC1α expression in VAT with CD68 expression in skeletal muscle and EMAT, implying a possible protective role of PGC1α against obesity-induced inflammation. CONCLUSION Unmasking the inter-tissue communication networks regarding PGC-1α expression in morbid obesity, will give more insight into its significant role in obesity-induced diseases. PGC1α could potentially represent a future preventive and therapeutic target against obesity-induced disease, probably through enhancing mitochondrial biogenesis and metabolism.
Collapse
Affiliation(s)
- Konstantinos Balampanis
- Department of Pathology, Medical School, University of Patras, 26500 Patras, Greece; Second Department of Internal Medicine, Research Unit and Diabetes Center, Attikon University Hospital, National and Kapodistrian University of Athens, Medical School, Rimini 1, Haidari, 12462 Athens, Greece.
| | - Athina Chasapi
- Department of Pathology, Medical School, University of Patras, 26500 Patras, Greece.
| | - Eleni Kourea
- Department of Pathology, Medical School, University of Patras, 26500 Patras, Greece.
| | - Anna Tanoglidi
- Department of Clinical Pathology, Akademiska University, Uppsala, Sweden.
| | - Erifili Hatziagelaki
- Second Department of Internal Medicine, Research Unit and Diabetes Center, Attikon University Hospital, National and Kapodistrian University of Athens, Medical School, Rimini 1, Haidari, 12462 Athens, Greece.
| | - Vaia Lambadiari
- Second Department of Internal Medicine, Research Unit and Diabetes Center, Attikon University Hospital, National and Kapodistrian University of Athens, Medical School, Rimini 1, Haidari, 12462 Athens, Greece.
| | - George Dimitriadis
- Second Department of Internal Medicine, Research Unit and Diabetes Center, Attikon University Hospital, National and Kapodistrian University of Athens, Medical School, Rimini 1, Haidari, 12462 Athens, Greece.
| | - George I Lambrou
- First Department of Pediatrics, Choremeio Research Laboratory, National and Kapodistrian University of Athens, Medical School, Thivon & Levadeias 8, Goudi, 11527 Athens, Greece.
| | - Fotios Kalfarentzos
- Department of Surgery, Medical School, University of Patras, 26500 Patras, Greece.
| | - Maria Melachrinou
- Department of Pathology, Medical School, University of Patras, 26500 Patras, Greece.
| | | |
Collapse
|
36
|
Nirwane A, Majumdar A. Understanding mitochondrial biogenesis through energy sensing pathways and its translation in cardio-metabolic health. Arch Physiol Biochem 2018; 124:194-206. [PMID: 29072101 DOI: 10.1080/13813455.2017.1391847] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Mitochondria play a pivotal role in physiological energy governance. Mitochondrial biogenesis comprises growth and division of pre-existing mitochondria, triggered by environmental stressors such as endurance exercise, caloric restriction, cold exposure and oxidative stress. For normal physiology, balance between energy intake, storage and expenditure is of utmost important for the coordinated regulation of energy homeostasis. In contrast, abnormalities in these regulations render the individual susceptible to cardiometabolic disorders. This review provides a comprehensive coverage and understanding on mitochondrial biogenesis achieved through energy-sensing pathways. This includes the complex coordination of nuclear, cytosolic and mitochondrial events involving energy sensors, transcription factors, coactivators and regulators. It focuses on the importance of mitochondrial biogenesis in cardiometabolic health. Lastly, converging on the benefits of caloric restriction and endurance exercise in achieving cardiometabolic health.
Collapse
Affiliation(s)
- Abhijit Nirwane
- a Department of Pharmacology , Bombay College of Pharmacy , Mumbai , India
- b Department of Pharmaceutical and Biomedical Sciences , University of Georgia , Athens , GA , USA
| | - Anuradha Majumdar
- a Department of Pharmacology , Bombay College of Pharmacy , Mumbai , India
| |
Collapse
|
37
|
Abstract
Obesity has been considered to be a chronic disease that requires medical prevention and treatment. Intriguingly, many factors, including adipose tissue dysfunction, mitochondrial dysfunction, alterations in the muscle fiber phenotype and in the gut microbiota composition, have been identified to be involved in the development of obesity and its associated metabolic disorders (in particular type 2 diabetes mellitus). In this narrative review, we will discuss our current understanding of the relationships of these factors and obesity development, and provide a summary of potential treatments to manage obesity. Level of Evidence Level V, narrative review.
Collapse
|
38
|
Zafaranieh S, Choobineh S, Soori R. The effect of 12 weeks of aerobic exercise on mitochondrial dynamics in cardiac myocytes of type 2 diabetic rats. SPORT SCIENCES FOR HEALTH 2018. [DOI: 10.1007/s11332-018-0430-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
39
|
The effect of differentiation and TGFβ on mitochondrial respiration and mitochondrial enzyme abundance in cultured primary human skeletal muscle cells. Sci Rep 2018; 8:737. [PMID: 29335583 PMCID: PMC5768688 DOI: 10.1038/s41598-017-18658-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 12/15/2017] [Indexed: 12/27/2022] Open
Abstract
Measuring mitochondrial respiration in cultured cells is a valuable tool to investigate the influence of physiological and disease-related factors on cellular metabolism; however, the details of the experimental workflow greatly influence the informative value of the results. Working with primary cells and cell types capable of differentiation can be particularly challenging. We present a streamlined workflow optimised for investigation of primary human skeletal muscle cells. We applied the workflow to differentiated and undifferentiated cells and we investigated the effect of TGFβ1 treatment. Differentiation of myoblasts to myotubes increased mitochondrial respiration and abundance of mitochondrial enzymes and mitochondrial marker proteins. Differentiation also induced qualitative changes in mitochondrial protein composition and respiration. TGFβ1 reduced complex IV protein MTCO1 abundance in both myoblasts and myotubes. In myoblasts, spare electron transport system (ETS) capacity was reduced due to a reduction in maximal oxygen consumption. In TGFβ1-treated myotubes, the reduction in spare ETS capacity is mainly a consequence of increased oxidative phosphorylation capacity and complex III protein UQCRC2. Taken together, our data shows that it is important to monitor muscle cell differentiation when mitochondrial function is studied. Our workflow is not only sensitive enough to detect physiological-sized differences, but also adequate to form mechanistic hypotheses.
Collapse
|
40
|
Huang H, Chi H, Liao D, Zou Y. Effects of coenzyme Q 10 on cardiovascular and metabolic biomarkers in overweight and obese patients with type 2 diabetes mellitus: a pooled analysis. Diabetes Metab Syndr Obes 2018; 11:875-886. [PMID: 30568475 PMCID: PMC6276825 DOI: 10.2147/dmso.s184301] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND The potential effects of coenzyme Q10 (CoQ10) supplementation in overweight/obese patients with type 2 diabetes mellitus are not fully established. In this article, we aimed to perform a pooled analysis to investigate the effects of CoQ10 intervention on cardiovascular disease (CVD) risk factors in overweight/obese patients with type 2 diabetes mellitus (T2DM). METHODS MEDLINE, Embase, and Cochrane databases were searched for randomized controlled trials that evaluated the changes in CVD risk factors among overweight and obese patients with T2DM following CoQ10 supplementation. Two investigators independently assessed articles for inclusion, extracted data, and assessed risk of bias. Major endpoints were synthesized as weighted mean differences (WMDs) with 95% CIs. Subgroup analyses were performed to check the consistency of effect sizes across groups. Publication bias and sensitivity analysis were also performed. RESULTS Fourteen eligible trials with 693 overweight/obese diabetic subjects were included for pooling. CoQ10 interventions significantly reduced fasting blood glucose (FBG; -0.59 mmol/L; 95% CI, -1.05 to -0.12; P=0.01), hemoglobin A1c (HbA1c; -0.28%; 95% CI-0.53 to -0.03; P=0.03), and triglyceride (TG) levels (0.17 mmol/L; 95% CI, -0.32 to -0.03; P=0.02). Subgroup analysis also showed that low-dose consumption of CoQ10 (<200 mg/d) effectively reduces the values of FBG, HbA1c, fasting blood insulin, homeostatic model assessment of insulin resistance, and TG. CoQ10 treatment was well tolerated, and no drug-related adverse reactions were reported. CONCLUSION Our findings provide substantial evidence that daily CoQ10 supplementation has beneficial effects on glucose control and lipid management in overweight and obese patients with T2DM.
Collapse
Affiliation(s)
- Haohai Huang
- Department of Clinical Pharmacy, Dongguan Third People's Hospital, Affiliated Dongguan Shilong People's Hospital of Southern Medical University, Dongguan, Guangdong, China
| | - Honggang Chi
- Department of Traditional Chinese Medicine, Scientific Research Platform, The Second Clinical Medical College, Guangdong Medical University, Dongguan, China,
| | - Dan Liao
- Department of Gynaecology & Obstetrics, Dongguan Third People's Hospital, Affiliated Dongguan Shilong People's Hospital of Southern Medical University, Dongguan, Guangdong, China,
| | - Ying Zou
- Department of Traditional Chinese Medicine, Scientific Research Platform, The Second Clinical Medical College, Guangdong Medical University, Dongguan, China,
- Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Guangdong Medical University, Dongguan, Guangdong, China,
| |
Collapse
|
41
|
Jones S, D'Silva A, Bhuva A, Lloyd G, Manisty C, Moon JC, Sharma S, Hughes AD. Improved Exercise-Related Skeletal Muscle Oxygen Consumption Following Uptake of Endurance Training Measured Using Near-Infrared Spectroscopy. Front Physiol 2017; 8:1018. [PMID: 29311956 PMCID: PMC5733097 DOI: 10.3389/fphys.2017.01018] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 11/24/2017] [Indexed: 11/20/2022] Open
Abstract
Skeletal muscle metabolic function is known to respond positively to exercise interventions. Developing non-invasive techniques that quantify metabolic adaptations and identifying interventions that impart successful response are ongoing challenges for research. Healthy non-athletic adults (18–35 years old) were enrolled in a study investigating physiological adaptations to a minimum of 16 weeks endurance training prior to undertaking their first marathon. Before beginning training, participants underwent measurements of skeletal muscle oxygen consumption using near-infrared spectroscopy (NIRS) at rest (resting muscleV˙O2) and immediately following a maximal exercise test (post-exercise muscleV˙O2). Exercise-related increase in muscleV˙O2 (ΔmV˙O2) was derived from these measurements and cardio-pulmonary peakV˙O2 measured by analysis of expired gases. All measurements were repeated within 3 weeks of participants completing following the marathon and marathon completion time recorded. MuscleV˙O2 was positively correlated with cardio-pulmonary peakV˙O2 (r = 0.63, p < 0.001). MuscleV˙O2 increased at follow-up (48% increase; p = 0.004) despite no change in cardio-pulmonary peakV˙O2 (0% change; p = 0.97). Faster marathon completion time correlated with higher cardio-pulmonary peakV˙O2 (rpartial = −0.58, p = 0.002) but not muscleV˙O2 (rpartial = 0.16, p = 0.44) after adjustment for age and sex [and adipose tissue thickness (ATT) for muscleV˙O2 measurements]. Skeletal muscle metabolic adaptions occur following training and completion of a first-time marathon; these can be identified non-invasively using NIRS. Although the cardio-pulmonary system is limiting for running performance, skeletal muscle changes can be detected despite minimal improvement in cardio-pulmonary function.
Collapse
Affiliation(s)
- Siana Jones
- Population Science and Experimental Medicine, Institute for Cardiovascular Science, University College London, London, United Kingdom
| | - Andrew D'Silva
- Cardiology Clinical and Academic Group, St. Georges University of London, London, United Kingdom
| | - Anish Bhuva
- Population Science and Experimental Medicine, Institute for Cardiovascular Science, University College London, London, United Kingdom.,Barts Heart Centre, St Bartholomew's Hospital, London, United Kingdom
| | - Guy Lloyd
- Population Science and Experimental Medicine, Institute for Cardiovascular Science, University College London, London, United Kingdom.,Barts Heart Centre, St Bartholomew's Hospital, London, United Kingdom
| | - Charlotte Manisty
- Population Science and Experimental Medicine, Institute for Cardiovascular Science, University College London, London, United Kingdom.,Barts Heart Centre, St Bartholomew's Hospital, London, United Kingdom
| | - James C Moon
- Population Science and Experimental Medicine, Institute for Cardiovascular Science, University College London, London, United Kingdom.,Barts Heart Centre, St Bartholomew's Hospital, London, United Kingdom
| | - Sanjay Sharma
- Cardiology Clinical and Academic Group, St. Georges University of London, London, United Kingdom
| | - Alun D Hughes
- Population Science and Experimental Medicine, Institute for Cardiovascular Science, University College London, London, United Kingdom
| |
Collapse
|
42
|
Dysregulation of mitochondrial function and biogenesis modulators in adipose tissue of obese children. Int J Obes (Lond) 2017; 42:618-624. [PMID: 29158541 DOI: 10.1038/ijo.2017.274] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 10/01/2017] [Accepted: 10/17/2017] [Indexed: 12/26/2022]
Abstract
BACKGROUND/OBJECTIVES We aimed to evaluate mitochondrial biogenesis (MB), structure, metabolism and dysfunction in abdominal adipose tissue from male pediatric patients with obesity. SUBJECTS/METHODS Samples were collected from five children with obesity (percentile ⩾95) and five eutrophic boys (percentile ⩾5/⩽85) (8-12 years old) following parental informed consent. We analyzed the expression of key genes involved in MB (sirtuin-1 (SIRT1), peroxisome proliferator-activated receptor-γ (PPARγ), PPARγ coactivator-1α (PGC1α), nuclear respiratory factors 1 and 2 (NRF1, NRF2) and mitochondrial transcription factor A (TFAM) and surrogates for mitochondrial function/structure/metabolism (porin, TOMM20, complex I and V, UCP1, UCP2, SIRT3, SOD2) by western blot. Citrate synthase (CS), complex I (CI) activity, adenosine triphosphate (ATP) levels, mitochondrial DNA (mtDNA) content and oxidative stress end points were also determined. RESULTS Most MB proteins were significantly decreased in samples from children with obesity except complex I, V and superoxide dismutase-2 (SOD2). Similarly, CS and CI activity showed a significant reduction, as well as ATP levels and mtDNA content. PPARγ, PGC1α, complex I and V and SOD2 were hyperacetylated compared with lean samples. Concurrently, in samples from children with obesity, we found decreased SOD2 activity and redox state imbalance highlighted by decreased reduced glutathione/oxidized glutathione (GSH/GSSG) ratio and significant increases in protein carbonylation. CONCLUSIONS Adipose tissue from children with obesity demonstrates a dysregulation of key modulators of MB and organelle structure, and displays hyperacetylation of key proteins and altered expression of upstream regulators of cell metabolism.
Collapse
|
43
|
Ju L, Tong W, Qiu M, Shen W, Sun J, Zheng S, Chen Y, Liu W, Tian J. Antioxidant MMCC ameliorates catch-up growth related metabolic dysfunction. Oncotarget 2017; 8:99931-99939. [PMID: 29245950 PMCID: PMC5725141 DOI: 10.18632/oncotarget.21965] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 09/29/2017] [Indexed: 11/25/2022] Open
Abstract
Postnatal catch-up growth may be related to reduce mitochondrial content and oxidation capacity in skeletal muscle. The aim of this study is to explore the effect and mechanism of antioxidant MitoQuinone mesylate beta cyclodextrin complex (MMCC) ameliorates catch-up growth related metabolic disorders. Catch-up growth mice were created by restricting maternal food intake during the last week of gestation and providing high fat diet after weaning. Low birthweight mice and normal birthweight controls were randomly subjected to normal fat diet, high fat diet and high fat diet with MMCC drinking from the 4th week. MMCC treatment for 21 weeks slowed down the catch up growth and ameliorated catch-up growth related obesity, glucose intolerance and insulin resistance. MMCC administration significantly inhibited the peroxidation of the membrane lipid and up-regulated the antioxidant enzymes Catalase and MnSOD. In addition, MMCC treatment effectively enhanced mitochondrial functions in skeletal muscle through the up-regulation of the ATP generation, and the promotion of mitochondrial replication and remodeling. To conclude, this study demonstrates that antioxidant MMCC effectively ameliorates catch-up growth related metabolic dysfunctions by increasing mitochondrial functions in skeletal muscle.
Collapse
Affiliation(s)
- Liping Ju
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenxin Tong
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Diabetes Complications and Metabolism, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Miaoyan Qiu
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weili Shen
- Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jichao Sun
- Laboratory of Endocrine and Metabolic Diseases, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Sheng Zheng
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Chen
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wentao Liu
- Key Laboratory of Shanghai Gastric Neoplasms, Department of Surgery, Ruijin Hospital, Shanghai Jiao Tong University, Shanghai Institute of Digestive Surgery, Shanghai, China
| | - Jingyan Tian
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Diabetes Complications and Metabolism, Beckman Research Institute of City of Hope, Duarte, CA, USA
| |
Collapse
|
44
|
Diabetes-Induced Dysfunction of Mitochondria and Stem Cells in Skeletal Muscle and the Nervous System. Int J Mol Sci 2017; 18:ijms18102147. [PMID: 29036909 PMCID: PMC5666829 DOI: 10.3390/ijms18102147] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 10/11/2017] [Indexed: 12/21/2022] Open
Abstract
Diabetes mellitus is one of the most common metabolic diseases spread all over the world, which results in hyperglycemia caused by the breakdown of insulin secretion or insulin action or both. Diabetes has been reported to disrupt the functions and dynamics of mitochondria, which play a fundamental role in regulating metabolic pathways and are crucial to maintain appropriate energy balance. Similar to mitochondria, the functions and the abilities of stem cells are attenuated under diabetic condition in several tissues. In recent years, several studies have suggested that the regulation of mitochondria functions and dynamics is critical for the precise differentiation of stem cells. Importantly, physical exercise is very useful for preventing the diabetic alteration by improving the functions of both mitochondria and stem cells. In the present review, we provide an overview of the diabetic alterations of mitochondria and stem cells and the preventive effects of physical exercise on diabetes, focused on skeletal muscle and the nervous system. We propose physical exercise as a countermeasure for the dysfunction of mitochondria and stem cells in several target tissues under diabetes complication and to improve the physiological function of patients with diabetes, resulting in their quality of life being maintained.
Collapse
|
45
|
Krabbendam IE, Honrath B, Culmsee C, Dolga AM. Mitochondrial Ca 2+-activated K + channels and their role in cell life and death pathways. Cell Calcium 2017; 69:101-111. [PMID: 28818302 DOI: 10.1016/j.ceca.2017.07.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Revised: 07/14/2017] [Accepted: 07/14/2017] [Indexed: 12/18/2022]
Abstract
Ca2+-activated K+ channels (KCa) are expressed at the plasma membrane and in cellular organelles. Expression of all KCa channel subtypes (BK, IK and SK) has been detected at the inner mitochondrial membrane of several cell types. Primary functions of these mitochondrial KCa channels include the regulation of mitochondrial ROS production, maintenance of the mitochondrial membrane potential and preservation of mitochondrial calcium homeostasis. These channels are therefore thought to contribute to cellular protection against oxidative stress through mitochondrial mechanisms of preconditioning. In this review, we summarize the current knowledge on mitochondrial KCa channels, and their role in mitochondrial function in relation to cell death and survival pathways. More specifically, we systematically discuss studies on the role of these mitochondrial KCa channels in pharmacological preconditioning, and according protective effects on ischemic insults to the brain and the heart.
Collapse
Affiliation(s)
- Inge E Krabbendam
- Faculty of Science and Engineering, Groningen Research Institute of Pharmacy, Department of Molecular Pharmacology, University of Groningen, 9713 AV Groningen, The Netherlands.
| | - Birgit Honrath
- Faculty of Science and Engineering, Groningen Research Institute of Pharmacy, Department of Molecular Pharmacology, University of Groningen, 9713 AV Groningen, The Netherlands; Institute of Pharmacology and Clinical Pharmacy, University of Marburg, 35043 Marburg, Germany.
| | - Carsten Culmsee
- Institute of Pharmacology and Clinical Pharmacy, University of Marburg, 35043 Marburg, Germany.
| | - Amalia M Dolga
- Faculty of Science and Engineering, Groningen Research Institute of Pharmacy, Department of Molecular Pharmacology, University of Groningen, 9713 AV Groningen, The Netherlands.
| |
Collapse
|
46
|
Nan J, Zhu W, Rahman M, Liu M, Li D, Su S, Zhang N, Hu X, Yu H, Gupta MP, Wang J. Molecular regulation of mitochondrial dynamics in cardiac disease. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:1260-1273. [DOI: 10.1016/j.bbamcr.2017.03.006] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 01/25/2017] [Accepted: 03/14/2017] [Indexed: 12/31/2022]
|
47
|
Jayanthy G, Roshana Devi V, Ilango K, Subramanian SP. Rosmarinic Acid Mediates Mitochondrial Biogenesis in Insulin Resistant Skeletal Muscle Through Activation of AMPK. J Cell Biochem 2017; 118:1839-1848. [PMID: 28059465 DOI: 10.1002/jcb.25869] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Accepted: 01/04/2017] [Indexed: 01/03/2023]
Abstract
Rosmarinic acid (RA), a polyphenol, is known to improve hepatic insulin sensitivity in experimental type 2 diabetes. However, its effect on skeletal muscle insulin resistance is meagerly understood. The present study was aimed to investigate the up- and downstream mediators of the molecular targets of RA in attenuating insulin resistance in the skeletal muscle both in vivo and in vitro. We found that supplementation of RA increased the expression of key genes involved in the mitochondrial biogenesis like PGC-1α, SIRT-1, and TFAM via activation of AMPK in the skeletal muscle of insulin resistant rats as well as in L6 myotubes. Further, RA treatment increased the glucose uptake and decreased the phosphorylation of serine IRS-1 while increasing the translocation of GLUT 4. Together, our findings evidenced that RA treatment significantly inhibit insulin resistance in skeletal muscle cells by enhancing mitochondrial biogenesis. J. Cell. Biochem. 118: 1839-1848, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Govindaraj Jayanthy
- Division of Molecular Biology, Interdisciplinary Institute of Indian System of Medicine, SRM University, Kattankulathur, Kancheepuram 603203, Tamil Nadu, India
| | - Vellai Roshana Devi
- Department of Biochemistry, University of Madras, Guindy Campus, Chennai 600025, Tamil Nadu, India
| | - Kaliappan Ilango
- Division of Molecular Biology, Interdisciplinary Institute of Indian System of Medicine, SRM University, Kattankulathur, Kancheepuram 603203, Tamil Nadu, India
| | | |
Collapse
|
48
|
Mohamed AA, Sabry S, Abdallah AM, Elazeem NAA, Refaey D, Algebaly HAF, Fath GAE, Omar H. Circulating adipokines in children with nonalcoholic fatty liver disease: possible noninvasive diagnostic markers. Ann Gastroenterol 2017; 30:457-463. [PMID: 28655985 PMCID: PMC5480001 DOI: 10.20524/aog.2017.0148] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Accepted: 03/27/2017] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The growing obesity pandemic is the leading cause for increasing prevalence of nonalcoholic fatty liver disease (NAFLD) in children. Histopathological evaluation of the liver remains the gold standard for NAFLD diagnosis, but it is an invasive procedure with a low but real risk of morbidity and mortality. The current study evaluated circulating chemerin and adiponectin as possible noninvasive diagnostic markers for NAFLD in obese non-diabetic children. METHODS A prospective case-control study was conducted, which included 101 obese children with biopsy-proven NAFLD and 57 age- and sex-matched controls. The overall mean age of the children was 10.08±3.12 years. All underwent a full clinical assessment, routine laboratory investigation, and abdominal ultrasound. Homeostatic model assessment-insulin resistance was calculated and circulating chemerin and adiponectin were evaluated using ELISA. RESULTS Elevated serum chemerin and decreased serum adiponectin were significantly associated with an increased likelihood of exhibiting NAFLD. Receiver operator characteristic curve analysis for differentiation of NAFLD patients from those in the control group demonstrated that chemerin, at a cutoff value of 186.7 ng/mL, yielded a sensitivity and specificity of 56.44% and 87.72% respectively (P<0.001), whereas adiponectin, at a cutoff value of 2.4 µg/mL, had a sensitivity and specificity of 74.26% and 3.51% respectively (P<0.001). Furthermore, body mass index, aspartate transaminase, alanine transaminase, triglycerides, and gamma-glutamyl transferase had significant positive correlations with chemerin and significant negative correlations with adiponectin (P≤0.001). CONCLUSION Circulating chemerin and adiponectin could serve as simple noninvasive diagnostic markers for NAFLD in non-diabetic obese children.
Collapse
Affiliation(s)
- Amal Ahmed Mohamed
- Department of Biochemistry, National Hepatology and Tropical Medicine Institute, Egypt (Amal Ahmed Mohmmed)
| | - Said Sabry
- Clinical Pathology, Damanhur National Medical Institute, Egypt (Said Sabry)
| | - Asmaa Mahmoud Abdallah
- Department of Clinical Nutrition, Faculty of Applied Medical Science, King Abdul-Aziz University, Jeddah, Kingdom of Saudi Arabia (Asmaa Mahmoud Abdallah)
| | - Naglaa Adly Abd Elazeem
- Medical Biochemistry Department, Faculty of Medicine, Beni Suef University (Naglaa Adly Abd Elazeem)
| | - Doaa Refaey
- Pediatric Department, Faculty of Medicine, Benha University, Egypt (Doaa Refaey)
| | | | - Gamal Abo El Fath
- Pediatric Department, Genetic Unit, Faculty of Medicine, Ain Shams University (Gamal Abo El Fath)
| | - Heba Omar
- Endemic Medicine and Hepatology Department, Faculty of Medicine, Cairo University (Heba Omar), Egypt
| |
Collapse
|
49
|
Yu L, Gong B, Duan W, Fan C, Zhang J, Li Z, Xue X, Xu Y, Meng D, Li B, Zhang M, Bin Zhang, Jin Z, Yu S, Yang Y, Wang H. Melatonin ameliorates myocardial ischemia/reperfusion injury in type 1 diabetic rats by preserving mitochondrial function: role of AMPK-PGC-1α-SIRT3 signaling. Sci Rep 2017; 7:41337. [PMID: 28120943 PMCID: PMC5264601 DOI: 10.1038/srep41337] [Citation(s) in RCA: 153] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 12/16/2016] [Indexed: 02/07/2023] Open
Abstract
Enhancing mitochondrial biogenesis and reducing mitochondrial oxidative stress have emerged as crucial therapeutic strategies to ameliorate diabetic myocardial ischemia/reperfusion (MI/R) injury. Melatonin has been reported to be a safe and potent cardioprotective agent. However, its role on mitochondrial biogenesis or reactive oxygen species (ROS) production in type 1 diabetic myocardium and the underlying mechanisms remain unknown. We hypothesize that melatonin ameliorates MI/R injury in type 1 diabetic rats by preserving mitochondrial function via AMPK-PGC-1α-SIRT3 signaling pathway. Both our in vivo and in vitro data showed that melatonin reduced MI/R injury by improving cardiac function, enhancing mitochondrial SOD activity, ATP production and oxidative phosphorylation complex (II, III and IV), reducing myocardial apoptosis and mitochondrial MDA, H2O2 generation. Importantly, melatonin also activated AMPK-PGC-1α-SIRT3 signaling and increased SOD2, NRF1 and TFAM expressions. However, these effects were abolished by Compound C (a specific AMPK signaling blocker) administration. Additionally, our cellular experiment showed that SIRT3 siRNA inhibited the cytoprotective effect of melatonin without affecting p-AMPK/AMPK ratio and PGC-1α expression. Taken together, we concluded that melatonin preserves mitochondrial function by reducing mitochondrial oxidative stress and enhancing its biogenesis, thus ameliorating MI/R injury in type 1 diabetic state. AMPK-PGC1α-SIRT3 axis plays an essential role in this process.
Collapse
Affiliation(s)
- Liming Yu
- Department of Cardiovascular Surgery, General Hospital of Shenyang Military Area Command, 83 Wenhua Road, Shenyang, Liaoning 110016, China.,Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an 710032, China
| | - Bing Gong
- Department of Thoracic and Cardiovascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, Jiangsu 210008, China
| | - Weixun Duan
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an 710032, China
| | - Chongxi Fan
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi'an, Shaanxi 710032, China
| | - Jian Zhang
- Department of Cardiovascular Surgery, General Hospital of Shenyang Military Area Command, 83 Wenhua Road, Shenyang, Liaoning 110016, China
| | - Zhi Li
- Department of Cardiovascular Surgery, General Hospital of Shenyang Military Area Command, 83 Wenhua Road, Shenyang, Liaoning 110016, China
| | - Xiaodong Xue
- Department of Cardiovascular Surgery, General Hospital of Shenyang Military Area Command, 83 Wenhua Road, Shenyang, Liaoning 110016, China
| | - Yinli Xu
- Department of Cardiovascular Surgery, General Hospital of Shenyang Military Area Command, 83 Wenhua Road, Shenyang, Liaoning 110016, China
| | - Dandan Meng
- Department of Cardiovascular Surgery, General Hospital of Shenyang Military Area Command, 83 Wenhua Road, Shenyang, Liaoning 110016, China
| | - Buying Li
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an 710032, China
| | - Meng Zhang
- Department of Natural Medicine, School of Pharmacy, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Bin Zhang
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an 710032, China
| | - Zhenxiao Jin
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an 710032, China
| | - Shiqiang Yu
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an 710032, China
| | - Yang Yang
- Department of Thoracic and Cardiovascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, Jiangsu 210008, China.,Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Huishan Wang
- Department of Cardiovascular Surgery, General Hospital of Shenyang Military Area Command, 83 Wenhua Road, Shenyang, Liaoning 110016, China
| |
Collapse
|
50
|
Supplementing the maternal diet of rats with butyrate enhances mitochondrial biogenesis in the skeletal muscles of weaned offspring. Br J Nutr 2017; 117:12-20. [DOI: 10.1017/s0007114516004402] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
AbstractThe present study aimed to investigate the effects of maternal dietary butyrate supplementation on energy metabolism and mitochondrial biogenesis in offspring skeletal muscle and the possible mediating mechanisms. Virgin female rats were randomly assigned to either control or butyrate diets (1 % butyrate sodium) throughout gestation and lactation. At the end of lactation (21 d), the offspring were killed by exsanguination from the abdominal aorta under anaesthesia. The results showed that maternal butyrate supplementation throughout gestation and lactation did not affect offspring body weight. However, the protein expressions of G-protein-coupled receptors (GPR) 43 and 41 were significantly enhanced in offspring skeletal muscle of the maternal butyrate-supplemented group. The ATP content, most of mitochondrial DNA-encoded gene expressions, the cytochrome c oxidase subunit 1 and 4 protein contents and the mitochondrial DNA copy number were significantly higher in the butyrate group than in the control group. Meanwhile, the protein expressions of type 1 myosin heavy chain, mitochondrial transcription factor A, PPAR-coactivator-1α (PGC-1α) and uncoupling protein 3 were significantly increased in the gastrocnemius muscle of the treatment group compared with the control group. These results indicate for the first time that maternal butyrate supplementation during the gestation and lactation periods influenced energy metabolism and mitochondrial biogenesis through the GPR and PGC-1α pathways in offspring skeletal muscle at weaning.
Collapse
|