1
|
Yuzefovych LV, Noh HL, Suk S, Schuler AM, Mulekar MS, Pastukh VM, Kim JK, Rachek LI. Mitochondria-Targeted DNA Repair Glycosylase hOGG1 Protects Against HFD-Induced Liver Oxidative Mitochondrial DNA Damage and Insulin Resistance in OGG1-Deficient Mice. Int J Mol Sci 2024; 25:12168. [PMID: 39596235 PMCID: PMC11595121 DOI: 10.3390/ijms252212168] [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: 09/13/2024] [Revised: 10/28/2024] [Accepted: 10/30/2024] [Indexed: 11/28/2024] Open
Abstract
8-oxoguanine DNA glycosylase-1 (OGG1) is a DNA glycosylase mediating the first step in base excision repair which removes 7,8-dihydro-8-oxoguanine (8-oxoG) and repairs oxidized nuclear and mitochondrial DNA. Previous studies showed that OGG1 deficiency results in an increased susceptibility to high-fat diet (HFD)-induced obesity and metabolic dysfunction in mice, suggesting a crucial role of OGG1 in metabolism. However, the tissue-specific mechanisms of how OGG1 deficiency leads to insulin resistance is unknown. Thus, in the current study, we used a hyperinsulinemic-euglycemic clamp to evaluate in-depth glucose metabolism in male wild-type (WT) mice and Ogg1-/- (Ogg1-KO) mice fed an HFD. Ogg1-KO mice fed HFD were more obese, with significantly lower hepatic insulin action compared to WT/HFD mice. Targeting human OGG1 to mitochondria protected against HFD-induced obesity, insulin resistance, oxidative mitochondrial DNA damage in the liver and showed decreased expression of liver gluconeogenic genes in Ogg1-KO mice, suggesting a putative protective mechanism. Additionally, several subunits of oxidative phosphorylation protein levels were noticeably increased in Ogg1-KO/Tg compared to Ogg1-KO mice fed an HFD which was associated with improved insulin signaling. Our findings demonstrate the crucial role of mitochondrial hOGG1 in HFD-induced insulin resistance and propose several protective mechanisms which can further direct the development of therapeutic treatment.
Collapse
Affiliation(s)
- Larysa V. Yuzefovych
- Departments of Pharmacology, Frederick P. Whiddon College of Medicine, University of South Alabama, Mobile, AL 36688, USA
| | - Hye Lim Noh
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Sujin Suk
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Anne Michele Schuler
- Departments of Microbiology, Frederick P. Whiddon College of Medicine, University of South Alabama, Mobile, AL 36688, USA
| | - Madhuri S. Mulekar
- Department of Mathematics and Statistics, College of Art and Science, University of South Alabama, Mobile, AL 36688, USA
| | - Viktor M. Pastukh
- Departments of Pharmacology, Frederick P. Whiddon College of Medicine, University of South Alabama, Mobile, AL 36688, USA
| | - Jason K. Kim
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Lyudmila I. Rachek
- Departments of Pharmacology, Frederick P. Whiddon College of Medicine, University of South Alabama, Mobile, AL 36688, USA
| |
Collapse
|
2
|
Cavestro C, Morra F, Legati A, D'Amato M, Nasca A, Iuso A, Lubarr N, Morrison JL, Wheeler PG, Serra‐Juhé C, Rodríguez‐Santiago B, Turón‐Viñas E, Prouteau C, Barth M, Hayflick SJ, Ghezzi D, Tiranti V, Di Meo I. Emerging variants, unique phenotypes, and transcriptomic signatures: an integrated study of COASY-associated diseases. Ann Clin Transl Neurol 2024; 11:1615-1629. [PMID: 38750253 PMCID: PMC11187879 DOI: 10.1002/acn3.52079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 04/17/2024] [Indexed: 06/20/2024] Open
Abstract
OBJECTIVE COASY, the gene encoding the bifunctional enzyme CoA synthase, which catalyzes the last two reactions of cellular de novo coenzyme A (CoA) biosynthesis, has been linked to two exceedingly rare autosomal recessive disorders, such as COASY protein-associated neurodegeneration (CoPAN), a form of neurodegeneration with brain iron accumulation (NBIA), and pontocerebellar hypoplasia type 12 (PCH12). We aimed to expand the phenotypic spectrum and gain insights into the pathogenesis of COASY-related disorders. METHODS Patients were identified through targeted or exome sequencing. To unravel the molecular mechanisms of disease, RNA sequencing, bioenergetic analysis, and quantification of critical proteins were performed on fibroblasts. RESULTS We identified five new individuals harboring novel COASY variants. While one case exhibited classical CoPAN features, the others displayed atypical symptoms such as deafness, language and autism spectrum disorders, brain atrophy, and microcephaly. All patients experienced epilepsy, highlighting its potential frequency in COASY-related disorders. Fibroblast transcriptomic profiling unveiled dysregulated expression in genes associated with mitochondrial respiration, responses to oxidative stress, transmembrane transport, various cellular signaling pathways, and protein translation, modification, and trafficking. Bioenergetic analysis revealed impaired mitochondrial oxygen consumption in COASY fibroblasts. Despite comparable total CoA levels to control cells, the amounts of mitochondrial 4'-phosphopantetheinylated proteins were significantly reduced in COASY patients. INTERPRETATION These results not only extend the clinical phenotype associated with COASY variants but also suggest a continuum between CoPAN and PCH12. The intricate interplay of altered cellular processes and signaling pathways provides valuable insights for further research into the pathogenesis of COASY-associated diseases.
Collapse
Affiliation(s)
- Chiara Cavestro
- Unit of Medical Genetics and NeurogeneticsFondazione IRCCS Istituto Neurologico Carlo BestaMilanItaly
| | - Francesca Morra
- Unit of Medical Genetics and NeurogeneticsFondazione IRCCS Istituto Neurologico Carlo BestaMilanItaly
| | - Andrea Legati
- Unit of Medical Genetics and NeurogeneticsFondazione IRCCS Istituto Neurologico Carlo BestaMilanItaly
| | - Marco D'Amato
- Unit of Medical Genetics and NeurogeneticsFondazione IRCCS Istituto Neurologico Carlo BestaMilanItaly
| | - Alessia Nasca
- Unit of Medical Genetics and NeurogeneticsFondazione IRCCS Istituto Neurologico Carlo BestaMilanItaly
| | - Arcangela Iuso
- Institute of Human Genetics, School of MedicineTechnical University of MunichMunichGermany
- Institute of NeurogenomicsHelmholtz Zentrum MünchenNeuherbergGermany
| | - Naomi Lubarr
- Department of NeurologyIcahn School of Medicine at Mount Sinai, Mount Sinai Beth IsraelNew YorkNew YorkUSA
| | | | | | - Clara Serra‐Juhé
- Genetics DepartmentHospital de la Santa Creu i Sant PauBarcelonaSpain
| | - Benjamín Rodríguez‐Santiago
- Genetics DepartmentHospital de la Santa Creu i Sant PauBarcelonaSpain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER)MadridSpain
- Genomic Instability Syndromes and DNA Repair Group and Join Research Unit on Genomic Medicine UAB‐Sant Pau Biomedical Research InstituteHospital de la Santa Creu i Sant PauBarcelonaSpain
| | - Eulalia Turón‐Viñas
- Child Neurology Unit, Pediatrics ServiceHospital de la Santa Creu i Sant PauBarcelonaSpain
| | | | - Magalie Barth
- Department of GeneticsUniversity Hospital of AngersAngersFrance
| | - Susan J. Hayflick
- Department of Molecular and Medical GeneticsOregon Health & Science UniversityPortlandOregonUSA
- Department of PediatricsOregon Health & Science UniversityPortlandOregonUSA
- Department of NeurologyOregon Health & Science UniversityPortlandOregonUSA
| | - Daniele Ghezzi
- Unit of Medical Genetics and NeurogeneticsFondazione IRCCS Istituto Neurologico Carlo BestaMilanItaly
- Department of Pathophysiology and TransplantationUniversity of MilanMilanItaly
| | - Valeria Tiranti
- Unit of Medical Genetics and NeurogeneticsFondazione IRCCS Istituto Neurologico Carlo BestaMilanItaly
| | - Ivano Di Meo
- Unit of Medical Genetics and NeurogeneticsFondazione IRCCS Istituto Neurologico Carlo BestaMilanItaly
| |
Collapse
|
3
|
Cilenti L, Di Gregorio J, Mahar R, Liu F, Ambivero CT, Periasamy M, Merritt ME, Zervos AS. Inactivation of mitochondrial MUL1 E3 ubiquitin ligase deregulates mitophagy and prevents diet-induced obesity in mice. Front Mol Biosci 2024; 11:1397565. [PMID: 38725872 PMCID: PMC11079312 DOI: 10.3389/fmolb.2024.1397565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 04/05/2024] [Indexed: 05/12/2024] Open
Abstract
Obesity is a growing epidemic affecting millions of people worldwide and a major risk factor for a multitude of chronic diseases and premature mortality. Accumulating evidence suggests that mitochondria have a profound role in diet-induced obesity and the associated metabolic changes, but the molecular mechanisms linking mitochondria to obesity remain poorly understood. Our studies have identified a new function for mitochondrial MUL1 E3 ubiquitin ligase, a protein known to regulate mitochondrial dynamics and mitophagy, in the control of energy metabolism and lipogenesis. Genetic deletion of Mul1 in mice impedes mitophagy and presents a metabolic phenotype that is resistant to high-fat diet (HFD)-induced obesity and metabolic syndrome. Several metabolic and lipidomic pathways are perturbed in the liver and white adipose tissue (WAT) of Mul1(-/-) animals on HFD, including the one driven by Stearoyl-CoA Desaturase 1 (SCD1), a pivotal regulator of lipid metabolism and obesity. In addition, key enzymes crucial for lipogenesis and fatty acid oxidation such as ACC1, FASN, AMPK, and CPT1 are also modulated in the absence of MUL1. The concerted action of these enzymes, in the absence of MUL1, results in diminished fat storage and heightened fatty acid oxidation. Our findings underscore the significance of MUL1-mediated mitophagy in regulating lipogenesis and adiposity, particularly in the context of HFD. Consequently, our data advocate the potential of MUL1 as a therapeutic target for drug development in the treatment of obesity, insulin resistance, NAFLD, and cardiometabolic diseases.
Collapse
Affiliation(s)
- Lucia Cilenti
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, Orlando, FL, United States
| | - Jacopo Di Gregorio
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, Orlando, FL, United States
| | - Rohit Mahar
- Department of Chemistry, Hemvati Nandan Bahuguna Garhwal University (A Central University), Srinagar Garhwal, Uttarakhand, India
| | - Fei Liu
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, Orlando, FL, United States
| | - Camilla T. Ambivero
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, Orlando, FL, United States
| | - Muthu Periasamy
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, Orlando, FL, United States
| | - Matthew E. Merritt
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL, United States
| | - Antonis S. Zervos
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, Orlando, FL, United States
| |
Collapse
|
4
|
Shen Y, Chen Y, Zhang S, Wu Z, Lu X, Liu W, Liu B, Zhou X. Smartphone-based digital phenotyping for genome-wide association study of intramuscular fat traits in longissimus dorsi muscle of pigs. Anim Genet 2024; 55:230-237. [PMID: 38290559 DOI: 10.1111/age.13401] [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: 12/11/2023] [Revised: 12/11/2023] [Accepted: 01/17/2024] [Indexed: 02/01/2024]
Abstract
Intramuscular fat (IMF) content and distribution significantly contribute to the eating quality of pork. However, the current methods used for measuring these traits are complex, time-consuming and costly. To simplify the measurement process, this study developed a smartphone application (App) called Pork IMF. This App serves as a rapid and portable phenotyping tool for acquiring pork images and extracting the image-based IMF traits through embedded deep-learning algorithms. Utilizing this App, we collected the IMF traits of the longissimus dorsi muscle in a crossbred population of Large White × Tongcheng pigs. Genome-wide association studies detected 13 and 16 SNPs that were significantly associated with IMF content and distribution, respectively, highlighting NR2F2, MCTP2, MTLN, ST3GAL5, NDUFAB1 and PID1 as candidate genes. Our research introduces a user-friendly digital phenotyping technology for quantifying IMF traits and suggests candidate genes and SNPs for genetic improvement of IMF traits in pigs.
Collapse
Affiliation(s)
- Yang Shen
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Yuxi Chen
- School of Computer Science and Artificial Intelligence, Wuhan University of Technology, Wuhan, China
| | - Shufeng Zhang
- School of Computer Science and Artificial Intelligence, Wuhan University of Technology, Wuhan, China
| | - Ze Wu
- School of Computer Science and Artificial Intelligence, Wuhan University of Technology, Wuhan, China
| | - Xiaoyu Lu
- School of Computer Science and Artificial Intelligence, Wuhan University of Technology, Wuhan, China
| | - Weizhen Liu
- School of Computer Science and Artificial Intelligence, Wuhan University of Technology, Wuhan, China
| | - Bang Liu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
- Hubei Hongshan Laboratory, Wuhan, China
| | - Xiang Zhou
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
- Hubei Hongshan Laboratory, Wuhan, China
- Shenzhen Institute of Nutrition and Health, Huazhong Agricultural University, Wuhan, China
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| |
Collapse
|
5
|
Peng H, Huang Y, Wei G, Pang Y, Yuan H, Zou X, Xie Y, Chen W. Testicular Toxicity in Rats Exposed to AlCl 3: a Proteomics Study. Biol Trace Elem Res 2024; 202:1084-1102. [PMID: 37382810 DOI: 10.1007/s12011-023-03745-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 06/22/2023] [Indexed: 06/30/2023]
Abstract
Aluminum contamination is a growing environmental and public health concern, and aluminum testicular toxicity has been reported in male rats; however, the underlying mechanisms of this toxicity are unclear. The objective of this study was to investigate the effects of exposure to aluminum chloride (AlCl3) on alterations in the levels of sex hormones (testosterone [T], luteinizing hormone [LH], and follicle-stimulating hormone [FSH]) and testicular damage. Additionally, the mechanisms of toxicity in the testes of AlCl3-exposed rats were analyzed by proteomics. Three different concentrations of AlCl3 were administered to rats. The results demonstrated a decrease in T, LH, and FSH levels with increasing concentrations of AlCl3 exposure. HE staining results revealed that the spermatogenic cells in the AlCl3-exposed rats were widened, disorganized, or absent, with increased severe tissue destruction at higher concentrations of AlCl3 exposure. Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) enrichment analyses revealed that differentially expressed proteins (DEPs) after AlCl3 exposure were primarily associated with various metabolic processes, sperm fibrous sheath, calcium-dependent protein binding, oxidative phosphorylation, and ribosomes. Subsequently, DEPs from each group were subjected to protein-protein interaction (PPI) analysis followed by the screening of interactional key DEPs. Western blot experiments validated the proteomics data, revealing the downregulation of sperm-related DEPs (AKAP4, ODF1, and OAZ3) and upregulation of regulatory ribosome-associated protein (UBA52) and mitochondrial ribosomal protein (MRPL32). These findings provide a basis for studying the mechanism of testicular toxicity due to AlCl3 exposure.
Collapse
Affiliation(s)
- Huixin Peng
- The Affiliated Hospital of You jiang Medical University for Nationalities, Baise, 533000, Guangxi, China
- Graduate School of You jiang Medical University for Nationalities, Baise, 533000, Guangxi, China
| | - Yanxin Huang
- The Affiliated Hospital of You jiang Medical University for Nationalities, Baise, 533000, Guangxi, China
- Graduate School of You jiang Medical University for Nationalities, Baise, 533000, Guangxi, China
| | - Guangji Wei
- The Affiliated Hospital of You jiang Medical University for Nationalities, Baise, 533000, Guangxi, China
- Graduate School of You jiang Medical University for Nationalities, Baise, 533000, Guangxi, China
| | - Yanfang Pang
- The Affiliated Hospital of You jiang Medical University for Nationalities, Baise, 533000, Guangxi, China
- Graduate School of You jiang Medical University for Nationalities, Baise, 533000, Guangxi, China
| | - Huixiong Yuan
- The Affiliated Hospital of You jiang Medical University for Nationalities, Baise, 533000, Guangxi, China
- Graduate School of You jiang Medical University for Nationalities, Baise, 533000, Guangxi, China
| | - Xiong Zou
- Guangxi Key Laboratory of reproductive health and birth defect prevention, Nanning, 530000, Guangxi, China
| | - Yu'an Xie
- Guangxi Key Laboratory of reproductive health and birth defect prevention, Nanning, 530000, Guangxi, China.
| | - Wencheng Chen
- The Affiliated Hospital of You jiang Medical University for Nationalities, Baise, 533000, Guangxi, China.
- Graduate School of You jiang Medical University for Nationalities, Baise, 533000, Guangxi, China.
| |
Collapse
|
6
|
Zhu J, Wang Y, Su Y, Zheng M, Cui H, Chen Z. RNA sequencing identifies key genes involved in intramuscular fat deposition in chickens at different developmental stages. BMC Genomics 2024; 25:219. [PMID: 38413888 PMCID: PMC10900564 DOI: 10.1186/s12864-023-09819-y] [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: 07/04/2023] [Accepted: 11/20/2023] [Indexed: 02/29/2024] Open
Abstract
BACKGROUND Intramuscular fat (IMF) is an important factor in meat quality, and triglyceride (TG) and Phospholipids (PLIP), as the main components of IMF, are of great significance to the improvement of meat quality. RESULTS In this study, we used 30 RNA sequences generated from the transcriptome of chicken breast muscle tissues at different developmental stages to construct a gene expression matrix to map RNA sequence reads to the chicken genome and identify the transcript of origin. We used weighted gene co-expression network analysis (WGCNA) and identified 27 co-expression modules, 10 of which were related to TG and PLIP. We identified 150 highly-connected hub genes related to TG and PLIP, respectively, which were found to be mainly enriched in the adipocytokine signaling pathway, MAPK signaling pathway, mTOR signaling pathway, FoxO signaling pathway, and TGF-beta signaling pathway. Additionally, using the BioMart database, we identified 134 and 145 candidate genes related to fat development in the TG-related module and PLIP-related module, respectively. Among them, RPS6KB1, BRCA1, CDK1, RPS3, PPARGC1A, ACSL1, NDUFAB1, NDUFA9, ATP5B and PRKAG2 were identified as candidate genes related to fat development and highly-connected hub genes in the module, suggesting that these ten genes may be important candidate genes affecting IMF deposition. CONCLUSIONS RPS6KB1, BRCA1, CDK1, RPS3, PPARGC1A, ACSL1, NDUFAB1, NDUFA9, ATP5B and PRKAG2 may be important candidate genes affecting IMF deposition. The purpose of this study was to identify the co-expressed gene modules related to chicken IMF deposition using WGCNA and determine key genes related to IMF deposition, so as to lay a foundation for further research on the molecular regulation mechanism underlying chicken fat deposition.
Collapse
Affiliation(s)
- Jinmei Zhu
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao, 266109, China
- State Key Laboratory of Animal Nutrition, Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Yongli Wang
- State Key Laboratory of Animal Nutrition, Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Yongchun Su
- Guangxi Jingling Agriculture and animal Husbandry Group Co., LTD, Nanning, 530049, China
| | - Maiqing Zheng
- State Key Laboratory of Animal Nutrition, Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Huanxian Cui
- State Key Laboratory of Animal Nutrition, Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.
| | - Zhiwu Chen
- Guangxi Jingling Agriculture and animal Husbandry Group Co., LTD, Nanning, 530049, China.
| |
Collapse
|
7
|
Wedan RJ, Longenecker JZ, Nowinski SM. Mitochondrial fatty acid synthesis is an emergent central regulator of mammalian oxidative metabolism. Cell Metab 2024; 36:36-47. [PMID: 38128528 PMCID: PMC10843818 DOI: 10.1016/j.cmet.2023.11.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/27/2023] [Accepted: 11/30/2023] [Indexed: 12/23/2023]
Abstract
Contrary to their well-known functions in nutrient breakdown, mitochondria are also important biosynthetic hubs and express an evolutionarily conserved mitochondrial fatty acid synthesis (mtFAS) pathway. mtFAS builds lipoic acid and longer saturated fatty acids, but its exact products, their ultimate destination in cells, and the cellular significance of the pathway are all active research questions. Moreover, why mitochondria need mtFAS despite their well-defined ability to import fatty acids is still unclear. The identification of patients with inborn errors of metabolism in mtFAS genes has sparked fresh research interest in the pathway. New mammalian models have provided insights into how mtFAS coordinates many aspects of oxidative mitochondrial metabolism and raise questions about its role in diseases such as obesity, diabetes, and heart failure. In this review, we discuss the products of mtFAS, their function, and the consequences of mtFAS impairment across models and in metabolic disease.
Collapse
Affiliation(s)
- Riley J Wedan
- Department of Metabolism and Nutritional Programming, The Van Andel Institute, Grand Rapids, MI 49503, USA; College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
| | - Jacob Z Longenecker
- Department of Metabolism and Nutritional Programming, The Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Sara M Nowinski
- Department of Metabolism and Nutritional Programming, The Van Andel Institute, Grand Rapids, MI 49503, USA.
| |
Collapse
|
8
|
Lei X, Hao Z, Wang H, Tang Z, Zhang Z, Yuan J. Identification of core genes, critical signaling pathways, and potential drugs for countering BPA-induced hippocampal neurotoxicity in male mice. Food Chem Toxicol 2023; 182:114195. [PMID: 37992956 DOI: 10.1016/j.fct.2023.114195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 10/09/2023] [Accepted: 11/16/2023] [Indexed: 11/24/2023]
Abstract
Although the neurotoxicity of the common chemical bisphenol A (BPA) to the mouse hippocampus has been often reported, the mechanism underlying BPA-induced depression-like behavior in mice remains unclear. We evaluated BPA's role in inducing depressive-like behavior by exposing male mice to different BPA concentrations (0, 0.01, 0.1, and 1 μg/mL) and using the forced swimming test (FST) and tail suspension test (TST). We aimed to identify critical gene and anti-BPA-neurotoxicity compounds using RNA sequencing combined with bioinformatics analysis. Our results showed that 1 μg/mL BPA exposure increased mouse immobility during the FST and TST. Based on BPA-induced hippocampal transcriptome changes, we identified NADH: ubiquinone oxidoreductase subunit AB1 (Ndufab1) as a critical and potential therapeutic target gene, and Ndufab1 mRNA and protein levels were downregulated in the BPA-exposed groups. Furthermore, molecular docking identified phenelzine as a compound that could counteract BPA-related neurotoxicity. Conclusively, our analyses confirmed that BPA triggers depressive behavior in male mice by downregulating Ndufab1 expression and suggested that phenelzine might reduce BPA-induced neurotoxicity.
Collapse
Affiliation(s)
- Xuepei Lei
- College of Life Sciences, Shanxi Agricultural University, Taigu, Shanxi, 030801, China
| | - Zhoujie Hao
- College of Life Sciences, Shanxi Agricultural University, Taigu, Shanxi, 030801, China
| | - Huimin Wang
- College of Life Sciences, Shanxi Agricultural University, Taigu, Shanxi, 030801, China
| | - Zhongwei Tang
- College of Life Sciences, Shanxi Agricultural University, Taigu, Shanxi, 030801, China
| | - Zhuo Zhang
- College of Life Sciences, Shanxi Agricultural University, Taigu, Shanxi, 030801, China
| | - Jianqin Yuan
- College of Life Sciences, Shanxi Agricultural University, Taigu, Shanxi, 030801, China; Shanxi Key Laboratory of Ecological Animal Sciences and Environmental Veterinary Medicine, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi, 030801, China.
| |
Collapse
|
9
|
Zeber-Lubecka N, Ciebiera M, Hennig EE. Polycystic Ovary Syndrome and Oxidative Stress-From Bench to Bedside. Int J Mol Sci 2023; 24:14126. [PMID: 37762427 PMCID: PMC10531631 DOI: 10.3390/ijms241814126] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/06/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Oxidative stress (OS) is a condition that occurs as a result of an imbalance between the production of reactive oxygen species (ROS) and the body's ability to detoxify and neutralize them. It can play a role in a variety of reproductive system conditions, including polycystic ovary syndrome (PCOS), endometriosis, preeclampsia, and infertility. In this review, we briefly discuss the links between oxidative stress and PCOS. Mitochondrial mutations may lead to impaired oxidative phosphorylation (OXPHOS), decreased adenosine triphosphate (ATP) production, and an increased production of ROS. These functional consequences may contribute to the metabolic and hormonal dysregulation observed in PCOS. Studies have shown that OS negatively affects ovarian follicles and disrupts normal follicular development and maturation. Excessive ROS may damage oocytes and granulosa cells within the follicles, impairing their quality and compromising fertility. Impaired OXPHOS and mitochondrial dysfunction may contribute to insulin resistance (IR) by disrupting insulin signaling pathways and impairing glucose metabolism. Due to dysfunctional OXPHOS, reduced ATP production, may hinder insulin-stimulated glucose uptake, leading to IR. Hyperandrogenism promotes inflammation and IR, both of which can increase the production of ROS and lead to OS. A detrimental feedback loop ensues as IR escalates, causing elevated insulin levels that exacerbate OS. Exploring the relations between OS and PCOS is crucial to fully understand the role of OS in the pathophysiology of PCOS and to develop effective treatment strategies to improve the quality of life of women affected by this condition. The role of antioxidants as potential therapies is also discussed.
Collapse
Affiliation(s)
- Natalia Zeber-Lubecka
- Department of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education, 01-813 Warsaw, Poland;
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Michał Ciebiera
- Second Department of Obstetrics and Gynecology, Centre of Postgraduate Medical Education, 01-813 Warsaw, Poland;
- Warsaw Institute of Women’s Health, 00-189 Warsaw, Poland
| | - Ewa E. Hennig
- Department of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education, 01-813 Warsaw, Poland;
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| |
Collapse
|
10
|
Liver-specific overexpression of lipoprotein lipase improves glucose metabolism in high-fat diet-fed mice. PLoS One 2022; 17:e0274297. [PMID: 36099304 PMCID: PMC9469954 DOI: 10.1371/journal.pone.0274297] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 08/25/2022] [Indexed: 11/24/2022] Open
Abstract
The liver is the main organ that regulates lipid and glucose metabolism. Ectopic lipid accumulation in the liver impairs insulin sensitivity and glucose metabolism. Lipoprotein lipase (LPL), mainly expressed in the adipose tissue and muscle, is a key enzyme that regulates lipid metabolism via the hydrolysis of triglyceride in chylomicrons and very-low-density lipoproteins. Here, we aimed to investigate whether the suppression level of hepatic lipid accumulation via overexpression of LPL in mouse liver leads to improved metabolism. To overexpress LPL in the liver, we generated an LPL-expressing adenovirus (Ad) vector using an improved Ad vector that exhibited considerably lower hepatotoxicity (Ad-LPL). C57BL/6 mice were treated with Ad vectors and simultaneously fed a high-fat diet (HFD). Lipid droplet formation in the liver decreased in Ad-LPL-treated mice relative to that in control Ad vector-treated mice. Glucose tolerance and insulin resistance were remarkably improved in Ad-LPL-treated mice compared to those in control Ad vector-treated mice. The expression levels of fatty acid oxidation-related genes, such as peroxisome proliferator-activated receptor α, carnitine palmitoyltransferase 1, and acyl-CoA oxidase 1, were 1.7–2.0-fold higher in Ad-LPL-treated mouse livers than that in control Ad-vector-treated mouse livers. Furthermore, hepatic LPL overexpression partly maintained mitochondrial content in HFD-fed mice. These results indicate that LPL overexpression in the livers of HFD-fed mice attenuates the accumulation of lipid droplets in the liver and improves glucose metabolism. These findings may enable the development of new drugs to treat metabolic syndromes such as type 2 diabetes mellitus and non-alcoholic fatty liver disease.
Collapse
|
11
|
Giannos P, Prokopidis K, Raleigh SM, Kelaiditi E, Hill M. Altered mitochondrial microenvironment at the spotlight of musculoskeletal aging and Alzheimer's disease. Sci Rep 2022; 12:11290. [PMID: 35788655 PMCID: PMC9253146 DOI: 10.1038/s41598-022-15578-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 06/27/2022] [Indexed: 11/23/2022] Open
Abstract
Emerging evidence has linked Alzheimer's disease (AD) onset with musculoskeletal aging via a muscle-brain crosstalk mediated by dysregulation of the mitochondrial microenvironment. This study investigated gene expression profiles from skeletal muscle tissues of older healthy adults to identify potential gene biomarkers whose dysregulated expression and protein interactome were involved in AD. Screening of the literature resulted in 12 relevant microarray datasets (GSE25941, GSE28392, GSE28422, GSE47881, GSE47969, GSE59880) in musculoskeletal aging and (GSE4757, GSE5281, GSE16759, GSE28146, GSE48350, GSE84422) in AD. Retrieved differentially expressed genes (DEGs) were used to construct two unique protein-protein interaction networks and clustering gene modules were identified. Overlapping module DEGs in the musculoskeletal aging and AD networks were ranked based on 11 topological algorithms and the five highest-ranked ones were considered as hub genes. The analysis revealed that the dysregulated expression of the mitochondrial microenvironment genes, NDUFAB1, UQCRC1, UQCRFS1, NDUFS3, and MRPL15, overlapped between both musculoskeletal aging and AD networks. Thus, these genes may have a potential role as markers of AD occurrence in musculoskeletal aging. Human studies are warranted to evaluate the functional role and prognostic value of these genes in aging populations with sarcopenia and AD.
Collapse
Affiliation(s)
- Panagiotis Giannos
- Society of Meta-research and Biomedical Innovation, London, UK. .,Department of Life Sciences, Faculty of Natural Sciences, Imperial College London, South Kensington, London, SW7 2AZ, UK.
| | - Konstantinos Prokopidis
- Society of Meta-research and Biomedical Innovation, London, UK.,Department of Musculoskeletal Biology, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Stuart M Raleigh
- Cardiovascular and Lifestyle Medicine Research Group, Centre for Sport, Exercise and Life Sciences, Coventry University, Coventry, UK
| | - Eirini Kelaiditi
- Faculty of Sport, Allied Health and Performance Science, St Mary's University Twickenham, Twickenham, UK
| | - Mathew Hill
- Centre for Sport, Exercise and Life Sciences, School of Life Sciences, Coventry University, Coventry, UK
| |
Collapse
|
12
|
Dumolt J, Powell TL, Jansson T, Rosario FJ. Normalization of maternal adiponectin in obese pregnant mice prevents programming of impaired glucose metabolism in adult offspring. FASEB J 2022; 36:e22383. [PMID: 35670755 DOI: 10.1096/fj.202200326r] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/16/2022] [Accepted: 05/19/2022] [Indexed: 11/11/2022]
Abstract
Infants born to obese mothers have a greater risk for childhood obesity and insulin resistance. However, the underlying biological mechanism remains elusive, which constitutes a significant roadblock for developing specific prevention strategies. Maternal adiponectin levels are lower in obese pregnant women, which is linked with increased placental nutrient transport and fetal overgrowth. We have previously reported that adiponectin supplementation to obese dams during the last four days of pregnancy prevented the development of obesity, glucose intolerance, muscle insulin resistance, and fatty liver in three months old offspring. In the present study, we tested the hypothesis that 6-9-month-old offspring of obese dams show glucose intolerance associated with muscle insulin resistance and mitochondrial dysfunction and that normalization of maternal adiponectin in obese pregnant mice prevents the development of this phenotype in the offspring. Male and female offspring of obese mice exhibited in vivo glucose intolerance and insulin resistance at 6 and 9 months of age. In gastrocnemius muscles ex vivo, male and female offspring of obese dams showed reduced phosphorylation of insulin receptor substrate 1Tyr-608 , AktThr-308 , and decreased Glut4 plasma membrane translocation upon insulin stimulation. These metabolic abnormalities in offspring born to obese mice were largely prevented by normalization of maternal adiponectin levels in late pregnancy. We provide evidence that low circulating maternal adiponectin is a critical mechanistic link between maternal obesity and the development of metabolic disease in offspring. Strategies aimed at improving maternal adiponectin levels may prevent long-term metabolic dysfunction in offspring of obese mothers.
Collapse
Affiliation(s)
- Jerad Dumolt
- Division of Reproductive Sciences, Department of OB/GYN, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Theresa L Powell
- Division of Reproductive Sciences, Department of OB/GYN, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.,Section of Neonatology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Thomas Jansson
- Division of Reproductive Sciences, Department of OB/GYN, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Fredrick J Rosario
- Division of Reproductive Sciences, Department of OB/GYN, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
13
|
Trumpff C, Owusu-Ansah E, Klein HU, Lee AJ, Petyuk V, Wingo TS, Wingo AP, Thambisetty M, Ferrucci L, Seyfried NT, Bennett DA, De Jager PL, Picard M. Mitochondrial respiratory chain protein co-regulation in the human brain. Heliyon 2022; 8:e09353. [PMID: 35600441 PMCID: PMC9118667 DOI: 10.1016/j.heliyon.2022.e09353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 03/12/2022] [Accepted: 04/27/2022] [Indexed: 11/09/2022] Open
Abstract
Mitochondrial respiratory chain (RC) function requires the stoichiometric interaction among dozens of proteins but their co-regulation has not been defined in the human brain. Here, using quantitative proteomics across three independent cohorts we systematically characterized the co-regulation patterns of mitochondrial RC proteins in the human dorsolateral prefrontal cortex (DLPFC). Whereas the abundance of RC protein subunits that physically assemble into stable complexes were correlated, indicating their co-regulation, RC assembly factors exhibited modest co-regulation. Within complex I, nuclear DNA-encoded subunits exhibited >2.5-times higher co-regulation than mitochondrial (mt)DNA-encoded subunits. Moreover, mtDNA copy number was unrelated to mtDNA-encoded subunits abundance, suggesting that mtDNA content is not limiting. Alzheimer's disease (AD) brains exhibited reduced abundance of complex I RC subunits, an effect largely driven by a 2-4% overall lower mitochondrial protein content. These findings provide foundational knowledge to identify molecular mechanisms contributing to age- and disease-related erosion of mitochondrial function in the human brain.
Collapse
Affiliation(s)
- Caroline Trumpff
- Department of Psychiatry, Division of Behavioral Medicine, Columbia University Irving Medical Center, New York, USA
| | - Edward Owusu-Ansah
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, USA
| | - Hans-Ulrich Klein
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, USA
| | - Annie J. Lee
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, USA
| | - Vladislav Petyuk
- Pacific Northwest National Laboratory, Richland, Washington State, USA
| | - Thomas S. Wingo
- Departments of Neurology and Human Genetics, Emory University, Atlanta, GA, USA
| | - Aliza P. Wingo
- Atlanta VA Medical Center, Decatur, GA, USA
- Department of Psychiatry, Emory University, Atlanta, GA, USA
| | - Madhav Thambisetty
- Clinical and Translational Neuroscience Section, Laboratory of Behavioral Neuroscience, National Institute on Aging Intramural Research Program, Baltimore, USA
| | - Luigi Ferrucci
- Longitudinal Study Section, National Institute on Aging, Baltimore, USA
| | | | - David A. Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois, USA
| | - Philip L. De Jager
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, USA
| | - Martin Picard
- Department of Psychiatry, Division of Behavioral Medicine, Columbia University Irving Medical Center, New York, USA
- Department of Neurology, H. Houston Merritt Center, Columbia Translational Neuroscience Initiative, Columbia University Irving Medical Center, New York, USA
- New York State Psychiatric Institute, New York, USA
| |
Collapse
|
14
|
Santambrogio P, Ripamonti M, Cozzi A, Raimondi M, Cavestro C, Di Meo I, Rubio A, Taverna S, Tiranti V, Levi S. Massive iron accumulation in PKAN-derived neurons and astrocytes: light on the human pathological phenotype. Cell Death Dis 2022; 13:185. [PMID: 35217637 PMCID: PMC8881507 DOI: 10.1038/s41419-022-04626-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/26/2022] [Accepted: 02/07/2022] [Indexed: 12/11/2022]
Abstract
Neurodegeneration associated with defective pantothenate kinase-2 (PKAN) is an early-onset monogenic autosomal-recessive disorder. The hallmark of the disease is the massive accumulation of iron in the globus pallidus brain region of patients. PKAN is caused by mutations in the PANK2 gene encoding the mitochondrial enzyme pantothenate kinase-2, whose function is to catalyze the first reaction of the CoA biosynthetic pathway. To date, the way in which this alteration leads to brain iron accumulation has not been elucidated. Starting from previously obtained hiPS clones, we set up a differentiation protocol able to generate inhibitory neurons. We obtained striatal-like medium spiny neurons composed of approximately 70-80% GABAergic neurons and 10-20% glial cells. Within this mixed population, we detected iron deposition in both PKAN cell types, however, the viability of PKAN GABAergic neurons was strongly affected. CoA treatment was able to reduce cell death and, notably, iron overload. Further differentiation of hiPS clones in a pure population of astrocytes showed particularly evident iron accumulation, with approximately 50% of cells positive for Perls staining. The analysis of these PKAN astrocytes indicated alterations in iron metabolism, mitochondrial morphology, respiratory activity, and oxidative status. Moreover, PKAN astrocytes showed signs of ferroptosis and were prone to developing a stellate phenotype, thus gaining neurotoxic features. This characteristic was confirmed in iPS-derived astrocyte and glutamatergic neuron cocultures, in which PKAN glutamatergic neurons were less viable in the presence of PKAN astrocytes. This newly generated astrocyte model is the first in vitro disease model recapitulating the human phenotype and can be exploited to deeply clarify the pathogenetic mechanisms underlying the disease.
Collapse
Affiliation(s)
| | - Maddalena Ripamonti
- IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Anna Cozzi
- IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marzia Raimondi
- IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Chiara Cavestro
- Medical Genetics and Neurogenetics Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Ivano Di Meo
- Medical Genetics and Neurogenetics Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Alicia Rubio
- IRCCS San Raffaele Scientific Institute, Milan, Italy
- Institute of Neuroscience, National Research Council, Milan, Italy
| | | | - Valeria Tiranti
- Medical Genetics and Neurogenetics Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy.
| | - Sonia Levi
- IRCCS San Raffaele Scientific Institute, Milan, Italy.
- Vita-Salute San Raffaele University, Milan, Italy.
| |
Collapse
|
15
|
Li H, Guan K, Liu D, Liu M. Identification of mitochondria-related hub genes in sarcopenia and functional regulation of MFG-E8 on ROS-mediated mitochondrial dysfunction and cell cycle arrest. Food Funct 2021; 13:624-638. [PMID: 34928287 DOI: 10.1039/d1fo02610k] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Sarcopenia has high prevalence in the elderly population, but the genes and pathways related to aging in elderly patients with sarcopenia are poorly understood. Milk fat globule epidermal growth factor 8 (MFG-E8) is a peripheral membrane glycoprotein isolated from the milk fat globule membrane (MFGM). It has been found to exhibit various nutritional effects, including antibacterial, anti-cancer, anti-oxidant, anti-sarcopenia, and improving brain development and cognitive effects. This study aimed to investigate key differentially expressed genes (DEGs) and pathways associated with the progression of sarcopenia using bioinformatics analysis and in vitro myoblast experiment. The gene expression profiles of GSE8479 and GSE9676, which includes 40 young normal samples and 55 elderly samples, were downloaded from the Gene Expression Omnibus Database (GEO). Over 3253 DEGs were identified in the young and elderly samples (adjusted p value <0.05). A total of 213 co-expressed significantly DEGs were identified with Venn diagrams, including 82 up-regulated DEGs and 131 down-regulated DEGs. Based on the analysis of Gene Ontology (GO), protein-protein interaction (PPI) networks and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway, 10 hub genes screened by our study have been proved to play a role in regulating the occurrence and development of aging-related sarcopenia mainly via metabolic pathways, Huntington's disease, Parkinson's disease, oxidative phosphorylation and non-alcoholic fatty liver disease pathways. To further verify the protective effect of MFG-E8 on oxidative stress injured myoblasts, the cell cycle distribution, cell viability and reactive oxygen species (ROS) production were measured. The protein and mRNA levels of Akt, extracellular regulated protein kinases (ERK), p21Cip1, p27Kip1, cyclin D1, cyclin E1, cyclin-dependent kinase (CDK) 2 and 4 were quantified using qRT-PCR and western blot analysis. The results indicated that MFG-E8 has potential anti-sarcopenia effects by promoting ERK and Akt activation-mediated cell proliferation and cell cycle progression in myoblasts.
Collapse
Affiliation(s)
- He Li
- College of Health Sciences, Jiangsu Normal University, Xuzhou 221116, Jiangsu, P.R. China. .,Jiangsu Province Engineering Research Center of Cardiovascular Drugs Targeting Endotheial Cells, Xuzhou 221116, Jiangsu, P.R. China
| | - Kaifang Guan
- College of Health Sciences, Jiangsu Normal University, Xuzhou 221116, Jiangsu, P.R. China. .,School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150090, Heilongjiang, P.R. China
| | - DanDan Liu
- College of Health Sciences, Jiangsu Normal University, Xuzhou 221116, Jiangsu, P.R. China.
| | - Min Liu
- School of Chemistry and Chemical Engineering, Guangxi University for Nationalities, Nanning 530008, Guangxi, P.R. China
| |
Collapse
|
16
|
Experimental models of lipid overload and their relevance in understanding skeletal muscle insulin resistance and pathological changes in mitochondrial oxidative capacity. Biochimie 2021; 196:182-193. [PMID: 34563603 DOI: 10.1016/j.biochi.2021.09.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 08/30/2021] [Accepted: 09/20/2021] [Indexed: 02/08/2023]
Abstract
It remains essential to decipher some of the pathological mechanisms that link obesity with deteriorating human health. Insulin resistance, due to enhanced free fatty acid substrate delivery, results in disrupted glucose homeostasis and altered mitochondrial oxidative capacity, which is a characteristic feature of an obese state. In fact, as a major site for regulating glucose homeostasis and energy production in response to insulin, the skeletal muscle has become an interesting target tissue to understand the impact of lipid overload on the development of insulin resistance and impaired mitochondrial respiratory function. In addition to systematically retrieving the discussed data, the current review brings an essential perspective in understanding the relevance of experimental models of lipid overload such as high fat diets in understanding the pathological link between insulin resistance and pathological changes in mitochondrial oxidative capacity. Importantly, inclusion of evidence from transgenic model highlights some of the unique molecular targets that are implicated in the development of insulin resistance and inefficient mitochondrial respiration processes within an obese state. Importantly, saturation with lipid products such as ceramides and diacylglycerols, especially within the skeletal muscle, appears to be instrumental in paving the path leading to worsening of metabolic complications. These metabolic consequences mostly interfere with the efficiency of the mitochondrial electron transport chain, leading to overproduction of toxic reactive oxygen species. Therefore, therapeutic agents that reverse the effects of lipid overload by improving insulin sensitivity and mitochondrial oxidative capacity are crucial for the management or even treatment of metabolic diseases.
Collapse
|
17
|
Human Mitoribosome Biogenesis and Its Emerging Links to Disease. Int J Mol Sci 2021; 22:ijms22083827. [PMID: 33917098 PMCID: PMC8067846 DOI: 10.3390/ijms22083827] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 12/20/2022] Open
Abstract
Mammalian mitochondrial ribosomes (mitoribosomes) synthesize a small subset of proteins, which are essential components of the oxidative phosphorylation machinery. Therefore, their function is of fundamental importance to cellular metabolism. The assembly of mitoribosomes is a complex process that progresses through numerous maturation and protein-binding events coordinated by the actions of several assembly factors. Dysregulation of mitoribosome production is increasingly recognized as a contributor to metabolic and neurodegenerative diseases. In recent years, mutations in multiple components of the mitoribosome assembly machinery have been associated with a range of human pathologies, highlighting their importance to cell function and health. Here, we provide a review of our current understanding of mitoribosome biogenesis, highlighting the key factors involved in this process and the growing number of mutations in genes encoding mitoribosomal RNAs, proteins, and assembly factors that lead to human disease.
Collapse
|
18
|
Ramírez-Camacho I, García-Niño W, Flores-García M, Pedraza-Chaverri J, Zazueta C. Alteration of mitochondrial supercomplexes assembly in metabolic diseases. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165935. [DOI: 10.1016/j.bbadis.2020.165935] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 08/10/2020] [Accepted: 08/11/2020] [Indexed: 01/05/2023]
|
19
|
Al-Attar R, Storey KB. RAGE against the stress: Mitochondrial suppression in hypometabolic hearts. Gene 2020; 761:145039. [PMID: 32777527 DOI: 10.1016/j.gene.2020.145039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/19/2020] [Accepted: 08/04/2020] [Indexed: 12/22/2022]
Abstract
The wood frog (Rana sylvatica) can tolerate full body freezing in winter. As a protective response, wood frogs dehydrate their cells and accumulate large quantities of glucose as an intracellular cryoprotectant. Freezing causes ischemia since blood delivery to organs is interrupted. Fascinatingly, wood frogs can tolerate dehydration, extreme hyperglycemia, and anoxia independently of freezing. In response to low oxygen levels, wood frogs strategically reduce their metabolic rates and allocate the finite amount of intracellular fuel available to pro-survival processes while reducing or interrupting all others. In this study, the involvement of advanced glycation end products (AGEs) and the high mobility group box 1 (HMGB1) protein in activating RAGE (AGE receptor) were investigated. The results show that freezing, anoxia and dehydration induced the expression of total HMGB1 and its acetylation in the heart. RAGE levels were induced in response to all stress conditions, which resulted in differential regulation of the ETS1 transcription factor. While the nuclear localization of total ETS1 was not affected, the DNA binding activity of total and its active form increased in response to freezing and dehydration but not in response to anoxia. Current results indicate that ETS1 acts as a transcriptional activator for peroxiredoxin 1 in response to freezing but acts as a transcriptional repressor of several nuclear-encoded mitochondrial genes in response to all stresses. Altogether, current results show that the HMGB1/RAGE axis may activate ETS1 and that this activation could result in both transcriptional activation and/or repression in a stress-dependent manner.
Collapse
Affiliation(s)
- Rasha Al-Attar
- Institude of Biochemistry and Department of Biology, Carleton University, Ottawa, ON K1S-5B6, Canada
| | - Kenneth B Storey
- Institude of Biochemistry and Department of Biology, Carleton University, Ottawa, ON K1S-5B6, Canada.
| |
Collapse
|
20
|
Nowinski SM, Solmonson A, Rusin SF, Maschek JA, Bensard CL, Fogarty S, Jeong MY, Lettlova S, Berg JA, Morgan JT, Ouyang Y, Naylor BC, Paulo JA, Funai K, Cox JE, Gygi SP, Winge DR, DeBerardinis RJ, Rutter J. Mitochondrial fatty acid synthesis coordinates oxidative metabolism in mammalian mitochondria. eLife 2020; 9:58041. [PMID: 32804083 PMCID: PMC7470841 DOI: 10.7554/elife.58041] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 08/15/2020] [Indexed: 12/13/2022] Open
Abstract
Cells harbor two systems for fatty acid synthesis, one in the cytoplasm (catalyzed by fatty acid synthase, FASN) and one in the mitochondria (mtFAS). In contrast to FASN, mtFAS is poorly characterized, especially in higher eukaryotes, with the major product(s), metabolic roles, and cellular function(s) being essentially unknown. Here we show that hypomorphic mtFAS mutant mouse skeletal myoblast cell lines display a severe loss of electron transport chain (ETC) complexes and exhibit compensatory metabolic activities including reductive carboxylation. This effect on ETC complexes appears to be independent of protein lipoylation, the best characterized function of mtFAS, as mutants lacking lipoylation have an intact ETC. Finally, mtFAS impairment blocks the differentiation of skeletal myoblasts in vitro. Together, these data suggest that ETC activity in mammals is profoundly controlled by mtFAS function, thereby connecting anabolic fatty acid synthesis with the oxidation of carbon fuels.
Collapse
Affiliation(s)
| | - Ashley Solmonson
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, United States
| | - Scott F Rusin
- Department of Cell Biology, Harvard University School of Medicine, Boston, United States
| | - J Alan Maschek
- Diabetes & Metabolism Research Center, Salt Lake City, United States.,Department of Nutrition and Integrative Physiology, Salt Lake City, United States.,Metabolomics, Proteomics and Mass Spectrometry Core Research Facilities University of Utah, Salt Lake City, United States
| | | | - Sarah Fogarty
- Department of Biochemistry, Salt Lake City, United States.,Howard Hughes Medical Institute, Salt Lake City, United States
| | - Mi-Young Jeong
- Department of Biochemistry, Salt Lake City, United States
| | | | - Jordan A Berg
- Department of Biochemistry, Salt Lake City, United States
| | - Jeffrey T Morgan
- Department of Biochemistry, Salt Lake City, United States.,Howard Hughes Medical Institute, Salt Lake City, United States
| | - Yeyun Ouyang
- Department of Biochemistry, Salt Lake City, United States
| | - Bradley C Naylor
- Metabolomics, Proteomics and Mass Spectrometry Core Research Facilities University of Utah, Salt Lake City, United States
| | - Joao A Paulo
- Department of Cell Biology, Harvard University School of Medicine, Boston, United States
| | - Katsuhiko Funai
- Diabetes & Metabolism Research Center, Salt Lake City, United States
| | - James E Cox
- Department of Biochemistry, Salt Lake City, United States.,Diabetes & Metabolism Research Center, Salt Lake City, United States.,Metabolomics, Proteomics and Mass Spectrometry Core Research Facilities University of Utah, Salt Lake City, United States
| | - Steven P Gygi
- Department of Cell Biology, Harvard University School of Medicine, Boston, United States
| | - Dennis R Winge
- Department of Biochemistry, Salt Lake City, United States.,Diabetes & Metabolism Research Center, Salt Lake City, United States.,Department of Internal Medicine, Salt Lake City, United States
| | - Ralph J DeBerardinis
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, United States.,Howard Hughes Medical Institute, Salt Lake City, United States
| | - Jared Rutter
- Department of Biochemistry, Salt Lake City, United States.,Diabetes & Metabolism Research Center, Salt Lake City, United States.,Howard Hughes Medical Institute, Salt Lake City, United States
| |
Collapse
|
21
|
Kastaniotis AJ, Autio KJ, R Nair R. Mitochondrial Fatty Acids and Neurodegenerative Disorders. Neuroscientist 2020; 27:143-158. [PMID: 32644907 DOI: 10.1177/1073858420936162] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Fatty acids in mitochondria, in sensu stricto, arise either as β-oxidation substrates imported via the carnitine shuttle or through de novo synthesis by the mitochondrial fatty acid synthesis (mtFAS) pathway. Defects in mtFAS or processes involved in the generation of the mtFAS product derivative lipoic acid (LA), including iron-sulfur cluster synthesis required for functional LA synthase, have emerged only recently as etiology for neurodegenerative disease. Intriguingly, mtFAS deficiencies very specifically affect CNS function, while LA synthesis and attachment defects have a pleiotropic presentation beyond neurodegeneration. Typical mtFAS defect presentations include optical atrophy, as well as basal ganglia defects associated with dystonia. The phenotype display of patients with mtFAS defects can resemble the presentation of disorders associated with coenzyme A (CoA) synthesis. A recent publication links these processes together based on the requirement of CoA for acyl carrier protein maturation. MtFAS defects, CoA synthesis- as well as Fe-S cluster-deficiencies share lack of LA as a common symptom.
Collapse
Affiliation(s)
| | - Kaija J Autio
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Remya R Nair
- Mammalian Genetics Unit, MRC Harwell Institute, Oxfordshire, UK
| |
Collapse
|