1
|
Mandujano-Lázaro G, Torres-Rojas MF, Ramírez-Moreno E, Marchat LA. Virtual screening combined with molecular docking for the !identification of new anti-adipogenic compounds. Sci Prog 2025; 108:368504251320313. [PMID: 39936374 PMCID: PMC11815789 DOI: 10.1177/00368504251320313] [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] [Indexed: 02/13/2025]
Abstract
Obesity is an important risk factor for diabetes, cardiovascular diseases, and cancer, reducing the quality of life and expectancy of millions of people. Consequently, obesity has turned into one of the most health public problems worldwide, which highlights the urgent need for new and safe treatments. Obesity is mainly related to excessive fat accumulation; therefore, proteins participating in white adipose tissue increase and dysfunction are considered pertinent and attractive targets for developing new methods that can help with body weight control. In this context, virtual screening of libraries containing a large number of molecules represents a valuable strategy for the identification of potential anti-adipogenic compounds with reduced costs and time production. Here, we review the scientific literature about the prediction of new ligands of specific proteins through molecular docking and virtual screening of chemical libraries, with the aim of proposing new potential anti-adipogenic molecules. First, we present the targets related to adipogenesis and adipocyte functions that were selected for the following studies: PPARγ, Crif1, SIRT1, ERβ, PC1, FTO, Mss51, and FABP4. Then, we describe the obtention of new ligands according to the characteristics of the virtual screening approach, i.e. a structure-based drug design (SBDD) or a ligand-based drug design (LBDD). Finally, the critical analysis of these computational strategies and the corresponding results points out the necessity of combining computational and in vitro or in vivo assays for the identification of effective new anti-adipogenic molecules for obesity control. It also evidences that translating molecular docking and virtual screening results into successful drug candidates for adipogenesis and obesity control remains a huge challenge.
Collapse
Affiliation(s)
- Gilberto Mandujano-Lázaro
- Laboratorio de Biomedicina Molecular 2, ENMH, Instituto Politécnico Nacional, Ciudad de México, México
| | - María F Torres-Rojas
- Laboratorio de Biomedicina Molecular 2, ENMH, Instituto Politécnico Nacional, Ciudad de México, México
| | - Esther Ramírez-Moreno
- Laboratorio de Biomedicina Molecular 2, ENMH, Instituto Politécnico Nacional, Ciudad de México, México
| | - Laurence A Marchat
- Laboratorio de Biomedicina Molecular 2, ENMH, Instituto Politécnico Nacional, Ciudad de México, México
| |
Collapse
|
2
|
Wang F, Huynh PM, An YA. Mitochondrial Function and Dysfunction in White Adipocytes and Therapeutic Implications. Compr Physiol 2024; 14:5581-5640. [PMID: 39382163 DOI: 10.1002/cphy.c230009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
For a long time, white adipocytes were thought to function as lipid storages due to the sizeable unilocular lipid droplet that occupies most of their space. However, recent discoveries have highlighted the critical role of white adipocytes in maintaining energy homeostasis and contributing to obesity and related metabolic diseases. These physiological and pathological functions depend heavily on the mitochondria that reside in white adipocytes. This article aims to provide an up-to-date overview of the recent research on the function and dysfunction of white adipocyte mitochondria. After briefly summarizing the fundamental aspects of mitochondrial biology, the article describes the protective role of functional mitochondria in white adipocyte and white adipose tissue health and various roles of dysfunctional mitochondria in unhealthy white adipocytes and obesity. Finally, the article emphasizes the importance of enhancing mitochondrial quantity and quality as a therapeutic avenue to correct mitochondrial dysfunction, promote white adipocyte browning, and ultimately improve obesity and its associated metabolic diseases. © 2024 American Physiological Society. Compr Physiol 14:5581-5640, 2024.
Collapse
Affiliation(s)
- Fenfen Wang
- Department of Anesthesiology, Critical Care, and Pain Medicine, Center for Perioperative Medicine, McGovern Medical School, UT Health Science Center at Houston, Houston, Texas, USA
| | - Phu M Huynh
- Department of Anesthesiology, Critical Care, and Pain Medicine, Center for Perioperative Medicine, McGovern Medical School, UT Health Science Center at Houston, Houston, Texas, USA
| | - Yu A An
- Department of Anesthesiology, Critical Care, and Pain Medicine, Center for Perioperative Medicine, McGovern Medical School, UT Health Science Center at Houston, Houston, Texas, USA
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, McGovern Medical School, UT Health Science Center at Houston, Houston, Texas, USA
- Department of Biochemistry and Molecular Biology, McGovern Medical School, UT Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
3
|
Cai J, Wang F, Shao M. The Emerging Importance of Mitochondria in White Adipocytes: Neither Last nor Least. Endocrinol Metab (Seoul) 2023; 38:493-503. [PMID: 37816498 PMCID: PMC10613775 DOI: 10.3803/enm.2023.1813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 09/05/2023] [Accepted: 09/14/2023] [Indexed: 10/12/2023] Open
Abstract
The growing recognition of mitochondria's crucial role in the regulation of white adipose tissue remodeling and energy balance underscores its significance. The marked metabolic diversity of mitochondria provides the molecular and cellular foundation for enabling adipose tissue plasticity in response to various metabolic cues. Effective control of mitochondrial function at the cellular level, not only in thermogenic brown and beige adipocytes but also in energy-storing white adipocytes, exerts a profound influence on adipose homeostasis. Furthermore, mitochondria play a pivotal role in intercellular communication within adipose tissue via production of metabolites with signaling properties. A more comprehensive understanding of mitochondrial regulation within white adipocytes will empower the development of targeted and efficacious strategies to enhance adipose function, leading to advancements in overall metabolic health.
Collapse
Affiliation(s)
- Juan Cai
- CAS Key Laboratory of Molecular Virology and Immunology, The Center for Microbes, Development and Health, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Fenfen Wang
- Department of Anesthesiology, Critical Care and Pain Medicine, Center for Perioperative Medicine, McGovern Medical School, UT Health Science Center at Houston, Houston, TX, USA
| | - Mengle Shao
- CAS Key Laboratory of Molecular Virology and Immunology, The Center for Microbes, Development and Health, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
4
|
Christensen IB, Blom I, Dohlmann TL, Finger F, Helge JW, Gerhart-Hines Z, Dela F, Larsen S. Effect of Simvastatin Treatment on Mitochondrial Function and Inflammatory Status of Human White Adipose Tissue. J Clin Endocrinol Metab 2023; 108:e916-e922. [PMID: 37161534 DOI: 10.1210/clinem/dgad259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 02/20/2023] [Accepted: 05/08/2023] [Indexed: 05/11/2023]
Abstract
BACKGROUND Statin therapy has shown pleiotropic effects affecting both mitochondrial function and inflammatory status. However, few studies have investigated the concurrent effects of statin exposure on mitochondrial function and inflammatory status in human subcutaneous white adipose tissue. OBJECTIVES In a cross-sectional study, we investigated the effects of simvastatin on mitochondrial function and inflammatory status in subcutaneous white adipose tissue of 55 human participants: 38 patients (19 females/19 males) in primary prevention with simvastatin (> 40 mg/d, > 3 mo) and 17 controls (9 females/8 males) with elevated plasma cholesterol. The 2 groups were matched on age, body mass index, and maximal oxygen consumption. METHODS Anthropometrics and fasting biochemical characteristics were measured. Mitochondrial respiratory capacity was assessed in white adipose tissue by high-resolution respirometry. Subcutaneous white adipose tissue expression of the inflammatory markers IL-6, chemokine (C-C motif) ligand 2 (CCL2), CCL-5, tumor necrosis factor-α, IL-10, and IL-4 was analyzed by quantitative PCR. RESULTS Simvastatin-treated patients showed lower plasma cholesterol (P < .0001), low-density lipoprotein (P < .0001), and triglyceride levels (P = .0116) than controls. Simvastatin-treated patients had a lower oxidative phosphorylation capacity of mitochondrial complex II (P = .0001 when normalized to wet weight, P < .0001 when normalized to citrate synthase activity [intrinsic]), and a lower intrinsic mitochondrial electron transport system capacity (P = .0004). Simvastatin-treated patients showed higher IL-6 expression than controls (P = .0202). CONCLUSION Simvastatin treatment was linked to mitochondrial respiratory capacity in human subcutaneous white adipose tissue, but no clear link was found between statin exposure, respiratory changes, and inflammatory status of adipose tissue.
Collapse
Affiliation(s)
- Ida Bager Christensen
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Ida Blom
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Tine Lovsø Dohlmann
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
- Department of Epidemiology Research, Statens Serum Institut, 2300 Copenhagen S, Denmark
| | - Fabian Finger
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Jørn W Helge
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Zachary Gerhart-Hines
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Flemming Dela
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
- Department of Geriatrics, Bispebjerg-Frederiksberg University Hospital, 2400 Copenhagen NV, Denmark
| | - Steen Larsen
- Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
- Clinical Research Centre, Medical University of Bialystok, 15-089 Białystok, Poland
| |
Collapse
|
5
|
Liu J, Lu W, Yan D, Guo J, Zhou L, Shi B, Su X. Mitochondrial respiratory complex I deficiency inhibits brown adipogenesis by limiting heme regulation of histone demethylation. Mitochondrion 2023; 72:22-32. [PMID: 37451354 DOI: 10.1016/j.mito.2023.07.004] [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: 11/14/2022] [Revised: 06/13/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
Mitochondrial functions play a crucial role in determining the metabolic and thermogenic status of brown adipocytes. Increasing evidence reveals that the mitochondrial oxidative phosphorylation (OXPHOS) system plays an important role in brown adipogenesis, but the mechanistic insights are limited. Herein, we explored the potential metabolic mechanisms leading to OXPHOS regulation of brown adipogenesis in pharmacological and genetic models of mitochondrial respiratory complex I deficiency. OXPHOS deficiency inhibits brown adipogenesis through disruption of the brown adipogenic transcription circuit without affecting ATP levels. Neither blockage of calcium signaling nor antioxidant treatment can rescue the suppressed brown adipogenesis. Metabolomics analysis revealed a decrease in levels of tricarboxylic acid cycle intermediates and heme. Heme supplementation specifically enhances respiratory complex I activity without affecting complex II and partially reverses the inhibited brown adipogenesis by OXPHOS deficiency. Moreover, the regulation of brown adipogenesis by the OXPHOS-heme axis may be due to the suppressed histone methylation status by increasing histone demethylation. In summary, our findings identified a heme-sensing retrograde signaling pathway that connects mitochondrial OXPHOS to the regulation of brown adipocyte differentiation and metabolic functions.
Collapse
Affiliation(s)
- Jingjing Liu
- Department of Biochemistry and Molecular Biology, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Wen Lu
- Department of Endocrinology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Dongyue Yan
- Department of Biochemistry and Molecular Biology, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Junyuan Guo
- Department of Biochemistry and Molecular Biology, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Li Zhou
- Department of Nutrition, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Bimin Shi
- Department of Endocrinology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Xiong Su
- Department of Biochemistry and Molecular Biology, Suzhou Medical College of Soochow University, Suzhou 215123, China.
| |
Collapse
|
6
|
Hasegawa K, Sakamaki Y, Tamaki M, Wakino S. PCK1 Protects against Mitoribosomal Defects in Diabetic Nephropathy in Mouse Models. J Am Soc Nephrol 2023; 34:1343-1365. [PMID: 37199399 PMCID: PMC10400109 DOI: 10.1681/asn.0000000000000156] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 05/09/2023] [Indexed: 05/19/2023] Open
Abstract
SIGNIFICANCE STATEMENT Renal gluconeogenesis plays an important role in the pathogenesis of diabetic nephropathy (DN). Proximal tubular phosphoenolpyruvate carboxykinase1 (PEPCK1) is the rate-limiting enzyme in gluconeogenesis. However, the functions of PEPCK1 have not been elucidated. We describe the novel role of PEPCK1 as a mitoribosomal protector using Pck1 transgenic (TG) mice and knockout mice. Pck1 blocks excessive glycolysis by suppressing the upregulation of excess HK2 (the rate-limiting enzyme of glycolysis). Notably, Pck1 overexpression retains mitoribosomal function and suppresses renal fibrosis. The renal and mitoribosomal protective roles of Pck1 may provide important clues for understanding DN pathogenesis and provide novel therapeutic targets. BACKGROUND Phosphoenolpyruvate carboxykinase (PEPCK) is part of the gluconeogenesis pathway, which maintains fasting glucose levels and affects renal physiology. PEPCK consists of two isoforms-PEPCK1 and PEPCK2-that the Pck1 and Pck2 genes encode. Gluconeogenesis increases in diabetic nephropathy (DN), escalating fasting and postprandial glucose levels. Sodium-glucose cotransporter-2 inhibitors increase hepatic and renal gluconeogenesis. We used genetically modified mice to investigate whether renal gluconeogenesis and Pck1 activity are renoprotective in DN. METHODS We investigated the expression of Pck1 in the proximal tubule (PTs) of streptozotocin (STZ)-treated diabetic mice. We studied the phenotypic changes in PT-specific transgenic (TG) mice and PT-specific Pck1 conditional knockout (CKO) mice. RESULTS The expression of Pck1 in PTs was downregulated in STZ-treated diabetic mice when they exhibited albuminuria. TG mice overexpressing Pck1 had improved albuminuria, concomitant with the mitigation of PT cell apoptosis and deposition of peritubular type IV collagen. Moreover, CKO mice exhibited PT cell apoptosis and type IV collagen deposition, findings also observed in STZ-treated mice. Renal fibrotic changes in CKO mice were associated with increasing defects in mitochondrial ribosomes (mitoribosomes). The TG mice were protected against STZ-induced mitoribosomal defects. CONCLUSION PCK1 preserves mitoribosomal function and may play a novel protective role in DN.
Collapse
Affiliation(s)
- Kazuhiro Hasegawa
- Department of Nephrology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Yusuke Sakamaki
- Department of Internal Medicine, Tokyo Dental College, Ichikawa General Hospital, Chiba, Japan
| | - Masanori Tamaki
- Department of Nephrology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Shu Wakino
- Department of Nephrology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| |
Collapse
|
7
|
Liu Y, Liu Z, Liang J, Sun C. ILC2s control obesity by regulating energy homeostasis and browning of white fat. Int Immunopharmacol 2023; 120:110272. [PMID: 37210911 DOI: 10.1016/j.intimp.2023.110272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 04/27/2023] [Accepted: 04/29/2023] [Indexed: 05/23/2023]
Abstract
Innate lymphoid cells (ILCs) have been a hot topic in recent research, they are widely distributed in vivo and play an important role in different tissues. The important role of group 2 innate lymphoid cells (ILC2s) in the conversion of white fat into beige fat has attracted widespread attention. Studies have shown that ILC2s regulate adipocyte differentiation and lipid metabolism. This article reviews the types and functions of ILCs, focusing on the relationship between differentiation, development and function of ILC2s, and elaborates on the relationship between peripheral ILC2s and browning of white fat and body energy homeostasis. This has important implications for the future treatment of obesity and related metabolic diseases.
Collapse
Affiliation(s)
- Yuexia Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China.
| | - Zunhai Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China.
| | - Juntong Liang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China.
| | - Chao Sun
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China.
| |
Collapse
|
8
|
Xu X, Zhao L, Terry PD, Chen J. Reciprocal Effect of Environmental Stimuli to Regulate the Adipogenesis and Osteogenesis Fate Decision in Bone Marrow-Derived Mesenchymal Stem Cells (BM-MSCs). Cells 2023; 12:1400. [PMID: 37408234 PMCID: PMC10216952 DOI: 10.3390/cells12101400] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/02/2023] [Accepted: 05/12/2023] [Indexed: 07/07/2023] Open
Abstract
Mesenchymal stem cells derived from bone marrow (BM-MSCs) can differentiate into adipocytes and osteoblasts. Various external stimuli, including environmental contaminants, heavy metals, dietary, and physical factors, are shown to influence the fate decision of BM-MSCs toward adipogenesis or osteogenesis. The balance of osteogenesis and adipogenesis is critical for the maintenance of bone homeostasis, and the interruption of BM-MSCs lineage commitment is associated with human health issues, such as fracture, osteoporosis, osteopenia, and osteonecrosis. This review focuses on how external stimuli shift the fate of BM-MSCs towards adipogenesis or osteogenesis. Future studies are needed to understand the impact of these external stimuli on bone health and elucidate the underlying mechanisms of BM-MSCs differentiation. This knowledge will inform efforts to prevent bone-related diseases and develop therapeutic approaches to treat bone disorders associated with various pathological conditions.
Collapse
Affiliation(s)
- Xinyun Xu
- Department of Nutrition, The University of Tennessee, Knoxville, TN 37996, USA
| | - Ling Zhao
- Department of Nutrition, The University of Tennessee, Knoxville, TN 37996, USA
| | - Paul D. Terry
- Department of Medicine, Graduate School of Medicine, The University of Tennessee, Knoxville, TN 37920, USA;
| | - Jiangang Chen
- Department of Public Health, The University of Tennessee, Knoxville, TN 37996, USA
| |
Collapse
|
9
|
Piao S, Lee I, Kim S, Park H, Nagar H, Choi SJ, Vu GH, Kim M, Lee EO, Jeon BH, Kim DW, Seo Y, Kim CS. CRIF1 siRNA-Encapsulated PLGA Nanoparticles Suppress Tumor Growth in MCF-7 Human Breast Cancer Cells. Int J Mol Sci 2023; 24:ijms24087453. [PMID: 37108616 PMCID: PMC10138627 DOI: 10.3390/ijms24087453] [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: 03/22/2023] [Revised: 04/11/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
Mitochondrial oxidative phosphorylation (OXPHOS) system dysfunction in cancer cells has been exploited as a target for anti-cancer therapeutic intervention. The downregulation of CR6-interacting factor 1 (CRIF1), an essential mito-ribosomal factor, can impair mitochondrial function in various cell types. In this study, we investigated whether CRIF1 deficiency induced by siRNA and siRNA nanoparticles could suppress MCF-7 breast cancer growth and tumor development, respectively. Our results showed that CRIF1 silencing decreased the assembly of mitochondrial OXPHOS complexes I and II, which induced mitochondrial dysfunction, mitochondrial reactive oxygen species (ROS) production, mitochondrial membrane potential depolarization, and excessive mitochondrial fission. CRIF1 inhibition reduced p53-induced glycolysis and apoptosis regulator (TIGAR) expression, as well as NADPH synthesis, leading to additional increases in ROS production. The downregulation of CRIF1 suppressed cell proliferation and inhibited cell migration through the induction of G0/G1 phase cell cycle arrest in MCF-7 breast cancer cells. Similarly, the intratumoral injection of CRIF1 siRNA-encapsulated PLGA nanoparticles inhibited tumor growth, downregulated the assembly of mitochondrial OXPHOS complexes I and II, and induced the expression of cell cycle protein markers (p53, p21, and p16) in MCF-7 xenograft mice. Thus, the inhibition of mitochondrial OXPHOS protein synthesis through CRIF1 deletion destroyed mitochondrial function, leading to elevated ROS levels and inducing antitumor effects in MCF-7 cells.
Collapse
Affiliation(s)
- Shuyu Piao
- Department of Physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Ikjun Lee
- Department of Physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Seonhee Kim
- Department of Physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Hyewon Park
- Department of Anatomy and Cell Biology & Medical Science, School of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Harsha Nagar
- Department of Physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Su-Jeong Choi
- Department of Physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Giang-Huong Vu
- Department of Physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Minsoo Kim
- Department of Physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Eun-Ok Lee
- Department of Physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Byeong-Hwa Jeon
- Department of Physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Dong Woon Kim
- Department of Anatomy and Cell Biology & Medical Science, School of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Youngduk Seo
- Department of Nuclear Medicine, Chungnam National University Sejong Hospital, Sejong 30099, Republic of Korea
| | - Cuk-Seong Kim
- Department of Physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| |
Collapse
|
10
|
Hwang B, Kwon MG, Cho MJ, Lee NK, Lee J, Lee JW, Oh KJ, Bae KH, Hwang JH, Min JK, Park JG. Hepatic PTP4A1 ameliorates high-fat diet-induced hepatosteatosis and hyperglycemia by the activation of the CREBH/FGF21 axis. Theranostics 2023; 13:1076-1090. [PMID: 36793871 PMCID: PMC9925322 DOI: 10.7150/thno.79434] [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: 09/30/2022] [Accepted: 01/18/2023] [Indexed: 02/04/2023] Open
Abstract
Precise regulation of kinases and phosphatases is crucial for human metabolic homeostasis. This study aimed to investigate the roles and molecular mechanisms of protein tyrosine phosphatase type IVA1 (PTP4A1) in regulating hepatosteatosis and glucose homeostasis. Method: Ptp4a1-/- mice, adeno-associated virus encoding Ptp4a1 under liver-specific promoter, adenovirus encoding Fgf21, and primary hepatocytes were used to evaluate PTP4A1-mediated regulation in the hepatosteatosis and glucose homeostasis. Glucose tolerance test, insulin tolerance test, 2-deoxyglucose uptake assay, and hyperinsulinemic-euglycemic clamp were performed to estimate glucose homeostasis in mice. The staining, including oil red O, hematoxylin & eosin, and BODIPY, and biochemical analysis for hepatic triglycerides were performed to assess hepatic lipids. Luciferase reporter assays, immunoprecipitation, immunoblots, quantitative real-time polymerase chain reaction, and immunohistochemistry staining were conducted to explore the underlying mechanism. Results: Here, we found that deficiency of PTP4A1 aggravated glucose homeostasis and hepatosteatosis in mice fed a high-fat (HF) diet. Increased lipid accumulation in hepatocytes of Ptp4a1-/- mice reduced the level of glucose transporter 2 on the plasma membrane of hepatocytes leading to a diminution of glucose uptake. PTP4A1 prevented hepatosteatosis by activating the transcription factor cyclic adenosine monophosphate-responsive element-binding protein H (CREBH)/fibroblast growth factor 21 (FGF21) axis. Liver-specific PTP4A1 or systemic FGF21 overexpression in Ptp4a1-/- mice fed an HF diet restored the disorder of hepatosteatosis and glucose homeostasis. Finally, liver-specific PTP4A1 expression ameliorated an HF diet-induced hepatosteatosis and hyperglycemia in wild-type mice. Conclusions: Hepatic PTP4A1 is critical for regulating hepatosteatosis and glucose homeostasis by activating the CREBH/FGF21 axis. Our current study provides a novel function of PTP4A1 in metabolic disorders; hence, modulating PTP4A1 may be a potential therapeutic strategy against hepatosteatosis-related diseases.
Collapse
Affiliation(s)
- Byungtae Hwang
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Min-Gi Kwon
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea.,Department of Bioscience, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Min Ji Cho
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Nam-Kyung Lee
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Jangwook Lee
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Jeong Woong Lee
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Kyoung-Jin Oh
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Kwang-Hee Bae
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Jung Hwan Hwang
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Jeong-Ki Min
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea.,Department of Bioscience, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Jong-Gil Park
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea.,Department of Bioscience, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| |
Collapse
|
11
|
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: 4] [Impact Index Per Article: 1.3] [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
|
12
|
Park JS, Yang S, Hwang SH, Choi J, Kwok SK, Kong YY, Youn J, Cho ML, Park SH. B cell-specific deletion of Crif1 drives lupus-like autoimmunity by activation of IL-17, IL-6, and pathogenic Tfh cells. Arthritis Rheumatol 2022; 74:1211-1222. [PMID: 35166061 DOI: 10.1002/art.42091] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 12/30/2021] [Accepted: 02/09/2022] [Indexed: 11/09/2022]
Abstract
OBJECTIVE CR6-interacting factor 1 (Crif1) is a nuclear transcriptional regulator and a mitochondrial inner membrane protein; however, its functions in B lymphocytes have been poorly defined. In this study, we investigated the effects of Crif1 on B-cell metabolic regulation, cell function, and autoimmune diseases. METHODS Using mice with B cell-specific deletion of Crif1 (Crif1ΔCD19 ), we assessed the relevance of Crif1 function for lupus disease parameters including anti-double-stranded DNA, cytokines, and kidney pathology. RNA sequencing was performed on B cells from Crif1ΔCD19 mice. The phenotypic and metabolic changes in immune cells were evaluated in Crif1ΔCD19 mice. Roquinsan/+ mice crossed with Crif1ΔCD19 mice were monitored to assess the functionality of Crif1-deficient B cells in lupus development. RESULTS Crif1ΔCD19 mice showed an autoimmune lupus-like phenotype, including high levels of autoantibodies to double-stranded DNA and severe lupus nephritis with increased mesangial hypercellularity. While loss of Crif1 in B cells showed impaired mitochondrial oxidative function, Crif1-deficient B cells promoted the production of IL-17 and IL-6 and was more potent in helping T cells develop into T follicular helper cells. In an autoimmune lupus mouse model, depletion of Crif1 in B cells exacerbated lupus severity and Crif1 overexpression prevented lupus development in Roquinsan/san mice. CONCLUSION These results showed that Crif1 was negatively correlated with disease severity, and overexpression of Crif1 ameliorated disease development. Our findings suggest that Crif1 is essential for preventing lupus development by maintaining B cell self-tolerance.
Collapse
Affiliation(s)
- Jin-Sil Park
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - SeungCheon Yang
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sun-Hee Hwang
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - JeongWon Choi
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seung-Ki Kwok
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Divison of Rheumatology, Department of Internal Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Young-Yun Kong
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Jeehee Youn
- Laboratory of Autoimmunology, Department of Anatomy and Cell Biology, College of Medicine, Hanyang University, Seoul, 04763, Korea
| | - Mi-La Cho
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Department of Medical Lifescience, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sung-Hwan Park
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Divison of Rheumatology, Department of Internal Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
13
|
Zhu Q, An YA, Scherer PE. Mitochondrial regulation and white adipose tissue homeostasis. Trends Cell Biol 2021; 32:351-364. [PMID: 34810062 DOI: 10.1016/j.tcb.2021.10.008] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/26/2021] [Accepted: 10/28/2021] [Indexed: 12/12/2022]
Abstract
The important role of mitochondria in the regulation of white adipose tissue (WAT) remodeling and energy balance is increasingly appreciated. The remarkable heterogeneity of the adipose tissue stroma provides a cellular basis to enable adipose tissue plasticity in response to various metabolic stimuli. Regulating mitochondrial function at the cellular level in adipocytes, in adipose progenitor cells (APCs), and in adipose tissue macrophages (ATMs) has a profound impact on adipose homeostasis. Moreover, mitochondria facilitate the cell-to-cell communication within WAT, as well as the crosstalk with other organs, such as the liver, the heart, and the pancreas. A better understanding of mitochondrial regulation in the diverse adipose tissue cell types allows us to develop more specific and efficient approaches to improve adipose function and achieve improvements in overall metabolic health.
Collapse
Affiliation(s)
- Qingzhang Zhu
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yu A An
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Philipp E Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Cell Biology, The University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
14
|
Lyu AR, Kim TH, Shin SA, Kim EH, Yu Y, Gajbhiye A, Kwon HC, Je AR, Huh YH, Park MJ, Park YH. Hearing Impairment in a Mouse Model of Diabetes Is Associated with Mitochondrial Dysfunction, Synaptopathy, and Activation of the Intrinsic Apoptosis Pathway. Int J Mol Sci 2021; 22:ijms22168807. [PMID: 34445504 PMCID: PMC8395823 DOI: 10.3390/ijms22168807] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 07/26/2021] [Accepted: 08/13/2021] [Indexed: 11/16/2022] Open
Abstract
Although previous studies continuously report an increased risk of hearing loss in diabetes patients, the impact of the disease on the inner ear remains unexplored. Herein, we examine the pathophysiology of diabetes-associated hearing impairment and cochlear synaptopathy in a mouse model of diabetes. Male B6.BKS(D)-Leprdb/J (db/db, diabetes) and heterozygote (db/+, control) mice were assigned into each experimental group (control vs. diabetes) based on the genotype and tested for hearing sensitivity every week from 6 weeks of age. Each cochlea was collected for histological and biological assays at 14 weeks of age. The diabetic mice exerted impaired hearing and a reduction in cochlear blood flow and C-terminal-binding protein 2 (CtBP2, a presynaptic ribbon marker) expression. Ultrastructural images revealed severely damaged mitochondria from diabetic cochlea accompanied by a reduction in Cytochrome c oxidase subunit 4 (COX4) and CR6-interacting factor 1 (CRIF1). The diabetic mice presented significantly decreased levels of platelet endothelial cell adhesion molecule (PECAM-1), B-cell lymphoma 2 (BCL-2), and procaspase-9, but not procaspase-8. Importantly, significant changes were not found in necroptotic programmed cell death markers (receptor-interacting serine/threonine-protein kinase 1, RIPK1; RIPK3; and mixed lineage kinase domain-like pseudokinase, MLKL) between the groups. Taken together, diabetic hearing loss is accompanied by synaptopathy, microangiopathy, damage to the mitochondrial structure/function, and activation of the intrinsic apoptosis pathway. Our results imply that mitochondrial dysfunction is deeply involved in diabetic hearing loss, and further suggests the potential benefits of therapeutic strategies targeting mitochondria.
Collapse
Affiliation(s)
- Ah-Ra Lyu
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, Chungnam National University, Daejeon 35015, Korea; (A.-R.L.); (E.-H.K.); (Y.Y.)
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Korea; (A.G.); (H.-C.K.)
| | - Tae-Hwan Kim
- Biomedical Convergence Research Center, Chungnam National University Hospital, Daejeon 35015, Korea;
| | - Sun-Ae Shin
- Brain Research Institute, College of Medicine, Chungnam National University, Daejeon 35015, Korea;
| | - Eung-Hyub Kim
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, Chungnam National University, Daejeon 35015, Korea; (A.-R.L.); (E.-H.K.); (Y.Y.)
| | - Yang Yu
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, Chungnam National University, Daejeon 35015, Korea; (A.-R.L.); (E.-H.K.); (Y.Y.)
| | - Akanksha Gajbhiye
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Korea; (A.G.); (H.-C.K.)
| | - Hyuk-Chan Kwon
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Korea; (A.G.); (H.-C.K.)
| | - A Reum Je
- Electron Microscopy Research Center, Korea Basic Science Institute, Cheongju 28119, Korea; (A.R.J.); (Y.H.H.)
| | - Yang Hoon Huh
- Electron Microscopy Research Center, Korea Basic Science Institute, Cheongju 28119, Korea; (A.R.J.); (Y.H.H.)
| | - Min Jung Park
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, Chungnam National University, Daejeon 35015, Korea; (A.-R.L.); (E.-H.K.); (Y.Y.)
- Brain Research Institute, College of Medicine, Chungnam National University, Daejeon 35015, Korea;
- Correspondence: (M.J.P.); (Y.-H.P.)
| | - Yong-Ho Park
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, Chungnam National University, Daejeon 35015, Korea; (A.-R.L.); (E.-H.K.); (Y.Y.)
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Korea; (A.G.); (H.-C.K.)
- Biomedical Convergence Research Center, Chungnam National University Hospital, Daejeon 35015, Korea;
- Brain Research Institute, College of Medicine, Chungnam National University, Daejeon 35015, Korea;
- Correspondence: (M.J.P.); (Y.-H.P.)
| |
Collapse
|
15
|
Petkevicius K, Bidault G, Virtue S, Newland SA, Dale M, Dugourd A, Saez-Rodriguez J, Mallat Z, Vidal-Puig A. Macrophage beta2-adrenergic receptor is dispensable for the adipose tissue inflammation and function. Mol Metab 2021; 48:101220. [PMID: 33774223 PMCID: PMC8086137 DOI: 10.1016/j.molmet.2021.101220] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/18/2021] [Accepted: 03/20/2021] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE Neuroimmune interactions between the sympathetic nervous system (SNS) and macrophages are required for the homeostasis of multiple tissues, including the adipose tissue. It has been proposed that the SNS maintains adipose tissue macrophages (ATMs) in an anti-inflammatory state via direct norepinephrine (NE) signaling to macrophages. This study aimed to investigate the physiological importance of this paradigm by utilizing a mouse model in which the adrenergic signaling from the SNS to macrophages, but not to other adipose tissue cells, was disrupted. METHODS We generated a macrophage-specific B2AR knockout mouse (Adrb2ΔLyz2) by crossing Adrb2fl/fl and Lyz2Cre/+ mice. We have previously shown that macrophages isolated from Adrb2ΔLyz2 animals do not respond to NE stimulation in vitro. Herein we performed a metabolic phenotyping of Adrb2ΔLyz2 mice on either chow or high-fat diet (HFD). We also assessed the adipose tissue function of Adrb2ΔLyz2 animals during fasting and cold exposure. Finally, we transplanted Adrb2ΔLyz2 bone marrow to low-density lipoprotein receptor (LDLR) knockout mice and investigated the development of atherosclerosis during Western diet feeding. RESULTS We demonstrated that SNS-associated ATMs have a transcriptional profile indicative of activated beta-2 adrenergic receptor (B2AR), the main adrenergic receptor isoform in myeloid cells. However, Adrb2ΔLyz2 mice have unaltered energy balance on a chow or HFD. Furthermore, Adrb2ΔLyz2 mice show similar levels of adipose tissue inflammation and function during feeding, fasting, or cold exposure, and develop insulin resistance during HFD at the same rate as controls. Finally, macrophage-specific B2AR deletion does not affect the development of atherosclerosis on an LDL receptor-null genetic background. CONCLUSIONS Overall, our data suggest that the SNS does not directly modulate the phenotype of adipose tissue macrophages in either lean mice or mouse models of cardiometabolic disease. Instead, sympathetic nerve activity exerts an indirect effect on adipose tissue macrophages through the modulation of adipocyte function.
Collapse
MESH Headings
- Adipocytes/metabolism
- Adipose Tissue, White/metabolism
- Animals
- Atherosclerosis/complications
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Bone Marrow Transplantation/methods
- Cells, Cultured
- Diet, High-Fat/adverse effects
- Diet, Western/adverse effects
- Disease Models, Animal
- Female
- Insulin Resistance/genetics
- Macrophages/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Obesity/complications
- Obesity/genetics
- Obesity/metabolism
- Panniculitis/genetics
- Panniculitis/metabolism
- Phenotype
- Receptors, Adrenergic, beta-2/genetics
- Receptors, Adrenergic, beta-2/metabolism
- Signal Transduction/genetics
- Sympathetic Nervous System/metabolism
Collapse
Affiliation(s)
- Kasparas Petkevicius
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, MDU MRC, Cambridge, United Kingdom.
| | - Guillaume Bidault
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, MDU MRC, Cambridge, United Kingdom
| | - Sam Virtue
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, MDU MRC, Cambridge, United Kingdom
| | - Stephen A Newland
- Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Martin Dale
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, MDU MRC, Cambridge, United Kingdom
| | - Aurelien Dugourd
- Joint Research Centre for Computational Biomedicine, Faculty of Medicine, RWTH Aachen University, Aachen, Germany; Institute for Computational Biomedicine, Heidelberg University, Faculty of Medicine & Heidelberg University Hospital, Heidelberg, Germany
| | - Julio Saez-Rodriguez
- Joint Research Centre for Computational Biomedicine, Faculty of Medicine, RWTH Aachen University, Aachen, Germany; Institute for Computational Biomedicine, Heidelberg University, Faculty of Medicine & Heidelberg University Hospital, Heidelberg, Germany
| | - Ziad Mallat
- Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Antonio Vidal-Puig
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, MDU MRC, Cambridge, United Kingdom; Wellcome Trust Sanger Institute, Hinxton, United Kingdom.
| |
Collapse
|
16
|
Na KR, Jeong JY, Shin JA, Chang YK, Suh KS, Lee KW, Choi DE. Mitochondrial Dysfunction in Podocytes Caused by CRIF1 Deficiency Leads to Progressive Albuminuria and Glomerular Sclerosis in Mice. Int J Mol Sci 2021; 22:ijms22094827. [PMID: 34063207 PMCID: PMC8124436 DOI: 10.3390/ijms22094827] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 04/26/2021] [Accepted: 04/26/2021] [Indexed: 12/25/2022] Open
Abstract
Recent studies have implicated mitochondrial disruption in podocyte dysfunction, which is a characteristic feature of primary and diabetic glomerular diseases. However, the mechanisms by which primary mitochondrial dysfunction in podocytes affects glomerular renal diseases are currently unknown. To investigate the role of mitochondrial oxidative phosphorylation (OxPhos) in podocyte dysfunction, glomerular function was examined in mice carrying a loss of function mutation of the gene encoding CR6-interacting factor-1 (CRIF1), which is essential for intramitochondrial production and the subsequent insertion of OxPhos polypeptides into the inner mitochondrial membrane. Homozygotic deficiency of CRIF1 in podocytes resulted in profound and progressive albuminuria from 3 weeks of age; the CRIF1-deficient mice also developed glomerular and tubulointerstitial lesions by 10 weeks of age. Furthermore, marked glomerular sclerosis and interstitial fibrosis were observed in homozygous CRIF1-deficient mice at 20 weeks of age. In cultured mouse podocytes, loss of CRIF1 resulted in OxPhos dysfunction and marked loss or abnormal aggregation of F-actin. These findings indicate that the OxPhos status determines the integrity of podocytes and their ability to maintain a tight barrier and control albuminuria. Analyses of the glomerular function of the podocyte-specific primary OxPhos dysfunction model mice demonstrate a link between podocyte mitochondrial dysfunction, progressive glomerular sclerosis, and tubulointerstitial diseases.
Collapse
Affiliation(s)
- Ki Ryang Na
- Department of Nephrology, Chungnam National University School of Medicine, Daejeon 35015, Korea; (K.R.N.); (J.Y.J.); (J.A.S.)
| | - Jin Young Jeong
- Department of Nephrology, Chungnam National University School of Medicine, Daejeon 35015, Korea; (K.R.N.); (J.Y.J.); (J.A.S.)
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Korea
| | - Jin Ah Shin
- Department of Nephrology, Chungnam National University School of Medicine, Daejeon 35015, Korea; (K.R.N.); (J.Y.J.); (J.A.S.)
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Korea
| | - Yoon-Kyung Chang
- Department of Nephrology, Daejeon St. Mary’s Hospital, Catholic University of Korea, Daejeon 34572, Korea;
| | - Kwang-Sun Suh
- Department of Pathology, Chungnam National University School of Medicine, Daejeon 35015, Korea;
| | - Kang Wook Lee
- Department of Nephrology, Chungnam National University School of Medicine, Daejeon 35015, Korea; (K.R.N.); (J.Y.J.); (J.A.S.)
- Correspondence: (K.W.L.); (D.E.C.)
| | - Dae Eun Choi
- Department of Nephrology, Chungnam National University School of Medicine, Daejeon 35015, Korea; (K.R.N.); (J.Y.J.); (J.A.S.)
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Korea
- Correspondence: (K.W.L.); (D.E.C.)
| |
Collapse
|
17
|
SIRT1 Activation Attenuates the Cardiac Dysfunction Induced by Endothelial Cell-Specific Deletion of CRIF1. Biomedicines 2021; 9:biomedicines9010052. [PMID: 33430144 PMCID: PMC7827654 DOI: 10.3390/biomedicines9010052] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/04/2021] [Accepted: 01/06/2021] [Indexed: 11/17/2022] Open
Abstract
The CR6-interacting factor1 (CRIF1) mitochondrial protein is indispensable for peptide synthesis and oxidative phosphorylation. Cardiomyocyte-specific deletion of CRIF1 showed impaired mitochondrial function and cardiomyopathy. We developed an endothelial cell-specific CRIF1 deletion mouse to ascertain whether dysfunctional endothelial CRIF1 influences cardiac function and is mediated by the antioxidant protein sirtuin 1 (SIRT1). We also examined the effect of the potent SIRT1 activator SRT1720 on cardiac dysfunction. Mice with endothelial cell-specific CRIF1 deletion showed an increased heart-to-body weight ratio, increased lethality, and markedly reduced fractional shortening of the left ventricle, resulting in severe cardiac dysfunction. Moreover, endothelial cell-specific CRIF1 deletion resulted in mitochondrial dysfunction, reduced ATP levels, inflammation, and excessive oxidative stress in heart tissues, associated with decreased SIRT1 expression. Intraperitoneal injection of SRT1720 ameliorated cardiac dysfunction by activating endothelial nitric oxide synthase, reducing oxidative stress, and inhibiting inflammation. Furthermore, the decreased endothelial junction-associated protein zonula occludens-1 in CRIF1-deleted mice was significantly recovered after SRT1720 treatment. Our results suggest that endothelial CRIF1 plays an important role in maintaining cardiac function, and that SIRT1 induction could be a therapeutic strategy for endothelial dysfunction-induced cardiac dysfunction.
Collapse
|
18
|
Dao T, Green AE, Kim YA, Bae SJ, Ha KT, Gariani K, Lee MR, Menzies KJ, Ryu D. Sarcopenia and Muscle Aging: A Brief Overview. Endocrinol Metab (Seoul) 2020; 35:716-732. [PMID: 33397034 PMCID: PMC7803599 DOI: 10.3803/enm.2020.405] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 11/30/2020] [Indexed: 12/11/2022] Open
Abstract
The world is facing the new challenges of an aging population, and understanding the process of aging has therefore become one of the most important global concerns. Sarcopenia is a condition which is defined by the gradual loss of skeletal muscle mass and function with age. In research and clinical practice, sarcopenia is recognized as a component of geriatric disease and is a current target for drug development. In this review we define this condition and provide an overview of current therapeutic approaches. We further highlight recent findings that describe key pathophysiological phenotypes of this condition, including alterations in muscle fiber types, mitochondrial function, nicotinamide adenine dinucleotide (NAD+) metabolism, myokines, and gut microbiota, in aged muscle compared to young muscle or healthy aged muscle. The last part of this review examines new therapeutic avenues for promising treatment targets. There is still no accepted therapy for sarcopenia in humans. Here we provide a brief review of the current state of research derived from various mouse models or human samples that provide novel routes for the development of effective therapeutics to maintain muscle health during aging.
Collapse
Affiliation(s)
- Tam Dao
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon,
Korea
| | - Alexander E. Green
- University of Ottawa Eric Poulin Centre for Neuromuscular Disease, Ottawa, ON,
Canada
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences University of Ottawa, Ottawa, ON,
Canada
| | - Yun A Kim
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon,
Korea
| | - Sung-Jin Bae
- Korean Medical Research Center for Healthy Aging, Pusan National University, Yangsan,
Korea
| | - Ki-Tae Ha
- Korean Medical Research Center for Healthy Aging, Pusan National University, Yangsan,
Korea
- Department of Korean Medical Science, Pusan National University School of Korean Medicine, Yangsan,
Korea
| | - Karim Gariani
- Service of Endocrinology, Diabetes, Nutrition and Therapeutic Patient Education, Geneva University Hospitals, Geneva,
Switzerland
- Faculty of Medicine, University of Geneva, Geneva,
Switzerland
| | - Mi-ra Lee
- Department of Social Welfare, Division of Public Service, Dong-Eui University, Busan,
Korea
- Mi-ra Lee, Department of Public Service, Dong-Eui University, 176 Eomgwang-ro, Busanjin-gu, Busan 47340, Korea, Tel: +82-51-890-2038, E-mail:
| | - Keir J. Menzies
- University of Ottawa Eric Poulin Centre for Neuromuscular Disease, Ottawa, ON,
Canada
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences University of Ottawa, Ottawa, ON,
Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON,
Canada
- Keir J. Menzies, Eric Poulin Centre for Neuromuscular Disease, Interdisciplinary School of Health Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada, Tel: +1-613-562-5800, E-mail:
| | - Dongryeol Ryu
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon,
Korea
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon,
Korea
- Samsung Biomedical Research Institute, Samsung Medical Center, Seoul,
Korea
- Corresponding authors: Dongryeol Ryu, Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Korea, Tel: +82-31-299-6138, E-mail:
| |
Collapse
|
19
|
The potential roles of NAD(P)H:quinone oxidoreductase 1 in the development of diabetic nephropathy and actin polymerization. Sci Rep 2020; 10:17735. [PMID: 33082368 PMCID: PMC7576596 DOI: 10.1038/s41598-020-74493-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 09/30/2020] [Indexed: 11/08/2022] Open
Abstract
Diabetic nephropathy (DN) is a major complication of diabetes mellitus. NAD(P)H:quinone oxidoreductase 1 (NQO1) is an antioxidant enzyme that has been involved in the progression of several kidney injuries. However, the roles of NQO1 in DN are still unclear. We investigated the effects of NQO1 deficiency in streptozotocin (STZ)-induced DN mice. NQO1 was upregulated in the glomerulus and podocytes under hyperglycemic conditions. NQO1 knockout (NKO) mice showed more severe changes in blood glucose and body weight than WT mice after STZ treatment. Furthermore, STZ-mediated pathological parameters including glomerular injury, blood urea nitrogen levels, and foot process width were more severe in NKO mice than WT mice. Importantly, urine albumin-to-creatinine ratio (ACR) was higher in healthy, non-treated NKO mice than WT mice. ACR response to STZ or LPS was dramatically increased in the urine of NKO mice compared to vehicle controls, while it maintained a normal range following treatment of WT mice. More importantly, we found that NQO1 can stimulate actin polymerization in an in vitro biochemical assay without directly the accumulation on F-actin. In summary, NQO1 has an important role against the development of DN pathogenesis and is a novel contributor in actin reorganization via stimulating actin polymerization.
Collapse
|
20
|
Expression analysis of mammalian mitochondrial ribosomal protein genes. Gene Expr Patterns 2020; 38:119147. [PMID: 32987154 DOI: 10.1016/j.gep.2020.119147] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 09/14/2020] [Accepted: 09/20/2020] [Indexed: 02/07/2023]
Abstract
Mitochondrial ribosomal proteins (MRPs) are essential components for the structural and functional integrity of the mitoribosome complex. Throughout evolution, the mammalian mitoribosome has acquired new Mrp genes to compensate for loss of ribosomal RNA. More than 80 MRPs have been identified in mammals. Here we document expression pattern of 79 Mrp genes during mouse development and adult tissues and find that these genes are consistently expressed throughout early embryogenesis with little stage or tissue specificity. Further investigation of the amino acid sequence reveals that this group of proteins has little to no protein similarity. Recent work has shown that the majority of Mrp genes are essential resulting in early embryonic lethality, suggesting no functional redundancy among the group. Taken together, these results indicate that the Mrp genes are not a gene family descended from a single ancestral gene, and that each MRP has unique and essential role in the mitoribosome complex. The lack of functional redundancy is surprising given the importance of the mitoribosome for cellular and organismal viability. Further, these data suggest that genomic variants in Mrp genes may be causative for early pregnancy loss and should be evaluated as clinically.
Collapse
|
21
|
Choi MJ, Jung SB, Lee SE, Kang SG, Lee JH, Ryu MJ, Chung HK, Chang JY, Kim YK, Hong HJ, Kim H, Kim HJ, Lee CH, Mardinoglu A, Yi HS, Shong M. An adipocyte-specific defect in oxidative phosphorylation increases systemic energy expenditure and protects against diet-induced obesity in mouse models. Diabetologia 2020; 63:837-852. [PMID: 31925461 DOI: 10.1007/s00125-019-05082-7] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 10/30/2019] [Indexed: 12/30/2022]
Abstract
AIMS/HYPOTHESIS Mitochondrial oxidative phosphorylation (OxPhos) is essential for energy production and survival. However, the tissue-specific and systemic metabolic effects of OxPhos function in adipocytes remain incompletely understood. METHODS We used adipocyte-specific Crif1 (also known as Gadd45gip1) knockout (AdKO) mice with decreased adipocyte OxPhos function. AdKO mice fed a normal chow or high-fat diet were evaluated for glucose homeostasis, weight gain and energy expenditure (EE). RNA sequencing of adipose tissues was used to identify the key mitokines affected in AdKO mice, which included fibroblast growth factor 21 (FGF21) and growth differentiation factor 15 (GDF15). For in vitro analysis, doxycycline was used to pharmacologically decrease OxPhos in 3T3L1 adipocytes. To identify the effects of GDF15 and FGF21 on the metabolic phenotype of AdKO mice, we generated AdKO mice with global Gdf15 knockout (AdGKO) or global Fgf21 knockout (AdFKO). RESULTS Under high-fat diet conditions, AdKO mice were resistant to weight gain and exhibited higher EE and improved glucose tolerance. In vitro pharmacological and in vivo genetic inhibition of OxPhos in adipocytes significantly upregulated mitochondrial unfolded protein response-related genes and secretion of mitokines such as GDF15 and FGF21. We evaluated the metabolic phenotypes of AdGKO and AdFKO mice, revealing that GDF15 and FGF21 differentially regulated energy homeostasis in AdKO mice. Both mitokines had beneficial effects on obesity and insulin resistance in the context of decreased adipocyte OxPhos, but only GDF15 regulated EE in AdKO mice. CONCLUSIONS/INTERPRETATION The present study demonstrated that the adipose tissue adaptive mitochondrial stress response affected systemic energy homeostasis via cell-autonomous and non-cell-autonomous pathways. We identified novel roles for adipose OxPhos and adipo-mitokines in the regulation of systemic glucose homeostasis and EE, which facilitated adaptation of an organism to local mitochondrial stress.
Collapse
Affiliation(s)
- Min Jeong Choi
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon, 35015, South Korea
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Saet-Byel Jung
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon, 35015, South Korea
| | - Seong Eun Lee
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon, 35015, South Korea
| | - Seul Gi Kang
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon, 35015, South Korea
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Ju Hee Lee
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon, 35015, South Korea
- Department of Internal Medicine, Chungnam National University Hospital, Daejeon, 35015, South Korea
| | - Min Jeong Ryu
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Hyo Kyun Chung
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon, 35015, South Korea
| | - Joon Young Chang
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon, 35015, South Korea
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Yong Kyung Kim
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon, 35015, South Korea
| | - Hyun Jung Hong
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon, 35015, South Korea
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Hail Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Hyun Jin Kim
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon, 35015, South Korea
- Department of Internal Medicine, Chungnam National University Hospital, Daejeon, 35015, South Korea
| | - Chul-Ho Lee
- Animal Model Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Adil Mardinoglu
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, UK
| | - Hyon-Seung Yi
- Department of Internal Medicine, Chungnam National University Hospital, Daejeon, 35015, South Korea.
| | - Minho Shong
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon, 35015, South Korea.
- Department of Internal Medicine, Chungnam National University Hospital, Daejeon, 35015, South Korea.
| |
Collapse
|
22
|
Lee JH, Park A, Oh KJ, Lee SC, Kim WK, Bae KH. The Role of Adipose Tissue Mitochondria: Regulation of Mitochondrial Function for the Treatment of Metabolic Diseases. Int J Mol Sci 2019; 20:ijms20194924. [PMID: 31590292 PMCID: PMC6801758 DOI: 10.3390/ijms20194924] [Citation(s) in RCA: 165] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 09/29/2019] [Accepted: 09/30/2019] [Indexed: 02/07/2023] Open
Abstract
: Mitochondria play a key role in maintaining energy homeostasis in metabolic tissues, including adipose tissues. The two main types of adipose tissues are the white adipose tissue (WAT) and the brown adipose tissue (BAT). WAT primarily stores excess energy, whereas BAT is predominantly responsible for energy expenditure by non-shivering thermogenesis through the mitochondria. WAT in response to appropriate stimuli such as cold exposure and β-adrenergic agonist undergoes browning wherein it acts as BAT, which is characterized by the presence of a higher number of mitochondria. Mitochondrial dysfunction in adipocytes has been reported to have strong correlation with metabolic diseases, including obesity and type 2 diabetes. Dysfunction of mitochondria results in detrimental effects on adipocyte differentiation, lipid metabolism, insulin sensitivity, oxidative capacity, and thermogenesis, which consequently lead to metabolic diseases. Recent studies have shown that mitochondrial function can be improved by using thiazolidinedione, mitochondria-targeted antioxidants, and dietary natural compounds; by performing exercise; and by controlling caloric restriction, thereby maintaining the metabolic homeostasis by inducing adaptive thermogenesis of BAT and browning of WAT. In this review, we focus on and summarize the molecular regulation involved in the improvement of mitochondrial function in adipose tissues so that strategies can be developed to treat metabolic diseases.
Collapse
Affiliation(s)
- Jae Ho Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
| | - Anna Park
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
| | - Kyoung-Jin Oh
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34141, Korea
| | - Sang Chul Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34141, Korea
| | - Won Kon Kim
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea.
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34141, Korea.
| | - Kwang-Hee Bae
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea.
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34141, Korea.
| |
Collapse
|
23
|
Woo CY, Jang JE, Lee SE, Koh EH, Lee KU. Mitochondrial Dysfunction in Adipocytes as a Primary Cause of Adipose Tissue Inflammation. Diabetes Metab J 2019; 43:247-256. [PMID: 30968618 PMCID: PMC6581541 DOI: 10.4093/dmj.2018.0221] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 01/19/2019] [Indexed: 12/17/2022] Open
Abstract
Adipose tissue inflammation is considered a major contributing factor in the development of obesity-associated insulin resistance and cardiovascular diseases. However, the cause of adipose tissue inflammation is presently unclear. The role of mitochondria in white adipocytes has long been neglected because of their low abundance. However, recent evidence suggests that mitochondria are essential for maintaining metabolic homeostasis in white adipocytes. In a series of recent studies, we found that mitochondrial function in white adipocytes is essential to the synthesis of adiponectin, which is the most abundant adipokine synthesized from adipocytes, with many favorable effects on metabolism, including improvement of insulin sensitivity and reduction of atherosclerotic processes and systemic inflammation. From these results, we propose a new hypothesis that mitochondrial dysfunction in adipocytes is a primary cause of adipose tissue inflammation and compared this hypothesis with a prevailing concept that "adipose tissue hypoxia" may underlie adipose tissue dysfunction in obesity. Recent studies have emphasized the role of the mitochondrial quality control mechanism in maintaining mitochondrial function. Future studies are warranted to test whether an inadequate mitochondrial quality control mechanism is responsible for mitochondrial dysfunction in adipocytes and adipose tissue inflammation.
Collapse
Affiliation(s)
- Chang Yun Woo
- Department of Internal Medicine, University of Ulsan College of Medicine, Seoul, Korea
| | - Jung Eun Jang
- Department of Internal Medicine, Inje University Haeundae Paik Hospital, Inje University College of Medicine, Busan, Korea
| | - Seung Eun Lee
- Department of Internal Medicine, Dongguk University Ilsan Hospital, Dongguk University College of Medicine, Goyang, Korea
| | - Eun Hee Koh
- Department of Internal Medicine, University of Ulsan College of Medicine, Seoul, Korea
| | - Ki Up Lee
- Department of Internal Medicine, University of Ulsan College of Medicine, Seoul, Korea.
| |
Collapse
|
24
|
Sustarsic EG, Ma T, Lynes MD, Larsen M, Karavaeva I, Havelund JF, Nielsen CH, Jedrychowski MP, Moreno-Torres M, Lundh M, Plucinska K, Jespersen NZ, Grevengoed TJ, Kramar B, Peics J, Hansen JB, Shamsi F, Forss I, Neess D, Keipert S, Wang J, Stohlmann K, Brandslund I, Christensen C, Jørgensen ME, Linneberg A, Pedersen O, Kiebish MA, Qvortrup K, Han X, Pedersen BK, Jastroch M, Mandrup S, Kjær A, Gygi SP, Hansen T, Gillum MP, Grarup N, Emanuelli B, Nielsen S, Scheele C, Tseng YH, Færgeman NJ, Gerhart-Hines Z. Cardiolipin Synthesis in Brown and Beige Fat Mitochondria Is Essential for Systemic Energy Homeostasis. Cell Metab 2018; 28:159-174.e11. [PMID: 29861389 PMCID: PMC6038052 DOI: 10.1016/j.cmet.2018.05.003] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 02/20/2018] [Accepted: 05/03/2018] [Indexed: 12/17/2022]
Abstract
Activation of energy expenditure in thermogenic fat is a promising strategy to improve metabolic health, yet the dynamic processes that evoke this response are poorly understood. Here we show that synthesis of the mitochondrial phospholipid cardiolipin is indispensable for stimulating and sustaining thermogenic fat function. Cardiolipin biosynthesis is robustly induced in brown and beige adipose upon cold exposure. Mimicking this response through overexpression of cardiolipin synthase (Crls1) enhances energy consumption in mouse and human adipocytes. Crls1 deficiency in thermogenic adipocytes diminishes inducible mitochondrial uncoupling and elicits a nuclear transcriptional response through endoplasmic reticulum stress-mediated retrograde communication. Cardiolipin depletion in brown and beige fat abolishes adipose thermogenesis and glucose uptake, which renders animals insulin resistant. We further identify a rare human CRLS1 variant associated with insulin resistance and show that adipose CRLS1 levels positively correlate with insulin sensitivity. Thus, adipose cardiolipin has a powerful impact on organismal energy homeostasis through thermogenic fat bioenergetics.
Collapse
Affiliation(s)
- Elahu G Sustarsic
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen 2200, Denmark; Department of Biomedical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Tao Ma
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen 2200, Denmark; Department of Biomedical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Matthew D Lynes
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Michael Larsen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Iuliia Karavaeva
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen 2200, Denmark; Department of Biomedical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Jesper F Havelund
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense 5230, Denmark
| | - Carsten H Nielsen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet, Copenhagen 2200, Denmark
| | | | - Marta Moreno-Torres
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense 5230, Denmark
| | - Morten Lundh
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen 2200, Denmark
| | - Kaja Plucinska
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen 2200, Denmark
| | - Naja Z Jespersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen 2200, Denmark; Centre of Inflammation and Metabolism and Centre for Physical Activity Research, Rigshospitalet, University Hospital of Copenhagen, Copenhagen 2200, Denmark
| | - Trisha J Grevengoed
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen 2200, Denmark
| | - Barbara Kramar
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen 2200, Denmark
| | - Julia Peics
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen 2200, Denmark; Department of Biomedical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Jakob B Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen 2200, Denmark; Department of Biomedical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Farnaz Shamsi
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Isabel Forss
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense 5230, Denmark
| | - Ditte Neess
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense 5230, Denmark
| | - Susanne Keipert
- Helmholtz Diabetes Center and German Diabetes Center (DZD), Helmholtz Zentrum München, Neuherberg 85764, Germany
| | - Jianing Wang
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Katharina Stohlmann
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen 2200, Denmark
| | - Ivan Brandslund
- Lillebaelt Hospital, Vejle 7100, Denmark; Institute of Regional Health Research, University of Southern Denmark, Odense 5230, Denmark
| | | | - Marit E Jørgensen
- Steno Diabetes Center, Gentofte 2820, Denmark; National Institute of Public Health, Southern Denmark University, Copenhagen 1353, Denmark
| | - Allan Linneberg
- Research Center for Prevention and Health, Glostrup 2600, Denmark; Center for Clinical Research and Prevention, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen 2200, Denmark
| | - Oluf Pedersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen 2200, Denmark
| | | | - Klaus Qvortrup
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Xianlin Han
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Bente Klarlund Pedersen
- Centre of Inflammation and Metabolism and Centre for Physical Activity Research, Rigshospitalet, University Hospital of Copenhagen, Copenhagen 2200, Denmark
| | - Martin Jastroch
- Helmholtz Diabetes Center and German Diabetes Center (DZD), Helmholtz Zentrum München, Neuherberg 85764, Germany
| | - Susanne Mandrup
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense 5230, Denmark
| | - Andreas Kjær
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet, Copenhagen 2200, Denmark
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen 2200, Denmark
| | - Matthew P Gillum
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen 2200, Denmark
| | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen 2200, Denmark
| | - Brice Emanuelli
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen 2200, Denmark
| | - Søren Nielsen
- Centre of Inflammation and Metabolism and Centre for Physical Activity Research, Rigshospitalet, University Hospital of Copenhagen, Copenhagen 2200, Denmark
| | - Camilla Scheele
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen 2200, Denmark; Centre of Inflammation and Metabolism and Centre for Physical Activity Research, Rigshospitalet, University Hospital of Copenhagen, Copenhagen 2200, Denmark
| | - Yu-Hua Tseng
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Nils J Færgeman
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense 5230, Denmark
| | - Zachary Gerhart-Hines
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen 2200, Denmark; Department of Biomedical Sciences, University of Copenhagen, Copenhagen 2200, Denmark.
| |
Collapse
|
25
|
Piao S, Lee JW, Nagar H, Jung SB, Choi S, Kim S, Lee I, Kim SM, Shin N, Lee YR, Lee SD, Park JB, Irani K, Won M, Hur GM, Jeon BH, Kim DW, Kim CS. CR6 interacting factor 1 deficiency promotes endothelial inflammation by SIRT1 downregulation. PLoS One 2018; 13:e0192693. [PMID: 29474366 PMCID: PMC5825004 DOI: 10.1371/journal.pone.0192693] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 01/29/2018] [Indexed: 11/29/2022] Open
Abstract
Aims CR6 interacting factor 1 (CRIF1) deficiency impairs mitochondrial oxidative phosphorylation complexes, contributing to increased mitochondrial and cellular reactive oxygen species (ROS) production. CRIF1 downregulation has also been revealed to decrease sirtuin 1 (SIRT1) expression and impair vascular function. Inhibition of SIRT1 disturbs oxidative energy metabolism and stimulates nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB)-induced inflammation. Therefore, we hypothesized that both CRIF1 deficiency-induced mitochondrial ROS production and SIRT1 reduction play stimulatory roles in vascular inflammation. Methods and results Plasma levels and mRNA expression of proinflammatory cytokines (tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and IL-6) were markedly elevated in endothelium-specific CRIF1-knockout mice and CRIF1-silenced endothelial cells, respectively. Moreover, CRIF1 deficiency-induced vascular adhesion molecule-1 (VCAM-1) expression was consistently attenuated by the antioxidant N-acetyl-cysteine and NF-κB inhibitor (BAY11). We next showed that siRNA-mediated CRIF1 downregulation markedly activated NF-κB. SIRT1 overexpression not only rescued CRIF1 deficiency-induced NF-κB activation but also decreased inflammatory cytokines (TNF-α, IL-1β, and IL-6) and VCAM-1 expression levels in endothelial cells. Conclusions These results strongly suggest that CRIF1 deficiency promotes endothelial cell inflammation by increasing VCAM-1 expression, elevating inflammatory cytokines levels, and activating the transcription factor NF-κB, all of which were inhibited by SIRT1 overexpression.
Collapse
Affiliation(s)
- Shuyu Piao
- Department of physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Jun Wan Lee
- Emergency ICU, Regional Emergency Center, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Harsha Nagar
- Department of physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Saet-byel Jung
- Department of Endocrinology, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Sujeong Choi
- Department of physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Seonhee Kim
- Department of physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Ikjun Lee
- Department of physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Sung-min Kim
- Department of physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Nara Shin
- Department of Anatomy & Medical Science, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Yu Ran Lee
- Department of physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Sang Do Lee
- Department of physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Jin Bong Park
- Department of physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Kaikobad Irani
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA United States of America
| | - Minho Won
- Department of Pharmacology, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Gang Min Hur
- Department of Pharmacology, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Byeong Hwa Jeon
- Department of physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Dong Woon Kim
- Department of Anatomy & Medical Science, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Cuk-Seong Kim
- Department of physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
- * E-mail:
| |
Collapse
|
26
|
Ma L, Fu Q, Xu B, Zhou H, Gao J, Shao X, Xiong J, Gu Q, Wen S, Li F, Shen L, Chen G, Fang H, Lyu J. Breast cancer-associated mitochondrial DNA haplogroup promotes neoplastic growth via ROS-mediated AKT activation. Int J Cancer 2017; 142:1786-1796. [PMID: 29226320 DOI: 10.1002/ijc.31207] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 11/28/2017] [Accepted: 12/04/2017] [Indexed: 12/13/2022]
Abstract
In the last decade, mitochondrial DNA (mtDNA) haplogroups have been associated with the occurrence of breast cancer. However, the underlying mechanism is not known. Combining a case-control study with a large cohort of women from Southern China with breast cancer and functional analyses with trans-mitochondrial technology, we demonstrate that the D5 haplogroup is associated with an increased risk of breast cancer [odds ratio (OR) = 2.789; 95% confidence interval (CI) [1.318, 5.901]; p = 0.007]. Furthermore, mitochondrial respiration, mitochondrial ATP content and membrane potential, were lower in both bone osteosarcoma and breast cancer cell models of cytoplasmic hybrids (cybrids) containing the mtDNA D5 haplogroup than in those with non-D5 haplogroups. Using in vitro and in vivo tumorigenicity assays, we found that cells with the D5 haplogroup were more susceptible to tumorigenesis compared to cells with non-D5 haplogroups. Mechanistically, the D5 haplogroup may promote tumorigenesis at least partially through activation of the v-AKT murine thymoma viral oncogene (AKT) via phosphorylation of threonine 308, which is mediated by increased reactive oxygen species generation in D5 cybrids. Our findings demonstrate that there is decreased mitochondrial function in cells with the D5 haplogroup compared to cells with non-D5 haplogroups, which may be associated with increased neoplastic growth in breast cancer.
Collapse
Affiliation(s)
- Lin Ma
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Key Clinical Laboratory of Henan Province, Zhengzhou, Henan, China
| | - Qingzi Fu
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Bing Xu
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Clinical Laboratory, Hangzhou Women's Hospital, Hangzhou, Zhejiang, China
| | - Huaibin Zhou
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jing Gao
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaoli Shao
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jingting Xiong
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qianru Gu
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Shumeng Wen
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Fengjie Li
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lijun Shen
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Guorong Chen
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Hezhi Fang
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jianxin Lyu
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
27
|
Yu HM, Chung HK, Kim KS, Lee JM, Hong JH, Park KS. PDE 5 inhibitor improves insulin sensitivity by enhancing mitochondrial function in adipocytes. Biochem Biophys Res Commun 2017; 493:631-636. [DOI: 10.1016/j.bbrc.2017.08.140] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Accepted: 08/24/2017] [Indexed: 01/07/2023]
|
28
|
Yan Q, Ahn SH, Medie FM, Sharma-Kuinkel BK, Park LP, Scott WK, Deshmukh H, Tsalik EL, Cyr DD, Woods CW, Yu CHA, Adams C, Qi R, Hansen B, Fowler VG. Candidate genes on murine chromosome 8 are associated with susceptibility to Staphylococcus aureus infection in mice and are involved with Staphylococcus aureus septicemia in humans. PLoS One 2017; 12:e0179033. [PMID: 28594911 PMCID: PMC5464679 DOI: 10.1371/journal.pone.0179033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 05/23/2017] [Indexed: 02/06/2023] Open
Abstract
We previously showed that chromosome 8 of A/J mice was associated with susceptibility to S. aureus infection. However, the specific genes responsible for this susceptibility are unknown. Chromosome substitution strain 8 (CSS8) mice, which have chromosome 8 from A/J but an otherwise C57BL/6J genome, were used to identify the genetic determinants of susceptibility to S. aureus on chromosome 8. Quantitative trait loci (QTL) mapping of S. aureus-infected N2 backcross mice (F1 [C8A] × C57BL/6J) identified a locus 83180780–88103009 (GRCm38/mm10) on A/J chromosome 8 that was linked to S. aureus susceptibility. All genes on the QTL (n~ 102) were further analyzed by three different strategies: 1) different expression in susceptible (A/J) and resistant (C57BL/6J) mice only in response to S. aureus, 2) consistently different expression in both uninfected and infected states between the two strains, and 3) damaging non-synonymous SNPs in either strain. Eleven candidate genes from the QTL region were significantly differently expressed in patients with S. aureus infection vs healthy human subjects. Four of these 11 genes also exhibited significantly different expression in S. aureus-challenged human neutrophils: Ier2, Crif1, Cd97 and Lyl1. CD97 ligand binding was evaluated within peritoneal neutrophils from A/J and C57BL/6J. CD97 from A/J had stronger CD55 but weaker integrin α5β1 ligand binding as compared with C57BL/6J. Because CD55/CD97 binding regulates immune cell activation and cytokine production, and integrin α5β1 is a membrane receptor for fibronectin, which is also bound by S. aureus, strain-specific differences could contribute to susceptibility to S. aureus. Down-regulation of Crif1 with siRNA was associated with increased host cell apoptosis among both naïve and S. aureus-infected bone marrow-derived macrophages. Specific genes in A/J chromosome 8, including Cd97 and Crif1, may play important roles in host defense against S. aureus.
Collapse
Affiliation(s)
- Qin Yan
- Division of Infectious Diseases & International Health, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Sun Hee Ahn
- Department of Biochemistry School of Dentistry, Chonnam National University, Bukgu, Gwangju, Korea
| | - Felix Mba Medie
- Division of Infectious Diseases & International Health, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Batu K. Sharma-Kuinkel
- Division of Infectious Diseases & International Health, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Lawrence P. Park
- Duke Global Health Institute, Duke University, Durham, North Carolina, United States of America
| | - William K. Scott
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida, United States of America
| | - Hitesh Deshmukh
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Ephraim L. Tsalik
- Division of Infectious Diseases & International Health, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, United States of America
- Emergency Medicine Service, Durham Veteran’s Affairs Medical Center, Durham, North Carolina, United States of America
- Duke Clinical Research Institute, Durham, North Carolina, United States of America
| | - Derek D. Cyr
- Duke Clinical Research Institute, Durham, North Carolina, United States of America
| | - Christopher W. Woods
- Division of Infectious Diseases & International Health, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke Clinical Research Institute, Durham, North Carolina, United States of America
- Section on Infectious Diseases, Durham Veteran’s Affairs Medical Center, Durham, North Carolina, United States of America
| | - Chen-Hsin Albert Yu
- Division of Infectious Diseases & International Health, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Carlton Adams
- Division of Infectious Diseases & International Health, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Robert Qi
- Division of Infectious Diseases & International Health, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Brenda Hansen
- Division of Infectious Diseases & International Health, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Vance G. Fowler
- Division of Infectious Diseases & International Health, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke Clinical Research Institute, Durham, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
29
|
Kang SG, Yi HS, Choi MJ, Ryu MJ, Jung S, Chung HK, Chang JY, Kim YK, Lee SE, Kim HW, Choi H, Kim DS, Lee JH, Kim KS, Kim HJ, Lee CH, Oike Y, Shong M. ANGPTL6 expression is coupled with mitochondrial OXPHOS function to regulate adipose FGF21. J Endocrinol 2017; 233:105-118. [PMID: 28184000 DOI: 10.1530/joe-16-0549] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 02/09/2017] [Indexed: 12/20/2022]
Abstract
Recent studies revealed that the inhibition of mitochondrial oxidative phosphorylation (OXPHOS) is coupled with the mitochondrial unfolded protein response, thereby stimulating the secretion of non-cell autonomous factors, which may control systemic energy metabolism and longevity. However, the nature and roles of non-cell autonomous factors induced in adipose tissue in response to reduced OXPHOS function remain to be clarified in mammals. CR6-interacting factor 1 (CRIF1) is an essential mitoribosomal protein for the intramitochondrial production of mtDNA-encoded OXPHOS subunits. Deficiency of CRIF1 impairs the proper formation of the OXPHOS complex, resulting in reduced function. To determine which secretory factors are induced in response to reduced mitochondrial OXPHOS function, we analyzed gene expression datasets in Crif1-depleted mouse embryonic fibroblasts. Crif1 deficiency preferentially increased the expression of angiopoietin-like 6 (Angptl6) and did not affect other members of the ANGPTL family. Moreover, treatment with mitochondrial OXPHOS inhibitors increased the expression of Angptl6 in cultured adipocytes. To confirm Angptl6 induction in vivo, we generated a murine model of reduced mitochondrial OXPHOS function using adipose tissue-specific Crif1-deficient mice and verified the upregulation of Angptl6 and fibroblast growth factor 21 (Fgf21) in white adipose tissue. Treatment with recombinant ANGPTL6 protein increased oxygen consumption and Pparα expression through the extracellular signal-regulated kinase/mitogen-activated protein kinase pathway in cultured adipocytes. Furthermore, the ANGPTL6-mediated increase in Pparα expression resulted in increased FGF21 expression, thereby promoting β-oxidation. In conclusion, mitochondrial OXPHOS function governs the expression of ANGPTL6, which is an essential factor for FGF21 production in adipose tissue and cultured adipocytes.
Collapse
Affiliation(s)
- Seul Gi Kang
- Research Center for Endocrine and Metabolic DiseasesChungnam National University School of Medicine, Daejeon, Korea
- Department of Medical ScienceChungnam National University School of Medicine, Daejeon, Korea
| | - Hyon-Seung Yi
- Research Center for Endocrine and Metabolic DiseasesChungnam National University School of Medicine, Daejeon, Korea
| | - Min Jeong Choi
- Research Center for Endocrine and Metabolic DiseasesChungnam National University School of Medicine, Daejeon, Korea
- Department of Medical ScienceChungnam National University School of Medicine, Daejeon, Korea
| | | | - Saetbyel Jung
- Research Center for Endocrine and Metabolic DiseasesChungnam National University School of Medicine, Daejeon, Korea
| | - Hyo Kyun Chung
- Research Center for Endocrine and Metabolic DiseasesChungnam National University School of Medicine, Daejeon, Korea
| | - Joon Young Chang
- Research Center for Endocrine and Metabolic DiseasesChungnam National University School of Medicine, Daejeon, Korea
- Department of Medical ScienceChungnam National University School of Medicine, Daejeon, Korea
| | - Yong Kyung Kim
- Research Center for Endocrine and Metabolic DiseasesChungnam National University School of Medicine, Daejeon, Korea
| | - Seong Eun Lee
- Research Center for Endocrine and Metabolic DiseasesChungnam National University School of Medicine, Daejeon, Korea
- Department of Medical ScienceChungnam National University School of Medicine, Daejeon, Korea
| | - Hyeon-Woo Kim
- Research Center for Endocrine and Metabolic DiseasesChungnam National University School of Medicine, Daejeon, Korea
- Department of Medical ScienceChungnam National University School of Medicine, Daejeon, Korea
| | - Hoil Choi
- Department of BiochemistryChungnam National University School of Medicine, Daejeon, Korea
| | - Dong Seok Kim
- Department of BiochemistryChungnam National University School of Medicine, Daejeon, Korea
| | - Ju Hee Lee
- Research Center for Endocrine and Metabolic DiseasesChungnam National University School of Medicine, Daejeon, Korea
| | - Koon Soon Kim
- Research Center for Endocrine and Metabolic DiseasesChungnam National University School of Medicine, Daejeon, Korea
| | - Hyun Jin Kim
- Research Center for Endocrine and Metabolic DiseasesChungnam National University School of Medicine, Daejeon, Korea
| | - Chul-Ho Lee
- Animal Model CenterKorea Research Institute of Bioscience and Biotechnology, Daejeon, Korea
| | - Yuichi Oike
- Department of Molecular GeneticsGraduate School of Medical Sciences, Kumamoto University, Chuo-ku, Kumamoto, Japan
| | - Minho Shong
- Research Center for Endocrine and Metabolic DiseasesChungnam National University School of Medicine, Daejeon, Korea
| |
Collapse
|
30
|
Targeted deletion of Crif1 in mouse epidermis impairs skin homeostasis and hair morphogenesis. Sci Rep 2017; 7:44828. [PMID: 28317864 PMCID: PMC5357846 DOI: 10.1038/srep44828] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 02/14/2017] [Indexed: 12/27/2022] Open
Abstract
The epidermis, which consists mainly of keratinocytes, acts as a physical barrier to infections by regulating keratinocyte proliferation and differentiation. Hair follicles undergo continuous cycling to produce new one. Therefore, optimum supply of energy from the mitochondria is essential for maintaining skin homeostasis and hair growth. CRIF1 is a mitochondrial protein that regulates mitoribosome-mediated synthesis and insertion of mitochondrial oxidative phosphorylation polypeptides into the mitochondrial membrane in mammals. Recent studies reveal that conditional knockout (cKO) of Crif1 in specific tissues of mice induced mitochondrial dysfunction. To determine whether the mitochondrial function of keratinocytes affects skin homeostasis and hair morphogenesis, we generated epidermis-specific Crif1 cKO mice. Deletion of Crif1 in epidermis resulted in impaired mitochondrial function and Crif1 cKO mice died within a week. Keratinocyte proliferation and differentiation were markedly inhibited in Crif1 cKO mice. Furthermore, hair follicle morphogenesis of Crif1 cKO mice was disrupted by down-regulation of Wnt/β-catenin signaling. These results demonstrate that mitochondrial function in keratinocytes is essential for maintaining epidermal homeostasis and hair follicle morphogenesis.
Collapse
|
31
|
Chung W, Ryu MJ, Heo JY, Lee S, Yoon S, Park H, Park S, Kim Y, Kim YH, Yoon SH, Shin YS, Lee WH, Ju X, Kweon GR, Ko Y. Sevoflurane Exposure during the Critical Period Affects Synaptic Transmission and Mitochondrial Respiration but Not Long-term Behavior in Mice. Anesthesiology 2017; 126:288-299. [DOI: 10.1097/aln.0000000000001470] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Abstract
Background
Anesthesia during the synaptogenic period induces dendritic spine formation, which may affect neurodevelopment. The authors, therefore, evaluated whether changes in synaptic transmission after dendritic spine formation induced by sevoflurane were associated with long-term behavioral changes. The effects of sevoflurane on mitochondrial function were also assessed to further understand the mechanism behind spinogenesis.
Methods
Postnatal day 16 to 17 mice were exposed to sevoflurane (2.5% for 2 h), and synaptic transmission was measured in the medial prefrontal cortex 6 h or 5 days later. The expression of postsynaptic proteins and mitochondrial function were measured after anesthesia. Long-term behavioral changes were assessed in adult mice.
Results
Sevoflurane increased the expression of excitatory postsynaptic proteins in male and female mice (n = 3 to 5 per group). Sevoflurane exposure in male mice transiently increased miniature excitatory postsynaptic current frequency (control: 8.53 ± 2.87; sevoflurane: 11.09 ± 2.58) but decreased miniature inhibitory postsynaptic current frequency (control: 10.18 ± 4.66; sevoflurane: 6.88 ± 2.15). Unexpectedly, sevoflurane increased miniature inhibitory postsynaptic current frequency (control: 1.81 ± 1.11; sevoflurane: 3.56 ± 1.74) in female mice (neurons, n = 10 to 21 per group). Sevoflurane also increased mitochondrial respiration in male mice (n = 5 to 8 per group). However, such changes from anesthesia during the critical period did not induce long-term behavioral consequences. Values are presented as mean ± SD.
Conclusions
Sevoflurane exposure during the critical period induces mitochondrial hyperactivity and transient imbalance of excitatory/inhibitory synaptic transmission, without long-lasting behavioral consequences. Further studies are needed to confirm sexual differences and to define the role of mitochondrial activity during anesthesia-induced spine formation.
Collapse
Affiliation(s)
- Woosuk Chung
- From the Department of Anesthesia and Pain Medicine, Chungnam National University, Daejeon, South Korea (W.C., S.L., S.Y., S.P., Y.H.K., S.H.Y., Y.S.S., W.H.L., Y.K.); Departments of Biochemistry (M.J.R., J.Y.H., G.R.K.) and Medical Science (J.Y.H., X.J.), Chungnam National University School of Medicine, Daejeon, South Korea; and Departments of Biological Sciences (H.P.) and Biomedical Sciences (
| | - Min Jeong Ryu
- From the Department of Anesthesia and Pain Medicine, Chungnam National University, Daejeon, South Korea (W.C., S.L., S.Y., S.P., Y.H.K., S.H.Y., Y.S.S., W.H.L., Y.K.); Departments of Biochemistry (M.J.R., J.Y.H., G.R.K.) and Medical Science (J.Y.H., X.J.), Chungnam National University School of Medicine, Daejeon, South Korea; and Departments of Biological Sciences (H.P.) and Biomedical Sciences (
| | - Jun Young Heo
- From the Department of Anesthesia and Pain Medicine, Chungnam National University, Daejeon, South Korea (W.C., S.L., S.Y., S.P., Y.H.K., S.H.Y., Y.S.S., W.H.L., Y.K.); Departments of Biochemistry (M.J.R., J.Y.H., G.R.K.) and Medical Science (J.Y.H., X.J.), Chungnam National University School of Medicine, Daejeon, South Korea; and Departments of Biological Sciences (H.P.) and Biomedical Sciences (
| | - Soomin Lee
- From the Department of Anesthesia and Pain Medicine, Chungnam National University, Daejeon, South Korea (W.C., S.L., S.Y., S.P., Y.H.K., S.H.Y., Y.S.S., W.H.L., Y.K.); Departments of Biochemistry (M.J.R., J.Y.H., G.R.K.) and Medical Science (J.Y.H., X.J.), Chungnam National University School of Medicine, Daejeon, South Korea; and Departments of Biological Sciences (H.P.) and Biomedical Sciences (
| | - Seunghwan Yoon
- From the Department of Anesthesia and Pain Medicine, Chungnam National University, Daejeon, South Korea (W.C., S.L., S.Y., S.P., Y.H.K., S.H.Y., Y.S.S., W.H.L., Y.K.); Departments of Biochemistry (M.J.R., J.Y.H., G.R.K.) and Medical Science (J.Y.H., X.J.), Chungnam National University School of Medicine, Daejeon, South Korea; and Departments of Biological Sciences (H.P.) and Biomedical Sciences (
| | - Haram Park
- From the Department of Anesthesia and Pain Medicine, Chungnam National University, Daejeon, South Korea (W.C., S.L., S.Y., S.P., Y.H.K., S.H.Y., Y.S.S., W.H.L., Y.K.); Departments of Biochemistry (M.J.R., J.Y.H., G.R.K.) and Medical Science (J.Y.H., X.J.), Chungnam National University School of Medicine, Daejeon, South Korea; and Departments of Biological Sciences (H.P.) and Biomedical Sciences (
| | - Sangil Park
- From the Department of Anesthesia and Pain Medicine, Chungnam National University, Daejeon, South Korea (W.C., S.L., S.Y., S.P., Y.H.K., S.H.Y., Y.S.S., W.H.L., Y.K.); Departments of Biochemistry (M.J.R., J.Y.H., G.R.K.) and Medical Science (J.Y.H., X.J.), Chungnam National University School of Medicine, Daejeon, South Korea; and Departments of Biological Sciences (H.P.) and Biomedical Sciences (
| | - Yangsik Kim
- From the Department of Anesthesia and Pain Medicine, Chungnam National University, Daejeon, South Korea (W.C., S.L., S.Y., S.P., Y.H.K., S.H.Y., Y.S.S., W.H.L., Y.K.); Departments of Biochemistry (M.J.R., J.Y.H., G.R.K.) and Medical Science (J.Y.H., X.J.), Chungnam National University School of Medicine, Daejeon, South Korea; and Departments of Biological Sciences (H.P.) and Biomedical Sciences (
| | - Yoon Hee Kim
- From the Department of Anesthesia and Pain Medicine, Chungnam National University, Daejeon, South Korea (W.C., S.L., S.Y., S.P., Y.H.K., S.H.Y., Y.S.S., W.H.L., Y.K.); Departments of Biochemistry (M.J.R., J.Y.H., G.R.K.) and Medical Science (J.Y.H., X.J.), Chungnam National University School of Medicine, Daejeon, South Korea; and Departments of Biological Sciences (H.P.) and Biomedical Sciences (
| | - Seok Hwa Yoon
- From the Department of Anesthesia and Pain Medicine, Chungnam National University, Daejeon, South Korea (W.C., S.L., S.Y., S.P., Y.H.K., S.H.Y., Y.S.S., W.H.L., Y.K.); Departments of Biochemistry (M.J.R., J.Y.H., G.R.K.) and Medical Science (J.Y.H., X.J.), Chungnam National University School of Medicine, Daejeon, South Korea; and Departments of Biological Sciences (H.P.) and Biomedical Sciences (
| | - Yong Sup Shin
- From the Department of Anesthesia and Pain Medicine, Chungnam National University, Daejeon, South Korea (W.C., S.L., S.Y., S.P., Y.H.K., S.H.Y., Y.S.S., W.H.L., Y.K.); Departments of Biochemistry (M.J.R., J.Y.H., G.R.K.) and Medical Science (J.Y.H., X.J.), Chungnam National University School of Medicine, Daejeon, South Korea; and Departments of Biological Sciences (H.P.) and Biomedical Sciences (
| | - Won Hyung Lee
- From the Department of Anesthesia and Pain Medicine, Chungnam National University, Daejeon, South Korea (W.C., S.L., S.Y., S.P., Y.H.K., S.H.Y., Y.S.S., W.H.L., Y.K.); Departments of Biochemistry (M.J.R., J.Y.H., G.R.K.) and Medical Science (J.Y.H., X.J.), Chungnam National University School of Medicine, Daejeon, South Korea; and Departments of Biological Sciences (H.P.) and Biomedical Sciences (
| | - Xianshu Ju
- From the Department of Anesthesia and Pain Medicine, Chungnam National University, Daejeon, South Korea (W.C., S.L., S.Y., S.P., Y.H.K., S.H.Y., Y.S.S., W.H.L., Y.K.); Departments of Biochemistry (M.J.R., J.Y.H., G.R.K.) and Medical Science (J.Y.H., X.J.), Chungnam National University School of Medicine, Daejeon, South Korea; and Departments of Biological Sciences (H.P.) and Biomedical Sciences (
| | - Gi Ryang Kweon
- From the Department of Anesthesia and Pain Medicine, Chungnam National University, Daejeon, South Korea (W.C., S.L., S.Y., S.P., Y.H.K., S.H.Y., Y.S.S., W.H.L., Y.K.); Departments of Biochemistry (M.J.R., J.Y.H., G.R.K.) and Medical Science (J.Y.H., X.J.), Chungnam National University School of Medicine, Daejeon, South Korea; and Departments of Biological Sciences (H.P.) and Biomedical Sciences (
| | - Youngkwon Ko
- From the Department of Anesthesia and Pain Medicine, Chungnam National University, Daejeon, South Korea (W.C., S.L., S.Y., S.P., Y.H.K., S.H.Y., Y.S.S., W.H.L., Y.K.); Departments of Biochemistry (M.J.R., J.Y.H., G.R.K.) and Medical Science (J.Y.H., X.J.), Chungnam National University School of Medicine, Daejeon, South Korea; and Departments of Biological Sciences (H.P.) and Biomedical Sciences (
| |
Collapse
|
32
|
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: 261] [Impact Index Per Article: 32.6] [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
|
33
|
Lai CS, Wu JC, Ho CT, Pan MH. Chemoprevention of obesity by dietary natural compounds targeting mitochondrial regulation. Mol Nutr Food Res 2016; 61. [DOI: 10.1002/mnfr.201600721] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 10/02/2016] [Accepted: 11/07/2016] [Indexed: 12/12/2022]
Affiliation(s)
- Ching-Shu Lai
- Department of Seafood Science; National Kaohsiung Marine University; Kaohsiung Taiwan
| | - Jia-Ching Wu
- Institute of Food Science and Technology; National Taiwan University; Taipei Taiwan
| | - Chi-Tang Ho
- Department of Food Science; Rutgers University; New Brunswick NJ USA
| | - Min-Hsiung Pan
- Institute of Food Science and Technology; National Taiwan University; Taipei Taiwan
- Department of Medical Research, China Medical University Hospital; China Medical University; Taichung Taiwan
- Department of Health and Nutrition Biotechnology; Asia University; Taichung Taiwan
| |
Collapse
|
34
|
Lee JH, Kang YE, Chang JY, Park KC, Kim HW, Kim JT, Kim HJ, Yi HS, Shong M, Chung HK, Kim KS. An engineered FGF21 variant, LY2405319, can prevent non-alcoholic steatohepatitis by enhancing hepatic mitochondrial function. Am J Transl Res 2016; 8:4750-4763. [PMID: 27904677 PMCID: PMC5126319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 10/23/2016] [Indexed: 06/06/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a prevalent obesity-related disease that affects large populations throughout the world due to excessive calorie intake and an increasingly sedentary lifestyle. Fibroblast growth factor 21 (FGF21) has recently emerged as a promising therapeutic candidate for the treatment of obesity and diabetes. FGF21 is a starvation-induced pleiotropic hormone with various beneficial metabolic effects, and pharmacological treatment in rodents has been shown to improve insulin sensitivity and decrease simple fatty liver disease. However, its effects on reversing the symptoms of advanced liver disease have yet to be validated. Here, we investigated the protective effects of the LY2405319 compound, an engineered FGF21 variant, in a non-alcoholic steatohepatitis (NASH) model using leptin-deficient ob/ob mice and a methionine- and choline-deficient (MCD) diet to induce steatohepatitis. LY2405319 treatment in ob/ob mice corroborated previous results showing that improvements in the metabolic parameters were due to increased mitochondrial oxygen consumption rate and fatty acid oxidation. LY2405319 treatment in ob/ob mice on an MCD diet significantly reduced the symptoms of steatohepatitis, as confirmed by Masson's trichrome staining intensity. Serum levels of AST and ALT were also reduced, suggesting an attenuation of liver injury, while detection of inflammatory markers showed decreased mRNA expression of TGF-β1 and type-I collagen, and decreased phosphorylation of NF-kB p65, JNK1/2, and p38. Collectively, these data show that LY2405319 treatment attenuated MCD diet-induced NASH progression. We propose that the LY2405319 compound is a potential therapeutic candidate for the treatment of advanced liver disease.
Collapse
Affiliation(s)
- Ju Hee Lee
- Department of Internal Medicine, Chungnam National University HospitalDaejeon 35015, Republic of Korea
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of MedicineDaejeon 35015, Republic of Korea
| | - Yea Eun Kang
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of MedicineDaejeon 35015, Republic of Korea
| | - Joon Young Chang
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of MedicineDaejeon 35015, Republic of Korea
- Department of Medical Science, Chungnam National University School of MedicineDaejeon 35015, Republic of Korea
| | - Ki Cheol Park
- Clinical Research Institute, Daejeon St. Mary’s Hospital, College of Medicine, The Catholic University of KoreaDaejeon 34943, Republic of Korea
| | - Hyeon-Woo Kim
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of MedicineDaejeon 35015, Republic of Korea
- Department of Medical Science, Chungnam National University School of MedicineDaejeon 35015, Republic of Korea
| | - Jung Tae Kim
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of MedicineDaejeon 35015, Republic of Korea
- Department of Medical Science, Chungnam National University School of MedicineDaejeon 35015, Republic of Korea
| | - Hyun Jin Kim
- Department of Internal Medicine, Chungnam National University HospitalDaejeon 35015, Republic of Korea
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of MedicineDaejeon 35015, Republic of Korea
| | - Hyon-Seung Yi
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of MedicineDaejeon 35015, Republic of Korea
| | - Minho Shong
- Department of Internal Medicine, Chungnam National University HospitalDaejeon 35015, Republic of Korea
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of MedicineDaejeon 35015, Republic of Korea
| | - Hyo Kyun Chung
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of MedicineDaejeon 35015, Republic of Korea
| | - Koon Soon Kim
- Department of Internal Medicine, Chungnam National University HospitalDaejeon 35015, Republic of Korea
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of MedicineDaejeon 35015, Republic of Korea
| |
Collapse
|
35
|
Yang H, Wu JW, Wang SP, Severi I, Sartini L, Frizzell N, Cinti S, Yang G, Mitchell GA. Adipose-Specific Deficiency of Fumarate Hydratase in Mice Protects Against Obesity, Hepatic Steatosis, and Insulin Resistance. Diabetes 2016; 65:3396-3409. [PMID: 27554470 PMCID: PMC5860441 DOI: 10.2337/db16-0136] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 08/16/2016] [Indexed: 01/08/2023]
Abstract
Obesity and type 2 diabetes are associated with impaired mitochondrial function in adipose tissue. To study the effects of primary deficiency of mitochondrial energy metabolism in fat, we generated mice with adipose-specific deficiency of fumarate hydratase (FH), an integral Krebs cycle enzyme (AFHKO mice). AFHKO mice have severe ultrastructural abnormalities of mitochondria, ATP depletion in white adipose tissue (WAT) and brown adipose tissue, low WAT mass with small adipocytes, and impaired thermogenesis with large unilocular brown adipocytes. AFHKO mice are strongly protected against obesity, insulin resistance, and fatty liver despite aging and high-fat feeding. AFHKO white adipocytes showed normal lipolysis but low triglyceride synthesis. ATP depletion in normal white adipocytes by mitochondrial toxins also decreased triglyceride synthesis, proportionally to ATP depletion, suggesting that reduced triglyceride synthesis may result nonspecifically from adipocyte energy deficiency. At thermoneutrality, protection from insulin resistance and hepatic steatosis was diminished. Taken together, the results show that under the cold stress of regular animal room conditions, adipocyte-specific FH deficiency in mice causes mitochondrial energy depletion in adipose tissues and protects from obesity, hepatic steatosis, and insulin resistance, suggesting that in cold-stressed animals, mitochondrial function in adipose tissue is a determinant of fat mass and insulin sensitivity.
Collapse
Affiliation(s)
- Hao Yang
- Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
- Division of Medical Genetics, Department of Pediatrics, Université de Montréal and CHU Sainte-Justine, Montreal, Quebec, Canada
| | - Jiang W Wu
- Division of Medical Genetics, Department of Pediatrics, Université de Montréal and CHU Sainte-Justine, Montreal, Quebec, Canada
| | - Shu P Wang
- Division of Medical Genetics, Department of Pediatrics, Université de Montréal and CHU Sainte-Justine, Montreal, Quebec, Canada
| | - Ilenia Severi
- Department of Experimental and Clinical Medicine, Center of Obesity, United Hospitals, University of Ancona (Università Politecnica Delle Marche), Ancona, Italy
| | - Loris Sartini
- Department of Experimental and Clinical Medicine, Center of Obesity, United Hospitals, University of Ancona (Università Politecnica Delle Marche), Ancona, Italy
| | - Norma Frizzell
- Department of Pharmacology, Physiology & Neuroscience, School of Medicine, University of South Carolina, Columbia, SC
| | - Saverio Cinti
- Department of Experimental and Clinical Medicine, Center of Obesity, United Hospitals, University of Ancona (Università Politecnica Delle Marche), Ancona, Italy
| | - Gongshe Yang
- Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Grant A Mitchell
- Division of Medical Genetics, Department of Pediatrics, Université de Montréal and CHU Sainte-Justine, Montreal, Quebec, Canada
| |
Collapse
|
36
|
Piao L, Jung I, Huh JY, Miyata T, Ha H. A novel plasminogen activator inhibitor-1 inhibitor, TM5441, protects against high-fat diet-induced obesity and adipocyte injury in mice. Br J Pharmacol 2016; 173:2622-32. [PMID: 27339909 DOI: 10.1111/bph.13541] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 06/11/2016] [Accepted: 06/16/2016] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Obesity is one of the most prevalent chronic diseases worldwide, and dysregulated adipocyte function plays an important role in obesity-associated metabolic disorder. The level of plasma plasminogen activator inhibitor-1 (PAI-1) is increased in obese subjects, and PAI-1 null mice show improved insulin sensitivity when subjected to high-fat and high-sucrose diet-induced metabolic stress, suggesting that a best-in-class PAI-1 inhibitor may become a novel therapeutic agent for obesity-associated metabolic syndrome. TM5441 is a novel orally active PAI-1 inhibitor that does not cause bleeding episodes. Hence, in the present study we examined the preventive effect of TM5441 on high-fat diet (HFD)-induced adipocyte dysfunction. EXPERIMENTAL APPROACH Ten-week-old C57BL/6J mice were fed a normal diet (18% of total calories from fat) or HFD (60% of total calories from fat) for 10 weeks, and TM5441 (20 mg·kg(-1) oral gavage) was administered daily with the initiation of HFD. KEY RESULTS TM5441 prevented HFD-induced body weight gain and systemic insulin resistance. TM5441 normalized HFD-induced dysregulated JNK and Akt phosphorylation, suggesting that it prevents the insulin resistance of adipocytes. TM5441 also attenuated the macrophage infiltration and increased expression of pro-inflammatory cytokines, such as inducible nitric oxide synthase, induced by the HFD. In addition, TM5441 prevented the HFD-induced down-regulation of genes involved in mitochondrial biogenesis and function, suggesting that it may prevent adipocyte inflammation and dysregulation by maintaining mitochondrial fitness. CONCLUSION AND IMPLICATIONS Our data suggest that TM5441 may become a novel therapeutic agent for obesity and obesity-related metabolic disorders.
Collapse
Affiliation(s)
- Lingjuan Piao
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Korea
| | - Inji Jung
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Korea
| | - Joo Young Huh
- Colleges of Pharmacy, Chonnam National University, Gwang-Ju, Korea
| | - Toshio Miyata
- United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hunjoo Ha
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Korea
| |
Collapse
|
37
|
Zhang X, Xiang L, Ran Q, Liu Y, Xiang Y, Xiao Y, Chen L, Li F, Zhong JF, Li Z. Crif1 Promotes Adipogenic Differentiation of Bone Marrow Mesenchymal Stem Cells After Irradiation by Modulating the PKA/CREB Signaling Pathway. Stem Cells 2016; 33:1915-26. [PMID: 25847389 DOI: 10.1002/stem.2019] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 02/12/2015] [Accepted: 02/28/2015] [Indexed: 12/22/2022]
Abstract
Dysfunction of the hematopoietic microenvironment is the main obstacle encountered during hematopoiesis reconstruction in patients with acute hematopoietic radiation syndrome. Bone marrow mesenchymal stem cells (BM-MSCs) play a crucial supporting role in hematopoiesis by maintaining the balance between adipogenic and osteogenic differentiation. In this study, we found that irradiation decreased the colony-forming efficiency of BM-MSCs and impaired the balance between adipogenic and osteogenic differentiation. Following irradiation, BM-MCSs became strongly predisposed to adipogenesis, as evidenced by increased oil red O staining and elevated mRNA and protein levels of the adipogenic markers and transcription factors PPARγ and AP2. Overexpression of the essential adipogenesis regulator Crif1 in BM-MSCs promoted adipogenesis after irradiation exposure by upregulating adipogenesis-related genes, including C/EBPβ, PPARγ, and AP2. We found that Crif1 promoted the phosphorylation of cAMP response element binding protein (CREB) through direct interaction with protein kinase A (PKA)-α. Phosphorylation of CREB was inhibited in Crif1-knockdown BM-MSCs even in the presence of a PKA agonist (db-cAMP) and could be suppressed in Crif1-overexpressing BM-MSCs by a PKAα inhibitor (H-89). These results suggest that Crif1 is an indispensable regulator of PKAα cat that modulates the PKA/CREB signaling pathway to promote adipogenic differentiation of BM-MSCs after irradiation.
Collapse
Affiliation(s)
- Xi Zhang
- Department of Hematology and, The Second Affiliated Hospital, Third Military Medical University, Chongqing, China
| | - Lixin Xiang
- Department of Blood Transfusion, The Second Affiliated Hospital, Third Military Medical University, Chongqing, China
| | - Qian Ran
- Department of Blood Transfusion, The Second Affiliated Hospital, Third Military Medical University, Chongqing, China
| | - Yao Liu
- Department of Hematology and, The Second Affiliated Hospital, Third Military Medical University, Chongqing, China
| | - Yang Xiang
- Department of Blood Transfusion, The Second Affiliated Hospital, Third Military Medical University, Chongqing, China
| | - Yanni Xiao
- Department of Blood Transfusion, The Second Affiliated Hospital, Third Military Medical University, Chongqing, China
| | - Li Chen
- Department of Blood Transfusion, The Second Affiliated Hospital, Third Military Medical University, Chongqing, China
| | - Fengjie Li
- Department of Blood Transfusion, The Second Affiliated Hospital, Third Military Medical University, Chongqing, China
| | - Jiang F Zhong
- Department of Pathology, University of Southern California, Keck School of Medicine, California, USA
| | - Zhongjun Li
- Department of Blood Transfusion, The Second Affiliated Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
38
|
Segregation of Naturally Occurring Mitochondrial DNA Variants in a Mini-Pig Model. Genetics 2016; 202:931-44. [PMID: 26819245 DOI: 10.1534/genetics.115.181321] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 01/17/2016] [Indexed: 11/18/2022] Open
Abstract
The maternally inherited mitochondrial genome (mtDNA) is present in multimeric form within cells and harbors sequence variants (heteroplasmy). While a single mtDNA variant at high load can cause disease, naturally occurring variants likely persist at low levels across generations of healthy populations. To determine how naturally occurring variants are segregated and transmitted, we generated a mini-pig model, which originates from the same maternal ancestor. Following next-generation sequencing, we identified a series of low-level mtDNA variants in blood samples from the female founder and her daughters. Four variants, ranging from 3% to 20%, were selected for validation by high-resolution melting analysis in 12 tissues from 31 animals across three generations. All four variants were maintained in the offspring, but variant load fluctuated significantly across the generations in several tissues, with sex-specific differences in heart and liver. Moreover, variant load was persistently reduced in high-respiratory organs (heart, brain, diaphragm, and muscle), which correlated significantly with higher mtDNA copy number. However, oocytes showed increased heterogeneity in variant load, which correlated with increased mtDNA copy number during in vitro maturation. Altogether, these outcomes show that naturally occurring mtDNA variants segregate and are maintained in a tissue-specific manner across generations. This segregation likely involves the maintenance of selective mtDNA variants during organogenesis, which can be differentially regulated in oocytes and preimplantation embryos during maturation.
Collapse
|
39
|
Kobori M, Takahashi Y, Sakurai M, Akimoto Y, Tsushida T, Oike H, Ippoushi K. Quercetin suppresses immune cell accumulation and improves mitochondrial gene expression in adipose tissue of diet-induced obese mice. Mol Nutr Food Res 2015; 60:300-12. [PMID: 26499876 PMCID: PMC5063128 DOI: 10.1002/mnfr.201500595] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 10/10/2015] [Accepted: 10/14/2015] [Indexed: 12/22/2022]
Abstract
SCOPE To examine the effect of dietary quercetin on the function of epididymal adipose tissue (EAT) in Western diet-induced obese mice. METHODS AND RESULTS C57BL/6J mice were fed a control diet; a Western diet high in fat, cholesterol, and sucrose; or the same Western diet containing 0.05% quercetin for 18 weeks. Supplementation with quercetin suppressed the increase in the number of macrophages, the decrease in the ratio of CD4(+) to CD8(+) T cells in EAT, and the elevation of plasma leptin and tumor necrosis factor α levels in mice fed the Western diet. Comprehensive gene expression analysis revealed that quercetin suppressed gene expression associated with the accumulation and activation of immune cells, including macrophages and lymphocytes in EAT. It also improved the expression of the oxidative stress-sensitive transcription factor NFκB, NADPH oxidases, and antioxidant enzymes. Quercetin markedly increased gene expression associated with mitochondrial oxidative phosphorylation and mitochondrial DNA content. CONCLUSION Quercetin most likely universally suppresses the accumulation and activation of immune cells, including antiinflammatory cells, whereas it specifically increased gene expression associated with mitochondrial oxidative phosphorylation. Suppression of oxidative stress and NFκB activity likely contributed to the prevention of the accumulation and activation of immune cells and resulting chronic inflammation.
Collapse
Affiliation(s)
- Masuko Kobori
- National Food Research Institute, National Agriculture and Food Research Organization, Tsukuba, Ibaraki, Japan
| | - Yumiko Takahashi
- National Food Research Institute, National Agriculture and Food Research Organization, Tsukuba, Ibaraki, Japan
| | - Mutsumi Sakurai
- National Food Research Institute, National Agriculture and Food Research Organization, Tsukuba, Ibaraki, Japan
| | - Yukari Akimoto
- National Food Research Institute, National Agriculture and Food Research Organization, Tsukuba, Ibaraki, Japan
| | - Tojiro Tsushida
- Department of Food Business, School of Food, Agricultural and Environmental Sciences, Miyagi University, Sendai, Miyagi, Japan
| | - Hideaki Oike
- National Food Research Institute, National Agriculture and Food Research Organization, Tsukuba, Ibaraki, Japan
| | - Katsunari Ippoushi
- National Food Research Institute, National Agriculture and Food Research Organization, Tsukuba, Ibaraki, Japan
| |
Collapse
|
40
|
Lim SJ, Scott A, Xiong XP, Vahidpour S, Karijolich J, Guo D, Pei S, Yu YT, Zhou R, Li WX. Requirement for CRIF1 in RNA interference and Dicer-2 stability. RNA Biol 2015; 11:1171-9. [PMID: 25483042 DOI: 10.4161/rna.34381] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
RNA interference (RNAi) is a eukaryotic gene-silencing system. Although the biochemistry of RNAi is relatively well defined, how this pathway is regulated remains incompletely understood. To identify genes involved in regulating the RNAi pathway, we screened for genetic mutations in Drosophila that alter the efficiency of RNAi. We identified the Drosophila homolog of the mammalian CR6-interacting factor 1 (CRIF1), also known as growth arrest and DNA-damage-inducible 45-gamma interacting protein (Gadd45GIP1), as a potential new regulator of the RNAi pathway. Loss-of-function mutants of Drosophila CRIF1 (dCRIF) are deficient in RNAi-mediated target gene knock-down, in the biogenesis of small interfering RNA (siRNA) molecules, and in antiviral immunity. Moreover, we show that dCRIF may function by interacting with, and stabilizing, the RNase III enzyme Dicer-2. Our results suggest that dCRIF may play an important role in regulating the RNAi pathway.
Collapse
Affiliation(s)
- Su Jun Lim
- a Department of Medicine ; University of California San Diego ; La Jolla , CA USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Schöttl T, Kappler L, Fromme T, Klingenspor M. Limited OXPHOS capacity in white adipocytes is a hallmark of obesity in laboratory mice irrespective of the glucose tolerance status. Mol Metab 2015; 4:631-42. [PMID: 26413469 PMCID: PMC4563017 DOI: 10.1016/j.molmet.2015.07.001] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 07/03/2015] [Accepted: 07/07/2015] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE Several human and rodent obesity studies speculate on a causal link between altered white adipocyte mitochondria in the obese state and changes in glucose homeostasis. We here aimed to dissect whether alterations in white adipocyte mitochondrial respiratory function are a specific phenomenon of obesity or impaired glucose tolerance or both. METHODS Mature white adipocytes were purified from posterior subcutaneous and intraabdominal epididymal fat of four murine obesity models characterized by either impaired or normal oral glucose tolerance. Bioenergetic profiles, including basal, leak, and maximal respiration, were generated using high-resolution respirometry. Cell respiratory control ratios were calculated to evaluate mitochondrial respiratory function. RESULTS Maximal respiration capacity and cell respiratory control ratios were diminished in white adipocytes of each of the four murine obesity models, both in the absence and the presence of impaired glucose tolerance. Limitation was more pronounced in adipocytes of intraabdominal versus subcutaneous fat. CONCLUSION Reduced mitochondrial respiratory capacity in white adipocytes is a hallmark of murine obesity irrespective of the glucose tolerance status. Impaired respiratory capacity in white adipocytes solely is not sufficient for the development of systemic glucose intolerance.
Collapse
Affiliation(s)
| | | | | | - Martin Klingenspor
- Molecular Nutritional Medicine, Technische Universität München, Else Kröner Fresenius Center for Nutritional Medicine, Freising, Germany
| |
Collapse
|
42
|
Lymphocyte-specific protein tyrosine kinase (Lck) interacts with CR6-interacting factor 1 (CRIF1) in mitochondria to repress oxidative phosphorylation. BMC Cancer 2015. [PMID: 26210498 PMCID: PMC4515320 DOI: 10.1186/s12885-015-1520-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Many cancer cells exhibit reduced mitochondrial respiration as part of metabolic reprogramming to support tumor growth. Mitochondrial localization of several protein tyrosine kinases is linked to this characteristic metabolic shift in solid tumors, but remains largely unknown in blood cancer. Lymphocyte-specific protein tyrosine kinase (Lck) is a key T-cell kinase and widely implicated in blood malignancies. The purpose of our study is to determine whether and how Lck contributes to metabolic shift in T-cell leukemia through mitochondrial localization. METHODS We compared the human leukemic T-cell line Jurkat with its Lck-deficient derivative Jcam cell line. Differences in mitochondrial respiration were measured by the levels of mitochondrial membrane potential, oxygen consumption, and mitochondrial superoxide. Detailed mitochondrial structure was visualized by transmission electron microscopy. Lck localization was evaluated by subcellular fractionation and confocal microscopy. Proteomic analysis was performed to identify proteins co-precipitated with Lck in leukemic T-cells. Protein interaction was validated by biochemical co-precipitation and confocal microscopy, followed by in situ proximity ligation assay microscopy to confirm close-range (<16 nm) interaction. RESULTS Jurkat cells have abnormal mitochondrial structure and reduced levels of mitochondrial respiration, which is associated with the presence of mitochondrial Lck and lower levels of mitochondrion-encoded electron transport chain proteins. Proteomics identified CR6-interacting factor 1 (CRIF1) as the novel Lck-interacting protein. Lck association with CRIF1 in Jurkat mitochondria was confirmed biochemically and by microscopy, but did not lead to CRIF1 tyrosine phosphorylation. Consistent with the role of CRIF1 in functional mitoribosome, shRNA-mediated silencing of CRIF1 in Jcam resulted in mitochondrial dysfunction similar to that observed in Jurkat. Reduced interaction between CRIF1 and Tid1, another key component of intramitochondrial translational machinery, in Jurkat further supports the role of mitochondrial Lck as a negative regulator of CRIF1 through competitive binding. CONCLUSIONS This is the first report demonstrating the role of mitochondrial Lck in metabolic reprogramming of leukemic cells. Mechanistically, it is distinct from other reported mitochondrial protein tyrosine kinases. In a kinase-independent manner, mitochondrial Lck interferes with mitochondrial translational machinery through competitive binding to CRIF1. These findings may reveal novel approaches in cancer therapy by targeting cancer cell metabolism.
Collapse
|
43
|
Koh EH, Kim AR, Kim H, Kim JH, Park HS, Ko MS, Kim MO, Kim HJ, Kim BJ, Yoo HJ, Kim SJ, Oh JS, Woo CY, Jang JE, Leem J, Cho MH, Lee KU. 11β-HSD1 reduces metabolic efficacy and adiponectin synthesis in hypertrophic adipocytes. J Endocrinol 2015; 225:147-58. [PMID: 25869616 DOI: 10.1530/joe-15-0117] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/07/2015] [Indexed: 12/23/2022]
Abstract
Mitochondrial dysfunction in hypertrophic adipocytes can reduce adiponectin synthesis. We investigated whether 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) expression is increased in hypertrophic adipocytes and whether this is responsible for mitochondrial dysfunction and reduced adiponectin synthesis. Differentiated 3T3L1 adipocytes were cultured for up to 21 days. The effect of AZD6925, a selective 11β-HSD1 inhibitor, on metabolism was examined. db/db mice were administered 600 mg/kg AZD6925 daily for 4 weeks via gastric lavage. Mitochondrial DNA (mtDNA) content, mRNA expression levels of 11 β -H sd1 and mitochondrial biogenesis factors, adiponectin synthesis, fatty acid oxidation (FAO), oxygen consumption rate and glycolysis were measured. Adipocyte hypertrophy in 3T3L1 cells exposed to a long duration of culture was associated with increased 11 β -Hsd1 mRNA expression and reduced mtDNA content, mitochondrial biogenesis factor expression and adiponectin synthesis. These cells displayed reduced mitochondrial respiration and increased glycolysis. Treatment of these cells with AZD6925 increased adiponectin synthesis and mitochondrial respiration. Inhibition of FAO by etomoxir blocked the AZD6925-induced increase in adiponectin synthesis, indicating that 11β-HSD1-mediated reductions in FAO are responsible for the reduction in adiponectin synthesis. The expression level of 11 β -Hsd1 was higher in adipose tissues of db/db mice. Administration of AZD6925 to db/db mice increased the plasma adiponectin level and adipose tissue FAO. In conclusion, increased 11β-HSD1 expression contributes to reduced mitochondrial respiration and adiponectin synthesis in hypertrophic adipocytes.
Collapse
Affiliation(s)
- Eun Hee Koh
- Department of Internal Medicine University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 138-736, Korea Biomedical Research Center Asan Institute for Life Sciences, Seoul 138-736, Korea Department of Biological Sciences Konkuk University, Seoul 143-701, Korea Department of Internal Medicine University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 138-736, Korea Biomedical Research Center Asan Institute for Life Sciences, Seoul 138-736, Korea Department of Biological Sciences Konkuk University, Seoul 143-701, Korea
| | - Ah-Ram Kim
- Department of Internal Medicine University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 138-736, Korea Biomedical Research Center Asan Institute for Life Sciences, Seoul 138-736, Korea Department of Biological Sciences Konkuk University, Seoul 143-701, Korea Department of Internal Medicine University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 138-736, Korea Biomedical Research Center Asan Institute for Life Sciences, Seoul 138-736, Korea Department of Biological Sciences Konkuk University, Seoul 143-701, Korea
| | - Hyunshik Kim
- Department of Internal Medicine University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 138-736, Korea Biomedical Research Center Asan Institute for Life Sciences, Seoul 138-736, Korea Department of Biological Sciences Konkuk University, Seoul 143-701, Korea
| | - Jin Hee Kim
- Department of Internal Medicine University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 138-736, Korea Biomedical Research Center Asan Institute for Life Sciences, Seoul 138-736, Korea Department of Biological Sciences Konkuk University, Seoul 143-701, Korea
| | - Hye-Sun Park
- Department of Internal Medicine University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 138-736, Korea Biomedical Research Center Asan Institute for Life Sciences, Seoul 138-736, Korea Department of Biological Sciences Konkuk University, Seoul 143-701, Korea
| | - Myoung Seok Ko
- Department of Internal Medicine University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 138-736, Korea Biomedical Research Center Asan Institute for Life Sciences, Seoul 138-736, Korea Department of Biological Sciences Konkuk University, Seoul 143-701, Korea
| | - Mi-Ok Kim
- Department of Internal Medicine University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 138-736, Korea Biomedical Research Center Asan Institute for Life Sciences, Seoul 138-736, Korea Department of Biological Sciences Konkuk University, Seoul 143-701, Korea
| | - Hyuk-Joong Kim
- Department of Internal Medicine University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 138-736, Korea Biomedical Research Center Asan Institute for Life Sciences, Seoul 138-736, Korea Department of Biological Sciences Konkuk University, Seoul 143-701, Korea Department of Internal Medicine University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 138-736, Korea Biomedical Research Center Asan Institute for Life Sciences, Seoul 138-736, Korea Department of Biological Sciences Konkuk University, Seoul 143-701, Korea
| | - Bum Joong Kim
- Department of Internal Medicine University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 138-736, Korea Biomedical Research Center Asan Institute for Life Sciences, Seoul 138-736, Korea Department of Biological Sciences Konkuk University, Seoul 143-701, Korea Department of Internal Medicine University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 138-736, Korea Biomedical Research Center Asan Institute for Life Sciences, Seoul 138-736, Korea Department of Biological Sciences Konkuk University, Seoul 143-701, Korea
| | - Hyun Ju Yoo
- Department of Internal Medicine University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 138-736, Korea Biomedical Research Center Asan Institute for Life Sciences, Seoul 138-736, Korea Department of Biological Sciences Konkuk University, Seoul 143-701, Korea
| | - Su Jung Kim
- Department of Internal Medicine University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 138-736, Korea Biomedical Research Center Asan Institute for Life Sciences, Seoul 138-736, Korea Department of Biological Sciences Konkuk University, Seoul 143-701, Korea
| | - Jin Sun Oh
- Department of Internal Medicine University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 138-736, Korea Biomedical Research Center Asan Institute for Life Sciences, Seoul 138-736, Korea Department of Biological Sciences Konkuk University, Seoul 143-701, Korea Department of Internal Medicine University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 138-736, Korea Biomedical Research Center Asan Institute for Life Sciences, Seoul 138-736, Korea Department of Biological Sciences Konkuk University, Seoul 143-701, Korea
| | - Chang-Yun Woo
- Department of Internal Medicine University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 138-736, Korea Biomedical Research Center Asan Institute for Life Sciences, Seoul 138-736, Korea Department of Biological Sciences Konkuk University, Seoul 143-701, Korea Department of Internal Medicine University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 138-736, Korea Biomedical Research Center Asan Institute for Life Sciences, Seoul 138-736, Korea Department of Biological Sciences Konkuk University, Seoul 143-701, Korea
| | - Jung Eun Jang
- Department of Internal Medicine University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 138-736, Korea Biomedical Research Center Asan Institute for Life Sciences, Seoul 138-736, Korea Department of Biological Sciences Konkuk University, Seoul 143-701, Korea Department of Internal Medicine University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 138-736, Korea Biomedical Research Center Asan Institute for Life Sciences, Seoul 138-736, Korea Department of Biological Sciences Konkuk University, Seoul 143-701, Korea
| | - Jaechan Leem
- Department of Internal Medicine University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 138-736, Korea Biomedical Research Center Asan Institute for Life Sciences, Seoul 138-736, Korea Department of Biological Sciences Konkuk University, Seoul 143-701, Korea Department of Internal Medicine University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 138-736, Korea Biomedical Research Center Asan Institute for Life Sciences, Seoul 138-736, Korea Department of Biological Sciences Konkuk University, Seoul 143-701, Korea
| | - Myung Hwan Cho
- Department of Internal Medicine University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 138-736, Korea Biomedical Research Center Asan Institute for Life Sciences, Seoul 138-736, Korea Department of Biological Sciences Konkuk University, Seoul 143-701, Korea
| | - Ki-Up Lee
- Department of Internal Medicine University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 138-736, Korea Biomedical Research Center Asan Institute for Life Sciences, Seoul 138-736, Korea Department of Biological Sciences Konkuk University, Seoul 143-701, Korea Department of Internal Medicine University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 138-736, Korea Biomedical Research Center Asan Institute for Life Sciences, Seoul 138-736, Korea Department of Biological Sciences Konkuk University, Seoul 143-701, Korea
| |
Collapse
|
44
|
Srivastava A, Shinn AS, Lee PJ, Mannam P. MKK3 mediates inflammatory response through modulation of mitochondrial function. Free Radic Biol Med 2015; 83:139-48. [PMID: 25697779 PMCID: PMC4441852 DOI: 10.1016/j.freeradbiomed.2015.01.035] [Citation(s) in RCA: 16] [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: 12/30/2014] [Revised: 01/23/2015] [Accepted: 01/23/2015] [Indexed: 12/30/2022]
Abstract
Mitochondria are increasingly recognized as drivers of inflammatory responses. MAP kinase kinase 3 (MKK3), a dual-specificity protein kinase, is activated in inflammation and in turn activates p38 MAP kinase signaling. Here we show that MKK3 influences mitochondrial function and acts as a critical mediator of inflammation. MKK3-deficient (MKK3(-/-)) mice and bone marrow-derived macrophages (BMDMs) secreted smaller amounts of cytokines than wild type (WT) after lipopolysaccharide (LPS) exposure. There was improved mitochondrial function, as measured by basal oxygen consumption rate, mitochondrial membrane potential, and ATP production, in MKK3(-/-) BMDMs. After LPS exposure, MKK3(-/-) BMDMs did not show a significant increase in cellular reactive oxygen species production or in mitochondrial superoxide compared to WT. Activation of two important inflammatory mediators, i.e., the nuclear translocation of NF-κB and caspase-1 activity (a key component of the inflammasome), was lower in MKK3(-/-) BMDMs. p38 and JNK activation was lower in MKK3(-/-) BMDMs compared to WT after exposure to LPS. Knockdown of MKK3 by siRNA in wild-type BMDMs improved mitochondrial membrane potential, reduced LPS-induced caspase-1 activation, and attenuated cytokine secretion. Our studies establish MKK3 as a regulator of mitochondrial function and inflammatory responses to LPS and suggest that MKK3 may be a therapeutic target in inflammatory disorders such as sepsis.
Collapse
Affiliation(s)
- Anup Srivastava
- Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520-8057, USA
| | - Amanda S Shinn
- Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520-8057, USA
| | - Patty J Lee
- Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520-8057, USA
| | - Praveen Mannam
- Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520-8057, USA.
| |
Collapse
|
45
|
Moreno-Navarrete JM, Ortega F, Moreno M, Xifra G, Ricart W, Fernández-Real JM. PRDM16 sustains white fat gene expression profile in human adipocytes in direct relation with insulin action. Mol Cell Endocrinol 2015; 405:84-93. [PMID: 25662275 DOI: 10.1016/j.mce.2015.01.042] [Citation(s) in RCA: 10] [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: 10/13/2014] [Revised: 01/22/2015] [Accepted: 01/29/2015] [Indexed: 01/08/2023]
Abstract
In the present study, we aimed to evaluate the possible role of PRDM16 in human adipocytes and in whole adipose tissue according to obesity and insulin sensitivity. PRDM16 knockdown (KD) had a dual behavior. While KD in preadipocytes led to enhanced gene expression markers of adipocyte differentiation, PRDM16 KD in fully differentiated adipocytes resulted in decreased adipogenic gene expression and insulin action. In line with KD in adipocytes, PRDM16 was positively associated with the expression of several genes involved in adipogenesis, insulin signaling, mitochondrial function and brown adipocyte-related markers in whole adipose tissue from two independent cohorts. PRDM16 was decreased in obese subjects in relation with the decrease of insulin sensitivity [HOM(AIR) (cohort 1) and M clamp value (cohort 2)]. Rosiglitazone (5 µmol/l) and metformin (5 mmol/l) led to increased PRDM16 mRNA and protein levels in isolated human adipocytes and in whole adipose tissue. In conclusion, PRDM16 might contribute to maintain adipose tissue "white fat" gene expression profile and systemic metabolic homeostasis.
Collapse
Affiliation(s)
- José María Moreno-Navarrete
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomèdica de Girona (IdIBGi), CIBEROBN (CB06/03/010), Instituto de Salud Carlos III (ISCIII), Girona 17007, Spain.
| | - Francisco Ortega
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomèdica de Girona (IdIBGi), CIBEROBN (CB06/03/010), Instituto de Salud Carlos III (ISCIII), Girona 17007, Spain
| | - María Moreno
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomèdica de Girona (IdIBGi), CIBEROBN (CB06/03/010), Instituto de Salud Carlos III (ISCIII), Girona 17007, Spain
| | - Gemma Xifra
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomèdica de Girona (IdIBGi), CIBEROBN (CB06/03/010), Instituto de Salud Carlos III (ISCIII), Girona 17007, Spain
| | - Wifredo Ricart
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomèdica de Girona (IdIBGi), CIBEROBN (CB06/03/010), Instituto de Salud Carlos III (ISCIII), Girona 17007, Spain
| | - José Manuel Fernández-Real
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomèdica de Girona (IdIBGi), CIBEROBN (CB06/03/010), Instituto de Salud Carlos III (ISCIII), Girona 17007, Spain.
| |
Collapse
|
46
|
Chung HK, Kim YK, Park JH, Ryu MJ, Chang JY, Hwang JH, Lee CH, Kim SH, Kim HJ, Kweon GR, Kim KS, Shong M. The indole derivative NecroX-7 improves nonalcoholic steatohepatitis in ob/ob mice through suppression of mitochondrial ROS/RNS and inflammation. Liver Int 2015; 35:1341-53. [PMID: 25443620 DOI: 10.1111/liv.12741] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 11/13/2014] [Indexed: 02/13/2023]
Abstract
BACKGROUND & AIMS Nonalcoholic steatohepatitis (NASH) is associated with cirrhosis and hepatocellular carcinoma. Reactive oxygen species (ROS) and reactive nitrogen species (RNS) play key roles in the development of the disease. However, the therapeutic target of NASH has not been fully defined and new treatments are needed. We investigated the protective effects of the antioxidant indole-derived NecroX-7 in a NASH mouse model using leptin-deficient ob/ob and methionine- and choline-deficient (MCD) diet-fed ob/ob mice. METHODS Six-week-old male mice were divided into three groups: ob/+ mice, ob/ob mice treated with vehicle and ob/ob mice treated daily with NecroX-7 (20 mg/kg) for 4 weeks. To study the effects of NecroX-7 in a fibrosis model, NASH was induced by feeding ob/ob mice an MCD diet. The effects of NecroX-7 on NASH progression were evaluated using biochemical, histological and molecular markers. RESULTS NecroX-7-treated ob/ob mice had a marked decrease in serum aspartate aminotransferase and alanine transaminase compared with vehicle-treated controls. Interestingly, hepatic steatosis and lipid peroxidation were significantly improved by NecroX-7 treatment. NecroX-7 inhibited tert-butylhydroperoxide- and H2 O2 -induced mitochondrial ROS/RNS in primary hepatocytes and attenuated mitochondrial dysfunction in vitro and in vivo. Furthermore, NecroX-7-treated mice exhibited fewer infiltrating macrophages and reduced hepatic tumour necrosis factor-alpha expression. Hepatic fibrosis in MCD-fed ob/ob mice was significantly decreased by NecroX-7 treatment. CONCLUSIONS NecroX-7 treatment improved hepatic steatosis and fibrosis in murine NASH models. These effects occurred through the suppression of whole-cell ROS/RNS and inflammatory responses and suggest that NecroX-7 has a potential therapeutic benefit in steatohepatitis.
Collapse
Affiliation(s)
- Hyo Kyun Chung
- Research Center for Endocrine & Metabolic Diseases, Chungnam National University School of Medicine, Daejeon, Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Kim YK, Joung KH, Ryu MJ, Kim SJ, Kim H, Chung HK, Lee MH, Lee SE, Choi MJ, Chang JY, Hong HJ, Kim KS, Lee SH, Kweon GR, Kim H, Lee CH, Kim HJ, Shong M. Disruption of CR6-interacting factor-1 (CRIF1) in mouse islet beta cells leads to mitochondrial diabetes with progressive beta cell failure. Diabetologia 2015; 58:771-80. [PMID: 25660120 DOI: 10.1007/s00125-015-3506-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 12/30/2014] [Indexed: 12/25/2022]
Abstract
AIM/HYPOTHESIS Although mitochondrial oxidative phosphorylation (OxPhos) dysfunction is believed to be responsible for beta cell dysfunction in insulin resistance and mitochondrial diabetes, the mechanisms underlying progressive beta cell failure caused by defective mitochondrial OxPhos are largely unknown. METHODS We examined the in vivo phenotypes of beta cell dysfunction in beta cell-specific Crif1 (also known as Gadd45gip1)-deficient mice. CR6-interacting factor-1 (CRIF1) is a mitochondrial protein essential for the synthesis and formation of the OxPhos complex in the inner mitochondrial membrane. RESULTS Crif1(beta-/-) mice exhibited impaired glucose tolerance with defective insulin secretion as early as 4 weeks of age without defects in islet structure. At 11 weeks of age, Crif1(beta-/-) mice displayed characteristic ultrastructural mitochondrial abnormalities as well as severe glucose intolerance. Furthermore, islet area and insulin content was decreased by approximately 50% compared with wild-type mice. Treatment with the glucoregulatory drug exenatide, a glucagon-like peptide-1 (GLP-1) agonist, was not sufficient to preserve beta cell function in Crif1(beta-/-) mice. CONCLUSIONS/INTERPRETATION Our results indicate that mitochondrial OxPhos dysfunction triggers progressive beta cell failure that is not halted by treatment with a GLP-1 agonist. The Crif1(beta-/-) mouse is a useful model for the study of beta cell failure caused by mitochondrial OxPhos dysfunction.
Collapse
Affiliation(s)
- Yong Kyung Kim
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, Daejeon, 301-721, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Nam M, Cooper MP. Role of Energy Metabolism in the Brown Fat Gene Program. Front Endocrinol (Lausanne) 2015; 6:104. [PMID: 26175716 PMCID: PMC4485181 DOI: 10.3389/fendo.2015.00104] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 06/15/2015] [Indexed: 12/20/2022] Open
Abstract
In murine and human brown adipose tissue (BAT), mitochondria are powerful generators of heat that safely metabolize fat, a feature that has great promise in the fight against obesity and diabetes. Recent studies suggest that the actions of mitochondria extend beyond their conventional role as generators of heat. There is mounting evidence that impaired mitochondrial respiratory capacity is accompanied by attenuated expression of Ucp1 and other BAT-selective genes, implying that mitochondria exert transcriptional control over the brown fat gene program. In this review, we discuss the current understanding of brown fat mitochondria, their potential role in transcriptional control of the brown fat gene program, and potential strategies to treat obesity in humans by leveraging thermogenesis in brown adipocytes.
Collapse
Affiliation(s)
- Minwoo Nam
- Division of Cardiovascular Medicine, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Marcus P. Cooper
- Division of Cardiovascular Medicine, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
- *Correspondence: Marcus P. Cooper, Division of Cardiovascular Medicine, Department of Medicine, University of Massachusetts Medical School, 368 Plantation Street, Albert Sherman Center, 7th Floor West, AS7-1053, Worcester, MA 01605, USA,
| |
Collapse
|
49
|
CRIF1 deficiency induces p66shc-mediated oxidative stress and endothelial activation. PLoS One 2014; 9:e98670. [PMID: 24906005 PMCID: PMC4048193 DOI: 10.1371/journal.pone.0098670] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 05/01/2014] [Indexed: 01/09/2023] Open
Abstract
Mitochondrial dysfunction has been implicated in the pathophysiology of various cardiovascular diseases. CRIF1 is a protein present in the mitochondria associated with large mitoribosomal subunits, and CRIF1 knockdown induces mitochondrial dysfunction and promotes ROS production. p66shc is a redox enzyme implicated in mitochondrial ROS generation and translation of oxidative signals and, therefore, is a key factor for oxidative stress in endothelial cells. In this study, we investigated whether mitochondrial dysfunction induced by CRIF1 knockdown induces p66shc stimulation and plays any role in mitochondrial dysfunction-induced endothelial activation. Knockdown of CRIF1 decreased the expression of mitochondrial oxidative phosphorylation (OXPHOS) complexes I, III and IV, leading to increased mitochondrial ROS (mtROS) and hyperpolarization of the mitochondrial membrane potential. Knockdown of CRIF1 also stimulated phosphorylation of p66shc and increased cytosolic ROS in endothelial cells. Furthermore, the expression of vascular cell adhesion molecule-1 and endoplasmic reticulum stress proteins were increased upon CRIF1 knockdown in endothelial cells. However, p66shc knockdown blunted the alteration in mitochondrial dynamics and ROS production in CRIF1 knockdown endothelial cells. In addition, p66shc knockdown reduced the CRIF1 knockdown-induced increases in adhesion between monocytes and endothelial cells. Taken together, these results suggest that CRIF1 knockdown partially induces endothelial activation via increased ROS production and phosphorylation of p66shc.
Collapse
|
50
|
Abstract
Obesity is now recognised as a low grade, chronic inflammatory disease that is linked to a myriad of disorders including cancer, cardiovascular disease and type 2 diabetes (T2D). With respect to T2D, work in the last decade has revealed that cells of the immune system are recruited to white adipose tissue beds (WAT), where they can secrete cytokines to modulate metabolism within WAT. As many of these cytokines are known to impair insulin action, blocking the recruitment of immune cells has been purported to have therapeutic utility for the treatment of obesity-induced T2D. As inflammation is critical for host defence, and energy consuming in nature, the blockade of inflammatory processes may, however, result in unwanted complications. In this review, we outline the immunological changes that occur within the WAT with respect to systemic glucose homeostasis. In particular, we focus on the role of major immune cell types in regulating nutrient homeostasis and potential initiating stimuli for WAT inflammation.
Collapse
Affiliation(s)
- H L Kammoun
- Cellular and Molecular Metabolism Laboratory, BakerIDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | | | | |
Collapse
|