1
|
Chen P, Yao L, Yuan M, Wang Z, Zhang Q, Jiang Y, Li L. Mitochondrial dysfunction: A promising therapeutic target for liver diseases. Genes Dis 2024; 11:101115. [PMID: 38299199 PMCID: PMC10828599 DOI: 10.1016/j.gendis.2023.101115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 07/15/2023] [Accepted: 08/10/2023] [Indexed: 02/02/2024] Open
Abstract
The liver is an important metabolic and detoxification organ and hence demands a large amount of energy, which is mainly produced by the mitochondria. Liver tissues of patients with alcohol-related or non-alcohol-related liver diseases contain ultrastructural mitochondrial lesions, mitochondrial DNA damage, disturbed mitochondrial dynamics, and compromised ATP production. Overproduction of mitochondrial reactive oxygen species induces oxidative damage to mitochondrial proteins and mitochondrial DNA, decreases mitochondrial membrane potential, triggers hepatocyte inflammation, and promotes programmed cell death, all of which impair liver function. Mitochondrial DNA may be a potential novel non-invasive biomarker of the risk of progression to liver cirrhosis and hepatocellular carcinoma in patients infected with the hepatitis B virus. We herein present a review of the mechanisms of mitochondrial dysfunction in the development of acute liver injury and chronic liver diseases, such as hepatocellular carcinoma, viral hepatitis, drug-induced liver injury, alcoholic liver disease, and non-alcoholic fatty liver disease. This review also discusses mitochondrion-centric therapies for treating liver diseases.
Collapse
Affiliation(s)
- Ping Chen
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Lichao Yao
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Mengqin Yuan
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Zheng Wang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Qiuling Zhang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Yingan Jiang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Lanjuan Li
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| |
Collapse
|
2
|
Tian J, Fan J, Zhang T. Mitochondria as a target for exercise-mitigated type 2 diabetes. J Mol Histol 2023; 54:543-557. [PMID: 37874501 DOI: 10.1007/s10735-023-10158-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 09/17/2023] [Indexed: 10/25/2023]
Abstract
Type 2 diabetes mellitus (T2DM) is one of most common metabolic diseases and continues to be a leading cause of death worldwide. Although great efforts have been made to elucidate the pathogenesis of diabetes, the underlying mechanism still remains unclear. Notably, overwhelming evidence has demonstrated that mitochondria are tightly correlated with the development of T2DM, and the defects of mitochondrial function in peripheral insulin-responsive tissues, such as skeletal muscle, liver and adipose tissue, are crucial drivers of T2DM. Furthermore, exercise training is considered as an effective stimulus for improving insulin sensitivity and hence is regarded as the best strategy to prevent and treat T2DM. Although the precise mechanisms by which exercise alleviates T2DM are not fully understood, mitochondria may be critical for the beneficial effects of exercise.
Collapse
Affiliation(s)
- Jingjing Tian
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai, China
| | - Jingcheng Fan
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai, China
| | - Tan Zhang
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China.
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai, China.
| |
Collapse
|
3
|
Paula VG, Sinzato YK, Gallego FQ, Cruz LL, Aquino AMD, Scarano WR, Corrente JE, Volpato GT, Damasceno DC. Intergenerational Hyperglycemia Impairs Mitochondrial Function and Follicular Development and Causes Oxidative Stress in Rat Ovaries Independent of the Consumption of a High-Fat Diet. Nutrients 2023; 15:4407. [PMID: 37892483 PMCID: PMC10609718 DOI: 10.3390/nu15204407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 09/16/2023] [Accepted: 09/20/2023] [Indexed: 10/29/2023] Open
Abstract
We analyzed the influence of maternal hyperglycemia and the post-weaning consumption of a high-fat diet on the mitochondrial function and ovarian development of the adult pups of diabetic rats. Female rats received citrate buffer (Control-C) or Streptozotocin (for diabetes induction-D) on postnatal day 5. These adult rats were mated to obtain female pups (O) from control dams (OC) or from diabetic dams (OD), and they received a standard diet (SD) or high-fat diet (HFD) from weaning to adulthood and were distributed into OC/SD, OC/HFD, OD/SD, and OD/HFD. In adulthood, the OGTT and AUC were performed. These rats were anesthetized and euthanized for sample collection. A high percentage of diabetic rats were found to be in the OD/HFD group (OD/HFD 40% vs. OC/SD 0% p < 0.05). Progesterone concentrations were lower in the experimental groups (OC/HFD 0.40 ± 0.04; OD/SD 0.30 ± 0.03; OD/HFD 0.24 ± 0.04 vs. OC/SD 0.45 ± 0.03 p < 0.0001). There was a lower expression of MFF (OD/SD 0.34 ± 0.33; OD/HFD 0.29 ± 0.2 vs. OC/SD 1.0 ± 0.41 p = 0.0015) and MFN2 in the OD/SD and OD/HFD groups (OD/SD 0.41 ± 0.21; OD/HFD 0.77 ± 0.18 vs. OC/SD 1.0 ± 0.45 p = 0.0037). The number of follicles was lower in the OD/SD and OD/HFD groups. A lower staining intensity for SOD and Catalase and higher staining intensity for MDA were found in ovarian cells in the OC/HFD, OD/SD, and OD/HFD groups. Fetal programming was responsible for mitochondrial dysfunction, ovarian reserve loss, and oxidative stress; the association of maternal diabetes with an HFD was responsible for the higher occurrence of diabetes in female adult pups.
Collapse
Affiliation(s)
- Verônyca Gonçalves Paula
- Laboratory of Experimental Research on Gynecology and Obstetrics, Gynecology, Postgraduate Course on Tocogynecology, Botucatu Medical School, São Paulo State University (Unesp), Botucatu 18618-687, SP, Brazil
| | - Yuri Karen Sinzato
- Laboratory of Experimental Research on Gynecology and Obstetrics, Gynecology, Postgraduate Course on Tocogynecology, Botucatu Medical School, São Paulo State University (Unesp), Botucatu 18618-687, SP, Brazil
| | - Franciane Quintanilha Gallego
- Laboratory of Experimental Research on Gynecology and Obstetrics, Gynecology, Postgraduate Course on Tocogynecology, Botucatu Medical School, São Paulo State University (Unesp), Botucatu 18618-687, SP, Brazil
| | - Larissa Lopes Cruz
- Laboratory of Experimental Research on Gynecology and Obstetrics, Gynecology, Postgraduate Course on Tocogynecology, Botucatu Medical School, São Paulo State University (Unesp), Botucatu 18618-687, SP, Brazil
| | - Ariana Musa de Aquino
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu 18618-689, SP, Brazil
| | - Wellerson Rodrigo Scarano
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu 18618-689, SP, Brazil
| | - José Eduardo Corrente
- Research Support Office, Botucatu Medical School, São Paulo State University (Unesp), Botucatu 18618-687, SP, Brazil
| | - Gustavo Tadeu Volpato
- Laboratory of System Physiology and Reproductive Toxicology, Institute of Biological and Health Sciences, Federal University of Mato Grosso (UFMT), Barra do Garças 78600-000, MG, Brazil
| | - Débora Cristina Damasceno
- Laboratory of Experimental Research on Gynecology and Obstetrics, Gynecology, Postgraduate Course on Tocogynecology, Botucatu Medical School, São Paulo State University (Unesp), Botucatu 18618-687, SP, Brazil
| |
Collapse
|
4
|
Sun X, Liu S, Cai J, Yang M, Li C, Tan M, He B. Mitochondrial Methionyl-tRNA Formyltransferase Deficiency Alleviates Metaflammation by Modulating Mitochondrial Activity in Mice. Int J Mol Sci 2023; 24:ijms24065999. [PMID: 36983072 PMCID: PMC10051599 DOI: 10.3390/ijms24065999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/03/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
Various studies have revealed the association of metabolic diseases with inflammation. Mitochondria are key organelles involved in metabolic regulation and important drivers of inflammation. However, it is uncertain whether the inhibition of mitochondrial protein translation results in the development of metabolic diseases, such that the metabolic benefits related to the inhibition of mitochondrial activity remain unclear. Mitochondrial methionyl-tRNA formyltransferase (Mtfmt) functions in the early stages of mitochondrial translation. In this study, we reveal that feeding with a high-fat diet led to the upregulation of Mtfmt in the livers of mice and that a negative correlation existed between hepatic Mtfmt gene expression and fasting blood glucose levels. A knockout mouse model of Mtfmt was generated to explore its possible role in metabolic diseases and its underlying molecular mechanisms. Homozygous knockout mice experienced embryonic lethality, but heterozygous knockout mice showed a global reduction in Mtfmt expression and activity. Moreover, heterozygous mice showed increased glucose tolerance and reduced inflammation, which effects were induced by the high-fat diet. The cellular assays showed that Mtfmt deficiency reduced mitochondrial activity and the production of mitochondrial reactive oxygen species and blunted nuclear factor-κB activation, which, in turn, downregulated inflammation in macrophages. The results of this study indicate that targeting Mtfmt-mediated mitochondrial protein translation to regulate inflammation might provide a potential therapeutic strategy for metabolic diseases.
Collapse
Affiliation(s)
- Xiaoxiao Sun
- Key Laboratory of Animal Physiology & Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Suyuan Liu
- Key Laboratory of Animal Physiology & Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Jiangxue Cai
- Key Laboratory of Animal Physiology & Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Miaoxin Yang
- Key Laboratory of Animal Physiology & Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Chenxuan Li
- Key Laboratory of Animal Physiology & Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Meiling Tan
- Key Laboratory of Animal Physiology & Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Bin He
- Key Laboratory of Animal Physiology & Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
5
|
Reynaud O, Wang J, Ayoub MB, Leduc-Gaudet JP, Mayaki D, Dulac M, Hussain SNA, Bergeron R, Gouspillou G. The impact of high-fat feeding and parkin overexpression on skeletal muscle mass, mitochondrial respiration, and H 2O 2 emission. Am J Physiol Cell Physiol 2023; 324:C366-C376. [PMID: 36571445 DOI: 10.1152/ajpcell.00388.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Obesity is a major risk factor for developing various health problems, including insulin resistance and type 2 diabetes. Although controversial, accumulation of mitochondrial dysfunction, and notably an increase in mitochondrial reactive oxygen species (ROS) production, was proposed as a key contributor leading to obesity-induced insulin resistance. Here, our goal was to investigate whether Parkin overexpression, a key regulator of the removal of dysfunctional mitochondria through mitophagy, could confer protection against obesity-induced mitochondrial dysfunction. To this end, intramuscular injections of adeno-associated viruses (AAVs) were performed to overexpress Parkin in limb muscle of 6-mo-old mice fed a control diet (CD) or a high-fat diet (HFD) for 12 wk. An AAV-expressing the green fluorescent protein (GFP) was used as control. HFD increased fat mass, altered glycemia, and resulted in insulin resistance. Parkin overexpression resulted in an increase in muscle mass in both CD and HFD mice. In CD mice, Parkin overexpression increased maximal mitochondrial respiration and lowered H2O2 emission. HFD increased mitochondrial respiration and, surprisingly, also lowered H2O2 emission. Parkin overexpression did not significantly impact mitochondrial function in HFD mice. Taken altogether, our results indicate that Parkin overexpression positively impacts muscle and mitochondrial health under basal conditions and challenges the notion that intrinsic mitochondrial dysfunction is involved in the development of insulin resistance caused by high-fat feeding.
Collapse
Affiliation(s)
- Olivier Reynaud
- Département des sciences biologiques, Faculté des Sciences, Université du Québec à Montréal, Montréal, Québec, Canada.,Département des sciences de l'activité physique, Faculté des Sciences, Université du Québec à Montréal, Montréal, Québec, Canada
| | - Jennifer Wang
- Département de médecine, Faculté de médecine, Université de Laval, Quebec City, Québec, Canada
| | - Marie-Belle Ayoub
- Département des sciences de l'activité physique, Faculté des Sciences, Université du Québec à Montréal, Montréal, Québec, Canada
| | - Jean-Philippe Leduc-Gaudet
- Meakins-Christie Laboratories and Translational Research in Respiratory Diseases Program, Department of Critical Care, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada.,Division of Experimental Medicine, Department of Medicine, McGill University, Montréal, Québec, Canada.,Venetian Institute of Molecular Medicine (VIMM) and Department of Biomedical Science, University of Padova, Padova, Italy
| | - Dominique Mayaki
- Meakins-Christie Laboratories and Translational Research in Respiratory Diseases Program, Department of Critical Care, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada.,Division of Experimental Medicine, Department of Medicine, McGill University, Montréal, Québec, Canada
| | - Maude Dulac
- Département des sciences biologiques, Faculté des Sciences, Université du Québec à Montréal, Montréal, Québec, Canada.,Département des sciences de l'activité physique, Faculté des Sciences, Université du Québec à Montréal, Montréal, Québec, Canada
| | - Sabah N A Hussain
- Meakins-Christie Laboratories and Translational Research in Respiratory Diseases Program, Department of Critical Care, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada.,Division of Experimental Medicine, Department of Medicine, McGill University, Montréal, Québec, Canada
| | - Raynald Bergeron
- École de kinésiologie et des sciences de l'activité physique, Faculté de médecine, Université de Montréal, Montréal, Québec, Canada
| | - Gilles Gouspillou
- Département des sciences de l'activité physique, Faculté des Sciences, Université du Québec à Montréal, Montréal, Québec, Canada.,Division of Experimental Medicine, Department of Medicine, McGill University, Montréal, Québec, Canada.,Centre de Recherche de l'Institut Universitaire de Gériatrie de Montréal, Montréal, Québec, Canada
| |
Collapse
|
6
|
Whitehall JC, Smith ALM, Greaves LC. Mitochondrial DNA Mutations and Ageing. Subcell Biochem 2023; 102:77-98. [PMID: 36600130 DOI: 10.1007/978-3-031-21410-3_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Mitochondria are subcellular organelles present in most eukaryotic cells which play a significant role in numerous aspects of cell biology. These include carbohydrate and fatty acid metabolism to generate cellular energy through oxidative phosphorylation, apoptosis, cell signalling, haem biosynthesis and reactive oxygen species production. Mitochondrial dysfunction is a feature of many human ageing tissues, and since the discovery that mitochondrial DNA mutations were a major underlying cause of changes in oxidative phosphorylation capacity, it has been proposed that they have a role in human ageing. However, there is still much debate on whether mitochondrial DNA mutations play a causal role in ageing or are simply a consequence of the ageing process. This chapter describes the structure of mammalian mitochondria, and the unique features of mitochondrial genetics, and reviews the current evidence surrounding the role of mitochondrial DNA mutations in the ageing process. It then focusses on more recent discoveries regarding the role of mitochondrial dysfunction in stem cell ageing and age-related inflammation.
Collapse
Affiliation(s)
- Julia C Whitehall
- Wellcome Centre for Mitochondrial Research, Biosciences Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - Anna L M Smith
- Wellcome Centre for Mitochondrial Research, Biosciences Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - Laura C Greaves
- Wellcome Centre for Mitochondrial Research, Biosciences Institute, Newcastle University, Newcastle Upon Tyne, UK.
| |
Collapse
|
7
|
Jiang Y, Xiang Y, Lin C, Zhang W, Yang Z, Xiang L, Xiao Y, Chen L, Ran Q, Li Z. Multifunctions of CRIF1 in cancers and mitochondrial dysfunction. Front Oncol 2022; 12:1009948. [PMID: 36263222 PMCID: PMC9574215 DOI: 10.3389/fonc.2022.1009948] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 09/14/2022] [Indexed: 11/23/2022] Open
Abstract
Sustaining proliferative signaling and enabling replicative immortality are two important hallmarks of cancer. The complex of cyclin-dependent kinase (CDK) and its cyclin plays a decisive role in the transformation of the cell cycle and is also critical in the initiation and progression of cancer. CRIF1, a multifunctional factor, plays a pivotal role in a series of cell biological progresses such as cell cycle, cell proliferation, and energy metabolism. CRIF1 is best known as a negative regulator of the cell cycle, on account of directly binding to Gadd45 family proteins or CDK2. In addition, CRIF1 acts as a regulator of several transcription factors such as Nur77 and STAT3 and partly determines the proliferation of cancer cells. Many studies showed that the expression of CRIF1 is significantly altered in cancers and potentially regarded as a tumor suppressor. This suggests that targeting CRIF1 would enhance the selectivity and sensitivity of cancer treatment. Moreover, CRIF1 might be an indispensable part of mitoribosome and is involved in the regulation of OXPHOS capacity. Further, CRIF1 is thought to be a novel target for the underlying mechanism of diseases with mitochondrial dysfunctions. In summary, this review would conclude the latest aspects of studies about CRIF1 in cancers and mitochondria-related diseases, shed new light on targeted therapy, and provide a more comprehensive holistic view.
Collapse
Affiliation(s)
- Yangzhou Jiang
- Laboratory of Radiation Biology, Laboratory Medicine Center, Department of Blood Transfusion, The Second Affiliated Hospital, Army Military Medical University, Chongqing, China
| | - Yang Xiang
- Laboratory of Radiation Biology, Laboratory Medicine Center, Department of Blood Transfusion, The Second Affiliated Hospital, Army Military Medical University, Chongqing, China
| | - Chuanchuan Lin
- Laboratory of Radiation Biology, Laboratory Medicine Center, Department of Blood Transfusion, The Second Affiliated Hospital, Army Military Medical University, Chongqing, China
| | - Weiwei Zhang
- Laboratory of Radiation Biology, Laboratory Medicine Center, Department of Blood Transfusion, The Second Affiliated Hospital, Army Military Medical University, Chongqing, China
| | - Zhenxing Yang
- Laboratory of Radiation Biology, Laboratory Medicine Center, Department of Blood Transfusion, The Second Affiliated Hospital, Army Military Medical University, Chongqing, China
| | - Lixin Xiang
- Laboratory of Radiation Biology, Laboratory Medicine Center, Department of Blood Transfusion, The Second Affiliated Hospital, Army Military Medical University, Chongqing, China
| | - Yanni Xiao
- Laboratory of Radiation Biology, Laboratory Medicine Center, Department of Blood Transfusion, The Second Affiliated Hospital, Army Military Medical University, Chongqing, China
| | - Li Chen
- Laboratory of Radiation Biology, Laboratory Medicine Center, Department of Blood Transfusion, The Second Affiliated Hospital, Army Military Medical University, Chongqing, China
| | - Qian Ran
- Laboratory of Radiation Biology, Laboratory Medicine Center, Department of Blood Transfusion, The Second Affiliated Hospital, Army Military Medical University, Chongqing, China
| | - Zhongjun Li
- Laboratory of Radiation Biology, Laboratory Medicine Center, Department of Blood Transfusion, The Second Affiliated Hospital, Army Military Medical University, Chongqing, China
- State Key Laboratory of Trauma, Burn and Combined Injuries, The Second Affiliated Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
8
|
Blackwood SJ, Horwath O, Moberg M, Pontén M, Apró W, Ekblom MM, Larsen FJ, Katz A. Extreme Variations in Muscle Fiber Composition Enable Detection of Insulin Resistance and Excessive Insulin Secretion. J Clin Endocrinol Metab 2022; 107:e2729-e2737. [PMID: 35405014 DOI: 10.1210/clinem/dgac221] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Indexed: 12/16/2022]
Abstract
CONTEXT Muscle fiber composition is associated with peripheral insulin action. OBJECTIVE We investigated whether extreme differences in muscle fiber composition are associated with alterations in peripheral insulin action and secretion in young, healthy subjects who exhibit normal fasting glycemia and insulinemia. METHODS Relaxation time following a tetanic contraction was used to identify subjects with a high or low expression of type I muscle fibers: group 1 (n = 11), area occupied by type I muscle fibers = 61.0 ± 11.8%, and group 2 (n = 8), type I area = 36.0 ± 4.9% (P < 0.001). Biopsies were obtained from the vastus lateralis muscle and analyzed for mitochondrial respiration on permeabilized fibers, muscle fiber composition, and capillary density. An intravenous glucose tolerance test was performed and indices of glucose tolerance, insulin sensitivity, and secretion were determined. RESULTS Glucose tolerance was similar between groups, whereas whole-body insulin sensitivity was decreased by ~50% in group 2 vs group 1 (P = 0.019). First-phase insulin release (area under the insulin curve during 10 minutes after glucose infusion) was increased by almost 4-fold in group 2 vs group 1 (P = 0.01). Whole-body insulin sensitivity was correlated with percentage area occupied by type I fibers (r = 0.54; P = 0.018) and capillary density in muscle (r = 0.61; P = 0.005) but not with mitochondrial respiration. Insulin release was strongly related to percentage area occupied by type II fibers (r = 0.93; P < 0.001). CONCLUSIONS Assessment of muscle contractile function in young healthy subjects may prove useful in identifying individuals with insulin resistance and enhanced glucose-stimulated insulin secretion prior to onset of clinical manifestations.
Collapse
Affiliation(s)
- Sarah J Blackwood
- Åstrand Laboratory, Department of Physiology, Nutrition and Biomechanics, The Swedish School of Sport and Health Sciences, Stockholm, Sweden
| | - Oscar Horwath
- Åstrand Laboratory, Department of Physiology, Nutrition and Biomechanics, The Swedish School of Sport and Health Sciences, Stockholm, Sweden
| | - Marcus Moberg
- Åstrand Laboratory, Department of Physiology, Nutrition and Biomechanics, The Swedish School of Sport and Health Sciences, Stockholm, Sweden
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Marjan Pontén
- Åstrand Laboratory, Department of Physiology, Nutrition and Biomechanics, The Swedish School of Sport and Health Sciences, Stockholm, Sweden
| | - William Apró
- Åstrand Laboratory, Department of Physiology, Nutrition and Biomechanics, The Swedish School of Sport and Health Sciences, Stockholm, Sweden
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Maria M Ekblom
- Department of Physical Activity and Health, The Swedish School of Sport and Health Sciences, Stockholm, Sweden
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Filip J Larsen
- Åstrand Laboratory, Department of Physiology, Nutrition and Biomechanics, The Swedish School of Sport and Health Sciences, Stockholm, Sweden
| | - Abram Katz
- Åstrand Laboratory, Department of Physiology, Nutrition and Biomechanics, The Swedish School of Sport and Health Sciences, Stockholm, Sweden
| |
Collapse
|
9
|
Mitochondrial protein import determines lifespan through metabolic reprogramming and de novo serine biosynthesis. Nat Commun 2022; 13:651. [PMID: 35115503 PMCID: PMC8814026 DOI: 10.1038/s41467-022-28272-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 01/13/2022] [Indexed: 02/04/2023] Open
Abstract
Sustained mitochondrial fitness relies on coordinated biogenesis and clearance. Both processes are regulated by constant targeting of proteins into the organelle. Thus, mitochondrial protein import sets the pace for mitochondrial abundance and function. However, our understanding of mitochondrial protein translocation as a regulator of longevity remains enigmatic. Here, we targeted the main protein import translocases and assessed their contribution to mitochondrial abundance and organismal physiology. We find that reduction in cellular mitochondrial load through mitochondrial protein import system suppression, referred to as MitoMISS, elicits a distinct longevity paradigm. We show that MitoMISS triggers the mitochondrial unfolded protein response, orchestrating an adaptive reprogramming of metabolism. Glycolysis and de novo serine biosynthesis are causatively linked to longevity, whilst mitochondrial chaperone induction is dispensable for lifespan extension. Our findings extent the pro-longevity role of UPRmt and provide insight, relevant to the metabolic alterations that promote or undermine survival and longevity. Mitochondrial function is linked to lifespan. Here the authors show that inhibition of mitochondrial protein import leads to a reduction in mitochondrial abundance and extends lifespan in Caenorhabditis elegans via activation of glycolysis and de novo serine biosynthesis.
Collapse
|
10
|
Wang D, Day EA, Townsend LK, Djordjevic D, Jørgensen SB, Steinberg GR. GDF15: emerging biology and therapeutic applications for obesity and cardiometabolic disease. Nat Rev Endocrinol 2021; 17:592-607. [PMID: 34381196 DOI: 10.1038/s41574-021-00529-7] [Citation(s) in RCA: 174] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/17/2021] [Indexed: 02/06/2023]
Abstract
Growth differentiation factor 15 (GDF15) is a member of the TGFβ superfamily whose expression is increased in response to cellular stress and disease as well as by metformin. Elevations in GDF15 reduce food intake and body mass in animal models through binding to glial cell-derived neurotrophic factor family receptor alpha-like (GFRAL) and the recruitment of the receptor tyrosine kinase RET in the hindbrain. This effect is largely independent of other appetite-regulating hormones (for example, leptin, ghrelin or glucagon-like peptide 1). Consistent with an important role for the GDF15-GFRAL signalling axis, some human genetic studies support an interrelationship with human obesity. Furthermore, findings in both mice and humans have shown that metformin and exercise increase circulating levels of GDF15. GDF15 might also exert anti-inflammatory effects through mechanisms that are not fully understood. These unique and distinct mechanisms for suppressing food intake and inflammation makes GDF15 an appealing candidate to treat many metabolic diseases, including obesity, type 2 diabetes mellitus, non-alcoholic fatty liver disease, cardiovascular disease and cancer cachexia. Here, we review the mechanisms regulating GDF15 production and secretion, GDF15 signalling in different cell types, and how GDF15-targeted pharmaceutical approaches might be effective in the treatment of metabolic diseases.
Collapse
Affiliation(s)
- Dongdong Wang
- Centre for Metabolism, Obesity and Diabetes Research and the Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Emily A Day
- Centre for Metabolism, Obesity and Diabetes Research and the Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Logan K Townsend
- Centre for Metabolism, Obesity and Diabetes Research and the Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Djordje Djordjevic
- Global Obesity and Liver Disease Research, Novo Nordisk A/S, Maaloev, Denmark
| | | | - Gregory R Steinberg
- Centre for Metabolism, Obesity and Diabetes Research and the Department of Medicine, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
11
|
Rodriguez-Cuenca S, Lelliot CJ, Campbell M, Peddinti G, Martinez-Uña M, Ingvorsen C, Dias AR, Relat J, Mora S, Hyötyläinen T, Zorzano A, Orešič M, Bjursell M, Bohlooly-Y M, Lindén D, Vidal-Puig A. Allostatic hypermetabolic response in PGC1α/β heterozygote mouse despite mitochondrial defects. FASEB J 2021; 35:e21752. [PMID: 34369602 DOI: 10.1096/fj.202100262rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 06/05/2021] [Accepted: 06/08/2021] [Indexed: 12/25/2022]
Abstract
Aging, obesity, and insulin resistance are associated with low levels of PGC1α and PGC1β coactivators and defective mitochondrial function. We studied mice deficient for PGC1α and PGC1β [double heterozygous (DH)] to investigate their combined pathogenic contribution. Contrary to our hypothesis, DH mice were leaner, had increased energy dissipation, a pro-thermogenic profile in BAT and WAT, and improved carbohydrate metabolism compared to wild types. WAT showed upregulation of mitochondriogenesis/oxphos machinery upon allelic compensation of PGC1α4 from the remaining allele. However, DH mice had decreased mitochondrial OXPHOS and biogenesis transcriptomes in mitochondria-rich organs. Despite being metabolically healthy, mitochondrial defects in DH mice impaired muscle fiber remodeling and caused qualitative changes in the hepatic lipidome. Our data evidence first the existence of organ-specific compensatory allostatic mechanisms are robust enough to drive an unexpected phenotype. Second, optimization of adipose tissue bioenergetics is sufficient to maintain a healthy metabolic phenotype despite a broad severe mitochondrial dysfunction in other relevant metabolic organs. Third, the decrease in PGC1s in adipose tissue of obese and diabetic patients is in contrast with the robustness of the compensatory upregulation in the adipose of the DH mice.
Collapse
Affiliation(s)
| | | | - Mark Campbell
- Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Gopal Peddinti
- VTT, Technical Research Center of Finland, Espoo, Finland
| | - Maite Martinez-Uña
- Department of Physiology, University of the Basque Country UPV/EHU, Bilbao, Spain
| | - Camilla Ingvorsen
- Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Ana Rita Dias
- Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Joana Relat
- Department of Nutrition, Food Science and Gastronomy, School of Pharmacy and Food Science, Food and Nutrition Torribera Campus, University of Barcelona (UB), Santa Coloma de Gramenet, Spain
- INSA-UB, Nutrition and Food Safety Research Institute, University of Barcelona, Barcelona, Spain
| | - Silvia Mora
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, The University of Liverpool, Liverpool, UK
| | | | - Antonio Zorzano
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Dept. Biochemistry and Molecular Biomedicine, University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Matej Orešič
- School of Science and Technology, Örebro University, Örebro, Sweden
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Mikael Bjursell
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | | | - Daniel Lindén
- Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
- Division of Endocrinology, Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Antonio Vidal-Puig
- Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
| |
Collapse
|
12
|
Gao H, Tripathi U, Trushin S, Okromelidze L, Pichurin NP, Wei L, Zhuang Y, Wang L, Trushina E. A genome-wide association study in human lymphoblastoid cells supports safety of mitochondrial complex I inhibitor. Mitochondrion 2021; 58:83-94. [PMID: 33610756 PMCID: PMC8743030 DOI: 10.1016/j.mito.2021.02.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 02/08/2021] [Indexed: 01/12/2023]
Abstract
Novel therapeutic strategies for Alzheimer's disease (AD) are of the greatest priority given the consistent failure of recent clinical trials focused on Aβ or pTau. Earlier, we demonstrated that mild mitochondrial complex I inhibitor CP2 blocks neurodegeneration and cognitive decline in multiple mouse models of AD. To evaluate the safety of CP2 in humans, we performed a genome-wide association study (GWAS) using 196 lymphoblastoid cell lines and identified 11 SNP loci and 64 mRNA expression probe sets that potentially associate with CP2 susceptibility. Using primary mouse neurons and pharmacokinetic study, we show that CP2 is generally safe at a therapeutic dose.
Collapse
Affiliation(s)
- Huanyao Gao
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Utkarsh Tripathi
- Department of Neurology, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Sergey Trushin
- Department of Neurology, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Lela Okromelidze
- Department of Neurology, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Nicholas P Pichurin
- Department of Neurology, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Lixuan Wei
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Yongxian Zhuang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Liewei Wang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Eugenia Trushina
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA; Department of Neurology, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA.
| |
Collapse
|
13
|
Wang L, Guo Y, Pan M, Li X, Huang D, Liu Y, Wu C, Zhang W, Mai K. Functions of Forkhead Box O on Glucose Metabolism in Abalone Haliotis discus hannai and Its Responses to High Levels of Dietary Lipid. Genes (Basel) 2021; 12:genes12020297. [PMID: 33672704 PMCID: PMC7924355 DOI: 10.3390/genes12020297] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 02/05/2021] [Accepted: 02/17/2021] [Indexed: 12/23/2022] Open
Abstract
The forkhead box O (FoxO) subfamily is a member of the forkhead transcription factor family. It has regulation functions in glucose metabolism in mammals and fish. In the present study, a gene of the foxo homolog in abalone Haliotis discus hannai was cloned. A conservative forkhead (FH) domain and a transactivation (FoxO-TAD) domain were identified. Abalone foxo-specific siRNA (small interfering RNA) was injected to investigate the functions of foxo on glucose metabolism. Knockdown of foxo inhibited expression of phosphoenolpyruvate carboxykinase (pepck) and significantly increased expressions of hexokinase (hk) and pyruvate kinase (pk), but it failed to inhibit the relative mRNA level of glucose-6-phosphatase (g6pase). Then, a 100-day feeding trial was conducted to investigate the response of foxo and glucose metabolism in abalone fed with 1.57% (LFD, low-fat diet), 3.82% (MFD, middle-fat diet) and 6.72% (HFD, high-fat diet) of dietary lipid, respectively. The insulin-signaling pathway (AKT) was depressed and FoxO was activated by the HFD, but it did not inhibit glycolysis (hk) or improved gluconeogenesis significantly (pepck and g6pase). At the same time, impaired hepatopancreas glycogen storage raised hemolymph glucose levels. In conclusion, abalone foxo can be regulated by dietary lipid and can regulate gluconeogenesis or glycolysis in response to changes of dietary lipid levels, in which glycogen metabolism plays an important role.
Collapse
Affiliation(s)
- Liu Wang
- The Key Laboratory of Aquaculture Nutrition and Feeds (Ministry of Agriculture and Rural Affairs), the Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao 266003, China; (L.W.); (Y.G.); (M.P.); (X.L.); (D.H.); (Y.L.); (K.M.)
| | - Yanlin Guo
- The Key Laboratory of Aquaculture Nutrition and Feeds (Ministry of Agriculture and Rural Affairs), the Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao 266003, China; (L.W.); (Y.G.); (M.P.); (X.L.); (D.H.); (Y.L.); (K.M.)
| | - Mingzhu Pan
- The Key Laboratory of Aquaculture Nutrition and Feeds (Ministry of Agriculture and Rural Affairs), the Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao 266003, China; (L.W.); (Y.G.); (M.P.); (X.L.); (D.H.); (Y.L.); (K.M.)
| | - Xinxin Li
- The Key Laboratory of Aquaculture Nutrition and Feeds (Ministry of Agriculture and Rural Affairs), the Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao 266003, China; (L.W.); (Y.G.); (M.P.); (X.L.); (D.H.); (Y.L.); (K.M.)
| | - Dong Huang
- The Key Laboratory of Aquaculture Nutrition and Feeds (Ministry of Agriculture and Rural Affairs), the Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao 266003, China; (L.W.); (Y.G.); (M.P.); (X.L.); (D.H.); (Y.L.); (K.M.)
| | - Yue Liu
- The Key Laboratory of Aquaculture Nutrition and Feeds (Ministry of Agriculture and Rural Affairs), the Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao 266003, China; (L.W.); (Y.G.); (M.P.); (X.L.); (D.H.); (Y.L.); (K.M.)
| | - Chenglong Wu
- School of Life Science, Huzhou University, 759 East 2nd Road, Huzhou 313000, China
- Correspondence: (C.W.); (W.Z.); Tel.: +86-532-8203-2145 (W.Z.)
| | - Wenbing Zhang
- The Key Laboratory of Aquaculture Nutrition and Feeds (Ministry of Agriculture and Rural Affairs), the Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao 266003, China; (L.W.); (Y.G.); (M.P.); (X.L.); (D.H.); (Y.L.); (K.M.)
- Correspondence: (C.W.); (W.Z.); Tel.: +86-532-8203-2145 (W.Z.)
| | - Kangsen Mai
- The Key Laboratory of Aquaculture Nutrition and Feeds (Ministry of Agriculture and Rural Affairs), the Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao 266003, China; (L.W.); (Y.G.); (M.P.); (X.L.); (D.H.); (Y.L.); (K.M.)
| |
Collapse
|
14
|
Szczepanowska K, Trifunovic A. Tune instead of destroy: How proteolysis keeps OXPHOS in shape. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2021; 1862:148365. [PMID: 33417924 DOI: 10.1016/j.bbabio.2020.148365] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 12/11/2020] [Accepted: 12/16/2020] [Indexed: 02/06/2023]
Abstract
Mitochondria are highly dynamic and stress-responsive organelles that are renewed, maintained and removed by a number of different mechanisms. Recent findings bring more evidence for the focused, defined, and regulatory function of the intramitochondrial proteases extending far beyond the traditional concepts of damage control and stress responses. Until recently, the macrodegradation processes, such as mitophagy, were promoted as the major regulator of OXPHOS remodelling and turnover. However, the spatiotemporal dynamics of the OXPHOS system can be greatly modulated by the intrinsic mitochondrial mechanisms acting apart from changes in the global mitochondrial dynamics. This, in turn, may substantially contribute to the shaping of the metabolic status of the cell.
Collapse
Affiliation(s)
- Karolina Szczepanowska
- Cologne Excellence Cluster on Cellular Stress Responses in Ageing-Associated Diseases (CECAD), Center for Molecular Medicine Cologne (CMMC), and Institute for Mitochondrial Diseases and Ageing, Medical Faculty, University of Cologne D-50931 Cologne, Germany; Institute for Mitochondrial Diseases and Ageing, Medical Faculty and Center for Molecular Medicine Cologne (CMMC), D-50931 Cologne, Germany.
| | - Aleksandra Trifunovic
- Cologne Excellence Cluster on Cellular Stress Responses in Ageing-Associated Diseases (CECAD), Center for Molecular Medicine Cologne (CMMC), and Institute for Mitochondrial Diseases and Ageing, Medical Faculty, University of Cologne D-50931 Cologne, Germany; Institute for Mitochondrial Diseases and Ageing, Medical Faculty and Center for Molecular Medicine Cologne (CMMC), D-50931 Cologne, Germany.
| |
Collapse
|
15
|
Abstract
Mounting evidence suggests a role for mitochondrial dysfunction in the pathogenesis of many diseases, including type 2 diabetes, aging, and ovarian failure. Because of the central role of mitochondria in energy production, heme biosynthesis, calcium buffering, steroidogenesis, and apoptosis signaling within cells, understanding the molecular mechanisms behind mitochondrial dysregulation and its potential implications in disease is critical. This review will take a journey through the past and summarize what is known about mitochondrial dysfunction in various disorders, focusing on metabolic alterations and reproductive abnormalities. Evidence is presented from studies in different human populations, and rodents with genetic manipulations of pathways known to affect mitochondrial function.
Collapse
Affiliation(s)
- Manasi Das
- VA San Diego Healthcare System, San Diego, California
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, California
| | - Consuelo Sauceda
- VA San Diego Healthcare System, San Diego, California
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, California
| | - Nicholas J G Webster
- VA San Diego Healthcare System, San Diego, California
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, California
- Moores Cancer Center, University of California, San Diego, La Jolla, California
| |
Collapse
|
16
|
Subramani E, Frigo DE. Mitochondria in Metabolic Syndrome, Reproduction and Transgenerational Inheritance-Ongoing Debates and Emerging Links. Endocrinology 2021; 162:bqaa182. [PMID: 33275660 PMCID: PMC7709213 DOI: 10.1210/endocr/bqaa182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Indexed: 12/27/2022]
Affiliation(s)
- Elavarasan Subramani
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Daniel E Frigo
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, Texas, USA
- Department of Biology and Biochemistry, University of Houston, Houston, Texas, USA
- The Houston Methodist Research Institute, Houston, Texas, USA
| |
Collapse
|
17
|
Genders AJ, Holloway GP, Bishop DJ. Are Alterations in Skeletal Muscle Mitochondria a Cause or Consequence of Insulin Resistance? Int J Mol Sci 2020; 21:ijms21186948. [PMID: 32971810 PMCID: PMC7554894 DOI: 10.3390/ijms21186948] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 09/18/2020] [Accepted: 09/18/2020] [Indexed: 12/14/2022] Open
Abstract
As a major site of glucose uptake following a meal, skeletal muscle has an important role in whole-body glucose metabolism. Evidence in humans and animal models of insulin resistance and type 2 diabetes suggests that alterations in mitochondrial characteristics accompany the development of skeletal muscle insulin resistance. However, it is unclear whether changes in mitochondrial content, respiratory function, or substrate oxidation are central to the development of insulin resistance or occur in response to insulin resistance. Thus, this review will aim to evaluate the apparent conflicting information placing mitochondria as a key organelle in the development of insulin resistance in skeletal muscle.
Collapse
Affiliation(s)
- Amanda J. Genders
- Institute for Health and Sport (iHeS), Victoria University, Melbourne 8001, Australia;
- Correspondence: ; Tel.: +61-3-9919-9556
| | - Graham P. Holloway
- Dept. Human Health and Nutritional Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada;
| | - David J. Bishop
- Institute for Health and Sport (iHeS), Victoria University, Melbourne 8001, Australia;
| |
Collapse
|
18
|
Sangwung P, Petersen KF, Shulman GI, Knowles JW. Mitochondrial Dysfunction, Insulin Resistance, and Potential Genetic Implications. Endocrinology 2020; 161:bqaa017. [PMID: 32060542 PMCID: PMC7341556 DOI: 10.1210/endocr/bqaa017] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Revised: 01/30/2020] [Accepted: 02/13/2020] [Indexed: 02/06/2023]
Abstract
Insulin resistance (IR) is fundamental to the development of type 2 diabetes (T2D) and is present in most prediabetic (preDM) individuals. Insulin resistance has both heritable and environmental determinants centered on energy storage and metabolism. Recent insights from human genetic studies, coupled with comprehensive in vivo and ex vivo metabolic studies in humans and rodents, have highlighted the critical role of reduced mitochondrial function as a predisposing condition for ectopic lipid deposition and IR. These studies support the hypothesis that reduced mitochondrial function, particularly in insulin-responsive tissues such as skeletal muscle, white adipose tissue, and the liver, is inextricably linked to tissue and whole body IR through the effects on cellular energy balance. Here we discuss these findings as well as address potential mechanisms that serve as the nexus between mitochondrial malfunction and IR.
Collapse
Affiliation(s)
- Panjamaporn Sangwung
- Stanford Division of Cardiovascular Medicine and Cardiovascular Institute, Stanford University, Stanford, California
- Stanford Diabetes Research Center, Stanford University, Stanford, California
| | - Kitt Falk Petersen
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, Connecticut
- Yale Diabetes Research Center, Yale School of Medicine, New Haven, Connecticut
| | - Gerald I Shulman
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, Connecticut
- Yale Diabetes Research Center, Yale School of Medicine, New Haven, Connecticut
| | - Joshua W Knowles
- Stanford Division of Cardiovascular Medicine and Cardiovascular Institute, Stanford University, Stanford, California
- Stanford Diabetes Research Center, Stanford University, Stanford, California
| |
Collapse
|
19
|
Ahmed F, Chehadé L, Garneau L, Caron A, Aguer C. The effects of acute BPA exposure on skeletal muscle mitochondrial function and glucose metabolism. Mol Cell Endocrinol 2020; 499:110580. [PMID: 31536778 DOI: 10.1016/j.mce.2019.110580] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 08/27/2019] [Accepted: 09/12/2019] [Indexed: 01/12/2023]
Abstract
Bisphenol A (BPA) is an environmental pollutant that has been associated with adverse health effects including skeletal muscle insulin resistance, a major contributor to the pathogenesis of type 2 diabetes (T2D). Early mitochondrial dysfunction and oxidative stress are linked to impaired glucose metabolism in skeletal muscle. In this study, we investigated the effects of BPA on skeletal muscle mitochondrial function and insulin sensitivity. L6 myotubes were treated with BPA (1 nM-105 nM) during the last 24 h of differentiation. Following exposure to 105 nM of BPA, resting and maximal oxygen consumption rates were decreased, whereas mitochondrial proton leak was increased. Overall metabolic activity, measured by redox ability, was decreased in L6 myotubes exposed to 105 nM of BPA. At this concentration, insulin-stimulated glucose uptake was increased, which corresponded to an increased phosphorylation of the insulin signaling protein Akt, and increased glycolysis measured by extracellular acidification rate (ECAR). Acute BPA exposure did not alter levels of oxidative stress markers in muscle cells, but significantly increased mitochondrial proton leak, which is known to be involved in decreased ROS production. The effects of BPA on glucose uptake, but not mitochondrial function, were reversed by the use of an estrogen receptor antagonist. These results suggest that acute exposure of L6 myotubes at only high concentrations of BPA alters glucose metabolism, which is likely a compensatory response to reduced mitochondrial energy production capacity.
Collapse
Affiliation(s)
- Fozia Ahmed
- Institut du Savoir Montfort, Ottawa, ON, K1K 0T2, Canada; Faculty of Medicine, Biochemistry, Microbiology and Immunology Department, University of Ottawa, Ottawa, ON, K1H 8L1, Canada.
| | - Lucia Chehadé
- Institut du Savoir Montfort, Ottawa, ON, K1K 0T2, Canada; Faculty of Medicine, Biochemistry, Microbiology and Immunology Department, University of Ottawa, Ottawa, ON, K1H 8L1, Canada.
| | - Léa Garneau
- Institut du Savoir Montfort, Ottawa, ON, K1K 0T2, Canada; Faculty of Medicine, Biochemistry, Microbiology and Immunology Department, University of Ottawa, Ottawa, ON, K1H 8L1, Canada.
| | - Audrey Caron
- Institut du Savoir Montfort, Ottawa, ON, K1K 0T2, Canada; Faculty of Medicine, Biochemistry, Microbiology and Immunology Department, University of Ottawa, Ottawa, ON, K1H 8L1, Canada.
| | - Céline Aguer
- Institut du Savoir Montfort, Ottawa, ON, K1K 0T2, Canada; Faculty of Medicine, Biochemistry, Microbiology and Immunology Department, University of Ottawa, Ottawa, ON, K1H 8L1, Canada; Faculty of Health Sciences, School of Human Kinetics, University of Ottawa, Ottawa, ON, K1N 6N5, Canada.
| |
Collapse
|
20
|
Langdahl JH, Frederiksen AL, Vissing J, Frost M, Yderstræde KB, Andersen PH. Mitochondrial mutation m.3243A>G associates with insulin resistance in non-diabetic carriers. Endocr Connect 2019; 8:829-837. [PMID: 31146262 PMCID: PMC6590205 DOI: 10.1530/ec-19-0118] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 05/30/2019] [Indexed: 01/08/2023]
Abstract
AIM This case-control study aimed to examine impairments in glucose metabolism in non-diabetic carriers of the mitochondrial mutation m.3243A>G by evaluating insulin secretion capacity and sensitivity. METHODS Glucose metabolism was investigated in 23 non-diabetic m.3243A>G carriers and age-, sex- and BMI-matched healthy controls with an extended 4-h oral glucose tolerance test (OGTT). Insulin sensitivity index and acute insulin response were estimated on the basis of the OGTT. This was accompanied by examination of body composition by dual-energy X-ray absorptiometry (DXA), maximum aerobic capacity and a Recent Physical Activity Questionnaire (RPAQ). RESULTS Fasting p-glucose, s-insulin and s-c-peptide levels did not differ between m.3243A>G carriers and controls. Insulin sensitivity index (BIGTT-S1) was significantly lower in the m.3243A>G carriers, but there was no difference in the acute insulin response between groups. P-lactate levels were higher in carriers throughout the OGTT. VO2max, but not BMI, waist and hip circumferences, lean and fat body mass%, MET or grip strength, was lower in mutation carriers. BIGTT-S1 remained lower in mutation carriers after adjustment for multiple confounding factors including VO2max in regression analyses. CONCLUSIONS Glucose metabolism in m.3243A>G carriers was characterized by reduced insulin sensitivity, which could represent the earliest phase in the pathogenesis of m.3243A>G-associated diabetes.
Collapse
Affiliation(s)
- Jakob Høgild Langdahl
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
- Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Endocrinology, Hospital of Southwest Jutland, Esbjerg, Denmark
- Correspondence should be addressed to J H Langdahl:
| | - Anja Lisbeth Frederiksen
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
- Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - John Vissing
- Copenhagen Neuromuscular Center, Department of Neurology, Rigshospitalet, Copenhagen, Denmark
| | - Morten Frost
- Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
- Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark
| | - Knud Bonnet Yderstræde
- Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
- Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark
| | - Per Heden Andersen
- Department of Endocrinology, Hospital of Southwest Jutland, Esbjerg, Denmark
| |
Collapse
|
21
|
Gong Y, Liu J, Xue Y, Zhuang Z, Qian S, Zhou W, Li X, Qian J, Ding G, Sun Z. Non-monotonic dose-response effects of arsenic on glucose metabolism. Toxicol Appl Pharmacol 2019; 377:114605. [PMID: 31170414 DOI: 10.1016/j.taap.2019.114605] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 05/29/2019] [Accepted: 05/31/2019] [Indexed: 01/08/2023]
Abstract
BACKGROUND Inorganic arsenic (iAs) is a widespread environmental toxin. In addition to being a human carcinogen, its effect on diabetes has started to gain recognition recently. Insulin is the key hormone regulating systemic glucose metabolism. The in vivo effect of iAs on insulin sensitivity has not been directly addressed. OBJECTIVES Here we use mouse models to dissect the dose-dependent effects of iAs on glucose metabolism in vivo. METHODS We performed hyperinsulinemic-euglycemic clamp, the gold standard analysis of systemic insulin sensitivity. We also performed dynamic metabolic testings and RNA-seq analysis. RESULTS We found that a low-dose exposure (0.25 ppm iAs in drinking water) caused glucose intolerance in adult male C57BL/6 mice, likely by disrupting glucose-induced insulin secretion without affecting peripheral insulin sensitivity. However, a higher-dose exposure (2.5 ppm iAs) had diminished effects on glucose tolerance despite disrupted pancreatic insulin secretion. Insulin Clamp analysis showed that 2.5 ppm iAs actually enhanced systemic insulin sensitivity by simultaneously enhancing insulin-stimulated glucose uptake in skeletal muscles and improved insulin-mediated suppression of endogenous glucose production. RNA-seq analysis of skeletal muscles revealed that 2.5 ppm iAs regulated expression of many genes involved in the metabolism of fatty acids, pyruvate, and amino acids. CONCLUSION These findings suggest that iAs has opposite glycemic effects on distinct metabolic tissues at different dose thresholds. Such non-monotonic dose-response effects of iAs on glucose tolerance shed light on the complex interactions between iAs and the systemic glucose metabolism, which could potentially help reconcile some of the conflicting results in human epidemiological studies.
Collapse
Affiliation(s)
- Yingyun Gong
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China; Department of Medicine-Endocrinology, Baylor College of Medicine, Houston, TX, United States of America
| | - Jidong Liu
- Department of Medicine-Endocrinology, Baylor College of Medicine, Houston, TX, United States of America
| | - Yanfeng Xue
- Department of Medicine-Endocrinology, Baylor College of Medicine, Houston, TX, United States of America
| | - Zhong Zhuang
- Department of Medicine-Endocrinology, Baylor College of Medicine, Houston, TX, United States of America
| | - Sichong Qian
- Department of Medicine-Endocrinology, Baylor College of Medicine, Houston, TX, United States of America
| | - Wenjun Zhou
- Department of Medicine-Endocrinology, Baylor College of Medicine, Houston, TX, United States of America
| | - Xin Li
- Department of Medicine-Endocrinology, Baylor College of Medicine, Houston, TX, United States of America
| | - Justin Qian
- Department of Medicine-Endocrinology, Baylor College of Medicine, Houston, TX, United States of America
| | - Guolian Ding
- Department of Medicine-Endocrinology, Baylor College of Medicine, Houston, TX, United States of America; The International Peace Maternity and Child Health Hospital, Institute of Embryo-Fetal Original Adult Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Zheng Sun
- Department of Medicine-Endocrinology, Baylor College of Medicine, Houston, TX, United States of America; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States of America.
| |
Collapse
|
22
|
Nilsson IAK. The anx/anx Mouse - A Valuable Resource in Anorexia Nervosa Research. Front Neurosci 2019; 13:59. [PMID: 30804742 PMCID: PMC6370726 DOI: 10.3389/fnins.2019.00059] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 01/21/2019] [Indexed: 01/31/2023] Open
Abstract
Animal models are invaluable resources in research concerning the neurobiology of anorexia nervosa (AN), to a large extent since valid clinical samples are rare. None of the existing models can capture all aspects of AN but they are able to mirror the core features of the disorder e.g., elective starvation, emaciation and premature death. The anorectic anx/anx mouse is of particular value for the understanding of the abnormal response to negative energy balance seen in AN. These mice appear normal at birth but gradually develops starvation and emaciation despite full access to food, and die prematurely around three weeks of age. Several changes in hypothalamic neuropeptidergic and -transmitter systems involved in regulating food intake and metabolism have been documented in the anx/anx mouse. These changes are accompanied by signs of inflammation and degeneration in the same hypothalamic regions; including activation of microglia cells and expression of major histocompatibility complex I by microglia and selective neuronal populations. These aberrances are likely related to the dysfunction of complex I (CI) in the oxidative phosphorylation system of the mitochondria, and subsequent increased oxidative stress, which also has been revealed in the hypothalamus of these mice. Interestingly, a similar CI dysfunction has been shown in leukocytes from patients with AN. In addition, a higher expression of the Neurotrophic Receptor Tyrosine Kinase 3 gene has been shown in the anx/anx hypothalamus. This agrees with AN being associated with specific variants of the genes for brain derived neurotrophic factor and Neurotrophic Receptor Tyrosine Kinase 2. The anx/anx mouse is also glucose intolerant and display pancreatic dysfunction related to increased levels of circulating free fatty acids (FFA) and pancreatic inflammation. An increased incidence of eating disorders has been reported for young diabetic women, and as well has increased levels of circulating FFAs in AN. Also similar to individuals with AN, the anx/anx mouse has reduced leptin and increased cholesterol levels in serum. Thus, the anx/anx mouse shares several characteristics with patients with AN, including emaciation, starvation, premature death, diabetic features, increased FFA and low leptin, and is therefore a unique resource in research on the (neuro)biology of AN.
Collapse
Affiliation(s)
- Ida A K Nilsson
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden.,Centre for Eating Disorders Innovation, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
23
|
Xu Z, Fu T, Guo Q, Sun W, Gan Z. Mitochondrial quality orchestrates muscle-adipose dialog to alleviate dietary obesity. Pharmacol Res 2018; 141:176-180. [PMID: 30583080 DOI: 10.1016/j.phrs.2018.12.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 12/12/2018] [Accepted: 12/20/2018] [Indexed: 01/14/2023]
Abstract
Skeletal muscle fitness is vital for human health and disease and is determined by the capacity for burning fuel, mitochondrial ATP production, and contraction. High quality mitochondria in skeletal muscle are essential for maintaining energy homeostasis in response to a myriad of physiologic or pathophysiological stresses. A sophisticated mitochondrial quality control system including mitochondrial autophagy, dynamics, and proteolysis has been identified, which maintains their functional integrity. In this review, we discuss recent studies highlighting mitochondrial quality control mechanisms that govern systemic metabolism by skeletal muscles. Increasing evidence suggests that mitochondria can "communicate" with the nucleus and triggers adaptive genomic re-programming during stress response. We focus on participation of the mitochondrial quality control system in the regulation of mitochondrial communications that drive the muscle to adipose dialog and suggest that muscle-specific regulation of mitochondrial quality impacts systemic homeostasis.
Collapse
Affiliation(s)
- Zhisheng Xu
- The State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Department of Spine Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center of Nanjing University, Nanjing 210061, China
| | - Tingting Fu
- The State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Department of Spine Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center of Nanjing University, Nanjing 210061, China
| | - Qiqi Guo
- The State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Department of Spine Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center of Nanjing University, Nanjing 210061, China
| | - Wanping Sun
- The State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Department of Spine Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center of Nanjing University, Nanjing 210061, China
| | - Zhenji Gan
- The State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Department of Spine Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center of Nanjing University, Nanjing 210061, China.
| |
Collapse
|
24
|
Fealy CE, Mulya A, Axelrod CL, Kirwan JP. Mitochondrial dynamics in skeletal muscle insulin resistance and type 2 diabetes. Transl Res 2018; 202:69-82. [PMID: 30153426 DOI: 10.1016/j.trsl.2018.07.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Revised: 07/08/2018] [Accepted: 07/23/2018] [Indexed: 01/09/2023]
Abstract
The traditional view of mitochondria as isolated, spherical, energy producing organelles, is undergoing a revolutionary change. Emerging data show that mitochondria form a dynamic reticulum that is regulated by cycles of fission and fusion. The discovery of proteins that modulate these activities has led to important advances in understanding human disease. Here, we review the latest evidence that connects the emerging field of mitochondrial dynamics to skeletal muscle insulin resistance and propose some potential mechanisms that may explain the long debated link between mitochondria and the development of type 2 diabetes.
Collapse
Affiliation(s)
- CiarÁn E Fealy
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Anny Mulya
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Christopher L Axelrod
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio; Integrated Physiology and Molecular Metabolism Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana
| | - John P Kirwan
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio; Integrated Physiology and Molecular Metabolism Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana.
| |
Collapse
|
25
|
Blackwood SJ, Hanya E, Katz A. Heating after intense repeated contractions inhibits glycogen accumulation in mouse EDL muscle: role of phosphorylase in postexercise glycogen metabolism. Am J Physiol Cell Physiol 2018; 315:C706-C713. [PMID: 30156860 DOI: 10.1152/ajpcell.00315.2018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The effects of heating on glycogen synthesis (incorporation of [14C]glucose into glycogen) and accumulation after intense repeated contractions were investigated. Isolated mouse extensor digitorum longus muscle (type II) was stimulated electrically to perform intense tetanic contractions at 25°C. After 120 min recovery at 25°C, glycogen accumulated to almost 80% of basal, whereas after recovery at 35°C, glycogen remained low (~25% of basal). Glycogen synthesis averaged 0.97 ± 0.07 µmol·30 min-1·g wet wt-1 during recovery at 25°C and 1.48 ± 0.08 during recovery at 35°C ( P < 0.001). There were no differences in phosphorylase and glycogen synthase total activities nor in phosphorylase fractional activity, whereas glycogen synthase fractional activity was increased by ~50% after recovery at 35°C vs. 25°C. Inorganic phosphate (Pi, substrate for phosphorylase) was markedly increased (~300% of basal) following contraction but returned to control levels after 120 min recovery at 25°C. In contrast, Pi remained elevated after recovery at 35°C (>2-fold higher than recovery at 25°C). Estimates of glycogen breakdown indicated that phosphorylase activity (either via inhibition at 25°C or activation at 35°C) was responsible for ~60% of glycogen accumulation during recovery at 25°C and ~45% during recovery at 35°C. These data demonstrate that despite the enhancing effect of heating on glycogen synthesis during recovery from intense contractions, glycogen accumulation is inhibited owing to Pi-mediated activation of phosphorylase. Thus phosphorylase can play a quantitatively important role in glycogen biogenesis during recovery from repeated contractions in isolated type II muscle.
Collapse
Affiliation(s)
- Sarah J Blackwood
- Department of Physical Therapy, School of Health Sciences, Ariel University, Ariel, Israel
| | - Ester Hanya
- Department of Physical Therapy, School of Health Sciences, Ariel University, Ariel, Israel
| | - Abram Katz
- Department of Physical Therapy, School of Health Sciences, Ariel University, Ariel, Israel
| |
Collapse
|
26
|
Petersen MC, Shulman GI. Mechanisms of Insulin Action and Insulin Resistance. Physiol Rev 2018; 98:2133-2223. [PMID: 30067154 PMCID: PMC6170977 DOI: 10.1152/physrev.00063.2017] [Citation(s) in RCA: 1460] [Impact Index Per Article: 243.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 03/22/2018] [Accepted: 03/24/2018] [Indexed: 12/15/2022] Open
Abstract
The 1921 discovery of insulin was a Big Bang from which a vast and expanding universe of research into insulin action and resistance has issued. In the intervening century, some discoveries have matured, coalescing into solid and fertile ground for clinical application; others remain incompletely investigated and scientifically controversial. Here, we attempt to synthesize this work to guide further mechanistic investigation and to inform the development of novel therapies for type 2 diabetes (T2D). The rational development of such therapies necessitates detailed knowledge of one of the key pathophysiological processes involved in T2D: insulin resistance. Understanding insulin resistance, in turn, requires knowledge of normal insulin action. In this review, both the physiology of insulin action and the pathophysiology of insulin resistance are described, focusing on three key insulin target tissues: skeletal muscle, liver, and white adipose tissue. We aim to develop an integrated physiological perspective, placing the intricate signaling effectors that carry out the cell-autonomous response to insulin in the context of the tissue-specific functions that generate the coordinated organismal response. First, in section II, the effectors and effects of direct, cell-autonomous insulin action in muscle, liver, and white adipose tissue are reviewed, beginning at the insulin receptor and working downstream. Section III considers the critical and underappreciated role of tissue crosstalk in whole body insulin action, especially the essential interaction between adipose lipolysis and hepatic gluconeogenesis. The pathophysiology of insulin resistance is then described in section IV. Special attention is given to which signaling pathways and functions become insulin resistant in the setting of chronic overnutrition, and an alternative explanation for the phenomenon of ‟selective hepatic insulin resistanceˮ is presented. Sections V, VI, and VII critically examine the evidence for and against several putative mediators of insulin resistance. Section V reviews work linking the bioactive lipids diacylglycerol, ceramide, and acylcarnitine to insulin resistance; section VI considers the impact of nutrient stresses in the endoplasmic reticulum and mitochondria on insulin resistance; and section VII discusses non-cell autonomous factors proposed to induce insulin resistance, including inflammatory mediators, branched-chain amino acids, adipokines, and hepatokines. Finally, in section VIII, we propose an integrated model of insulin resistance that links these mediators to final common pathways of metabolite-driven gluconeogenesis and ectopic lipid accumulation.
Collapse
Affiliation(s)
- Max C Petersen
- Departments of Internal Medicine and Cellular & Molecular Physiology, Howard Hughes Medical Institute, Yale University School of Medicine , New Haven, Connecticut
| | - Gerald I Shulman
- Departments of Internal Medicine and Cellular & Molecular Physiology, Howard Hughes Medical Institute, Yale University School of Medicine , New Haven, Connecticut
| |
Collapse
|
27
|
Ruegsegger GN, Creo AL, Cortes TM, Dasari S, Nair KS. Altered mitochondrial function in insulin-deficient and insulin-resistant states. J Clin Invest 2018; 128:3671-3681. [PMID: 30168804 DOI: 10.1172/jci120843] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Diabetes profoundly alters fuel metabolism; both insulin deficiency and insulin resistance are characterized by inefficient mitochondrial coupling and excessive production of reactive oxygen species (ROS) despite their association with normal to high oxygen consumption. Altered mitochondrial function in diabetes can be traced to insulin's pivotal role in maintaining mitochondrial proteome abundance and quality by enhancing mitochondrial biogenesis and preventing proteome damage and degradation, respectively. Although insulin enhances gene transcription, it also induces decreases in amino acids. Thus, if amino acid depletion is not corrected, increased transcription will not result in enhanced translation of transcripts to proteins. Mitochondrial biology varies among tissues, and although most studies in humans are performed in skeletal muscle, abnormalities have been reported in multiple organs in preclinical models of diabetes. Nutrient excess, especially fat excess, alters mitochondrial physiology by driving excess ROS emission that impairs insulin action. Excessive ROS irreversibly damages DNA and proteome with adverse effects on cellular functions. In insulin-resistant people, aerobic exercise stimulates both mitochondrial biogenesis and efficiency concurrent with enhancement of insulin action. This Review discusses the association between both insulin-deficient and insulin-resistant diabetes and alterations in mitochondrial proteome homeostasis and function that adversely affect cellular functions, likely contributing to many diabetic complications.
Collapse
|
28
|
Gan Z, Fu T, Kelly DP, Vega RB. Skeletal muscle mitochondrial remodeling in exercise and diseases. Cell Res 2018; 28:969-980. [PMID: 30108290 DOI: 10.1038/s41422-018-0078-7] [Citation(s) in RCA: 134] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Accepted: 07/27/2018] [Indexed: 12/18/2022] Open
Abstract
Skeletal muscle fitness and plasticity is an important determinant of human health and disease. Mitochondria are essential for maintaining skeletal muscle energy homeostasis by adaptive re-programming to meet the demands imposed by a myriad of physiologic or pathophysiological stresses. Skeletal muscle mitochondrial dysfunction has been implicated in the pathogenesis of many diseases, including muscular dystrophy, atrophy, type 2 diabetes, and aging-related sarcopenia. Notably, exercise counteracts the effects of many chronic diseases on skeletal muscle mitochondrial function. Recent studies have revealed a finely tuned regulatory network that orchestrates skeletal muscle mitochondrial biogenesis and function in response to exercise and in disease states. In addition, increasing evidence suggests that mitochondria also serve to "communicate" with the nucleus and mediate adaptive genomic re-programming. Here we review the current state of knowledge relevant to the dynamic remodeling of skeletal muscle mitochondria in response to exercise and in disease states.
Collapse
Affiliation(s)
- Zhenji Gan
- The State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Department of Spine Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center of Nanjing University, 210061, Nanjing, China.
| | - Tingting Fu
- The State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Department of Spine Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center of Nanjing University, 210061, Nanjing, China
| | - Daniel P Kelly
- Cardiovascular Institute and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | - Rick B Vega
- Translational Research Institute for Metabolism and Diabetes, Florida Hospital, Orlando, FL, 32804, USA.
| |
Collapse
|
29
|
Metcalfe LK, Smith GC, Turner N. Defining lipid mediators of insulin resistance - controversies and challenges. J Mol Endocrinol 2018; 62:JME-18-0023. [PMID: 30068522 DOI: 10.1530/jme-18-0023] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 07/04/2018] [Accepted: 07/31/2018] [Indexed: 12/31/2022]
Abstract
Essential elements of all cells, lipids play important roles in energy production, signalling and as structural components. Despite these critical functions, excessive availability and intracellular accumulation of lipid is now recognised as a major factor contributing to many human diseases, including obesity and diabetes. In the context of these metabolic disorders, ectopic deposition of lipid has been proposed to have deleterious effects of insulin action. While this relationship has been recognised for some time now, there is currently no unifying mechanism to explain how lipids precipitate the development of insulin resistance. This review summarises the evidence linking specific lipid molecules to the induction of insulin resistance, describing some of the current controversies and challenges for future studies in this field.
Collapse
Affiliation(s)
- Louise K Metcalfe
- L Metcalfe, Department of Pharmacology, School of Medical Sciences, UNSW Australia, Kensington, Australia
| | - Greg C Smith
- G Smith, Department of Pharmacology, School of Medical Sciences, UNSW Australia, Kensington, Australia
| | - Nigel Turner
- N Turner, Department of Pharmacology, School of Medical Sciences, University of New South Wales, Sydney, Australia
| |
Collapse
|
30
|
Siasos G, Tsigkou V, Kosmopoulos M, Theodosiadis D, Simantiris S, Tagkou NM, Tsimpiktsioglou A, Stampouloglou PK, Oikonomou E, Mourouzis K, Philippou A, Vavuranakis M, Stefanadis C, Tousoulis D, Papavassiliou AG. Mitochondria and cardiovascular diseases-from pathophysiology to treatment. ANNALS OF TRANSLATIONAL MEDICINE 2018; 6:256. [PMID: 30069458 DOI: 10.21037/atm.2018.06.21] [Citation(s) in RCA: 172] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Mitochondria are the source of cellular energy production and are present in different types of cells. However, their function is especially important for the heart due to the high demands in energy which is achieved through oxidative phosphorylation. Mitochondria form large networks which regulate metabolism and the optimal function is achieved through the balance between mitochondrial fusion and mitochondrial fission. Moreover, mitochondrial function is upon quality control via the process of mitophagy which removes the damaged organelles. Mitochondrial dysfunction is associated with the development of numerous cardiac diseases such as atherosclerosis, ischemia-reperfusion (I/R) injury, hypertension, diabetes, cardiac hypertrophy and heart failure (HF), due to the uncontrolled production of reactive oxygen species (ROS). Therefore, early control of mitochondrial dysfunction is a crucial step in the therapy of cardiac diseases. A number of anti-oxidant molecules and medications have been used but the results are inconsistent among the studies. Eventually, the aim of future research is to design molecules which selectively target mitochondrial dysfunction and restore the capacity of cellular anti-oxidant enzymes.
Collapse
Affiliation(s)
- Gerasimos Siasos
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece.,Division of Cardiovascular, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Vasiliki Tsigkou
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Marinos Kosmopoulos
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Dimosthenis Theodosiadis
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Spyridon Simantiris
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Nikoletta Maria Tagkou
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Athina Tsimpiktsioglou
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Panagiota K Stampouloglou
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Evangelos Oikonomou
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Konstantinos Mourouzis
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Anastasios Philippou
- Department of Experimental Physiology, Medical School, National and Kapodistrian University of Athens, Greece
| | - Manolis Vavuranakis
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | | | - Dimitris Tousoulis
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Athanasios G Papavassiliou
- Department of Biological Chemistry, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| |
Collapse
|
31
|
Hanya E, Katz A. Increased temperature accelerates glycogen synthesis and delays fatigue in isolated mouse muscle during repeated contractions. Acta Physiol (Oxf) 2018; 223:e13027. [PMID: 29297989 DOI: 10.1111/apha.13027] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 12/27/2017] [Accepted: 12/28/2017] [Indexed: 01/05/2023]
Abstract
AIM Elevated glycogen content in muscle delays fatigue during exercise. We examined if increasing muscle temperature during recovery from exercise affects glycogen synthesis and muscle performance during a subsequent bout of exercise. METHODS Isolated mouse extensor digitorum longus muscles were stimulated electrically to perform repeated tetanic contractions until force decreased to 40% of initial at 25°C. Thereafter, muscles recovered for 120 minutes at 25°C (control), 120 minutes at 35°C or 60 minutes at 35°C followed by 60 minutes at 25°C. After recovery, muscles were again stimulated to fatigue at 25°C. RESULTS In the control group, the number of contractions in the second run was slightly less than during the first run (92 ± 5%). Following recovery for 120 minutes at 35°C, the number of contractions was similar to the first run (98 ± 6%). Allowing recovery for 120 minutes at 35°C in the presence of the antioxidant N-acetylcysteine also did not alter the number of contractions in the second run (98 ± 3%). However, recovery for 60 minutes at 35°C followed by 60 minutes at 25°C resulted in an increase in the number of contractions during the second run (110 ± 2%, P < .001). Incorporation of [14 C]glucose into glycogen (glycogen synthesis) during recovery was 1.7-fold higher at 35°C vs 25°C (1.44 ± 0.08 μmol (30 min)-1 (g wet muscle)-1 vs 0.84 ± 0.04; P < .001). CONCLUSION These data demonstrate that, under the conditions studied, elevating muscle temperature for 60 minutes following a bout of repeated contractions delays muscle fatigue during a subsequent bout of repeated contractions and this is associated with enhanced glycogen synthesis in isolated muscle.
Collapse
Affiliation(s)
- E. Hanya
- Department of Physical Therapy; School of Health Sciences; Ariel University; Ariel Israel
| | - A. Katz
- Department of Physical Therapy; School of Health Sciences; Ariel University; Ariel Israel
| |
Collapse
|
32
|
Becker C, Kukat A, Szczepanowska K, Hermans S, Senft K, Brandscheid CP, Maiti P, Trifunovic A. CLPP deficiency protects against metabolic syndrome but hinders adaptive thermogenesis. EMBO Rep 2018; 19:e45126. [PMID: 29588285 PMCID: PMC5934779 DOI: 10.15252/embr.201745126] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 02/26/2018] [Accepted: 02/27/2018] [Indexed: 12/21/2022] Open
Abstract
Mitochondria are fundamental for cellular metabolism as they are both a source and a target of nutrient intermediates originating from converging metabolic pathways, and their role in the regulation of systemic metabolism is increasingly recognized. Thus, maintenance of mitochondrial homeostasis is indispensable for a functional energy metabolism of the whole organism. Here, we report that loss of the mitochondrial matrix protease CLPP results in a lean phenotype with improved glucose homeostasis. Whole-body CLPP-deficient mice are protected from diet-induced obesity and insulin resistance, which was not present in mouse models with either liver- or muscle-specific depletion of CLPP However, CLPP ablation also leads to a decline in brown adipocytes function leaving mice unable to cope with a cold-induced stress due to non-functional adaptive thermogenesis. These results demonstrate a critical role for CLPP in different metabolic stress conditions such as high-fat diet feeding and cold exposure providing tools to understand pathologies with deregulated Clpp expression and novel insights into therapeutic approaches against metabolic dysfunctions linked to mitochondrial diseases.
Collapse
Affiliation(s)
- Christina Becker
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Institute for Mitochondrial Diseases and Aging, Medical Faculty, University of Cologne, Cologne, Germany
| | - Alexandra Kukat
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Institute for Mitochondrial Diseases and Aging, Medical Faculty, University of Cologne, Cologne, Germany
| | - Karolina Szczepanowska
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Institute for Mitochondrial Diseases and Aging, Medical Faculty, University of Cologne, Cologne, Germany
| | - Steffen Hermans
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Institute for Mitochondrial Diseases and Aging, Medical Faculty, University of Cologne, Cologne, Germany
| | - Katharina Senft
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Institute for Mitochondrial Diseases and Aging, Medical Faculty, University of Cologne, Cologne, Germany
| | - Christoph Paul Brandscheid
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Institute for Mitochondrial Diseases and Aging, Medical Faculty, University of Cologne, Cologne, Germany
| | - Priyanka Maiti
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Institute for Mitochondrial Diseases and Aging, Medical Faculty, University of Cologne, Cologne, Germany
| | - Aleksandra Trifunovic
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Institute for Mitochondrial Diseases and Aging, Medical Faculty, University of Cologne, Cologne, Germany
- Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| |
Collapse
|
33
|
Agerholm M, Dall M, Jensen BAH, Prats C, Madsen S, Basse AL, Graae AS, Risis S, Goldenbaum J, Quistorff B, Larsen S, Vienberg SG, Treebak JT. Perturbations of NAD + salvage systems impact mitochondrial function and energy homeostasis in mouse myoblasts and intact skeletal muscle. Am J Physiol Endocrinol Metab 2018; 314:E377-E395. [PMID: 29208611 DOI: 10.1152/ajpendo.00213.2017] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Nicotinamide adenine dinucleotide (NAD+) can be synthesized by nicotinamide phosphoribosyltransferase (NAMPT). We aimed to determine the role of NAMPT in maintaining NAD+ levels, mitochondrial function, and metabolic homeostasis in skeletal muscle cells. We generated stable Nampt knockdown (sh Nampt KD) C2C12 cells using a shRNA lentiviral approach. Moreover, we applied gene electrotransfer to express Cre recombinase in tibialis anterior muscle of floxed Nampt mice. In sh Nampt KD C2C12 myoblasts, Nampt and NAD+ levels were reduced by 70% and 50%, respectively, and maximal respiratory capacity was reduced by 25%. Moreover, anaerobic glycolytic flux increased by 55%, and 2-deoxyglucose uptake increased by 25% in sh Nampt KD cells. Treatment with the NAD+ precursor nicotinamide riboside restored NAD+ levels in sh Nampt cells and increased maximal respiratory capacity by 18% and 32% in control and sh Nampt KD cells, respectively. Expression of Cre recombinase in muscle of floxed Nampt mice reduced NAMPT and NAD+ levels by 38% and 43%, respectively. Glucose uptake increased by 40%, and mitochondrial complex IV respiration was compromised by 20%. Hypoxia-inducible factor (HIF)-1α-regulated genes and histone H3 lysine 9 (H3K9) acetylation, a known sirtuin 6 (SIRT6) target, were increased in shNampt KD cells. Thus, we propose that the shift toward glycolytic metabolism observed, at least in part, is mediated by the SIRT6/HIF1α axis. Our findings suggest that NAMPT plays a key role for maintaining NAD+ levels in skeletal muscle and that NAMPT deficiency compromises oxidative phosphorylation capacity and alters energy homeostasis in this tissue.
Collapse
Affiliation(s)
- Marianne Agerholm
- Section of Integrative Physiology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen , Copenhagen , Denmark
| | - Morten Dall
- Section of Integrative Physiology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen , Copenhagen , Denmark
| | - Benjamin A H Jensen
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen , Copenhagen , Denmark
| | - Clara Prats
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Søren Madsen
- Section of Integrative Physiology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen , Copenhagen , Denmark
| | - Astrid L Basse
- Section of Integrative Physiology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen , Copenhagen , Denmark
| | - Anne-Sofie Graae
- Section of Integrative Physiology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen , Copenhagen , Denmark
| | - Steve Risis
- Section of Integrative Physiology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen , Copenhagen , Denmark
| | - Julie Goldenbaum
- Section of Integrative Physiology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen , Copenhagen , Denmark
| | - Bjørn Quistorff
- Section for Translational Metabolic Physiology, Novo Nordisk Foundation Center for Basic Metabolic Research, and Department of Biomedical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Steen Larsen
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Sara G Vienberg
- Section of Integrative Physiology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen , Copenhagen , Denmark
| | - Jonas T Treebak
- Section of Integrative Physiology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen , Copenhagen , Denmark
| |
Collapse
|
34
|
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disorder without a cure. Most AD cases are sporadic where age represents the greatest risk factor. Lack of understanding of the disease mechanism hinders the development of efficacious therapeutic approaches. The loss of synapses in the affected brain regions correlates best with cognitive impairment in AD patients and has been considered as the early mechanism that precedes neuronal loss. Oxidative stress has been recognized as a contributing factor in aging and in the progression of multiple neurodegenerative diseases including AD. Increased production of reactive oxygen species (ROS) associated with age- and disease-dependent loss of mitochondrial function, altered metal homeostasis, and reduced antioxidant defense directly affect synaptic activity and neurotransmission in neurons leading to cognitive dysfunction. In addition, molecular targets affected by ROS include nuclear and mitochondrial DNA, lipids, proteins, calcium homeostasis, mitochondrial dynamics and function, cellular architecture, receptor trafficking and endocytosis, and energy homeostasis. Abnormal cellular metabolism in turn could affect the production and accumulation of amyloid-β (Aβ) and hyperphosphorylated Tau protein, which independently could exacerbate mitochondrial dysfunction and ROS production, thereby contributing to a vicious cycle. While mounting evidence implicates ROS in the AD etiology, clinical trials with antioxidant therapies have not produced consistent results. In this review, we will discuss the role of oxidative stress in synaptic dysfunction in AD, innovative therapeutic strategies evolved based on a better understanding of the complexity of molecular mechanisms of AD, and the dual role ROS play in health and disease.
Collapse
Affiliation(s)
- Eric Tönnies
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Eugenia Trushina
- Department of Neurology, Mayo Clinic, Rochester, MN, USA.,Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
35
|
Saitoh S, Okano S, Nohara H, Nakano H, Shirasawa N, Naito A, Yamamoto M, Kelly VP, Takahashi K, Tanaka T, Nakajima M, Nakajima O. 5-aminolevulinic acid (ALA) deficiency causes impaired glucose tolerance and insulin resistance coincident with an attenuation of mitochondrial function in aged mice. PLoS One 2018; 13:e0189593. [PMID: 29364890 PMCID: PMC5783358 DOI: 10.1371/journal.pone.0189593] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 11/28/2017] [Indexed: 12/25/2022] Open
Abstract
In vertebrates, the initial step in heme biosynthesis is the production of 5-aminolevulinic acid (ALA) by ALA synthase (ALAS). ALA formation is believed to be the rate-limiting step for cellular heme production. Recently, several cohort studies have demonstrated the potential of ALA as a treatment for individuals with prediabetes and type-2 diabetes mellitus. These studies imply that a mechanism exists by which ALA or heme can control glucose metabolism. The ALAS1 gene encodes a ubiquitously expressed isozyme. Mice heterozygous null for ALAS1 (A1+/-s) experience impaired glucose tolerance (IGT) and insulin resistance (IR) beyond 20-weeks of age (aged A1+/-s). IGT and IR were remedied in aged A1+/-s by the oral administration of ALA for 1 week. However, the positive effect of ALA proved to be reversible and was lost upon termination of ALA administration. In the skeletal muscle of aged A1+/-s an attenuation of mitochondrial function is observed, coinciding with IGT and IR. Oral administration of ALA for 1-week brought about only a partial improvement in mitochondrial activity however, a 6-week period of ALA treatment was sufficient to remedy mitochondrial function. Studies on differentiated C2C12 myocytes indicate that the impairment of glucose metabolism is a cell autonomous effect and that ALA deficiency ultimately leads to heme depletion. This sequela is evidenced by a reduction of glucose uptake in C2C12 cells following the knockdown of ALAS1 or the inhibition of heme biosynthesis by succinylacetone. Our data provide in vivo proof that ALA deficiency attenuates mitochondrial function, and causes IGT and IR in an age-dependent manner. The data reveals an unexpected metabolic link between heme and glucose that is relevant to the pathogenesis of IGT/IR.
Collapse
Affiliation(s)
- Shinichi Saitoh
- Research Center for Molecular Genetics, Institute for Promotion of Medical Science Research, Yamagata University Faculty of Medicine, Yamagata, Yamagata, Japan
| | - Satoshi Okano
- Research Center for Molecular Genetics, Institute for Promotion of Medical Science Research, Yamagata University Faculty of Medicine, Yamagata, Yamagata, Japan
| | - Hidekazu Nohara
- Research Center for Molecular Genetics, Institute for Promotion of Medical Science Research, Yamagata University Faculty of Medicine, Yamagata, Yamagata, Japan
| | - Hiroshi Nakano
- Research Center for Molecular Genetics, Institute for Promotion of Medical Science Research, Yamagata University Faculty of Medicine, Yamagata, Yamagata, Japan
| | - Nobuyuki Shirasawa
- Department of Anatomy and Structural Science, Yamagata University Faculty of Medicine, Yamagata, Yamagata, Japan
| | - Akira Naito
- Department of Anatomy and Structural Science, Yamagata University Faculty of Medicine, Yamagata, Yamagata, Japan
| | - Masayuki Yamamoto
- Department of Medical Biochemistry, Tohoku University School of Medicine, Sendai, Japan
| | - Vincent P. Kelly
- School of Biochemistry & Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | | | | | | | - Osamu Nakajima
- Research Center for Molecular Genetics, Institute for Promotion of Medical Science Research, Yamagata University Faculty of Medicine, Yamagata, Yamagata, Japan
| |
Collapse
|
36
|
Mitohormesis, an Antiaging Paradigm. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 340:35-77. [DOI: 10.1016/bs.ircmb.2018.05.002] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
37
|
Mitochondrial (Dys) Function in Inflammaging: Do MitomiRs Influence the Energetic, Oxidative, and Inflammatory Status of Senescent Cells? Mediators Inflamm 2017; 2017:2309034. [PMID: 29445253 PMCID: PMC5763118 DOI: 10.1155/2017/2309034] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 11/20/2017] [Indexed: 12/19/2022] Open
Abstract
A relevant feature of aging is chronic low-grade inflammation, termed inflammaging, a key process promoting the development of all major age-related diseases. Senescent cells can acquire the senescence-associated (SA) secretory phenotype (SASP), characterized by the secretion of proinflammatory factors fuelling inflammaging. Cellular senescence is also accompanied by a deep reshaping of microRNA expression and by the modulation of mitochondria activity, both master regulators of the SASP. Here, we synthesize novel findings regarding the role of mitochondria in the SASP and in the inflammaging process and propose a network linking nuclear-encoded SA-miRNAs to mitochondrial gene regulation and function in aging cells. In this conceptual structure, SA-miRNAs can translocate to mitochondria (SA-mitomiRs) and may affect the energetic, oxidative, and inflammatory status of senescent cells. We discuss the potential role of several of SA-mitomiRs (i.e., let-7b, miR-1, miR-130a-3p, miR-133a, miR-146a-5p, miR-181c-5p, and miR-378-5p), using miR-146a as a proof-of-principle model. Finally, we propose a comprehensive, metabolic, and epigenetic view of the senescence process, in order to amplify the range of possible approaches to target inflammaging, with the ultimate goal of decelerating the aging rate, postponing or blunting the development of age-related diseases.
Collapse
|
38
|
Hill S, Deepa SS, Sataranatarajan K, Premkumar P, Pulliam D, Liu Y, Soto VY, Fischer KE, Van Remmen H. Sco2 deficient mice develop increased adiposity and insulin resistance. Mol Cell Endocrinol 2017; 455:103-114. [PMID: 28428045 PMCID: PMC5592144 DOI: 10.1016/j.mce.2017.03.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 03/18/2017] [Accepted: 03/20/2017] [Indexed: 10/19/2022]
Abstract
Cytochrome c oxidase (COX) is an essential transmembrane protein complex (Complex IV) in the mitochondrial respiratory electron chain. Mutations in genes responsible for the assembly of COX are associated with Leigh syndrome, cardiomyopathy, spinal muscular atrophy and other fatal metabolic disorders in humans. Previous studies have shown that mice lacking the COX assembly protein Surf1 (Surf1-/- mice) paradoxically show a number of beneficial metabolic phenotypes including increased insulin sensitivity, upregulation of mitochondrial biogenesis, induction of stress response pathways and increased lifespan. To determine whether these effects are specific to the Surf1 mutation or a more general effect of reduced COX activity, we asked whether a different mutation causing reduced COX activity would have similar molecular and physiologic changes. Sco2 knock-in/knock-out (KI/KO) mice in which one allele of the Sco2 gene that encodes a copper chaperone required for COX activity is deleted and the second allele is mutated, have previously been shown to be viable despite a 30-60% reduction in COX activity. In contrast to the Surf1-/- mice, we show that Sco2 KI/KO mice have increased fat mass, associated with reduced β-oxidation and increased adipogenesis markers, reduced insulin receptor beta (IR-β levels in adipose tissue, reduced muscle glucose transporter 4 (Glut4) levels and a impaired response to the insulin tolerance test consistent with insulin resistance. COX activity and protein are reduced approximately 50% in adipose tissue from the Sco2 KI/KO mice. Consistent with the increase in adipose tissue mass, the Sco2 KI/KO mice also show increased hepatosteatosis, elevated serum and liver triglyceride and increased serum cholesterol levels compared to wild-type controls. In contrast to the Surf1-/- mice, which show increased mitochondrial number, upregulation of the mitochondrial unfolded protein response (UPRMT) pathway and no significant change in mitochondrial respiration in several tissues, Sco2 KI/KO mice do not upregulate the UPRMT, and tissue oxygen consumption and levels of several proteins involved in mitochondrial function are reduced in adipose tissue compared to wild type mice. Thus, the metabolic effects of the Sco2 and Surf1-/- mutations are opposite, despite comparable changes in COX activity, illuminating the complex impact of mitochondrial dysfunction on physiology and pointing to an important role for complex IV in regulating metabolism.
Collapse
Affiliation(s)
- Shauna Hill
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 N.E. 13th Street, Oklahoma City, OK 73104, United States; Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, United States
| | - Sathyaseelan S Deepa
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 N.E. 13th Street, Oklahoma City, OK 73104, United States
| | - Kavithalakshmi Sataranatarajan
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 N.E. 13th Street, Oklahoma City, OK 73104, United States
| | - Pavithra Premkumar
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 N.E. 13th Street, Oklahoma City, OK 73104, United States
| | - Daniel Pulliam
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 N.E. 13th Street, Oklahoma City, OK 73104, United States; Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, United States
| | - Yuhong Liu
- Department of Biology, University of Alabama at Birmingham, 1720 2nd Avenue South, CH 464, Birmingham, AL 35294, United States
| | - Vanessa Y Soto
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, United States
| | - Kathleen E Fischer
- Department of Biology, University of Alabama at Birmingham, 1720 2nd Avenue South, CH 464, Birmingham, AL 35294, United States
| | - Holly Van Remmen
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 N.E. 13th Street, Oklahoma City, OK 73104, United States; Oklahoma City VA Medical Center, Oklahoma City, OK, United States.
| |
Collapse
|
39
|
Bergström U, Lindfors C, Svedberg M, Johansen JE, Häggkvist J, Schalling M, Wibom R, Katz A, Nilsson IAK. Reduced metabolism in the hypothalamus of the anorectic anx/anx mouse. J Endocrinol 2017; 233:15-24. [PMID: 28130409 DOI: 10.1530/joe-16-0383] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 01/27/2017] [Indexed: 12/13/2022]
Abstract
The anorectic anx/anx mouse exhibits a mitochondrial complex I dysfunction that is related to aberrant expression of hypothalamic neuropeptides and transmitters regulating food intake. Hypothalamic activity, i.e. neuronal firing and transmitter release, is dependent on glucose utilization and energy metabolism. To better understand the role of hypothalamic activity in anorexia, we assessed carbohydrate and high-energy phosphate metabolism, in vivo and in vitro, in the anx/anx hypothalamus. In the fasted state, hypothalamic glucose uptake in the anx/anx mouse was reduced by ~50% of that seen in wild-type (wt) mice (P < 0.05). Under basal conditions, anx/anx hypothalamus ATP and glucose 6-P contents were similar to those in wt hypothalamus, whereas phosphocreatine was elevated (~2-fold; P < 0.001) and lactate was reduced (~35%; P < 0.001). The anx/anx hypothalamus had elevated total AMPK (~25%; P < 0.05) and GLUT4 (~60%; P < 0.01) protein contents, whereas GLUT1 and GLUT3 were similar to that of wt hypothalamus. Interestingly, the activation state of AMPK (ratio of phosphorylated AMPK/total AMPK) was significantly decreased in hypothalamus of the anx/anx mouse (~60% of that in wt; P < 0.05). Finally, during metabolic stress (ischemia), accumulation of lactate (measure of glycolysis) and IMP and AMP (breakdown products of ATP) were ~50% lower in anx/anx vs wt hypothalamus. These data demonstrate that carbohydrate and high-energy phosphate utilization in the anx/anx hypothalamus are diminished under basal and stress conditions. The decrease in hypothalamic metabolism may contribute to the anorectic behavior of the anx/anx mouse, i.e. its inability to regulate food intake in accordance with energy status.
Collapse
Affiliation(s)
- Ulrika Bergström
- Department of Molecular Medicine and SurgeryKarolinska Institutet, Stockholm, Sweden
- Center for Molecular MedicineKarolinska University Hospital, Stockholm, Sweden
| | - Charlotte Lindfors
- Department of Molecular Medicine and SurgeryKarolinska Institutet, Stockholm, Sweden
- Center for Molecular MedicineKarolinska University Hospital, Stockholm, Sweden
| | - Marie Svedberg
- Department of Clinical NeuroscienceCenter for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden
| | - Jeanette E Johansen
- Department of Molecular Medicine and SurgeryKarolinska Institutet, Stockholm, Sweden
- Center for Molecular MedicineKarolinska University Hospital, Stockholm, Sweden
| | - Jenny Häggkvist
- Department of Clinical NeuroscienceCenter for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden
| | - Martin Schalling
- Department of Molecular Medicine and SurgeryKarolinska Institutet, Stockholm, Sweden
- Center for Molecular MedicineKarolinska University Hospital, Stockholm, Sweden
| | - Rolf Wibom
- Department of Medical Biochemistry and BiophysicsKarolinska Institutet, Stockholm, Sweden
| | - Abram Katz
- Department of Physical TherapyAriel University, Ariel, Israel
| | - Ida A K Nilsson
- Department of Molecular Medicine and SurgeryKarolinska Institutet, Stockholm, Sweden
- Center for Molecular MedicineKarolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
40
|
Di Meo S, Iossa S, Venditti P. Skeletal muscle insulin resistance: role of mitochondria and other ROS sources. J Endocrinol 2017; 233:R15-R42. [PMID: 28232636 DOI: 10.1530/joe-16-0598] [Citation(s) in RCA: 193] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 01/31/2017] [Indexed: 12/12/2022]
Abstract
At present, obesity is one of the most important public health problems in the world because it causes several diseases and reduces life expectancy. Although it is well known that insulin resistance plays a pivotal role in the development of type 2 diabetes mellitus (the more frequent disease in obese people) the link between obesity and insulin resistance is yet a matter of debate. One of the most deleterious effects of obesity is the deposition of lipids in non-adipose tissues when the capacity of adipose tissue is overwhelmed. During the last decade, reduced mitochondrial function has been considered as an important contributor to 'toxic' lipid metabolite accumulation and consequent insulin resistance. More recent reports suggest that mitochondrial dysfunction is not an early event in the development of insulin resistance, but rather a complication of the hyperlipidemia-induced reactive oxygen species (ROS) production in skeletal muscle, which might promote mitochondrial alterations, lipid accumulation and inhibition of insulin action. Here, we review the literature dealing with the mitochondria-centered mechanisms proposed to explain the onset of obesity-linked IR in skeletal muscle. We conclude that the different pathways leading to insulin resistance may act synergistically because ROS production by mitochondria and other sources can result in mitochondrial dysfunction, which in turn can further increase ROS production leading to the establishment of a harmful positive feedback loop.
Collapse
Affiliation(s)
- Sergio Di Meo
- Department of BiologyUniversity of Naples 'Federico II', Naples, Italy
| | - Susanna Iossa
- Department of BiologyUniversity of Naples 'Federico II', Naples, Italy
| | - Paola Venditti
- Department of BiologyUniversity of Naples 'Federico II', Naples, Italy
| |
Collapse
|
41
|
Spahis S, Borys JM, Levy E. Metabolic Syndrome as a Multifaceted Risk Factor for Oxidative Stress. Antioxid Redox Signal 2017; 26:445-461. [PMID: 27302002 DOI: 10.1089/ars.2016.6756] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
SIGNIFICANCE Metabolic syndrome (MetS) is associated with a greater risk of diabetes and cardiovascular diseases. It is estimated that this multifactorial condition affects 20%-30% of the world's population. A detailed understanding of MetS mechanisms is crucial for the development of effective prevention strategies and adequate intervention tools that could curb its increasing prevalence and limit its comorbidities, particularly in younger age groups. With advances in basic redox biology, oxidative stress (OxS) involvement in the complex pathophysiology of MetS has become widely accepted. Nevertheless, its clear association with and causative effects on MetS require further elucidation. Recent Advances: Although a better understanding of the causes, risks, and effects of MetS is essential, studies suggest that oxidant/antioxidant imbalance is a key contributor to this condition. OxS is now understood to be a major underlying mechanism for mitochondrial dysfunction, ectopic lipid accumulation, and gut microbiota impairment. CRITICAL ISSUES Further studies, particularly in the field of translational research, are clearly required to understand and control the production of reactive oxygen species (ROS) levels, especially in the mitochondria, since the various therapeutic trials conducted to date have not targeted this major ROS-generating system, aimed to delay MetS onset, or prevent its progression. FUTURE DIRECTIONS Multiple relevant markers need to be identified to clarify the role of ROS in the etiology of MetS. Future clinical trials should provide important proof of concept for the effectiveness of antioxidants as useful therapeutic approaches to simultaneously counteract mitochondrial OxS, alleviate MetS symptoms, and prevent complications. Antioxid. Redox Signal. 26, 445-461.
Collapse
Affiliation(s)
- Schohraya Spahis
- 1 Research Center , Ste-Justine MUHC, Montreal, Canada .,2 Department of Nutrition, Université de Montréal , Montreal, Canada
| | | | - Emile Levy
- 1 Research Center , Ste-Justine MUHC, Montreal, Canada .,2 Department of Nutrition, Université de Montréal , Montreal, Canada .,3 EPODE International Network , Paris, France
| |
Collapse
|
42
|
Bénit P, Pelhaître A, Saunier E, Bortoli S, Coulibaly A, Rak M, Schiff M, Kroemer G, Zeviani M, Rustin P. Paradoxical Inhibition of Glycolysis by Pioglitazone Opposes the Mitochondriopathy Caused by AIF Deficiency. EBioMedicine 2017; 17:75-87. [PMID: 28229909 PMCID: PMC5360583 DOI: 10.1016/j.ebiom.2017.02.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 02/14/2017] [Accepted: 02/14/2017] [Indexed: 12/13/2022] Open
Abstract
Mice with the hypomorphic AIF-Harlequin mutation exhibit a highly heterogeneous mitochondriopathy that mostly affects respiratory chain complex I, causing a cerebral pathology that resembles that found in patients with AIF loss-of-function mutations. Here we describe that the antidiabetic drug pioglitazone (PIO) can improve the phenotype of a mouse Harlequin (Hq) subgroup, presumably due to an inhibition of glycolysis that causes an increase in blood glucose levels. This glycolysis-inhibitory PIO effect was observed in cultured astrocytes from Hq mice, as well as in human skin fibroblasts from patients with AIF mutation. Glycolysis inhibition by PIO resulted from direct competitive inhibition of glyceraldehyde-3-phosphate dehydrogenase (GAPDH). Moreover, GAPDH protein levels were reduced in the cerebellum and in the muscle from Hq mice that exhibited an improved phenotype upon PIO treatment. Altogether, our results suggest that excessive glycolysis participates to the pathogenesis of mitochondriopathies and that pharmacological inhibition of glycolysis may have beneficial effects in this condition.
Collapse
Affiliation(s)
- Paule Bénit
- INSERM UMR 1141, PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Alice Pelhaître
- INSERM UMR 1141, PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Elise Saunier
- INSERM UMR 1124, Centre Universitaire des Saints-Pères, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Sylvie Bortoli
- INSERM UMR 1124, Centre Universitaire des Saints-Pères, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Assetou Coulibaly
- INSERM UMR 1141, PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Malgorzata Rak
- INSERM UMR 1141, PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Manuel Schiff
- INSERM UMR 1141, PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France; Reference Center for Inherited Metabolic Diseases, Hôpital Robert Debré, Assistance Publique - Hôpitaux de Paris, 48 Boulevard Sérurier, 75019 Paris, France
| | - Guido Kroemer
- Equipe11 labellisée Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France; INSERM U1138, Centre de Recherche des Cordeliers, Paris, France; Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France; Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France; Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm 17176, Sweden
| | - Massimo Zeviani
- MRC-Mitochondrial Biology Unit, Cambridge, Cambridgeshire, United Kingdom
| | - Pierre Rustin
- INSERM UMR 1141, PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France.
| |
Collapse
|
43
|
Chung HK, Ryu D, Kim KS, Chang JY, Kim YK, Yi HS, Kang SG, Choi MJ, Lee SE, Jung SB, Ryu MJ, Kim SJ, Kweon GR, Kim H, Hwang JH, Lee CH, Lee SJ, Wall CE, Downes M, Evans RM, Auwerx J, Shong M. Growth differentiation factor 15 is a myomitokine governing systemic energy homeostasis. J Cell Biol 2017; 216:149-165. [PMID: 27986797 PMCID: PMC5223607 DOI: 10.1083/jcb.201607110] [Citation(s) in RCA: 245] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 10/09/2016] [Accepted: 11/30/2016] [Indexed: 01/06/2023] Open
Abstract
Reduced mitochondrial electron transport chain activity promotes longevity and improves energy homeostasis via cell-autonomous and -non-autonomous factors in multiple model systems. This mitohormetic effect is thought to involve the mitochondrial unfolded protein response (UPRmt), an adaptive stress-response pathway activated by mitochondrial proteotoxic stress. Using mice with skeletal muscle-specific deficiency of Crif1 (muscle-specific knockout [MKO]), an integral protein of the large mitoribosomal subunit (39S), we identified growth differentiation factor 15 (GDF15) as a UPRmt-associated cell-non-autonomous myomitokine that regulates systemic energy homeostasis. MKO mice were protected against obesity and sensitized to insulin, an effect associated with elevated GDF15 secretion after UPRmt activation. In ob/ob mice, administration of recombinant GDF15 decreased body weight and improved insulin sensitivity, which was attributed to elevated oxidative metabolism and lipid mobilization in the liver, muscle, and adipose tissue. Thus, GDF15 is a potent mitohormetic signal that safeguards against the onset of obesity and insulin resistance.
Collapse
Affiliation(s)
- Hyo Kyun Chung
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon 301-721, South Korea
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon 34134, South Korea
| | - Dongryeol Ryu
- Laboratory for Integrative and Systems Physiology, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Koon Soon Kim
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon 301-721, South Korea
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon 34134, South Korea
| | - Joon Young Chang
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon 301-721, South Korea
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon 34134, South Korea
| | - Yong Kyung Kim
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon 301-721, South Korea
| | - Hyon-Seung Yi
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon 301-721, South Korea
| | - Seul Gi Kang
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon 301-721, South Korea
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon 34134, South Korea
| | - Min Jeong Choi
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon 301-721, South Korea
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon 34134, South Korea
| | - Seong Eun Lee
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon 301-721, South Korea
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon 34134, South Korea
| | - Saet-Byel Jung
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon 301-721, South Korea
| | - Min Jeong Ryu
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon 301-721, South Korea
| | - Soung Jung Kim
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon 301-721, South Korea
| | - Gi Ryang Kweon
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon 34134, South Korea
| | - Hail Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 305-338, South Korea
| | - Jung Hwan Hwang
- Animal Model Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-764, South Korea
| | - Chul-Ho Lee
- Animal Model Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-764, South Korea
| | - Se-Jin Lee
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | | | - Michael Downes
- Gene Expression Laboratory, Salk Institute, La Jolla, CA 92037
| | - Ronald M Evans
- Gene Expression Laboratory, Salk Institute, La Jolla, CA 92037
| | - Johan Auwerx
- Laboratory for Integrative and Systems Physiology, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Minho Shong
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon 301-721, South Korea
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon 34134, South Korea
| |
Collapse
|
44
|
Gonzalez-Franquesa A, Patti ME. Insulin Resistance and Mitochondrial Dysfunction. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 982:465-520. [DOI: 10.1007/978-3-319-55330-6_25] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
45
|
Swerdlow RH. Bioenergetics and metabolism: a bench to bedside perspective. J Neurochem 2016; 139 Suppl 2:126-135. [PMID: 26968700 PMCID: PMC5851778 DOI: 10.1111/jnc.13509] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Revised: 12/02/2015] [Accepted: 12/11/2015] [Indexed: 12/13/2022]
Abstract
'Metabolism' refers to the vast collection of chemical processes that occur within a living organism. Within this broad designation, one can identify metabolism events that relate specifically to energy homeostasis, whether they occur at the subcellular, cellular, organ, or whole organism level. This review operationally refers to this type of metabolism as 'energy metabolism' or 'bioenergetics.' Changes in energy metabolism/bioenergetics have been linked to brain aging and a number of neurodegenerative diseases, and research suggests mitochondria may uniquely contribute to this. Interventions that manipulate energy metabolism/bioenergetic function and mitochondria may have therapeutic potential and efforts intended to accomplish this are playing out at basic, translational, and clinical levels. This review follows evolving views of energy metabolism's role in neurodegenerative diseases but especially Alzheimer's disease, with an emphasis on the bench-to-bedside process whose ultimate goal is to develop therapeutic interventions. It further considers challenges encountered during this process, which include linking basic concepts to a medical question at the initial research stage, adapting conceptual knowledge gained to a disease-associated application in the translational stage, extending what has been learned to the clinical arena, and maintaining support for the research at each of these fundamentally linked but functionally distinct stages. A bench-to-bedside biomedical research process is discussed that moves through conceptual, basic, translational, and clinical levels. For example, herein a case was made that bioenergetics is a valid Alzheimer's disease therapeutic target. Following this, a fundamental strategy for manipulating bioenergetics was defined, potential implications studied, and the approach extended to the clinical arena. This article is part of the 60th Anniversary special issue.
Collapse
Affiliation(s)
- Russell H Swerdlow
- University of Kansas Alzheimer's Disease Center and the departments of Neurology, Molecular and Integrative Physiology, and Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, USA.
| |
Collapse
|
46
|
Affourtit C. Mitochondrial involvement in skeletal muscle insulin resistance: A case of imbalanced bioenergetics. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2016; 1857:1678-93. [PMID: 27473535 DOI: 10.1016/j.bbabio.2016.07.008] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 06/19/2016] [Accepted: 07/23/2016] [Indexed: 12/16/2022]
Abstract
Skeletal muscle insulin resistance in obesity associates with mitochondrial dysfunction, but the causality of this association is controversial. This review evaluates mitochondrial models of nutrient-induced muscle insulin resistance. It transpires that all models predict that insulin resistance arises as a result of imbalanced cellular bioenergetics. The nature and precise origin of the proposed insulin-numbing molecules differ between models but all species only accumulate when metabolic fuel supply outweighs energy demand. This observation suggests that mitochondrial deficiency in muscle insulin resistance is not merely owing to intrinsic functional defects, but could instead be an adaptation to nutrient-induced changes in energy expenditure. Such adaptive effects are likely because muscle ATP supply is fully driven by energy demand. This market-economic control of myocellular bioenergetics offers a mechanism by which insulin-signalling deficiency can cause apparent mitochondrial dysfunction, as insulin resistance lowers skeletal muscle anabolism and thus dampens ATP demand and, consequently, oxidative ATP synthesis.
Collapse
Affiliation(s)
- Charles Affourtit
- School of Biomedical and Healthcare Sciences, Plymouth University Peninsula Schools of Medicine and Dentistry, Plymouth University, Drake Circus, PL4 8AA Plymouth, UK.
| |
Collapse
|
47
|
Nisr RB, Affourtit C. Palmitate-induced changes in energy demand cause reallocation of ATP supply in rat and human skeletal muscle cells. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2016; 1857:1403-1411. [PMID: 27154056 DOI: 10.1016/j.bbabio.2016.04.286] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 04/16/2016] [Accepted: 04/28/2016] [Indexed: 01/22/2023]
Abstract
Mitochondrial dysfunction has been associated with obesity-related muscle insulin resistance, but the causality of this association is controversial. The notion that mitochondrial oxidative capacity may be insufficient to deal appropriately with excessive nutrient loads is for example disputed. Effective mitochondrial capacity is indirectly, but largely determined by ATP-consuming processes because skeletal muscle energy metabolism is mostly controlled by ATP demand. Probing the bioenergetics of rat and human myoblasts in real time we show here that the saturated fatty acid palmitate lowers the rate and coupling efficiency of oxidative phosphorylation under conditions it causes insulin resistance. Stearate affects the bioenergetic parameters similarly, whereas oleate and linoleate tend to decrease the rate but not the efficiency of ATP synthesis. Importantly, we reveal that palmitate influences how oxidative ATP supply is used to fuel ATP-consuming processes. Direct measurement of newly made protein demonstrates that palmitate lowers the rate of de novo protein synthesis by more than 30%. The anticipated decrease of energy demand linked to protein synthesis is confirmed by attenuated cycloheximide-sensitivity of mitochondrial respiratory activity used to make ATP. This indirect measure of ATP turnover indicates that palmitate lowers ATP supply reserved for protein synthesis by at least 40%. This decrease is also provoked by stearate, oleate and linoleate, albeit to a lesser extent. Moreover, palmitate lowers ATP supply for sodium pump activity by 60-70% and, in human cells, decreases ATP supply for DNA/RNA synthesis by almost three-quarters. These novel fatty acid effects on energy expenditure inform the 'mitochondrial insufficiency' debate.
Collapse
Affiliation(s)
- Raid B Nisr
- School of Biomedical and Healthcare Sciences, Plymouth University, Drake Circus, PL4 8AA, Plymouth, UK
| | - Charles Affourtit
- School of Biomedical and Healthcare Sciences, Plymouth University, Drake Circus, PL4 8AA, Plymouth, UK.
| |
Collapse
|
48
|
The Role of Organelle Stresses in Diabetes Mellitus and Obesity: Implication for Treatment. Anal Cell Pathol (Amst) 2015; 2015:972891. [PMID: 26613076 PMCID: PMC4646985 DOI: 10.1155/2015/972891] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Accepted: 10/08/2015] [Indexed: 12/17/2022] Open
Abstract
The type 2 diabetes pandemic in recent decades is a huge global health threat. This pandemic is primarily attributed to the surplus of nutrients and the increased prevalence of obesity worldwide. In contrast, calorie restriction and weight reduction can drastically prevent type 2 diabetes, indicating a central role of nutrient excess in the development of diabetes. Recently, the molecular links between excessive nutrients, organelle stress, and development of metabolic disease have been extensively studied. Specifically, excessive nutrients trigger endoplasmic reticulum stress and increase the production of mitochondrial reactive oxygen species, leading to activation of stress signaling pathway, inflammatory response, lipogenesis, and pancreatic beta-cell death. Autophagy is required for clearance of hepatic lipid clearance, alleviation of pancreatic beta-cell stress, and white adipocyte differentiation. ROS scavengers, chemical chaperones, and autophagy activators have demonstrated promising effects for the treatment of insulin resistance and diabetes in preclinical models. Further results from clinical trials are eagerly awaited.
Collapse
|
49
|
Zamora M, Pardo R, Villena JA. Pharmacological induction of mitochondrial biogenesis as a therapeutic strategy for the treatment of type 2 diabetes. Biochem Pharmacol 2015. [PMID: 26212547 DOI: 10.1016/j.bcp.2015.06.032] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Defects in mitochondrial oxidative function have been associated with the onset of type 2 diabetes. Although the causal relationship between mitochondrial dysfunction and diabetes has not been fully established, numerous studies indicate that improved glucose homeostasis achieved via lifestyle interventions, such as exercise or calorie restriction, is tightly associated with increased mitochondrial biogenesis and oxidative function. Therefore, it is conceivable that potentiating mitochondrial biogenesis by pharmacological means could constitute an efficacious therapeutic strategy that would particularly benefit those diabetic patients who cannot adhere to comprehensive programs based on changes in lifestyle or that require a relatively rapid improvement in their diabetic status. In this review, we discuss several pharmacological targets and drugs that modulate mitochondrial biogenesis as well as their potential use as treatments for insulin resistance and diabetes.
Collapse
Affiliation(s)
- Mònica Zamora
- Cell Biology Group, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Rosario Pardo
- Laboratory of Metabolism and Obesity, Vall d'Hebron-Institut de Recerca, Universitat Autònoma de Barcelona, CIBER on Diabetes and Associated Metabolic Diseases (CIBERDEM), Barcelona, Spain
| | - Josep A Villena
- Laboratory of Metabolism and Obesity, Vall d'Hebron-Institut de Recerca, Universitat Autònoma de Barcelona, CIBER on Diabetes and Associated Metabolic Diseases (CIBERDEM), Barcelona, Spain.
| |
Collapse
|
50
|
Lee MS. Effect of mitochondrial stress on systemic metabolism. Ann N Y Acad Sci 2015; 1350:61-5. [DOI: 10.1111/nyas.12822] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 05/20/2015] [Accepted: 05/21/2015] [Indexed: 12/30/2022]
Affiliation(s)
- Myung-Shik Lee
- Severans Biomedical Research Institute and Department of Internal Medicine; Yonsei University College of Medicine; Seoul Republic of Korea
| |
Collapse
|