Skip Navigation
Skip to contents

Diabetes Metab J : Diabetes & Metabolism Journal

Search
OPEN ACCESS

Search

Page Path
HOME > Search
39 "Mitochondria"
Filter
Filter
Article category
Keywords
Publication year
Authors
Funded articles
Original Articles
Basic Research
Article image
PDZD8 Augments Endoplasmic Reticulum-Mitochondria Contact and Regulates Ca2+ Dynamics and Cypd Expression to Induce Pancreatic β-Cell Death during Diabetes
Yongxin Liu, Yongqing Wei, Xiaolong Jin, Hongyu Cai, Qianqian Chen, Xiujuan Zhang
Received August 12, 2023  Accepted March 26, 2024  Published online July 29, 2024  
DOI: https://doi.org/10.4093/dmj.2023.0275    [Epub ahead of print]
  • 1,489 View
  • 81 Download
AbstractAbstract PDFSupplementary MaterialPubReader   ePub   
Background
Diabetes mellitus (DM) is a chronic metabolic disease that poses serious threats to human physical and mental health worldwide. The PDZ domain-containing 8 (PDZD8) protein mediates mitochondria-associated endoplasmic reticulum (ER) membrane (MAM) formation in mammals. We explored the role of PDZD8 in DM and investigated its potential mechanism of action.
Methods
High-fat diet (HFD)- and streptozotocin-induced mouse DM and palmitic acid (PA)-induced insulin 1 (INS-1) cell models were constructed. PDZD8 expression was detected using immunohistochemistry, quantitative real-time polymerase chain reaction (qRT-PCR), and Western blotting. MAM formation, interactions between voltage-dependent anion-selective channel 1 (VDAC1) and inositol 1,4,5-triphosphate receptor type 1 (IP3R1), pancreatic β-cell apoptosis and proliferation were detected using transmission electron microscopy (TEM), proximity ligation assay (PLA), terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay, immunofluorescence staining, and Western blotting. The mitochondrial membrane potential, cell apoptosis, cytotoxicity, and subcellular Ca2+ localization in INS-1 cells were detected using a JC-1 probe, flow cytometry, and an lactate dehydrogenase kit.
Results
PDZD8 expression was up-regulated in the islets of HFD mice and PA-treated pancreatic β-cells. PDZD8 knockdown markedly shortened MAM perimeter, suppressed the expression of MAM-related proteins IP3R1, glucose-regulated protein 75 (GRP75), and VDAC1, inhibited the interaction between VDAC1 and IP3R1, alleviated mitochondrial dysfunction and ER stress, reduced the expression of ER stress-related proteins, and decreased apoptosis while increased proliferation of pancreatic β-cells. Additionally, PDZD8 knockdown alleviated Ca2+ flow into the mitochondria and decreased cyclophilin D (Cypd) expression. Cypd overexpression alleviated the promoting effect of PDZD8 knockdown on the apoptosis of β-cells.
Conclusion
PDZD8 knockdown inhibited pancreatic β-cell death in DM by alleviated ER-mitochondria contact and the flow of Ca2+ into the mitochondria.
Basic Research
Article image
DGAT2 Plays a Crucial Role to Control ESRRA-PROX1 Transcriptional Network to Maintain Hepatic Mitochondrial Sustainability
Yoseob Lee, Yeseong Hwang, Minki Kim, Hyeonuk Jeon, Seyeon Joo, Sungsoon Fang, Jae-Woo Kim
Diabetes Metab J. 2024;48(5):901-914.   Published online April 22, 2024
DOI: https://doi.org/10.4093/dmj.2023.0368
  • 2,240 View
  • 132 Download
AbstractAbstract PDFSupplementary MaterialPubReader   ePub   
Background
Diacylglycerol O-acyltransferase 2 (DGAT2) synthesizes triacylglycerol (TG) from diacylglycerol; therefore, DGAT2 is considered as a therapeutic target for steatosis. However, the consequence of inhibiting DGAT2 is not fully investigated due to side effects including lethality and lipotoxicity. In this article, we observed the role of DGAT2 in hepatocarcinoma.
Methods
The role of DGAT2 is analyzed via loss-of-function assay. DGAT2 knockdown (KD) and inhibitor treatment on HepG2 cell line was analyzed. Cumulative analysis of cell metabolism with bioinformatic data were assessed, and further compared with different cohorts of liver cancer patients and non-alcoholic fatty liver disease (NAFLD) patients to elucidate how DGAT2 is regulating cancer metabolism.
Results
Mitochondrial function is suppressed in DGAT2 KD HepG2 cell along with the decreased lipid droplets. In the aspect of the cancer, DGAT2 KD upregulates cell proliferation. Analyzing transcriptome of NAFLD and hepatocellular carcinoma (HCC) patients highlights negatively correlating expression patterns of 73 lipid-associated genes including DGAT2. Cancer patients with the lower DGAT2 expression face lower survival rate. DGAT2 KD cell and patients’ transcriptome show downregulation in estrogen- related receptor alpha (ESRRA) via integrated system for motif activity response analysis (ISMARA), with increased dimerization with corepressor prospero homeobox 1 (PROX1).
Conclusion
DGAT2 sustains the stability of mitochondria in hepatoma via suppressing ESRRA-PROX1 transcriptional network and hinders HCC from shifting towards glycolytic metabolism, which lowers cell proliferation.
Brief Report
Genetics
Article image
Clinical Characteristics of Diabetes in People with Mitochondrial DNA 3243A>G Mutation in Korea
Eun Hoo Rho, Sang Ik Baek, Heerah Lee, Moon-Woo Seong, Jong-Hee Chae, Kyong Soo Park, Soo Heon Kwak
Diabetes Metab J. 2024;48(3):482-486.   Published online February 1, 2024
DOI: https://doi.org/10.4093/dmj.2023.0078
  • 1,683 View
  • 239 Download
  • 2 Crossref
AbstractAbstract PDFSupplementary MaterialPubReader   ePub   
Maternally inherited diabetes and deafness (MIDD) is a rare mitochondrial disorder primarily resulting from m.3243A>G mutation. The clinical characteristics of MIDD exhibit significant heterogeneity. Our study aims to delineate these characteristics and determine the potential correlation with m.3243A>G heteroplasmy levels. This retrospective, descriptive study encompassed patients with confirmed m.3243A>G mutation and diabetes mellitus at Seoul National University Hospital. Our cohort comprises 40 patients with MIDD, with a mean age at study enrollment of 33.3±12.9 years and an average % of heteroplasmy of 30.0%± 14.6% in the peripheral blood. The most prevalent comorbidity was hearing loss (90%), followed by albuminuria (61%), seizure (38%), and stroke (33%). We observed a significant negative correlation between % of heteroplasmy and age at diabetes diagnosis. These clinical features can aid in the suspicion of MIDD and further consideration of genetic testing for m.3243A>G mutation.

Citations

Citations to this article as recorded by  
  • Clinical Characteristics of Diabetes in People with Mitochondrial DNA 3243A>G Mutation in Korea (Diabetes Metab J 2024;48:482-6)
    Eun Hoo Rho, Soo Heon Kwak
    Diabetes & Metabolism Journal.2024; 48(4): 818.     CrossRef
  • MIDD Patients Should Not Be Confused with MELAS Patients, Even Though Both Carry the m.3243A>G Variant
    Josef Finsterer, Sounira Mehri
    Diabetes & Metabolism Journal.2024; 48(4): 816.     CrossRef
Reviews
Basic Research
Article image
Mitochondrial Stress and Mitokines: Therapeutic Perspectives for the Treatment of Metabolic Diseases
Benyuan Zhang, Joon Young Chang, Min Hee Lee, Sang-Hyeon Ju, Hyon-Seung Yi, Minho Shong
Diabetes Metab J. 2024;48(1):1-18.   Published online January 3, 2024
DOI: https://doi.org/10.4093/dmj.2023.0115
  • 3,452 View
  • 325 Download
  • 1 Web of Science
  • 1 Crossref
AbstractAbstract PDFPubReader   ePub   
Mitochondrial stress and the dysregulated mitochondrial unfolded protein response (UPRmt) are linked to various diseases, including metabolic disorders, neurodegenerative diseases, and cancer. Mitokines, signaling molecules released by mitochondrial stress response and UPRmt, are crucial mediators of inter-organ communication and influence systemic metabolic and physiological processes. In this review, we provide a comprehensive overview of mitokines, including their regulation by exercise and lifestyle interventions and their implications for various diseases. The endocrine actions of mitokines related to mitochondrial stress and adaptations are highlighted, specifically the broad functions of fibroblast growth factor 21 and growth differentiation factor 15, as well as their specific actions in regulating inter-tissue communication and metabolic homeostasis. Finally, we discuss the potential of physiological and genetic interventions to reduce the hazards associated with dysregulated mitokine signaling and preserve an equilibrium in mitochondrial stress-induced responses. This review provides valuable insights into the mechanisms underlying mitochondrial regulation of health and disease by exploring mitokine interactions and their regulation, which will facilitate the development of targeted therapies and personalized interventions to improve health outcomes and quality of life.

Citations

Citations to this article as recorded by  
  • Mitochondria: fundamental characteristics, challenges, and impact on aging
    Runyu Liang, Luwen Zhu, Yongyin Huang, Jia Chen, Qiang Tang
    Biogerontology.2024;[Epub]     CrossRef
Basic Research
Article image
Mitochondrial-Encoded Peptide MOTS-c, Diabetes, and Aging-Related Diseases
Byung Soo Kong, Changhan Lee, Young Min Cho
Diabetes Metab J. 2023;47(3):315-324.   Published online February 24, 2023
DOI: https://doi.org/10.4093/dmj.2022.0333
  • 8,446 View
  • 334 Download
  • 5 Web of Science
  • 5 Crossref
AbstractAbstract PDFPubReader   ePub   
Mitochondria are complex metabolic organelles with manifold pathophysiological implications in diabetes. Currently published mitochondrial-encoded peptides, which are expressed from the mitochondrial open reading frame of the 12S ribosomal RNA type-c (MOTS-c), 16S rRNA (humanin and short humanin like peptide 1-6 [SHLP1-6]), or small human mitochondrial open reading frame over serine tRNA (SHMOOSE) are associated with regulation of cellular metabolism and insulin action in age-related diseases, such as type 2 diabetes mellitus. This review focuses mainly on recent advances in MOTS-c research with regards to diabetes, including both type 1 and type 2. The emerging understanding of MOTS-c in diabetes may provide insight into the development of new therapies for diabetes and other age or senescence-related diseases.

Citations

Citations to this article as recorded by  
  • Mitochondrial-derived peptides: Antidiabetic functions and evolutionary perspectives
    Satadeepa Kal, Sumana Mahata, Suborno Jati, Sushil K. Mahata
    Peptides.2024; 172: 171147.     CrossRef
  • Mitochondrial Stress and Mitokines: Therapeutic Perspectives for the Treatment of Metabolic Diseases
    Benyuan Zhang, Joon Young Chang, Min Hee Lee, Sang-Hyeon Ju, Hyon-Seung Yi, Minho Shong
    Diabetes & Metabolism Journal.2024; 48(1): 1.     CrossRef
  • Mitochondrial bioenergetics, metabolism, and beyond in pancreatic β-cells and diabetes
    Alejandra María Rivera Nieves, Brian Michael Wauford, Accalia Fu
    Frontiers in Molecular Biosciences.2024;[Epub]     CrossRef
  • Haplotype variability in mitochondrial rRNA predisposes to metabolic syndrome
    Petr Pecina, Kristýna Čunátová, Vilma Kaplanová, Guillermo Puertas-Frias, Jan Šilhavý, Kateřina Tauchmannová, Marek Vrbacký, Tomáš Čajka, Ondřej Gahura, Markéta Hlaváčková, Viktor Stránecký, Stanislav Kmoch, Michal Pravenec, Josef Houštěk, Tomáš Mráček, A
    Communications Biology.2024;[Epub]     CrossRef
  • Pyrroloquinoline Quinone Alleviates Mitochondria Damage in Radiation-Induced Lung Injury in a MOTS-c-Dependent Manner
    Yanli Zhang, Jianfeng Huang, Shengpeng Li, Junlin Jiang, Jiaojiao Sun, Dan Chen, Qingfeng Pang, Yaxian Wu
    Journal of Agricultural and Food Chemistry.2024; 72(38): 20944.     CrossRef
Basic Research
Article image
The Link between Mitochondrial Dysfunction and Sarcopenia: An Update Focusing on the Role of Pyruvate Dehydrogenase Kinase 4
Min-Ji Kim, Ibotombi Singh Sinam, Zerwa Siddique, Jae-Han Jeon, In-Kyu Lee
Diabetes Metab J. 2023;47(2):153-163.   Published online January 12, 2023
DOI: https://doi.org/10.4093/dmj.2022.0305
  • 6,442 View
  • 447 Download
  • 10 Web of Science
  • 11 Crossref
AbstractAbstract PDFPubReader   ePub   
Sarcopenia, defined as a progressive loss of muscle mass and function, is typified by mitochondrial dysfunction and loss of mitochondrial resilience. Sarcopenia is associated not only with aging, but also with various metabolic diseases characterized by mitochondrial dyshomeostasis. Pyruvate dehydrogenase kinases (PDKs) are mitochondrial enzymes that inhibit the pyruvate dehydrogenase complex, which controls pyruvate entry into the tricarboxylic acid cycle and the subsequent adenosine triphosphate production required for normal cellular activities. PDK4 is upregulated in mitochondrial dysfunction-related metabolic diseases, especially pathologic muscle conditions associated with enhanced muscle proteolysis and aberrant myogenesis. Increases in PDK4 are associated with perturbation of mitochondria-associated membranes and mitochondrial quality control, which are emerging as a central mechanism in the pathogenesis of metabolic disease-associated muscle atrophy. Here, we review how mitochondrial dysfunction affects sarcopenia, focusing on the role of PDK4 in mitochondrial homeostasis. We discuss the molecular mechanisms underlying the effects of PDK4 on mitochondrial dysfunction in sarcopenia and show that targeting mitochondria could be a therapeutic target for treating sarcopenia.

Citations

Citations to this article as recorded by  
  • Synthesis, activatory effects, molecular docking and ADME studies as rabbit muscle pyruvate kinase activators of ureido phenyl substituted 1,4-dihydropyridine derivatives
    Mustafa Oğuzhan Kaya, Tuna Demirci, Ümit Çalışır, Oğuzhan Özdemir, Yeşim Kaya, Mustafa Arslan
    Research on Chemical Intermediates.2024; 50(1): 437.     CrossRef
  • Unraveling the causes of sarcopenia: Roles of neuromuscular junction impairment and mitochondrial dysfunction
    Yanmei Miao, Leiyu Xie, Jiamei Song, Xing Cai, Jinghe Yang, Xinglong Ma, Shaolin Chen, Peng Xie
    Physiological Reports.2024;[Epub]     CrossRef
  • Metabolic clues to aging: exploring the role of circulating metabolites in frailty, sarcopenia and vascular aging related traits and diseases
    Zonghao Qian, Yuzhen Huang, Yucong Zhang, Ni Yang, Ziwei Fang, Cuntai Zhang, Le Zhang
    Frontiers in Genetics.2024;[Epub]     CrossRef
  • Inhibition of Pyruvate Dehydrogenase Kinase 4 Protects Cardiomyocytes from lipopolysaccharide-Induced Mitochondrial Damage by Reducing Lactate Accumulation
    Tangtian Chen, Qiumin Xie, Bin Tan, Qin Yi, Han Xiang, Rui Wang, Qin Zhou, Bolin He, Jie Tian, Jing Zhu, Hao Xu
    Inflammation.2024; 47(4): 1356.     CrossRef
  • Effect of resistance training plus enriched probiotic supplement on sestrin2, oxidative stress, and mitophagy markers in elderly male Wistar rats
    Majid Mohabbat, Hamid Arazi
    Scientific Reports.2024;[Epub]     CrossRef
  • Comparison between single-muscle evaluation and cross-sectional area muscle evaluation for predicting the prognosis in patients with oral squamous cell carcinoma: a retrospective cohort study
    Hirotaka Takayama, Takuya Yoshimura, Hajime Suzuki, Yuka Hirano, Masahiro Tezuka, Takayuki Ishida, Kiyohide Ishihata, Marie Amitani, Haruka Amitani, Yasunori Nakamura, Yasushi Imamura, Akio Inui, Norifumi Nakamura
    Frontiers in Oncology.2024;[Epub]     CrossRef
  • Resveratrol and Vitamin D: Eclectic Molecules Promoting Mitochondrial Health in Sarcopenia
    Cristina Russo, Maria Stella Valle, Floriana D’Angeli, Sofia Surdo, Lucia Malaguarnera
    International Journal of Molecular Sciences.2024; 25(14): 7503.     CrossRef
  • Unveiling the crucial role of peroxisomal acyl-CoA metabolism in muscle atrophy: Insights from genetic models and therapeutic interventions
    Songling Jiang, Han Sol Kim, Ji Hyun Ryu, Yong-Soo Lee, Dong-Ik Kim, Eun-Jung Jin
    Nano Today.2024; 58: 102417.     CrossRef
  • Dichloroacetate Prevents Sepsis Associated Encephalopathy by Inhibiting Microglia Pyroptosis through PDK4/NLRP3
    Xuliang Huang, Yuhao Zheng, Nan Wang, Mingming Zhao, Jinhui Liu, Wen Lin, Ye Zhu, Xiaofan Xie, Ya Lv, Junlu Wang, Yunchang Mo
    Inflammation.2024;[Epub]     CrossRef
  • The MyoGravity project to study real microgravity effects on human muscle precursor cells and tissue
    Ester Sara Di Filippo, Sara Chiappalupi, Stefano Falone, Vincenza Dolo, Fernanda Amicarelli, Silvia Marchianò, Adriana Carino, Gabriele Mascetti, Giovanni Valentini, Sara Piccirillo, Michele Balsamo, Marco Vukich, Stefano Fiorucci, Guglielmo Sorci, Stefan
    npj Microgravity.2024;[Epub]     CrossRef
  • Neuroprotective Effects and Therapeutic Potential of Dichloroacetate: Targeting Metabolic Disorders in Nervous System Diseases
    Yue Zhang, Meiyan Sun, Hongxiang Zhao, Zhengyan Wang, Yanan Shi, Jianxin Dong, Kaifang Wang, Xi Wang, Xingyue Li, Haiyan Qi, Xiaoyong Zhao
    International Journal of Nanomedicine.2023; Volume 18: 7559.     CrossRef
Sulwon Lecture 2021
Basic Research
Article image
Exercise, Mitohormesis, and Mitochondrial ORF of the 12S rRNA Type-C (MOTS-c)
Tae Kwan Yoon, Chan Hee Lee, Obin Kwon, Min-Seon Kim
Diabetes Metab J. 2022;46(3):402-413.   Published online May 25, 2022
DOI: https://doi.org/10.4093/dmj.2022.0092
  • 6,478 View
  • 256 Download
  • 12 Web of Science
  • 13 Crossref
AbstractAbstract PDFPubReader   ePub   
Low levels of mitochondrial stress are beneficial for organismal health and survival through a process known as mitohormesis. Mitohormetic responses occur during or after exercise and may mediate some salutary effects of exercise on metabolism. Exercise-related mitohormesis involves reactive oxygen species production, mitochondrial unfolded protein response (UPRmt), and release of mitochondria-derived peptides (MDPs). MDPs are a group of small peptides encoded by mitochondrial DNA with beneficial metabolic effects. Among MDPs, mitochondrial ORF of the 12S rRNA type-c (MOTS-c) is the most associated with exercise. MOTS-c expression levels increase in skeletal muscles, systemic circulation, and the hypothalamus upon exercise. Systemic MOTS-c administration increases exercise performance by boosting skeletal muscle stress responses and by enhancing metabolic adaptation to exercise. Exogenous MOTS-c also stimulates thermogenesis in subcutaneous white adipose tissues, thereby enhancing energy expenditure and contributing to the anti-obesity effects of exercise training. This review briefly summarizes the mitohormetic mechanisms of exercise with an emphasis on MOTS-c.

Citations

Citations to this article as recorded by  
  • Mitochondrial-derived peptides: Antidiabetic functions and evolutionary perspectives
    Satadeepa Kal, Sumana Mahata, Suborno Jati, Sushil K. Mahata
    Peptides.2024; 172: 171147.     CrossRef
  • Beneficial Effects of Low-Grade Mitochondrial Stress on Metabolic Diseases and Aging
    Se Hee Min, Gil Myoung Kang, Jae Woo Park, Min-Seon Kim
    Yonsei Medical Journal.2024; 65(2): 55.     CrossRef
  • Roles of Myokines and Muscle-Derived Extracellular Vesicles in Musculoskeletal Deterioration under Disuse Conditions
    Jie Zhang, Yunfang Gao, Jiangwei Yan
    Metabolites.2024; 14(2): 88.     CrossRef
  • The current insights of mitochondrial hormesis in the occurrence and treatment of bone and cartilage degeneration
    Wacili Da, Quan Chen, Bin Shen
    Biological Research.2024;[Epub]     CrossRef
  • Mitochondria-derived peptide is an effective target for treating streptozotocin induced painful diabetic neuropathy through induction of activated protein kinase/peroxisome proliferator-activated receptor gamma coactivator 1alpha -mediated mitochondrial b
    Lingfei Xu, Xihui Tang, Long Yang, Min Chang, Yuqing Xu, Qingsong Chen, Chen Lu, Su Liu, Jinhong Jiang
    Molecular Pain.2024;[Epub]     CrossRef
  • MOTS-c is an effective target for treating cancer-induced bone pain through the induction of AMPK-mediated mitochondrial biogenesis
    Long Yang, Miaomiao Li, Yucheng Liu, Yang Bai, Tianyu Yin, Yangyang Chen, Jinhong Jiang, Su Liu
    Acta Biochimica et Biophysica Sinica.2024;[Epub]     CrossRef
  • Adipose thermogenic mechanisms by cold, exercise and intermittent fasting: Similarities, disparities and the application in treatment
    Linshan Chen, Longhua Liu
    Clinical Nutrition.2024; 43(9): 2043.     CrossRef
  • Antifragility and antiinflammaging: Can they play a role for a healthy longevity?
    Fabiola Olivieri, Francesco Prattichizzo, Fabrizia Lattanzio, Anna Rita Bonfigli, Liana Spazzafumo
    Ageing Research Reviews.2023; 84: 101836.     CrossRef
  • MOTS-c: A promising mitochondrial-derived peptide for therapeutic exploitation
    Yuejun Zheng, Zilin Wei, Tianhui Wang
    Frontiers in Endocrinology.2023;[Epub]     CrossRef
  • MOTS-c: A potential anti-pulmonary fibrosis factor derived by mitochondria
    Zewei Zhang, Dongmei Chen, Kaili Du, Yaping Huang, Xingzhe Li, Quwen Li, Xiaoting Lv
    Mitochondrion.2023; 71: 76.     CrossRef
  • Mitochondrial-Encoded Peptide MOTS-c, Diabetes, and Aging-Related Diseases
    Byung Soo Kong, Changhan Lee, Young Min Cho
    Diabetes & Metabolism Journal.2023; 47(3): 315.     CrossRef
  • MOTS-c Serum Concentration Positively Correlates with Lower-Body Muscle Strength and Is Not Related to Maximal Oxygen Uptake—A Preliminary Study
    Remigiusz Domin, Michał Pytka, Mikołaj Żołyński, Jan Niziński, Marcin Rucinski, Przemysław Guzik, Jacek Zieliński, Marek Ruchała
    International Journal of Molecular Sciences.2023; 24(19): 14951.     CrossRef
  • Unique Properties of Apicomplexan Mitochondria
    Ian M. Lamb, Ijeoma C. Okoye, Michael W. Mather, Akhil B. Vaidya
    Annual Review of Microbiology.2023; 77(1): 541.     CrossRef
Original Article
Metabolic Risk/Epidemiology
Effect of Low-Dose Persistent Organic Pollutants on Mitochondrial Function: Human and in Vitro Evidence
Se-A Kim, Hoyul Lee, Sung-Mi Park, Mi-Jin Kim, Yu-Mi Lee, Young-Ran Yoon, Hyun-Kyung Lee, Hyo-Bang Moon, In-Kyu Lee, Duk-Hee Lee
Diabetes Metab J. 2022;46(4):592-604.   Published online January 26, 2022
DOI: https://doi.org/10.4093/dmj.2021.0132
  • 5,989 View
  • 253 Download
  • 7 Web of Science
  • 9 Crossref
AbstractAbstract PDFSupplementary MaterialPubReader   ePub   
Background
Chronic exposure to low-dose persistent organic pollutants (POPs) can induce mitochondrial dysfunction. This study evaluated the association between serum POP concentrations and oxygen consumption rate (OCR) as a marker of mitochondrial function in humans and in vitro cells.
Methods
Serum concentrations of organochlorine pesticides (OCPs) and polychlorinated biphenyls (PCBs) were measured in 323 adults. The OCRs of platelets and peripheral blood mononuclear cells (PBMCs) were assessed in 20 mL of fresh blood using a Seahorse XF analyzer. Additionally, the in vitro effects of Arochlor-1254, β-hexachlorocyclohexane, and p,p´-dichlorodiphenyltrichloroethane at concentrations of 0.1 pM to 100 nM were evaluated in human platelets, human PBMCs, and Jurkat T-cells.
Results
The association between serum POP concentrations and OCR differed depending on the cell type. As serum OCP concentrations increased, basal platelet OCR levels decreased significantly; according to the OCP quintiles of summary measure, they were 8.6, 9.6, 8.2, 8.0, and 7.1 pmol/min/μg (P trend=0.005). Notably, the basal PBMC OCR levels decreased remarkably as the serum PCB concentration increased. PBMC OCR levels were 46.5, 34.3, 29.1, 16.5, and 13.1 pmol/min/μg according to the PCB quintiles of summary measure (P trend <0.001), and this inverse association was consistently observed in all subgroups stratified by age, sex, obesity, type 2 diabetes mellitus, and hypertension, respectively. In vitro experimental studies have also demonstrated that chronic exposure to low-dose POPs could decrease OCR levels.
Conclusion
The findings from human and in vitro studies suggest that chronic exposure to low-dose POPs can induce mitochondrial dysfunction by impairing oxidative phosphorylation.

Citations

Citations to this article as recorded by  
  • Persistent Organic Pollutants released from decomposed adipose tissue affect mitochondrial enzyme function in the brain and eyes other than the liver
    Dongshin Yang, Eun Ko, Hwayeon Lim, Hyojin Lee, Kitae Kim, Moonsung Choi, Sooim Shin
    Environmental Science and Pollution Research.2024; 31(7): 10648.     CrossRef
  • Obesity paradox can be a fact: unveiling the hidden role of adipose tissue
    Duk-Hee Lee
    European Heart Journal.2024; 45(24): 2167.     CrossRef
  • Reduced bioenergetics and mitochondrial fragmentation in human primary cytotrophoblasts induced by an EGFR-targeting chemical mixture
    Anita A. Waye, Elvis Ticiani, Zinat Sharmin, Vanessa Perez Silos, Thilini Perera, Alex Tu, Irina A. Buhimschi, Carlos A. Murga-Zamalloa, Ying S. Hu, Almudena Veiga-Lopez
    Chemosphere.2024; 364: 143301.     CrossRef
  • Can lipophilic pollutants in adipose tissue explain weight change‐related risk in type 2 diabetes mellitus?
    Duk‐Hee Lee, In‐Kyu Lee
    Journal of Diabetes Investigation.2023; 14(4): 528.     CrossRef
  • Mitochondrial and metabolic features of salugenesis and the healing cycle
    Robert K. Naviaux
    Mitochondrion.2023; 70: 131.     CrossRef
  • Obesity Paradox in Sepsis: Role of Adipose Tissue in Storing Mitochondrial Toxins
    Duk-Hee Lee
    Critical Care Medicine.2023; 51(8): e172.     CrossRef
  • Human Preadipocytes Differentiated under Hypoxia following PCB126 Exposure during Proliferation: Effects on Differentiation, Glucose Uptake and Adipokine Profile
    Zeinab El Amine, Jean-François Mauger, Pascal Imbeault
    Cells.2023; 12(18): 2326.     CrossRef
  • Is micronucleus assay in oral exfoliated cells a useful biomarker for biomonitoring populations exposed to pesticides? A systematic review with meta-analysis
    Ingra Tais Malacarne, Wilton Mitsunari Takeshita, Daniel Vitor de Souza, Barbara dos Anjos Rosario, Milena de Barros Viana, Ana Claudia Muniz Renno, Daisy Maria Favero Salvadori, Daniel Araki Ribeiro
    Environmental Science and Pollution Research.2022; 29(43): 64392.     CrossRef
  • Comment on: Obesity is Associated with Improved Postoperative Overall Survival, Independent of Skeletal Muscle Mass in Lung Adenocarcinoma by Lee et al.
    Duk‐Hee Lee
    Journal of Cachexia, Sarcopenia and Muscle.2022; 13(5): 2576.     CrossRef
Review
Basic Research
Article image
Mitochondrial TFAM as a Signaling Regulator between Cellular Organelles: A Perspective on Metabolic Diseases
Jin-Ho Koh, Yong-Woon Kim, Dae-Yun Seo, Tae-Seo Sohn
Diabetes Metab J. 2021;45(6):853-865.   Published online November 22, 2021
DOI: https://doi.org/10.4093/dmj.2021.0138
  • 8,029 View
  • 311 Download
  • 18 Web of Science
  • 21 Crossref
Graphical AbstractGraphical Abstract AbstractAbstract PDFPubReader   ePub   
Tissues actively involved in energy metabolism are more likely to face metabolic challenges from bioenergetic substrates and are susceptible to mitochondrial dysfunction, leading to metabolic diseases. The mitochondria receive signals regarding the metabolic states in cells and transmit them to the nucleus or endoplasmic reticulum (ER) using calcium (Ca2+) for appropriate responses. Overflux of Ca2+ in the mitochondria or dysregulation of the signaling to the nucleus and ER could increase the incidence of metabolic diseases including insulin resistance and type 2 diabetes mellitus. Mitochondrial transcription factor A (Tfam) may regulate Ca2+ flux via changing the mitochondrial membrane potential and signals to other organelles such as the nucleus and ER. Since Tfam is involved in metabolic function in the mitochondria, here, we discuss the contribution of Tfam in coordinating mitochondria-ER activities for Ca2+ flux and describe the mechanisms by which Tfam affects mitochondrial Ca2+ flux in response to metabolic challenges.

Citations

Citations to this article as recorded by  
  • Targeted metabolomics reveals the aberrant energy status in diabetic peripheral neuropathy and the neuroprotective mechanism of traditional Chinese medicine JinMaiTong
    Bingjia Zhao, Qian Zhang, Yiqian He, Weifang Cao, Wei Song, Xiaochun Liang
    Journal of Pharmaceutical Analysis.2024; 14(2): 225.     CrossRef
  • Mitochondrial damage‐associated molecular patterns: A new insight into metabolic inflammation in type 2 diabetes mellitus
    Yan Wang, Jingwu Wang, Si‐Yu Tao, Zhengting Liang, Rong xie, Nan‐nan Liu, Ruxue Deng, Yuelin Zhang, Deqiang Deng, Guangjian Jiang
    Diabetes/Metabolism Research and Reviews.2024;[Epub]     CrossRef
  • Altered Energy Metabolism, Mitochondrial Dysfunction, and Redox Imbalance Influencing Reproductive Performance in Granulosa Cells and Oocyte During Aging
    Hiroshi Kobayashi, Chiharu Yoshimoto, Sho Matsubara, Hiroshi Shigetomi, Shogo Imanaka
    Reproductive Sciences.2024; 31(4): 906.     CrossRef
  • The Protective Mechanism of TFAM on Mitochondrial DNA and its Role in Neurodegenerative Diseases
    Ying Song, Wenjun Wang, Beibei Wang, Qiwen Shi
    Molecular Neurobiology.2024; 61(7): 4381.     CrossRef
  • When Our Best Friend Becomes Our Worst Enemy: The Mitochondrion in Trauma, Surgery, and Critical Illness
    May-Kristin Torp, Kåre-Olav Stensløkken, Jarle Vaage
    Journal of Intensive Care Medicine.2024;[Epub]     CrossRef
  • Attenuating mitochondrial dysfunction and morphological disruption with PT320 delays dopamine degeneration in MitoPark mice
    Vicki Wang, Kuan-Yin Tseng, Tung-Tai Kuo, Eagle Yi-Kung Huang, Kuo-Lun Lan, Zi-Rong Chen, Kuo-Hsing Ma, Nigel H. Greig, Jin Jung, Ho-II Choi, Lars Olson, Barry J. Hoffer, Yuan-Hao Chen
    Journal of Biomedical Science.2024;[Epub]     CrossRef
  • The role of mitochondrial damage-associated molecular patterns in acute pancreatitis
    Yan Zhou, Xiaoyi Huang, Yinglu Jin, Minhao Qiu, Peter C. Ambe, Zarrin Basharat, Wandong Hong
    Biomedicine & Pharmacotherapy.2024; 175: 116690.     CrossRef
  • TFAM is an autophagy receptor that limits inflammation by binding to cytoplasmic mitochondrial DNA
    Hao Liu, Cien Zhen, Jianming Xie, Zhenhuan Luo, Lin Zeng, Guojun Zhao, Shaohua Lu, Haixia Zhuang, Hualin Fan, Xia Li, Zhaojie Liu, Shiyin Lin, Huilin Jiang, Yuqian Chen, Jiahao Cheng, Zhiyu Cao, Keyu Dai, Jinhua Shi, Zhaohua Wang, Yongquan Hu, Tian Meng,
    Nature Cell Biology.2024; 26(6): 878.     CrossRef
  • Disrupted mitochondrial transcription factor A expression promotes mitochondrial dysfunction and enhances ocular surface inflammation by activating the absent in melanoma 2 inflammasome
    Yaqiong Li, Lei Tian, Siyuan Li, Xiaoniao Chen, Fengyang Lei, Jiayu Bao, Qianru Wu, Ya Wen, Ying Jie
    Free Radical Biology and Medicine.2024; 222: 106.     CrossRef
  • Prolonged Hypoxic Exposure Impairs Endothelial Functions: Possible Mechanism of HIF-1α Signaling
    Junyoung Hong, Junchul Shin
    Exercise Science.2024; 33(2): 168.     CrossRef
  • Protein Arginine Methyltransferases: Emerging Targets in Cardiovascular and Metabolic Disease
    Yan Zhang, Shibo Wei, Eun-Ju Jin, Yunju Jo, Chang-Myung Oh, Gyu-Un Bae, Jong-Sun Kang, Dongryeol Ryu
    Diabetes & Metabolism Journal.2024; 48(4): 487.     CrossRef
  • Development of a Competitive Nutrient-Based T-Cell Immunotherapy Designed to Block the Adaptive Warburg Effect in Acute Myeloid Leukemia
    Huynh Cao, Jeffrey Xiao, David J. Baylink, Vinh Nguyen, Nathan Shim, Jae Lee, Dave J. R. Mallari, Samiksha Wasnik, Saied Mirshahidi, Chien-Shing Chen, Hisham Abdel-Azim, Mark E. Reeves, Yi Xu
    Biomedicines.2024; 12(10): 2250.     CrossRef
  • Effects of the anti-inflammatory drug celecoxib on cell death signaling in human colon cancer
    Ryuto Maruyama, Yuki Kiyohara, Yasuhiro Kudo, Tomoyasu Sugiyama
    Naunyn-Schmiedeberg's Archives of Pharmacology.2023; 396(6): 1171.     CrossRef
  • gp130 Activates Mitochondrial Dynamics for Hepatocyte Survival in a Model of Steatohepatitis
    Daria Shunkina, Anastasia Dakhnevich, Egor Shunkin, Olga Khaziakhmatova, Valeria Shupletsova, Maria Vulf, Alexandra Komar, Elena Kirienkova, Larisa Litvinova
    Biomedicines.2023; 11(2): 396.     CrossRef
  • Pharmacological Activation of Rev-erbα Attenuates Doxorubicin-Induced Cardiotoxicity by PGC-1α Signaling Pathway
    Runmei Zou, Shuo Wang, Hong Cai, Yuwen Wang, Cheng Wang, Vivek Pandey
    Cardiovascular Therapeutics.2023; 2023: 1.     CrossRef
  • Protective Effect of Ergothioneine against 7-Ketocholesterol-Induced Mitochondrial Damage in hCMEC/D3 Human Brain Endothelial Cells
    Damien Meng-Kiat Leow, Irwin Kee-Mun Cheah, Zachary Wei-Jie Fong, Barry Halliwell, Wei-Yi Ong
    International Journal of Molecular Sciences.2023; 24(6): 5498.     CrossRef
  • Effect of PPARγ on oxidative stress in diabetes-related dry eye
    Jing Wang, Shuangping Chen, Xiuxiu Zhao, Qian Guo, Ruibo Yang, Chen Zhang, Yue Huang, Lechong Ma, Shaozhen Zhao
    Experimental Eye Research.2023; 231: 109498.     CrossRef
  • Chiisanoside Mediates the Parkin/ZNF746/PGC-1α Axis by Downregulating MiR-181a to Improve Mitochondrial Biogenesis in 6-OHDA-Caused Neurotoxicity Models In Vitro and In Vivo: Suggestions for Prevention of Parkinson’s Disease
    Yu-Ling Hsu, Hui-Jye Chen, Jia-Xin Gao, Ming-Yang Yang, Ru-Huei Fu
    Antioxidants.2023; 12(9): 1782.     CrossRef
  • TBBPA causes apoptosis in grass carp hepatocytes involving destroyed ER-mitochondrial function
    Dongxu Han, Naixi Yang, Huanyi Liu, Yujie Yao, Shiwen Xu
    Chemosphere.2023; 341: 139974.     CrossRef
  • Impact of Roux-en-Y Gastric Bypass on Mitochondrial Biogenesis and Dynamics in Leukocytes of Obese Women
    Zaida Abad-Jiménez, Teresa Vezza, Sandra López-Domènech, Meylin Fernández-Reyes, Francisco Canet, Carlos Morillas, Segundo Ángel Gómez-Abril, Celia Bañuls, Víctor M. Víctor, Milagros Rocha
    Antioxidants.2022; 11(7): 1302.     CrossRef
  • The Effects of Galgunhwanggumhwangryun-tang on Glucose and Energy Metabolism in C2C12 Myotubes
    Jihong Oh, Song-Yi Han, Soo Kyoung Lim, Hojun Kim
    Journal of Korean Medicine for Obesity Research.2022; 22(2): 93.     CrossRef
Original Article
Basic Research
Article image
Umbilical Cord-Mesenchymal Stem Cell-Conditioned Medium Improves Insulin Resistance in C2C12 Cell
Kyung-Soo Kim, Yeon Kyung Choi, Mi Jin Kim, Jung Wook Hwang, Kyunghoon Min, Sang Youn Jung, Soo-Kyung Kim, Yong-Soo Choi, Yong-Wook Cho
Diabetes Metab J. 2021;45(2):260-269.   Published online July 10, 2020
DOI: https://doi.org/10.4093/dmj.2019.0191
  • 10,850 View
  • 219 Download
  • 10 Web of Science
  • 10 Crossref
Graphical AbstractGraphical Abstract AbstractAbstract PDFSupplementary MaterialPubReader   ePub   
Background

Umbilical cord-mesenchymal stem cell-conditioned medium (UC-MSC-CM) has emerged as a promising cell-free therapy. The aim of this study was to explore the therapeutic effects of UC-MSC-CM on insulin resistance in C2C12 cell.

Methods

Insulin resistance was induced by palmitate. Effects of UC-MSC-CM on insulin resistance were evaluated using glucose uptake, glucose transporter type 4 (GLUT4) translocation, the insulin-signaling pathway, and mitochondrial contents and functions in C2C12 cell.

Results

Glucose uptake was improved by UC-MSC-CM. UC-MSC-CM treatment increased only in membranous GLUT4 expression, not in cytosolic GLUT4 expression. It restored the insulin-signaling pathway in insulin receptor substrate 1 and protein kinase B. Mitochondrial contents evaluated by mitochondrial transcription factor A, mitochondrial DNA copy number, and peroxisome proliferator-activated receptor gamma coactivator 1-alpha were increased by UC-MSC-CM. In addition, UC-MSC-CM significantly decreased mitochondrial reactive oxygen species and increased fatty acid oxidation and mitochondrial membrane potential. There was no improvement in adenosine triphosphate (ATP) contents, but ATP synthesis was improved by UC-MSC-CM. Cytokine and active factor analysis of UC-MSC-CM showed that it contained many regulators inhibiting insulin resistance.

Conclusion

UC-MSC-CM improves insulin resistance with multiple mechanisms in C2C12 cell.

Citations

Citations to this article as recorded by  
  • Wharton’s Jelly Mesenchymal Stem Cell Conditioned Medium Ameliorates Diabetes‐Induced Testicular Damage and Sperm Abnormalities by Mitigating Oxidative Stress, Apoptosis, and Inflammation
    Mojtaba Sargazi, Narges Karbalaei, Saied Karbalay-Doust, Sara Keshtgar, Zohre Aghaei, Pasquale Marrazzo
    Stem Cells International.2024;[Epub]     CrossRef
  • Secretome Derived from Mesenchymal Stem/Stromal Cells: A Promising Strategy for Diabetes and its Complications
    ling li, Siyu Hua, Lianghui You, Tianying Zhong
    Current Stem Cell Research & Therapy.2024; 19(10): 1328.     CrossRef
  • Neurotransmitters in Type 2 Diabetes and the Control of Systemic and Central Energy Balance
    Amnah Al-Sayyar, Maha M. Hammad, Michayla R. Williams, Mohammed Al-Onaizi, Jehad Abubaker, Fawaz Alzaid
    Metabolites.2023; 13(3): 384.     CrossRef
  • Neuroprotective Effect of Wharton’s Jelly-Derived Mesenchymal Stem Cell-Conditioned Medium (WJMSC-CM) on Diabetes-Associated Cognitive Impairment by Improving Oxidative Stress, Neuroinflammation, and Apoptosis
    Zohre Aghaei, Narges Karbalaei, Mohammad Reza Namavar, Masoud Haghani, Mahboobeh Razmkhah, Mahdi Khorsand Ghaffari, Marzieh Nemati, Andrea Ballini
    Stem Cells International.2023; 2023: 1.     CrossRef
  • Mesenchymal-Stem Cell-Derived Conditioned Media Versus Exosomes in the Treatment of Rat Model of Polycystic Ovary: An Attempt to Understand the Underlying Mechanisms (Biochemical and Histological Study)
    Soha Abd-elkawy Abd-elwahab, Noura Hassan Khamis, Rehab Ahmed Rifaai, Nashwa Fathy Gamal El-Tahawy, Randa Ahmed Ibrahim
    Microscopy and Microanalysis.2023; 29(3): 1244.     CrossRef
  • Therapeutic Potential of Mesenchymal Stem Cell‐Derived Conditioned Medium for Diabetes Mellitus and Related Complications
    Basak Isildar, Serbay Ozkan, Meral Koyuturk
    Advanced Therapeutics.2023;[Epub]     CrossRef
  • Treatment of type 2 diabetes mellitus with stem cells and antidiabetic drugs: a dualistic and future-focused approach
    Priyamvada Amol Arte, Kanchanlata Tungare, Mustansir Bhori, Renitta Jobby, Jyotirmoi Aich
    Human Cell.2023; 37(1): 54.     CrossRef
  • Perinatal Stem Cell Therapy to Treat Type 1 Diabetes Mellitus: A Never-Say-Die Story of Differentiation and Immunomodulation
    Francesca Paris, Valeria Pizzuti, Pasquale Marrazzo, Andrea Pession, Francesco Alviano, Laura Bonsi
    International Journal of Molecular Sciences.2022; 23(23): 14597.     CrossRef
  • Mesenchymal Stem Cell-Derived Apoptotic Bodies: Biological Functions and Therapeutic Potential
    Huixue Tang, Huikun Luo, Zihan Zhang, Di Yang
    Cells.2022; 11(23): 3879.     CrossRef
  • Human umbilical cord mesenchymal stem cells in type 2 diabetes mellitus: the emerging therapeutic approach
    Andreia Gomes, Pedro Coelho, Raquel Soares, Raquel Costa
    Cell and Tissue Research.2021; 385(3): 497.     CrossRef
Sulwon Lecture 2019
Pathophysiology
The Role of Growth Differentiation Factor 15 in Energy Metabolism
Joon Young Chang, Hyun Jung Hong, Seul Gi Kang, Jung Tae Kim, Ben Yuan Zhang, Minho Shong
Diabetes Metab J. 2020;44(3):363-371.   Published online June 29, 2020
DOI: https://doi.org/10.4093/dmj.2020.0087
  • 9,515 View
  • 246 Download
  • 17 Web of Science
  • 16 Crossref
AbstractAbstract PDFPubReader   

Growth differentiation factor 15 (GDF15) is receiving great interest beyond its role as an aging and disease-related biomarker. Recent discovery of its receptor, glial cell line-derived neurotrophic factor (GDNF) family receptor α-like (GFRAL), suggests a central role in appetite regulation. However, there is also considerable evidence that GDF15 may have peripheral activity through an as-of-yet undiscovered mode of action. This raises the question as to whether increased GDF15 induction during pathophysiologic conditions also suppresses appetite. The present review will briefly introduce the discovery of GDF15 and describe the different contexts under which GDF15 is induced, focusing on its induction during mitochondrial dysfunction. We will further discuss the metabolic role of GDF15 under various pathophysiological conditions and conclude with possible therapeutic applications.

Citations

Citations to this article as recorded by  
  • Growth differentiation factor-15 is an IFN-γ regulated mediator of infection-induced weight loss and the hepatic FGF21 response
    Jojo Reyes, Yanlin Zhao, Krushang Pandya, George S. Yap
    Brain, Behavior, and Immunity.2024; 116: 24.     CrossRef
  • Biomarkers of disability worsening in inactive primary progressive multiple sclerosis
    Maria-Elizabeth Baeva, Isabelle Tottenham, Marcus Koch, Carlos Camara-Lemarroy
    Journal of Neuroimmunology.2024; 387: 578268.     CrossRef
  • Tricarboxylic Acid Cycle Intermediates and Individual Ageing
    Natalia Kurhaluk
    Biomolecules.2024; 14(3): 260.     CrossRef
  • The implicated role of GDF15 in gastrointestinal cancer
    Yingqi Xue, Yan Zhang, Yale Su, Jiangqi Zhao, Daoquan Yu, Yunju Jo, Jongkil Joo, Hyun Joo Lee, Dongryeol Ryu, Shibo Wei
    European Journal of Clinical Investigation.2024;[Epub]     CrossRef
  • Correlation Between Growth Differentiation Factor-15 and Peripheral Neuropathy in Patients with Type 2 Diabetes Mellitus
    Yue Li, Yuhui Wang, Yonghong Cao, Xinxiu Zhang, Wu Dai, Yiran Zhao, Lei Zhang, Xiaofang Han
    Diabetes, Metabolic Syndrome and Obesity.2024; Volume 17: 3019.     CrossRef
  • A long‐acting GDF15 analog causes robust, sustained weight loss and reduction of food intake in an obese nonhuman primate model
    Songmao Zheng, David Polidori, Yuanping Wang, Brian Geist, Xiefan Lin‐Schmidt, Jennifer L. Furman, Serena Nelson, Andrea R. Nawrocki, Simon A. Hinke
    Clinical and Translational Science.2023; 16(8): 1431.     CrossRef
  • GDF15 enhances body weight and adiposity reduction in obese mice by leveraging the leptin pathway
    Samuel N. Breit, Rakesh Manandhar, Hong-Ping Zhang, Michelle Lee-Ng, David A. Brown, Vicky Wang-Wei Tsai
    Cell Metabolism.2023; 35(8): 1341.     CrossRef
  • Serum growth differentiation factor-15 (GDF-15) is a biomarker of cardiac manifestations in children with COVID-19
    Sally Raafat Ishak, Mona Mostafa El Ganzoury, Eman Mahmoud Fouda, Maha Ahmad Anwar, Amany Moustafa Kamal, Heba Mostafa Hamza, Nehad Ahmed Bakry
    European Journal of Medical Research.2023;[Epub]     CrossRef
  • Understanding the molecular basis of anorexia and tissue wasting in cancer cachexia
    Eunbyul Yeom, Kweon Yu
    Experimental & Molecular Medicine.2022; 54(4): 426.     CrossRef
  • Investigating the combination of plasma amyloid-beta and geroscience biomarkers on the incidence of clinically meaningful cognitive decline in older adults
    Wan-Hsuan Lu, Kelly Virecoulon Giudici, John E. Morley, Sophie Guyonnet, Angelo Parini, Geetika Aggarwal, Andrew D. Nguyen, Yan Li, Randall J. Bateman, Bruno Vellas, Philipe de Souto Barreto, Bruno Vellas, Sophie Guyonnet, Isabelle Carrié, Lauréane Brigit
    GeroScience.2022; 44(3): 1489.     CrossRef
  • The Potential Role of Growth Differentiation Factor 15 in COVID-19: A Corollary Subjective Effect or Not?
    Ahmad O. Babalghith, Hayder M. Al-kuraishy, Ali I. Al-Gareeb, Michel De Waard, Jean-Marc Sabatier, Hebatallah M. Saad, Gaber El-Saber Batiha
    Diagnostics.2022; 12(9): 2051.     CrossRef
  • Metformin and growth differentiation factor 15 (GDF15) in type 2 diabetes mellitus: A hidden treasure
    Hayder M. Al‐kuraishy, Ali I. Al‐Gareeb, Athanasios Alexiou, Marios Papadakis, Eman Hassan Nadwa, Sarah M. Albogami, Mohammed Alorabi, Hebatallah M. Saad, Gaber El‐Saber Batiha
    Journal of Diabetes.2022; 14(12): 806.     CrossRef
  • Effects of Exercise Intervention on Mitochondrial Stress Biomarkers in Metabolic Syndrome Patients: A Randomized Controlled Trial
    Jae Seung Chang, Jun Namkung
    International Journal of Environmental Research and Public Health.2021; 18(5): 2242.     CrossRef
  • Neurological & psychological aspects of Barth syndrome: Clinical manifestations and potential pathogenic mechanisms
    Melissa Olivar-Villanueva, Mindong Ren, Colin K.L. Phoon
    Mitochondrion.2021; 61: 188.     CrossRef
  • The metabolic role of spermidine in obesity: Evidence from cells to community
    Yanee Choksomngam, Sintip Pattanakuhar, Nipon Chattipakorn, Siriporn C. Chattipakorn
    Obesity Research & Clinical Practice.2021; 15(4): 315.     CrossRef
  • The Role of GDF15 as a Myomitokine
    Kornelia Johann, Maximilian Kleinert, Susanne Klaus
    Cells.2021; 10(11): 2990.     CrossRef
Review
Basic Research
Mitochondrial Mechanisms in Diabetic Cardiomyopathy
Johannes Gollmer, Andreas Zirlik, Heiko Bugger
Diabetes Metab J. 2020;44(1):33-53.   Published online February 21, 2020
DOI: https://doi.org/10.4093/dmj.2019.0185
  • 11,547 View
  • 250 Download
  • 66 Web of Science
  • 70 Crossref
AbstractAbstract PDFPubReader   

Mitochondrial medicine is increasingly discussed as a promising therapeutic approach, given that mitochondrial defects are thought to contribute to many prevalent diseases and their complications. In individuals with diabetes mellitus (DM), defects in mitochondrial structure and function occur in many organs throughout the body, contributing both to the pathogenesis of DM and complications of DM. Diabetic cardiomyopathy (DbCM) is increasingly recognized as an underlying cause of increased heart failure in DM, and several mitochondrial mechanisms have been proposed to contribute to the development of DbCM. Well established mechanisms include myocardial energy depletion due to impaired adenosine triphosphate (ATP) synthesis and mitochondrial uncoupling, and increased mitochondrial oxidative stress. A variety of upstream mechanisms of impaired ATP regeneration and increased mitochondrial reactive oxygen species have been proposed, and recent studies now also suggest alterations in mitochondrial dynamics and autophagy, impaired mitochondrial Ca2+ uptake, decreased cardiac adiponectin action, increased O-GlcNAcylation, and impaired activity of sirtuins to contribute to mitochondrial defects in DbCM, among others. In the current review, we present and discuss the evidence that underlies both established and recently proposed mechanisms that are thought to contribute to mitochondrial dysfunction in DbCM.

Citations

Citations to this article as recorded by  
  • SIRT1: a promising therapeutic target in type 2 diabetes mellitus
    Ainaz Mihanfar, Maryam Akbarzadeh, Saber Ghazizadeh Darband, Shirin Sadighparvar, Maryam Majidinia
    Archives of Physiology and Biochemistry.2024; 130(1): 13.     CrossRef
  • Effect of exercise on improving myocardial mitochondrial function in decreasing diabetic cardiomyopathy
    Feng Zhang, Jian jian Lin, Hao nan Tian, Jun Wang
    Experimental Physiology.2024; 109(2): 190.     CrossRef
  • Cardioprotective effects of asiaticoside against diabetic cardiomyopathy: Activation of the AMPK/Nrf2 pathway
    Chennian Xu, Lin Xia, Dengyue Xu, Yang Liu, Ping Jin, Mengen Zhai, Yu Mao, Yiwei Wang, Anguo Wen, Jian Yang, Lifang Yang
    Journal of Cellular and Molecular Medicine.2024;[Epub]     CrossRef
  • Mitochondrial dynamics and the significance of its disturbances in the development of childhood diseases. Part II. Cardiological and endocrinological aspects
    V. S. Sukhorukov, T. I. Baranich, A. V. Egorova, E. N. Fedorova, K. A. Skvortsova, D. A. Kharlamov, A. I. Krapivkin
    Rossiyskiy Vestnik Perinatologii i Pediatrii (Russian Bulletin of Perinatology and Pediatrics).2024; 69(2): 12.     CrossRef
  • Ferroptosis: Mechanisms and role in diabetes mellitus and its complications
    Pan Liu, Zhengdong Zhang, Yichen Cai, Zhaoying Li, Qian Zhou, Qiu Chen
    Ageing Research Reviews.2024; 94: 102201.     CrossRef
  • Salidroside Alleviates Myocardial Ischemia Reperfusion by Balancing Mitochondrial Homeostasis via Nrf2
    Tingxu Yan, Xu Li, Xin Wang, Bosai He, Ying Jia, Wei Xiao, Rong He
    Journal of Food Biochemistry.2024; 2024: 1.     CrossRef
  • AGO2 Protects Against Diabetic Cardiomyopathy by Activating Mitochondrial Gene Translation
    Jiabing Zhan, Kunying Jin, Rong Xie, Jiahui Fan, Yuyan Tang, Chen Chen, Huaping Li, Dao Wen Wang
    Circulation.2024; 149(14): 1102.     CrossRef
  • Pathophysiological aspects of morphological changes in the structure of atrioventricular valves leafl ets in diabetes mellitus
    V. V. Krylov, M. R. Ragimov, I. V. Misnikova
    Clinical Medicine (Russian Journal).2024; 102(2): 109.     CrossRef
  • Promoting mitochondrial dynamics by inhibiting the PINK1–PRKN pathway to relieve diabetic nephropathy
    Jun-yi Zhu, Joyce van de Leemput, Zhe Han
    Disease Models & Mechanisms.2024;[Epub]     CrossRef
  • SGLT2 inhibitors: from glucose-lowering to cardiovascular benefits
    Alberto Preda, Fabrizio Montecucco, Federico Carbone, Giovanni G Camici, Thomas F Lüscher, Simon Kraler, Luca Liberale
    Cardiovascular Research.2024; 120(5): 443.     CrossRef
  • Glucagon-like peptide-1 receptor agonists and sodium-glucose cotransporter 2 inhibitors, anti-diabetic drugs in heart failure and cognitive impairment: potential mechanisms of the protective effects
    Maria Antonietta Riemma, Elena Mele, Maria Donniacuo, Marialucia Telesca, Gabriella Bellocchio, Giuseppe Castaldo, Francesco Rossi, Antonella De Angelis, Donato Cappetta, Konrad Urbanek, Liberato Berrino
    Frontiers in Pharmacology.2024;[Epub]     CrossRef
  • IL-37 ameliorates myocardial fibrosis by regulating mtDNA-enriched vesicle release in diabetic cardiomyopathy mice
    Qingyu Huang, Tongqing Chen, Jian Li, Yiming Wang, Huairui Shi, Yifei Yu, Qingwei Ji, Xiaoyan Shen, Tao Sun, Haiming Shi, Xinping Luo, Bo Jin, Yan You, Bangwei Wu
    Journal of Translational Medicine.2024;[Epub]     CrossRef
  • Lysophosphatidylcholine trigger myocardial injury in diabetic cardiomyopathy via the TLR4/ZNF480/AP-1/NF-kB pathway
    Nannan Liu, Yang Chen, Tian An, Siyu Tao, Bohan Lv, Jinfang Dou, Ruxue Deng, Xianjie Zhen, Yuelin Zhang, Caizhong Lu, Zhongsheng Chang, Guangjian Jiang
    Heliyon.2024; 10(13): e33601.     CrossRef
  • Improvement of Cardiac Function and Subcellular Defects Due to Chronic Diabetes upon Treatment with Sarpogrelate
    Paramjit S. Tappia, Vijayan Elimban, Anureet K. Shah, Ramesh K. Goyal, Naranjan S. Dhalla
    Journal of Cardiovascular Development and Disease.2024; 11(7): 215.     CrossRef
  • Progress of Mitochondrial Kinetics in Ejection Fraction Preserving Heart Failure
    梦茹 胡
    Advances in Clinical Medicine.2024; 14(07): 945.     CrossRef
  • Molecular Basis of Cardiomyopathies in Type 2 Diabetes
    Silvia Giardinelli, Giovanni Meliota, Donatella Mentino, Gabriele D’Amato, Maria Felicia Faienza
    International Journal of Molecular Sciences.2024; 25(15): 8280.     CrossRef
  • Exendin-4 exhibits cardioprotective effects against high glucose-induced mitochondrial abnormalities: Potential role of GLP-1 receptor and mTOR signaling
    Warisara Parichatikanond, Sudhir Pandey, Supachoke Mangmool
    Biochemical Pharmacology.2024; 229: 116552.     CrossRef
  • Therapeutic potential of finerenone for diabetic cardiomyopathy: focus on the mechanisms
    Jing Wang, Haojie Xue, Jinyu He, Li Deng, Julong Tian, Yang Jiang, Jian Feng
    Diabetology & Metabolic Syndrome.2024;[Epub]     CrossRef
  • Cellular interplay between cardiomyocytes and non-myocytes in diabetic cardiomyopathy
    Ren Jie Phang, Rebecca H Ritchie, Derek J Hausenloy, Jarmon G Lees, Shiang Y Lim
    Cardiovascular Research.2023; 119(3): 668.     CrossRef
  • Mitochondrial miRNA as epigenomic signatures: Visualizing aging-associated heart diseases through a new lens
    Jasvinder Singh Bhatti, Naina Khullar, Rajesh Vijayvergiya, Umashanker Navik, Gurjit Kaur Bhatti, P. Hemachandra Reddy
    Ageing Research Reviews.2023; 86: 101882.     CrossRef
  • Eucommiae Folium and Active Compounds Protect Against Mitochondrial Dysfunction-Calcium Overload in Epileptic Hippocampal Neurons Through the Hypertrophic Cardiomyopathy Pathway
    Shuai-nan Zhang, Hong-mei Li, Qi Liu, Xu-zhao Li, Wu-de Yang, Ying Zhou
    Neurochemical Research.2023; 48(9): 2674.     CrossRef
  • Speckle-tracking echocardiography provides sensitive measurements of subtle early alterations associated with cardiac dysfunction in T2DM rats
    Yanchao Qi, Zhiyan Chen, Bingyan Guo, Zhe Liu, Lijie Wang, Suyun Liu, Lixiang Xue, Meifang Ma, Yajuan Yin, Yongjun Li, Gang Liu
    BMC Cardiovascular Disorders.2023;[Epub]     CrossRef
  • Novel insights into the role of mitochondria in diabetic cardiomyopathy: molecular mechanisms and potential treatments
    Fumin Zhi, Qian Zhang, Li Liu, Xing Chang, Hongtao Xu
    Cell Stress and Chaperones.2023; 28(6): 641.     CrossRef
  • Transcription factor EB: A potential integrated network regulator in metabolic-associated cardiac injury
    Weixing Wen, Haoxiao Zheng, Weiwen Li, Guolin Huang, Peng Chen, Xiaolin Zhu, Yue Cao, Jiahuan Li, Xiaohui Huang, Yuli Huang
    Metabolism.2023; 147: 155662.     CrossRef
  • Role of STIM1 in the Regulation of Cardiac Energy Substrate Preference
    Panpan Liu, Zhuli Yang, Youjun Wang, Aomin Sun
    International Journal of Molecular Sciences.2023; 24(17): 13188.     CrossRef
  • Mitochondrial dysfunction at the crossroad of cardiovascular diseases and cancer
    Carmine Rocca, Teresa Soda, Ernestina Marianna De Francesco, Marco Fiorillo, Francesco Moccia, Giuseppe Viglietto, Tommaso Angelone, Nicola Amodio
    Journal of Translational Medicine.2023;[Epub]     CrossRef
  • Triarylphosphonium compounds as effective vectors for mitochondria-targeted delivery systems: decoration strategies and prospects for clinical application
    T. N. Pashirova, A. V. Nemtarev, E. B. Souto, V. F. Mironov
    Russian Chemical Reviews.2023; 92(10): RCR5095.     CrossRef
  • Moderate- and High-Intensity Endurance Training Alleviate Diabetes-Induced Cardiac Dysfunction in Rats
    Sarah D’Haese, Maxim Verboven, Lize Evens, Dorien Deluyker, Ivo Lambrichts, BO Eijnde, Dominique Hansen, Virginie Bito
    Nutrients.2023; 15(18): 3950.     CrossRef
  • Alpha-lipoic acid enhances ischemic postconditioning-mediated improvement of myocardial infarction and apoptosis in diabetic rats with ischemia/reperfusion injury
    Sanaz Gholami, Reza Badalzadeh, Alireza Alihemmati
    Canadian Journal of Physiology and Pharmacology.2023; 101(12): 682.     CrossRef
  • Concurrent diabetes and heart failure: interplay and novel therapeutic approaches
    Qutuba G Karwi, Kim L Ho, Simran Pherwani, Ezra B Ketema, Qiuyu Sun, Gary D Lopaschuk
    Cardiovascular Research.2022; 118(3): 686.     CrossRef
  • Ca2+ mishandling and mitochondrial dysfunction: a converging road to prediabetic and diabetic cardiomyopathy
    Carolina Jaquenod De Giusti, Julieta Palomeque, Alicia Mattiazzi
    Pflügers Archiv - European Journal of Physiology.2022; 474(1): 33.     CrossRef
  • Double-edge sword roles of iron in driving energy production versus instigating ferroptosis
    Shuping Zhang, Wei Xin, Gregory J. Anderson, Ruibin Li, Ling Gao, Shuguang Chen, Jiajun Zhao, Sijin Liu
    Cell Death & Disease.2022;[Epub]     CrossRef
  • Differential remodelling of mitochondrial subpopulations and mitochondrial dysfunction are a feature of early stage diabetes
    Bodour S. Rajab, Sarah Kassab, Connor D. Stonall, Hussam Daghistani, Stephen Gibbons, Mamas Mamas, David Smith, Aleksandr Mironov, Zainab AlBalawi, Yin Hua Zhang, Florence Baudoin, Min Zi, Sukhpal Prehar, Elizabeth J. Cartwright, Ashraf Kitmitto
    Scientific Reports.2022;[Epub]     CrossRef
  • GlyNAC (Glycine and N-Acetylcysteine) Supplementation Improves Impaired Mitochondrial Fuel Oxidation and Lowers Insulin Resistance in Patients with Type 2 Diabetes: Results of a Pilot Study
    Rajagopal V. Sekhar
    Antioxidants.2022; 11(1): 154.     CrossRef
  • Glucose-derived posttranslational modification in cardiovascular disease
    Michael Lehrke, Julia Moellmann, Florian Kahles, Nikolaus Marx
    Molecular Aspects of Medicine.2022; 86: 101084.     CrossRef
  • Mitochondrial Implications in Cardiovascular Aging and Diseases: The Specific Role of Mitochondrial Dynamics and Shifts
    Anastasia V. Poznyak, Tatiana V. Kirichenko, Evgeny E. Borisov, Nikolay K. Shakhpazyan, Andrey G. Kartuesov, Alexander N. Orekhov
    International Journal of Molecular Sciences.2022; 23(6): 2951.     CrossRef
  • Editorial: Management of Diabetes and its Complications: A Focus on Endothelial Dysfunction
    Shanhu Qiu, Jianhua Ma, Tongzhi Wu
    Frontiers in Endocrinology.2022;[Epub]     CrossRef
  • Alternative autophagy: mechanisms and roles in different diseases
    Hong Feng, Nian Wang, Nan Zhang, Hai-han Liao
    Cell Communication and Signaling.2022;[Epub]     CrossRef
  • Mitochondria-Mediated Cardiovascular Benefits of Sodium-Glucose Co-Transporter 2 Inhibitors
    Siarhei A. Dabravolski, Alexander D. Zhuravlev, Andrey G. Kartuesov, Evgeny E. Borisov, Vasily N. Sukhorukov, Alexander N. Orekhov
    International Journal of Molecular Sciences.2022; 23(10): 5371.     CrossRef
  • Animal Models of Dysregulated Cardiac Metabolism
    Heiko Bugger, Nikole J. Byrne, E. Dale Abel
    Circulation Research.2022; 130(12): 1965.     CrossRef
  • Mitochondrial Dynamics and Mitophagy in Cardiometabolic Disease
    Jianguo Lin, Jinlong Duan, Qingqing Wang, Siyu Xu, Simin Zhou, Kuiwu Yao
    Frontiers in Cardiovascular Medicine.2022;[Epub]     CrossRef
  • Perspectives for Forkhead box transcription factors in diabetic cardiomyopathy: Their therapeutic potential and possible effects of salvianolic acids
    Ronghui Han, Hemeng Huang, Weiyi Xia, Jingjin Liu, Hui Luo, Jing Tang, Zhengyuan Xia
    Frontiers in Cardiovascular Medicine.2022;[Epub]     CrossRef
  • Molecular and cellular mechanisms in diabetic heart failure: Potential therapeutic targets
    Misganaw Asmamaw Mengstie, Endeshaw Chekol Abebe, Awgichew Behaile Teklemariam, Anemut Tilahun Mulu, Assefa Agegnehu Teshome, Edgeit Abebe Zewde, Zelalem Tilahun Muche, Muluken Teshome Azezew
    Frontiers in Endocrinology.2022;[Epub]     CrossRef
  • Effect of Chronic Treatment with Uridine on Cardiac Mitochondrial Dysfunction in the C57BL/6 Mouse Model of High-Fat Diet–Streptozotocin-Induced Diabetes
    Natalia V. Belosludtseva, Vlada S. Starinets, Irina B. Mikheeva, Maxim N. Belosludtsev, Mikhail V. Dubinin, Galina D. Mironova, Konstantin N. Belosludtsev
    International Journal of Molecular Sciences.2022; 23(18): 10633.     CrossRef
  • FGF21–Sirtuin 3 Axis Confers the Protective Effects of Exercise Against Diabetic Cardiomyopathy by Governing Mitochondrial Integrity
    Leigang Jin, Leiluo Geng, Lei Ying, Lingling Shu, Kevin Ye, Ranyao Yang, Yan Liu, Yao Wang, Yin Cai, Xue Jiang, Qin Wang, Xingqun Yan, Boya Liao, Jie Liu, Fuyu Duan, Gary Sweeney, Connie Wai Hong Woo, Yu Wang, Zhengyuan Xia, Qizhou Lian, Aimin Xu
    Circulation.2022; 146(20): 1537.     CrossRef
  • Novel Insights Into Molecular Mechanism of Mitochondria in Diabetic Cardiomyopathy
    Jing Bai, Chuanbin Liu, Pingjun Zhu, Yang Li
    Frontiers in Physiology.2021;[Epub]     CrossRef
  • Mitochondrial Dysfunction Increases Arrhythmic Triggers and Substrates; Potential Anti-arrhythmic Pharmacological Targets
    Khalil Saadeh, Ibrahim Talal Fazmin
    Frontiers in Cardiovascular Medicine.2021;[Epub]     CrossRef
  • Prostaglandin E receptor subtype 4 protects against diabetic cardiomyopathy by modulating cardiac fatty acid metabolism via FOXO1/CD36 signalling
    Fan Ying, Hao Liu, Eva Hoi Ching Tang, Ishan Lakhani, Ningning Liu, Zhengyuan Xia, Shiming Liu
    Biochemical and Biophysical Research Communications.2021; 548: 196.     CrossRef
  • Study of long non-coding RNA and mitochondrial dysfunction in diabetic rats
    Haytham K. Sultan, Wael M. El-Ayat, Azza H. AbouGhalia, Noha N. Lasheen, Amr S. Moustafa
    Tissue and Cell.2021; 71: 101516.     CrossRef
  • A Role of Glucose Overload in Diabetic Cardiomyopathy in Nonhuman Primates
    Xiu Wang, Shi Jin, Weina Hu, Gaetano Santulli
    Journal of Diabetes Research.2021; 2021: 1.     CrossRef
  • Dysregulation of circulating miRNAs promotes the pathogenesis of diabetes-induced cardiomyopathy
    Uzair Ahmed, Usman Ali Ashfaq, Muhammad Qasim, Imtiaz Ahmad, Hafiz Usman Ahmad, Muhammad Tariq, Muhammad Shareef Masoud, Saba Khaliq, Muhammad Younas Khan Barozai
    PLOS ONE.2021; 16(4): e0250773.     CrossRef
  • MicroRNAs Regulating Mitochondrial Function in Cardiac Diseases
    Guang-Qiong Zhang, Sheng-Quan Wang, Yan Chen, Ling-Yun Fu, Yi-Ni Xu, Ling Li, Ling Tao, Xiang-Chun Shen
    Frontiers in Pharmacology.2021;[Epub]     CrossRef
  • Effects of omega-3 fatty acids and metformin combination on diabetic cardiomyopathy in rats through autophagic pathway
    Salma M. Eraky, Nehal M. Ramadan
    The Journal of Nutritional Biochemistry.2021; 97: 108798.     CrossRef
  • Adropin Alleviates Myocardial Fibrosis in Diabetic Cardiomyopathy Rats: A Preliminary Study
    Mao Liu, Jiao Ai, Zhuang Shuai, Kai Tang, Zongyu Li, Yin Huang
    Frontiers in Cardiovascular Medicine.2021;[Epub]     CrossRef
  • Fibrosis of the diabetic heart: Clinical significance, molecular mechanisms, and therapeutic opportunities
    Izabela Tuleta, Nikolaos G. Frangogiannis
    Advanced Drug Delivery Reviews.2021; 176: 113904.     CrossRef
  • Effect of the MPT Pore Inhibitor Alisporivir on the Development of Mitochondrial Dysfunction in the Heart Tissue of Diabetic Mice
    Natalia V. Belosludtseva, Vlada S. Starinets, Irina B. Mikheeva, Dmitriy A. Serov, Maxim E. Astashev, Maxim N. Belosludtsev, Mikhail V. Dubinin, Konstantin N. Belosludtsev
    Biology.2021; 10(9): 839.     CrossRef
  • Characterisation of the Myocardial Mitochondria Structural and Functional Phenotype in a Murine Model of Diabetic Cardiomyopathy
    Alex M. Parker, Mitchel Tate, Darnel Prakoso, Minh Deo, Andrew M. Willis, David M. Nash, Daniel G. Donner, Simon Crawford, Helen Kiriazis, Cesare Granata, Melinda T. Coughlan, Miles J. De Blasio, Rebecca H. Ritchie
    Frontiers in Physiology.2021;[Epub]     CrossRef
  • Radiation‑induced dysfunction of energy metabolism in the heart results in the fibrosis of cardiac tissues
    Peng Xu, Yali Yi, Yijing Luo, Zhicheng Liu, Yilin Xu, Jing Cai, Zhimin Zeng, Anwen Liu
    Molecular Medicine Reports.2021;[Epub]     CrossRef
  • Heart Failure and Diabetes: Perspective of a Dangerous Association
    Liliana E. Favaloro, Roxana D. Ratto, Carla Musso
    Current Hypertension Reviews.2021; 17(2): 85.     CrossRef
  • CTRP9 Mediates Protective Effects in Cardiomyocytes via AMPK- and Adiponectin Receptor-Mediated Induction of Anti-Oxidant Response
    Bernd Niemann, Ling Li, Dorothee Siegler, Benedikt H. Siegler, Fabienne Knapp, Jakob Hanna, Muhammad Aslam, Michael Kracht, Rainer Schulz, Susanne Rohrbach
    Cells.2020; 9(5): 1229.     CrossRef
  • The Hippo Pathway Orchestrates Mitochondrial Quality Control: A Novel Focus on Cardiovascular Diseases
    Ying Tan, Cai Lei, Huifang Tang, Xiao Zhu, Guanghui Yi
    DNA and Cell Biology.2020; 39(9): 1494.     CrossRef
  • Adeno-Associated Viral Transfer of Glyoxalase-1 Blunts Carbonyl and Oxidative Stresses in Hearts of Type 1 Diabetic Rats
    Fadhel A. Alomar, Abdullah Al-Rubaish, Fahad Al-Muhanna, Amein K. Al-Ali, JoEllyn McMillan, Jaipaul Singh, Keshore R. Bidasee
    Antioxidants.2020; 9(7): 592.     CrossRef
  • Loss of function of transcription factor EB remodels lipid metabolism and cell death pathways in the cardiomyocyte
    Purvi C. Trivedi, Jordan J. Bartlett, Angella Mercer, Logan Slade, Marc Surette, Andrea Ballabio, Stephane Flibotte, Bahira Hussein, Brian Rodrigues, Petra C. Kienesberger, Thomas Pulinilkunnil
    Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease.2020; 1866(10): 165832.     CrossRef
  • Rescue of myocardial energetic dysfunction in diabetes through the correction of mitochondrial hyperacetylation by honokiol
    Matthew Kerr, Jack J. Miller, Dharendra Thapa, Sophie Stiewe, Kerstin N. Timm, Claudia N. Montes Aparicio, Iain Scott, Damian J. Tyler, Lisa C. Heather
    JCI Insight.2020;[Epub]     CrossRef
  • Diabetes Mellitus, Mitochondrial Dysfunction and Ca2+-Dependent Permeability Transition Pore
    Konstantin N. Belosludtsev, Natalia V. Belosludtseva, Mikhail V. Dubinin
    International Journal of Molecular Sciences.2020; 21(18): 6559.     CrossRef
  • Effects of Standardized Green Tea Extract and Its Main Component, EGCG, on Mitochondrial Function and Contractile Performance of Healthy Rat Cardiomyocytes
    Rocchina Vilella, Gianluca Sgarbi, Valeria Naponelli, Monia Savi, Leonardo Bocchi, Francesca Liuzzi, Riccardo Righetti, Federico Quaini, Caterina Frati, Saverio Bettuzzi, Giancarlo Solaini, Donatella Stilli, Federica Rizzi, Alessandra Baracca
    Nutrients.2020; 12(10): 2949.     CrossRef
  • Diabetic cardiomyopathy: definition, diagnosis criteria, treatment directions and prevention of heart failure
    N. A. Koziolova, P. G. Karavaev, A. S. Veklich
    South Russian Journal of Therapeutic Practice.2020; 1(2): 93.     CrossRef
  • Adiponectin protects HL-1 cardiomyocytes against rotenone-induced cytotoxicity through AMPK activation
    Biao Li, Baojian Zhang, Na Liu, Keke Wu, Yingxu Ma, Wanyun Zuo, Zuodong Ning, Yaozhong Liu, Chao Sun, Yichao Xiao, Tao Tu, Qiming Liu
    Toxicology Letters.2020; 335: 82.     CrossRef
  • Nutrient Sensor mTOR and OGT: Orchestrators of Organelle Homeostasis in Pancreatic β-Cells
    Nicholas Esch, Seokwon Jo, Mackenzie Moore, Emilyn U. Alejandro, Yingke Xu
    Journal of Diabetes Research.2020; 2020: 1.     CrossRef
  • Where Does Metformin Stand in Modern Day Management of Type 2 Diabetes?
    Ehtasham Ahmad, Jack Sargeant, Francesco Zaccardi, Kamlesh Khunti, David Webb, Melanie Davies
    Pharmaceuticals.2020; 13(12): 427.     CrossRef
Original Article
Basic Research
Article image
Inhibition of Ceramide Accumulation in Podocytes by Myriocin Prevents Diabetic Nephropathy
Chang-Yun Woo, Ji Yeon Baek, Ah-Ram Kim, Chung Hwan Hong, Ji Eun Yoon, Hyoun Sik Kim, Hyun Ju Yoo, Tae-Sik Park, Ranjan Kc, Ki-Up Lee, Eun Hee Koh
Diabetes Metab J. 2020;44(4):581-591.   Published online November 4, 2019
DOI: https://doi.org/10.4093/dmj.2019.0063
  • 7,108 View
  • 174 Download
  • 32 Web of Science
  • 34 Crossref
AbstractAbstract PDFPubReader   ePub   
Background

Ceramides are associated with metabolic complications including diabetic nephropathy in patients with diabetes. Recent studies have reported that podocytes play a pivotal role in the progression of diabetic nephropathy. Also, mitochondrial dysfunction is known to be an early event in podocyte injury. Thus, we tested the hypothesis that ceramide accumulation in podocytes induces mitochondrial damage through reactive oxygen species (ROS) production in patients with diabetic nephropathy.

Methods

We used Otsuka Long Evans Tokushima Fatty (OLETF) rats and high-fat diet (HFD)-fed mice. We fed the animals either a control- or a myriocin-containing diet to evaluate the effects of the ceramide. Also, we assessed the effects of ceramide on intracellular ROS generation and on podocyte autophagy in cultured podocytes.

Results

OLETF rats and HFD-fed mice showed albuminuria, histologic features of diabetic nephropathy, and podocyte injury, whereas myriocin treatment effectively treated these abnormalities. Cultured podocytes exposed to agents predicted to be risk factors (high glucose, high free fatty acid, and angiotensin II in combination [GFA]) showed an increase in ceramide accumulation and ROS generation in podocyte mitochondria. Pretreatment with myriocin reversed GFA-induced mitochondrial ROS generation and prevented cell death. Myriocin-pretreated cells were protected from GFA-induced disruption of mitochondrial integrity.

Conclusion

We showed that mitochondrial ceramide accumulation may result in podocyte damage through ROS production. Therefore, this signaling pathway could become a pharmacological target to abate the development of diabetic kidney disease.

Citations

Citations to this article as recorded by  
  • Interplay of lipid metabolism and inflammation in podocyte injury
    Zilv Luo, Zhaowei Chen, Jijia Hu, Guohua Ding
    Metabolism.2024; 150: 155718.     CrossRef
  • A review of the mechanisms of abnormal ceramide metabolism in type 2 diabetes mellitus, Alzheimer’s disease, and their co-morbidities
    Yun Pan, Jieying Li, Panjie Lin, Lihua Wan, Yiqian Qu, Lingyong Cao, Lei Wang
    Frontiers in Pharmacology.2024;[Epub]     CrossRef
  • Ceramides and mitochondrial homeostasis
    Song Ding, Guorui Li, Tinglv Fu, Tianyu Zhang, Xiao Lu, Ning Li, Qing Geng
    Cellular Signalling.2024; 117: 111099.     CrossRef
  • The Influence of Soy Isoflavones and Soy Isoflavones with Inulin on Kidney Morphology, Fatty Acids, and Associated Parameters in Rats with and without Induced Diabetes Type 2
    Kamila Misiakiewicz-Has, Dominika Maciejewska-Markiewicz, Dagmara Szypulska-Koziarska, Agnieszka Kolasa, Barbara Wiszniewska
    International Journal of Molecular Sciences.2024; 25(10): 5418.     CrossRef
  • Deletion of IRE1α in podocytes exacerbates diabetic nephropathy in mice
    Andrey V. Cybulsky, Joan Papillon, Julie Guillemette, José R. Navarro-Betancourt, Chen-Fang Chung, Takao Iwawaki, I. George Fantus
    Scientific Reports.2024;[Epub]     CrossRef
  • Roles of Mitochondrial Dysfunction in Diabetic Kidney Disease: New Perspectives from Mechanism to Therapy
    Yichen Yang, Jiahui Liu, Qiling Shi, Buyu Guo, Hanbing Jia, Yuxuan Yang, Songbo Fu
    Biomolecules.2024; 14(6): 733.     CrossRef
  • Lipidomic study of kidney in a mouse model with urine flow obstruction
    Divyavani Gowda, Md. Abdul Masum, Siddabasave Gowda B. Gowda, Chandra Shekhar, Md. Zahir Uddin Rubel, Shunnosuke Kira, Osamu Ichii, Yasuhiro Kon, Hitoshi Chiba, Shu-Ping Hui
    Scientific Reports.2024;[Epub]     CrossRef
  • Sphingolipids and Chronic Kidney Disease
    Zrinka Šakić, Armin Atić, Slavica Potočki, Nikolina Bašić-Jukić
    Journal of Clinical Medicine.2024; 13(17): 5050.     CrossRef
  • Reduced sphingolipid biosynthesis modulates proteostasis networks to enhance longevity
    Nathaniel L. Hepowit, Eric Blalock, Sangderk Lee, Kimberly M. Bretland, Jason A. MacGurn, Robert C. Dickson
    Aging.2023; 15(2): 472.     CrossRef
  • Protective effect of natural products in the metabolic-associated kidney diseases via regulating mitochondrial dysfunction
    Peng Liu, Yao Chen, Jing Xiao, Wenhui Zhu, Xiaoming Yan, Ming Chen
    Frontiers in Pharmacology.2023;[Epub]     CrossRef
  • BCAA insufficiency leads to premature ovarian insufficiency via ceramide‐induced elevation of ROS
    Xiao Guo, Yuemeng Zhu, Lu Guo, Yiwen Qi, Xiaocheng Liu, Jinhui Wang, Jiangtao Zhang, Linlin Cui, Yueyang Shi, Qichu Wang, Cenxi Liu, Guangxing Lu, Yilian Liu, Tao Li, Shangyu Hong, Yingying Qin, Xuelian Xiong, Hao Wu, Lin Huang, He Huang, Chao Gu, Bin Li,
    EMBO Molecular Medicine.2023;[Epub]     CrossRef
  • Chinese herbal medicine and its active compounds in attenuating renal injury via regulating autophagy in diabetic kidney disease
    Peng Liu, Wenhui Zhu, Yang Wang, Guijie Ma, Hailing Zhao, Ping Li
    Frontiers in Endocrinology.2023;[Epub]     CrossRef
  • Integrated gas chromatography‐mass spectrometry and ultra‐high‐performance liquid chromatography‐mass spectrometry renal metabolomics and lipidomics deciphered the metabolic regulation mechanism of Gushudan on kidney‐yang‐deficiency‐syndrome rats
    Qing Lu, Jing Zhang, Ling Xin, Yanwei Lou, Feng Qin, Longshan Zhao, Zhili Xiong
    Journal of Separation Science.2023;[Epub]     CrossRef
  • Advances in the pharmacological study of Chinese herbal medicine to alleviate diabetic nephropathy by improving mitochondrial oxidative stress
    Ming Chen, Yao Chen, Wenhui Zhu, Xiaoming Yan, Jing Xiao, Peiqing Zhang, Peng Liu, Ping Li
    Biomedicine & Pharmacotherapy.2023; 165: 115088.     CrossRef
  • Rodent models to study type 1 and type 2 diabetes induced human diabetic nephropathy
    Amit Talukdar, Mandira Basumatary
    Molecular Biology Reports.2023; 50(9): 7759.     CrossRef
  • Art2 mediates selective endocytosis of methionine transporters during adaptation to sphingolipid depletion
    Nathaniel L. Hepowit, Bradley Moon, Adam C. Ebert, Robert C. Dickson, Jason A. MacGurn
    Journal of Cell Science.2023;[Epub]     CrossRef
  • Kidney lipid dysmetabolism and lipid droplet accumulation in chronic kidney disease
    Alla Mitrofanova, Sandra Merscher, Alessia Fornoni
    Nature Reviews Nephrology.2023; 19(10): 629.     CrossRef
  • Research progress of autophagy in pathogenesis of diabetes nephropathy
    Shengnan Zeng, Ying Li
    Diabetic Nephropathy.2023; 3(3): 51.     CrossRef
  • Associations of plasma sphingolipids with measures of insulin sensitivity, β-cell function, and incident diabetes in Japanese Americans
    Ji Cheol Bae, Pandora L. Wander, Rozenn N. Lemaitre, Amanda M. Fretts, Colleen M. Sitlani, Hai H. Bui, Melissa K. Thomas, Donna Leonetti, Wilfred Y. Fujimoto, Edward J. Boyko, Kristina M. Utzschneider
    Nutrition, Metabolism and Cardiovascular Diseases.2023;[Epub]     CrossRef
  • Lipidomic approaches to dissect dysregulated lipid metabolism in kidney disease
    Judy Baek, Chenchen He, Farsad Afshinnia, George Michailidis, Subramaniam Pennathur
    Nature Reviews Nephrology.2022; 18(1): 38.     CrossRef
  • Podocyte Bioenergetics in the Development of Diabetic Nephropathy: The Role of Mitochondria
    Irena Audzeyenka, Agnieszka Bierżyńska, Abigail C Lay
    Endocrinology.2022;[Epub]     CrossRef
  • Acylcarnitines: Can They Be Biomarkers of Diabetic Nephropathy?
    Xiaodie Mu, Min Yang, Peiyao Ling, Aihua Wu, Hua Zhou, Jingting Jiang
    Diabetes, Metabolic Syndrome and Obesity: Targets and Therapy.2022; Volume 15: 247.     CrossRef
  • Research Progress on Natural Products’ Therapeutic Effects on Atrial Fibrillation by Regulating Ion Channels
    Jinshan He, Sicong Li, Yumeng Ding, Yujia Tong, Xuebin Li, Simona Saponara
    Cardiovascular Therapeutics.2022; 2022: 1.     CrossRef
  • Mechanisms of podocyte injury and implications for diabetic nephropathy
    Federica Barutta, Stefania Bellini, Gabriella Gruden
    Clinical Science.2022; 136(7): 493.     CrossRef
  • A Rheostat of Ceramide and Sphingosine-1-Phosphate as a Determinant of Oxidative Stress-Mediated Kidney Injury
    Norishi Ueda
    International Journal of Molecular Sciences.2022; 23(7): 4010.     CrossRef
  • Implications of Sphingolipid Metabolites in Kidney Diseases
    Shamroop kumar Mallela, Sandra Merscher, Alessia Fornoni
    International Journal of Molecular Sciences.2022; 23(8): 4244.     CrossRef
  • Role of ceramides in the pathogenesis of diabetes mellitus and its complications
    Nawajes Mandal, Richard Grambergs, Koushik Mondal, Sandip K. Basu, Faiza Tahia, Sam Dagogo-Jack
    Journal of Diabetes and its Complications.2021; 35(2): 107734.     CrossRef
  • Rotten to the Cortex: Ceramide-Mediated Lipotoxicity in Diabetic Kidney Disease
    Rebekah J. Nicholson, Marcus G. Pezzolesi, Scott A. Summers
    Frontiers in Endocrinology.2021;[Epub]     CrossRef
  • Enhancing lifespan of budding yeast by pharmacological lowering of amino acid pools
    Nathaniel L. Hepowit, Jessica K. A. Macedo, Lyndsay E. A. Young, Ke Liu, Ramon C. Sun, Jason A. MacGurn, Robert C. Dickson
    Aging.2021; 13(6): 7846.     CrossRef
  • New insights into renal lipid dysmetabolism in diabetic kidney disease
    Alla Mitrofanova, George Burke, Sandra Merscher, Alessia Fornoni
    World Journal of Diabetes.2021; 12(5): 524.     CrossRef
  • Excessively Enlarged Mitochondria in the Kidneys of Diabetic Nephropathy
    Kiyoung Kim, Eun-Young Lee
    Antioxidants.2021; 10(5): 741.     CrossRef
  • Mechanistic insights into the role of serum-glucocorticoid kinase 1 in diabetic nephropathy: A systematic review
    Saba Noor, Taj Mohammad, Gulam M. Ashraf, Joviana Farhat, Anwar L. Bilgrami, Mathew Suji Eapen, Sukhwinder Singh Sohal, Dharmendra Kumar Yadav, Md Imtaiyaz Hassan
    International Journal of Biological Macromolecules.2021; 193: 562.     CrossRef
  • The Updates of Podocyte Lipid Metabolism in Proteinuric Kidney Disease
    Yu Sun, Sijia Cui, Yunfeng Hou, Fan Yi
    Kidney Diseases.2021; 7(6): 438.     CrossRef
  • Saturated fatty acids induce insulin resistance in podocytes through inhibition of IRS1 via activation of both IKKβ and mTORC1
    Benoit Denhez, Marina Rousseau, Crysta Spino, David-Alexandre Dancosst, Marie-Ève Dumas, Andréanne Guay, Farah Lizotte, Pedro Geraldes
    Scientific Reports.2020;[Epub]     CrossRef
Review
Others
Mitochondrial Toxins and Healthy Lifestyle Meet at the Crossroad of Hormesis
Yu-Mi Lee, Duk-Hee Lee
Diabetes Metab J. 2019;43(5):568-577.   Published online October 24, 2019
DOI: https://doi.org/10.4093/dmj.2019.0143
  • 7,007 View
  • 98 Download
  • 14 Web of Science
  • 13 Crossref
AbstractAbstract PDFPubReader   

Mitochondrial function is crucial for the maintenance of cellular homeostasis under physiological and stress conditions. Thus, chronic exposure to environmental chemicals that affect mitochondrial function can have harmful effects on humans. We argue that the concept of hormesis should be revisited to explain the non-linear responses to mitochondrial toxins at a low-dose range and develop practical methods to protect humans from the negative effects of mitochondrial toxins. Of the most concern to humans are lipophilic chemical mixtures and heavy metals, owing to their physical properties. Even though these chemicals tend to demonstrate no safe level in humans, a non-linear dose-response has been also observed. Stress response activation, i.e., hormesis, can explain this non-linearity. Recently, hormesis has reemerged as a unifying concept because diverse stressors can induce similar stress responses. Besides potentially harmful environmental chemicals, healthy lifestyle interventions such as exercise, calorie restriction (especially glucose), cognitive stimulation, and phytochemical intake also activate stress responses. This conceptual link can lead to the development of practical methods that counterbalance the harm of mitochondrial toxins. Unlike chemical hormesis with its safety issues, the activation of stress responses via lifestyle modification can be safely used to combat the negative effects of mitochondrial toxins.

Citations

Citations to this article as recorded by  
  • Polyethylene terephthalate (PET) micro- and nanoplastic particles affect the mitochondrial efficiency of human brain vascular pericytes without inducing oxidative stress
    Sean M. Gettings, William Timbury, Anna Dmochowska, Riddhi Sharma, Rebecca McGonigle, Lewis E. MacKenzie, Guillaume Miquelard-Garnier, Nora Bourbia
    NanoImpact.2024; 34: 100508.     CrossRef
  • The current insights of mitochondrial hormesis in the occurrence and treatment of bone and cartilage degeneration
    Wacili Da, Quan Chen, Bin Shen
    Biological Research.2024;[Epub]     CrossRef
  • Neurodegenerative disorders, metabolic icebergs, and mitohormesis
    Matthew C. L. Phillips, Martin Picard
    Translational Neurodegeneration.2024;[Epub]     CrossRef
  • Rules of Heliogeomagnetics Diversely Coordinating Biological Rhythms and Promoting Human Health
    Kuniaki Otsuka, Germaine Cornelissen, Andi Weydahl, Denis Gubin, Larry A. Beaty, Masatoshi Murase
    Applied Sciences.2023; 13(2): 951.     CrossRef
  • Can lipophilic pollutants in adipose tissue explain weight change‐related risk in type 2 diabetes mellitus?
    Duk‐Hee Lee, In‐Kyu Lee
    Journal of Diabetes Investigation.2023; 14(4): 528.     CrossRef
  • Hormetic Effects of Cerium, Lanthanum and Their Combination at Sub-micromolar Concentrations in Sea Urchin Sperm
    Giovanni Pagano, Antonios Apostolos Brouziotis, Daniel Lyons, Ivana Čarapar, Rahime Oral, Serkan Tez, Philippe J. Thomas, Franca Tommasi, Giovanni Libralato, Marco Guida, Marco Trifuoggi
    Bulletin of Environmental Contamination and Toxicology.2023;[Epub]     CrossRef
  • Mitochondria: It is all about energy
    Amaloha Casanova, Anne Wevers, Santiago Navarro-Ledesma, Leo Pruimboom
    Frontiers in Physiology.2023;[Epub]     CrossRef
  • Type 2 Diabetes Induced by Changes in Proteomic Profiling of Zebrafish Chronically Exposed to a Mixture of Organochlorine Pesticides at Low Concentrations
    Yan Gao, Hyojin Lee, Sangkyu Lee, Ki-Tae Kim
    International Journal of Environmental Research and Public Health.2022; 19(9): 4991.     CrossRef
  • Effect of Low-Dose Persistent Organic Pollutants on Mitochondrial Function: Human and in Vitro Evidence
    Se-A Kim, Hoyul Lee, Sung-Mi Park, Mi-Jin Kim, Yu-Mi Lee, Young-Ran Yoon, Hyun-Kyung Lee, Hyo-Bang Moon, In-Kyu Lee, Duk-Hee Lee
    Diabetes & Metabolism Journal.2022; 46(4): 592.     CrossRef
  • Can Environmental Pollutants Be a Factor Linking Obesity and COVID-19?
    Duk-Hee Lee
    Journal of Korean Medical Science.2021;[Epub]     CrossRef
  • Intensive weight loss and cognition: The dynamics of persistent organic pollutants in adipose tissue can explain the unexpected results from the Action for Health in Diabetes (Look AHEAD) study
    Yu‐Mi Lee, Sun‐Hee Park, Duk‐Hee Lee
    Alzheimer's & Dementia.2020; 16(4): 696.     CrossRef
  • Lipophilic Environmental Chemical Mixtures Released During Weight‐Loss: The Need to Consider Dynamics
    Duk‐Hee Lee, David R Jacobs, Lars Lind, P. Monica Lind
    BioEssays.2020;[Epub]     CrossRef
  • Environmental toxicology and ecotoxicology: How clean is clean? Rethinking dose-response analysis
    Evgenios Agathokleous, Edward J. Calabrese
    Science of The Total Environment.2020; 746: 138769.     CrossRef
Sulwon Lecture 2018
Pathophysiology
Mitochondrial Dysfunction in Adipocytes as a Primary Cause of Adipose Tissue Inflammation
Chang-Yun Woo, Jung Eun Jang, Seung Eun Lee, Eun Hee Koh, Ki-Up Lee
Diabetes Metab J. 2019;43(3):247-256.   Published online March 27, 2019
DOI: https://doi.org/10.4093/dmj.2018.0221
  • 9,655 View
  • 274 Download
  • 74 Web of Science
  • 74 Crossref
AbstractAbstract PDFPubReader   

Adipose tissue inflammation is considered a major contributing factor in the development of obesity-associated insulin resistance and cardiovascular diseases. However, the cause of adipose tissue inflammation is presently unclear. The role of mitochondria in white adipocytes has long been neglected because of their low abundance. However, recent evidence suggests that mitochondria are essential for maintaining metabolic homeostasis in white adipocytes. In a series of recent studies, we found that mitochondrial function in white adipocytes is essential to the synthesis of adiponectin, which is the most abundant adipokine synthesized from adipocytes, with many favorable effects on metabolism, including improvement of insulin sensitivity and reduction of atherosclerotic processes and systemic inflammation. From these results, we propose a new hypothesis that mitochondrial dysfunction in adipocytes is a primary cause of adipose tissue inflammation and compared this hypothesis with a prevailing concept that “adipose tissue hypoxia” may underlie adipose tissue dysfunction in obesity. Recent studies have emphasized the role of the mitochondrial quality control mechanism in maintaining mitochondrial function. Future studies are warranted to test whether an inadequate mitochondrial quality control mechanism is responsible for mitochondrial dysfunction in adipocytes and adipose tissue inflammation.

Citations

Citations to this article as recorded by  
  • Prolonged Endurance Exercise Increases Macrophage Content and Mitochondrial Respiration in Adipose Tissue in Trained Men
    Ronni Eg Sahl, Ioanna Patsi, Mikkel Thunestvedt Hansen, Tue Rømer, Jacob Frandsen, Hanne Kruuse Rasmusen, Arthur Ingersen, Steen Seier Poulsen, Flemming Dela, Steen Larsen, Jørn Wulff Helge
    The Journal of Clinical Endocrinology & Metabolism.2024; 109(2): e799.     CrossRef
  • The Role of Ion-Transporting Proteins in Human Disease
    Yoshinori Marunaka
    International Journal of Molecular Sciences.2024; 25(3): 1726.     CrossRef
  • The Role of Obesity in Type 2 Diabetes Mellitus—An Overview
    Preethi Chandrasekaran, Ralf Weiskirchen
    International Journal of Molecular Sciences.2024; 25(3): 1882.     CrossRef
  • The Metabolic Syndrome, a Human Disease
    Marià Alemany
    International Journal of Molecular Sciences.2024; 25(4): 2251.     CrossRef
  • Inflammation‐mediated metabolic regulation in adipose tissue
    Shujie Xu, Feng Lu, Jianhua Gao, Yi Yuan
    Obesity Reviews.2024;[Epub]     CrossRef
  • Sleeve Gastrectomy Reduces Oxidative Stress and Reverses Mitochondrial Dysfunction Associated with Metabolic Syndrome
    Micaela M. Rossi, Franco J. Signorini, Tomas A. Castillo, María P. Scribano Parada, Federico Moser, Maria dC Baez
    Obesity Surgery.2024; 34(6): 2042.     CrossRef
  • The relationships between high-fat diet and metabolic syndrome: Potential mechanisms
    Chao Tang, Yuxin Wang, Zeyu Xu, Dan Chen, Jingguo Xu, Duo Yang, Li Zhang, Jun Liu, Juan Kan
    Food Bioscience.2024; 59: 104261.     CrossRef
  • Obesity, Dietary Patterns, and Hormonal Balance Modulation: Gender-Specific Impacts
    Elisa Mazza, Ersilia Troiano, Yvelise Ferro, Fabrizia Lisso, Martina Tosi, Ettore Turco, Roberta Pujia, Tiziana Montalcini
    Nutrients.2024; 16(11): 1629.     CrossRef
  • Mitochondrial quality control in human health and disease
    Bo-Hao Liu, Chen-Zhen Xu, Yi Liu, Zi-Long Lu, Ting-Lv Fu, Guo-Rui Li, Yu Deng, Guo-Qing Luo, Song Ding, Ning Li, Qing Geng
    Military Medical Research.2024;[Epub]     CrossRef
  • Adalimumab Treatment Effects on Inflammation and Adipose Tissue Mitochondrial Respiration in Hidradenitis Suppurativa
    Ronni Eg Sahl, Axel Illeris Poggi, Valdemar Wendelboe Nielsen, Yiqiu Yao, Ioanna Patsi, Steen Seier Poulsen, Flemming Dela, Steen Larsen, Simon Francis Thomsen, Jørn Wulff Helge
    Endocrinology, Diabetes & Metabolism.2024;[Epub]     CrossRef
  • Could very low-calorie ketogenic diets turn off low grade inflammation in obesity? Emerging evidence
    Luigi Barrea, Massimiliano Caprio, Mikiko Watanabe, Giuseppe Cammarata, Alessandra Feraco, Giovanna Muscogiuri, Ludovica Verde, Annamaria Colao, Silvia Savastano
    Critical Reviews in Food Science and Nutrition.2023; 63(26): 8320.     CrossRef
  • The emergent role of mitochondrial RNA modifications in metabolic alterations
    Hatim Boughanem, Yvonne Böttcher, João Tomé‐Carneiro, María‐Carmen López de las Hazas, Alberto Dávalos, Akin Cayir, Manuel Macias‐González
    WIREs RNA.2023;[Epub]     CrossRef
  • Age‐associated adipose tissue inflammation promotes monocyte chemotaxis and enhances atherosclerosis
    Jianrui Song, Diana Farris, Paola Ariza, Smriti Moorjani, Mita Varghese, Muriel Blin, Judy Chen, Daniel Tyrrell, Min Zhang, Kanakadurga Singer, Morgan Salmon, Daniel R. Goldstein
    Aging Cell.2023;[Epub]     CrossRef
  • Obesity, diabetes mellitus, and cardiometabolic risk: An Obesity Medicine Association (OMA) Clinical Practice Statement (CPS) 2023
    Harold Edward Bays, Shagun Bindlish, Tiffany Lowe Clayton
    Obesity Pillars.2023; 5: 100056.     CrossRef
  • A role of STING signaling in obesity-induced lung inflammation
    Yong Qi, Zhuhua Wu, Dan Chen, Li Zhu, Yunlei Yang
    International Journal of Obesity.2023; 47(4): 325.     CrossRef
  • Estrogens in Adipose Tissue Physiology and Obesity-Related Dysfunction
    Alina Kuryłowicz
    Biomedicines.2023; 11(3): 690.     CrossRef
  • White Adipose Tissue Dysfunction: Pathophysiology and Emergent Measurements
    Natalia Santillana, Camila Astudillo-Guerrero, Amanda D’Espessailles, Gonzalo Cruz
    Nutrients.2023; 15(7): 1722.     CrossRef
  • Pleiotropic and multi-systemic actions of physical exercise on PGC-1α signaling during the aging process
    Ivo Vieira de Sousa Neto, Ana Paula Pinto, Vitor Rosetto Muñoz, Rita de Cássia Marqueti, José Rodrigo Pauli, Eduardo Rochete Ropelle, Adelino Sanchez Ramos da Silva
    Ageing Research Reviews.2023; 87: 101935.     CrossRef
  • The impact of metabolic endotoxaemia on the browning process in human adipocytes
    Farah Omran, Alice M. Murphy, Awais Z. Younis, Ioannis Kyrou, Jana Vrbikova, Vojtech Hainer, Petra Sramkova, Martin Fried, Graham Ball, Gyanendra Tripathi, Sudhesh Kumar, Philip G. McTernan, Mark Christian
    BMC Medicine.2023;[Epub]     CrossRef
  • Molecular Mechanisms of Obesity-Induced Development of Insulin Resistance and Promotion of Amyloid-β Accumulation: Dietary Therapy Using Weak Organic Acids via Improvement of Lowered Interstitial Fluid pH
    Yoshinori Marunaka
    Biomolecules.2023; 13(5): 779.     CrossRef
  • From Obesity-Induced Low-Grade Inflammation to Lipotoxicity and Mitochondrial Dysfunction: Altered Multi-Crosstalk between Adipose Tissue and Metabolically Active Organs
    Gina Cavaliere, Fabiano Cimmino, Giovanna Trinchese, Angela Catapano, Lidia Petrella, Margherita D’Angelo, Lucio Lucchin, Maria Pina Mollica
    Antioxidants.2023; 12(6): 1172.     CrossRef
  • Receptor for the Advanced Glycation End Products (RAGE) Pathway in Adipose Tissue Metabolism
    Klaudia Gutowska, Krzysztof Czajkowski, Alina Kuryłowicz
    International Journal of Molecular Sciences.2023; 24(13): 10982.     CrossRef
  • Aerobic and Resistance Training Attenuate Differently Knee Joint Damage Caused by a High-Fat–High-Sucrose Diet in a Rat Model
    Nada Abughazaleh, Kevin Boldt, Jaqueline Lourdes Rios, Stela Marcia Mattiello, Kelsey H. Collins, Ruth-Anne Seerattan, Walter Herzog
    CARTILAGE.2023;[Epub]     CrossRef
  • Exercise mitigates age-related metabolic diseases by improving mitochondrial dysfunction
    Dandan Jia, Zhenjun Tian, Ru Wang
    Ageing Research Reviews.2023; 91: 102087.     CrossRef
  • Mitochondrial dynamics and metabolism across skin cells: implications for skin homeostasis and aging
    Ines Martic, Federica Papaccio, Barbara Bellei, Maria Cavinato
    Frontiers in Physiology.2023;[Epub]     CrossRef
  • Diabetes Mellitus, Energy Metabolism, and COVID-19
    Caterina Conte, Elisa Cipponeri, Michael Roden
    Endocrine Reviews.2023;[Epub]     CrossRef
  • Influence of Breastfeeding on the State of Meta-Inflammation in Obesity—A Narrative Review
    Dominika Mazur, Małgorzata Satora, Anna K. Rekowska, Zuzanna Kabała, Aleksandra Łomża, Żaneta Kimber-Trojnar, Bożena Leszczyńska-Gorzelak
    Current Issues in Molecular Biology.2023; 45(11): 9003.     CrossRef
  • AGER-1 Long Non-Coding RNA Levels Correlate with the Expression of the Advanced Glycosylation End-Product Receptor, a Regulator of the Inflammatory Response in Visceral Adipose Tissue of Women with Obesity and Type 2 Diabetes Mellitus
    Klaudia Gutowska, Krzysztof Koźniewski, Michał Wąsowski, Marta Izabela Jonas, Zbigniew Bartoszewicz, Wojciech Lisik, Maurycy Jonas, Artur Binda, Paweł Jaworski, Wiesław Tarnowski, Bartłomiej Noszczyk, Monika Puzianowska-Kuźnicka, Krzysztof Czajkowski, Ali
    International Journal of Molecular Sciences.2023; 24(24): 17447.     CrossRef
  • Pharmacological treatment with FGF21 strongly improves plasma cholesterol metabolism to reduce atherosclerosis
    Cong Liu, Milena Schönke, Enchen Zhou, Zhuang Li, Sander Kooijman, Mariëtte R Boon, Mikael Larsson, Kristina Wallenius, Niek Dekker, Louise Barlind, Xiao-Rong Peng, Yanan Wang, Patrick C N Rensen
    Cardiovascular Research.2022; 118(2): 489.     CrossRef
  • Obesity-Related Adipose Tissue Remodeling in the Light of Extracellular Mitochondria Transfer
    Simon Lecoutre, Karine Clément, Isabelle Dugail
    International Journal of Molecular Sciences.2022; 23(2): 632.     CrossRef
  • IL-4 polarized human macrophage exosomes control cardiometabolic inflammation and diabetes in obesity
    Tuan Anh Phu, Martin Ng, Ngan K. Vu, Laura Bouchareychas, Robert L. Raffai
    Molecular Therapy.2022; 30(6): 2274.     CrossRef
  • Insulin-inducible THRSP maintains mitochondrial function and regulates sphingolipid metabolism in human adipocytes
    Maria A. Ahonen, Marcus Höring, Van Dien Nguyen, Sami Qadri, Juuso H. Taskinen, Meghana Nagaraj, Martin Wabitsch, Pamela Fischer-Posovszky, You Zhou, Gerhard Liebisch, P. A. Nidhina Haridas, Hannele Yki-Järvinen, Vesa M. Olkkonen
    Molecular Medicine.2022;[Epub]     CrossRef
  • Modulation of adipose inflammation by cellular retinoic acid-binding protein 1
    Chin-Wen Wei, Jennifer Nhieu, Yu-Lung Lin, Li-Na Wei
    International Journal of Obesity.2022; 46(10): 1759.     CrossRef
  • The Role of Adipokines in Pancreatic Cancer
    Qi Wang, Huizhi Wang, Yuntao Ding, Mengtian Wan, Min Xu
    Frontiers in Oncology.2022;[Epub]     CrossRef
  • Epigenetic Reprogramming of the Inflammatory Response in Obesity and Type 2 Diabetes
    Federica Zatterale, Gregory Alexander Raciti, Immacolata Prevenzano, Alessia Leone, Michele Campitelli, Veronica De Rosa, Francesco Beguinot, Luca Parrillo
    Biomolecules.2022; 12(7): 982.     CrossRef
  • Cellular Metabolism and Bioenergetic Function in Human Fibroblasts and Preadipocytes of Type 2 Familial Partial Lipodystrophy
    Cristina Algieri, Chiara Bernardini, Fabiana Trombetti, Elisa Schena, Augusta Zannoni, Monica Forni, Salvatore Nesci
    International Journal of Molecular Sciences.2022; 23(15): 8659.     CrossRef
  • Shared pathobiology identifies AMPK as a therapeutic target for obesity and autosomal dominant polycystic kidney disease
    Ioan-Andrei Iliuta, Xuewen Song, Lauren Pickel, Amirreza Haghighi, Ravi Retnakaran, James Scholey, Hoon-Ki Sung, Gregory R. Steinberg, York Pei
    Frontiers in Molecular Biosciences.2022;[Epub]     CrossRef
  • Hypoxia as a Double-Edged Sword to Combat Obesity and Comorbidities
    Ruwen Wang, Qin Sun, Xianmin Wu, Yiyin Zhang, Xiaorui Xing, Kaiqing Lin, Yue Feng, Mingqi Wang, Yibing Wang, Ru Wang
    Cells.2022; 11(23): 3735.     CrossRef
  • Macrophage and Adipocyte Mitochondrial Dysfunction in Obesity-Induced Metabolic Diseases
    Liwen Wang, Jie Hu, Haiyan Zhou
    The World Journal of Men's Health.2021; 39(4): 606.     CrossRef
  • ESRRA (estrogen related receptor alpha) is a critical regulator of intestinal homeostasis through activation of autophagic flux via gut microbiota
    Sup Kim, June-Young Lee, Seul Gi Shin, Jin Kyung Kim, Prashanta Silwal, Young Jae Kim, Na-Ri Shin, Pil Soo Kim, Minho Won, Sang-Hee Lee, Soo Yeon Kim, Miwa Sasai, Masahiro Yamamoto, Jin-Man Kim, Jin-Woo Bae, Eun-Kyeong Jo
    Autophagy.2021; 17(10): 2856.     CrossRef
  • GDF15 as a central mediator for integrated stress response and a promising therapeutic molecule for metabolic disorders and NASH
    Kook Hwan Kim, Myung-Shik Lee
    Biochimica et Biophysica Acta (BBA) - General Subjects.2021; 1865(3): 129834.     CrossRef
  • The Influence of Obesity and Associated Fatty Acids on Placental Inflammation
    Alison J. Eastman, Rebecca E. Moore, Steven D. Townsend, Jennifer A. Gaddy, David M. Aronoff
    Clinical Therapeutics.2021; 43(2): 265.     CrossRef
  • Targeting the G protein-coupled estrogen receptor (GPER) in obesity and diabetes
    Geetanjali Sharma, Eric R. Prossnitz
    Endocrine and Metabolic Science.2021; 2: 100080.     CrossRef
  • Changes in Body Composition Are Associated with Metabolic Changes and the Risk of Metabolic Syndrome
    Yun Hwan Oh, Seulggie Choi, Gyeongsil Lee, Joung Sik Son, Kyae Hyung Kim, Sang Min Park
    Journal of Clinical Medicine.2021; 10(4): 745.     CrossRef
  • N6-Adenosine Methylation (m6A) RNA Modification: an Emerging Role in Cardiovascular Diseases
    Ye-shi Chen, Xin-ping Ouyang, Xiao-hua Yu, Petr Novák, Le Zhou, Ping-ping He, Kai Yin
    Journal of Cardiovascular Translational Research.2021; 14(5): 857.     CrossRef
  • From Metabolic Syndrome to Neurological Diseases: Role of Autophagy
    Jessica Maiuolo, Micaela Gliozzi, Vincenzo Musolino, Cristina Carresi, Federica Scarano, Saverio Nucera, Miriam Scicchitano, Francesca Bosco, Stefano Ruga, Maria Caterina Zito, Roberta Macri, Rosamaria Bulotta, Carolina Muscoli, Vincenzo Mollace
    Frontiers in Cell and Developmental Biology.2021;[Epub]     CrossRef
  • Absent Exercise-Induced Improvements in Fat Oxidation in Women With Polycystic Ovary Syndrome After High-Intensity Interval Training
    Sofie Lionett, Ida Almenning Kiel, Ragnhild Røsbjørgen, Stian Lydersen, Steen Larsen, Trine Moholdt
    Frontiers in Physiology.2021;[Epub]     CrossRef
  • Roles of interstitial fluid pH and weak organic acids in development and amelioration of insulin resistance
    Yoshinori Marunaka
    Biochemical Society Transactions.2021; 49(2): 715.     CrossRef
  • The Role of Mitochondrial Adaptation and Metabolic Flexibility in the Pathophysiology of Obesity and Insulin Resistance: an Updated Overview
    Dimitrios Tsilingiris, Evangelia Tzeravini, Chrysi Koliaki, Maria Dalamaga, Alexander Kokkinos
    Current Obesity Reports.2021; 10(3): 191.     CrossRef
  • Obesity-Related Inflammation and Endothelial Dysfunction in COVID-19: Impact on Disease Severity
    Andrea De Lorenzo, Vanessa Estato, Hugo C Castro-Faria-Neto, Eduardo Tibirica
    Journal of Inflammation Research.2021; Volume 14: 2267.     CrossRef
  • Thermogenic Fat: Development, Physiological Function, and Therapeutic Potential
    Bruna B. Brandão, Ankita Poojari, Atefeh Rabiee
    International Journal of Molecular Sciences.2021; 22(11): 5906.     CrossRef
  • Metabolic Syndrome in an Aging Society – Role of Oxidant-Antioxidant Imbalance and Inflammation Markers in Disentangling Atherosclerosis
    Sylwia Dziegielewska-Gesiak
    Clinical Interventions in Aging.2021; Volume 16: 1057.     CrossRef
  • Recruitment and remodeling of peridroplet mitochondria in human adipose tissue
    Rebeca Acín-Perez, Anton Petcherski, Michaela Veliova, Ilan Y. Benador, Essam A. Assali, Georgia Colleluori, Saverio Cinti, Alexandra J. Brownstein, Siyouneh Baghdasarian, Masha J. Livhits, Michael W. Yeh, Karthickeyan Chella Krishnan, Laurent Vergnes, Na
    Redox Biology.2021; 46: 102087.     CrossRef
  • New Insights Into Mitochondrial Dysfunction at Disease Susceptibility Loci in the Development of Type 2 Diabetes
    Hannah Maude, Winston Lau, Nikolas Maniatis, Toby Andrew
    Frontiers in Endocrinology.2021;[Epub]     CrossRef
  • Effects of sleeve gastrectomy on bone mass, microstructure of femurs and bone metabolism associated serum factors in obese rats
    Ying Xue, Ran Li, Yong Zhao, Ling Li, Yun Zhou
    BMC Endocrine Disorders.2021;[Epub]     CrossRef
  • The cyclin dependent kinase inhibitor Roscovitine prevents diet-induced metabolic disruption in obese mice
    Nabil Rabhi, Kathleen Desevin, Briana Noel Cortez, Ryan Hekman, Jean Z. Lin, Andrew Emili, Stephen R. Farmer
    Scientific Reports.2021;[Epub]     CrossRef
  • Reliability and variation in mitochondrial respiration in human adipose tissue
    Ronni Eg Sahl, Eva Frederikke Høy Helms, Malte Schmücker, Mathias Flensted-Jensen, Arthur Ingersen, Thomas Morville, Flemming Dela, Jørn Wulff Helge, Steen Larsen
    Adipocyte.2021; 10(1): 605.     CrossRef
  • Inhibition of protein tyrosine phosphatase improves mitochondrial bioenergetics and dynamics, reduces oxidative stress, and enhances adipogenic differentiation potential in metabolically impaired progenitor stem cells
    Katarzyna Kornicka-Garbowska, Lynda Bourebaba, Michael Röcken, Krzysztof Marycz
    Cell Communication and Signaling.2021;[Epub]     CrossRef
  • microRNAs in Human Adipose Tissue Physiology and Dysfunction
    Alina Kurylowicz
    Cells.2021; 10(12): 3342.     CrossRef
  • Aging, obese-insulin resistance, and bone remodeling
    Napatsorn Imerb, Chanisa Thonusin, Nipon Chattipakorn, Siriporn C. Chattipakorn
    Mechanisms of Ageing and Development.2020; 191: 111335.     CrossRef
  • Chronic Adipose Tissue Inflammation Linking Obesity to Insulin Resistance and Type 2 Diabetes
    Federica Zatterale, Michele Longo, Jamal Naderi, Gregory Alexander Raciti, Antonella Desiderio, Claudia Miele, Francesco Beguinot
    Frontiers in Physiology.2020;[Epub]     CrossRef
  • Is Mitochondrial Dysfunction a Common Root of Noncommunicable Chronic Diseases?
    Alexis Diaz-Vegas, Pablo Sanchez-Aguilera, James R Krycer, Pablo E Morales, Matías Monsalves-Alvarez, Mariana Cifuentes, Beverly A Rothermel, Sergio Lavandero
    Endocrine Reviews.2020;[Epub]     CrossRef
  • Inflammatory Signaling and Brown Fat Activity
    Farah Omran, Mark Christian
    Frontiers in Endocrinology.2020;[Epub]     CrossRef
  • Omega-3 fatty acids as regulators of brown/beige adipose tissue: from mechanisms to therapeutic potential
    Marta Fernández-Galilea, Elisa Félix-Soriano, Ignacio Colón-Mesa, Xavier Escoté, Maria J. Moreno-Aliaga
    Journal of Physiology and Biochemistry.2020; 76(2): 251.     CrossRef
  • Anti-Inflammatory Strategies Targeting Metaflammation in Type 2 Diabetes
    Alina Kuryłowicz, Krzysztof Koźniewski
    Molecules.2020; 25(9): 2224.     CrossRef
  • Obese Adipose Tissue Secretion Induces Inflammation in Preadipocytes: Role of Toll-Like Receptor-4
    Mariana Renovato-Martins, Catharina Moreira-Nunes, Georgia C. Atella, Christina Barja-Fidalgo, João Alfredo de Moraes
    Nutrients.2020; 12(9): 2828.     CrossRef
  • Diabetes and Metabolism Journal in 2020: Good to Great
    In-Kyung Jeong
    Diabetes & Metabolism Journal.2020; 44(1): 1.     CrossRef
  • The Effect of Silibinin on Protein Expression Profile in White Adipose Tissue of Obese Mice
    Fei Wang, Shuchun Chen, Luping Ren, Yichao Wang, Zelin Li, Tiantian Song, He Zhang, Qiwen Yang
    Frontiers in Pharmacology.2020;[Epub]     CrossRef
  • Beneficial Effects of Bariatric Surgery-Induced by Weight Loss on the Proteome of Abdominal Subcutaneous Adipose Tissue
    Bárbara María Varela-Rodríguez, Paula Juiz-Valiña, Luis Varela, Elena Outeiriño-Blanco, Susana Belén Bravo, María Jesús García-Brao, Enrique Mena, José Francisco Noguera, Javier Valero-Gasalla, Fernando Cordido, Susana Sangiao-Alvarellos
    Journal of Clinical Medicine.2020; 9(1): 213.     CrossRef
  • Impact of Skeletal Muscle Mass on Metabolic Health
    Gyuri Kim, Jae Hyeon Kim
    Endocrinology and Metabolism.2020; 35(1): 1.     CrossRef
  • Sea buckthorn (Hippophae rhamnoides L.) oil enhances proliferation, adipocytes differentiation and insulin sensitivity in 3T3-L1 cells
    Ting Zhang, Xuze Qin, Yuxin Cao, Jianxin Zhang, Junxing Zhao
    Food Science and Biotechnology.2020; 29(11): 1511.     CrossRef
  • Adipose tissue secretory profile and cardiometabolic risk in obesity
    Pengcheng Zhang, Daniels Konja, Yu Wang
    Endocrine and Metabolic Science.2020; 1(3-4): 100061.     CrossRef
  • Mitochondrial Dynamics in the Brain Are Associated With Feeding, Glucose Homeostasis, and Whole-Body Metabolism
    Jessica L. Haigh, Lauryn E. New, Beatrice M. Filippi
    Frontiers in Endocrinology.2020;[Epub]     CrossRef
  • Adipogenesis: A Necessary but Harmful Strategy
    Mohammed El Hafidi, Mabel Buelna-Chontal, Fausto Sánchez-Muñoz, Roxana Carbó
    International Journal of Molecular Sciences.2019; 20(15): 3657.     CrossRef

Diabetes Metab J : Diabetes & Metabolism Journal
Close layer
TOP