1
|
Ferrero G, Cardamone MD, Luca F, Bourk E, Ricci L, Liu W, Gao Y, Burrone G, Muhammad A, Chan S, Smith E, Fan TYC, Cutrupi S, Garcia-Bassets I, De Bortoli M, Rosenfeld MG, Perissi V. Nonproteolytic ubiquitination regulates chromatin occupancy by the NCoR/SMRT/HDAC3 corepressor complex in MCF-7 breast cancer cells. Proc Natl Acad Sci U S A 2025; 122:e2502805122. [PMID: 40305047 PMCID: PMC12067245 DOI: 10.1073/pnas.2502805122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 03/31/2025] [Indexed: 05/02/2025] Open
Abstract
Tight regulation of gene expression is achieved through the coordinated action of transcription factors and cofactors that often can act as both repressors and activators in response to regulatory signals, with their activity modulated by context-specific signal transduction pathways that also impinge on their transient and cyclical recruitment to chromatin. However, the mechanisms underlying the intricate interplay between the regulatory strategies controlling cofactors' activity and localization across subcellar domains remain poorly understood. Here, we investigated the role of G-Protein Pathway Suppressor 2 (GPS2), a transcriptional cofactor critical for maintaining cellular homeostasis via regulation of mitochondrial biogenesis, stress response, lipid metabolism, insulin signaling, and inflammation, in MCF-7 breast cancer cells. By integration of biochemical assays with genome-wide RNA sequencing and Chromatin immunoprecipitation-Seq analyses, we show that nuclear GPS2 is required for licensing histone deacetylase 3 recruitment to chromatin via restricted ubiquitination by tumor necrosis factor receptor-associated factor 6 (TRAF6), an E3 ubiquitin ligase previously shown to regulate the switch from repressive to activating functions of the nuclear receptor corepressor (NCoR)/silencing mediator of retinoic acid and thyroid hormone receptor (SMRT) complex and here unexpectedly found to translocate to the nucleus in response to IL-1β stimulation. Nuclear TRAF6 is recruited to chromatin via direct interaction with the corepressors NCoR/SMRT, and TRAF6-mediated ubiquitination of TGF-beta activated kinase 1 (MAP3K7) binding protein 2 (TAB2), a facultative component of the NCoR/SMRT complex, contributes to corepressor clearance from target regulatory regions. Together, these results reveal an exquisite mechanism for coordinating the local regulation of cofactor activity with proinflammatory signaling pathways.
Collapse
Affiliation(s)
- Giulio Ferrero
- Department of Clinical and Biological Science, University of Torino, Orbassano (Torino)10043, Italy
| | - Maria Dafne Cardamone
- Department of Biochemistry and Cell Biology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA02118
| | - Francesca Luca
- Department of Clinical and Biological Science, University of Torino, Orbassano (Torino)10043, Italy
- Department of Biochemistry and Cell Biology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA02118
| | - Eliot Bourk
- Department of Medicine, School of Medicine, University of California San Diego, La Jolla, CA92037
| | - Laura Ricci
- Department of Clinical and Biological Science, University of Torino, Orbassano (Torino)10043, Italy
- Department of Medicine, School of Medicine, University of California San Diego, La Jolla, CA92037
| | - Wen Liu
- Department of Medicine, School of Medicine, University of California San Diego, La Jolla, CA92037
| | - Yuan Gao
- Department of Biochemistry and Cell Biology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA02118
| | - Giulia Burrone
- Department of Clinical and Biological Science, University of Torino, Orbassano (Torino)10043, Italy
- Department of Biochemistry and Cell Biology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA02118
- Department of Computer Science, University of Torino, Torino10149, Italy
| | - Akhirah Muhammad
- Department of Biochemistry and Cell Biology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA02118
| | - Stefanie Chan
- Department of Biochemistry and Cell Biology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA02118
| | - Emma Smith
- Department of Biochemistry and Cell Biology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA02118
| | - Ting-Yu Claire Fan
- Department of Biochemistry and Cell Biology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA02118
| | - Santina Cutrupi
- Department of Clinical and Biological Science, University of Torino, Orbassano (Torino)10043, Italy
| | - Ivan Garcia-Bassets
- Department of Medicine, School of Medicine, University of California San Diego, La Jolla, CA92037
| | - Michele De Bortoli
- Department of Clinical and Biological Science, University of Torino, Orbassano (Torino)10043, Italy
| | - Michael G. Rosenfeld
- Department of Medicine, School of Medicine, University of California San Diego, La Jolla, CA92037
| | - Valentina Perissi
- Department of Biochemistry and Cell Biology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA02118
| |
Collapse
|
2
|
Islam MS, Wei P, Suzauddula M, Nime I, Feroz F, Acharjee M, Pan F. The interplay of factors in metabolic syndrome: understanding its roots and complexity. Mol Med 2024; 30:279. [PMID: 39731011 DOI: 10.1186/s10020-024-01019-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 11/29/2024] [Indexed: 12/29/2024] Open
Abstract
Metabolic syndrome (MetS) is an indicator and diverse endocrine syndrome that combines different metabolic defects with clinical, physiological, biochemical, and metabolic factors. Obesity, visceral adiposity and abdominal obesity, dyslipidemia, insulin resistance (IR), elevated blood pressure, endothelial dysfunction, and acute or chronic inflammation are the risk factors associated with MetS. Abdominal obesity, a hallmark of MetS, highlights dysfunctional fat tissue and increased risk for cardiovascular disease and diabetes. Insulin, a vital peptide hormone, regulates glucose metabolism throughout the body. When cells become resistant to insulin's effects, it disrupts various molecular pathways, leading to IR. This condition is linked to a range of disorders, including obesity, diabetes, fatty liver disease, cardiovascular disease, and polycystic ovary syndrome. Atherogenic dyslipidemia is characterized by three key factors: high levels of small, low-dense lipoprotein (LDL) particles and triglycerides, alongside low levels of high-density lipoprotein (HDL), the "good" cholesterol. Such a combination is a major player in MetS, where IR is a driving force. Atherogenic dyslipidemia contributes significantly to the development of atherosclerosis, which can lead to cardiovascular disease. On top of that, genetic alteration and lifestyle factors such as diet and exercise influence the complexity and progression of MetS. To enhance our understanding and consciousness, it is essential to understand the fundamental pathogenesis of MetS. This review highlights current advancements in MetS research including the involvement of gut microbiome, epigenetic regulation, and metabolomic profiling for early detection of Mets. In addition, this review emphasized the epidemiology and fundamental pathogenesis of MetS, various risk factors, and their preventive measures. The goal of this effort is to deepen understanding of MetS and encourage further research to develop effective strategies for preventing and managing complex metabolic diseases.
Collapse
Affiliation(s)
- Md Sharifull Islam
- Center for Cancer Immunology, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Department of Microbiology, Stamford University Bangladesh, 51, Siddeswari Road, Dhaka, 1217, Bangladesh
| | - Ping Wei
- Center for Cancer Immunology, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Department of Pediatric Otolaryngology Head and Neck Surgery, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Md Suzauddula
- Department of Food Nutrition Dietetics and Health, Kansas State University, Manhattan, KS, 66506, USA
| | - Ishatur Nime
- Key Laboratory of Environment Correlative Dietology, College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Farahnaaz Feroz
- Department of Microbiology, Stamford University Bangladesh, 51, Siddeswari Road, Dhaka, 1217, Bangladesh
| | - Mrityunjoy Acharjee
- Department of Microbiology, Stamford University Bangladesh, 51, Siddeswari Road, Dhaka, 1217, Bangladesh
| | - Fan Pan
- Center for Cancer Immunology, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
3
|
Yang Y, Wu J, Zhou W, Ji G, Dang Y. Protein posttranslational modifications in metabolic diseases: basic concepts and targeted therapies. MedComm (Beijing) 2024; 5:e752. [PMID: 39355507 PMCID: PMC11442990 DOI: 10.1002/mco2.752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 10/03/2024] Open
Abstract
Metabolism-related diseases, including diabetes mellitus, obesity, hyperlipidemia, and nonalcoholic fatty liver disease, are becoming increasingly prevalent, thereby posing significant threats to human health and longevity. Proteins, as the primary mediators of biological activities, undergo various posttranslational modifications (PTMs), including phosphorylation, ubiquitination, acetylation, methylation, and SUMOylation, among others, which substantially diversify their functions. These modifications are crucial in the physiological and pathological processes associated with metabolic disorders. Despite advancements in the field, there remains a deficiency in contemporary summaries addressing how these modifications influence processes of metabolic disease. This review aims to systematically elucidate the mechanisms through which PTM of proteins impact the progression of metabolic diseases, including diabetes, obesity, hyperlipidemia, and nonalcoholic fatty liver disease. Additionally, the limitations of the current body of research are critically assessed. Leveraging PTMs of proteins provides novel insights and therapeutic targets for the prevention and treatment of metabolic disorders. Numerous drugs designed to target these modifications are currently in preclinical or clinical trials. This review also provides a comprehensive summary. By elucidating the intricate interplay between PTMs and metabolic pathways, this study advances understanding of the molecular mechanisms underlying metabolic dysfunction, thereby facilitating the development of more precise and effective disease management strategies.
Collapse
Affiliation(s)
- Yunuo Yang
- Institute of Digestive DiseasesChina‐Canada Center of Research for Digestive Diseases (ccCRDD)Shanghai University of Traditional Chinese MedicineShanghaiChina
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine)ShanghaiChina
| | - Jiaxuan Wu
- Institute of Digestive DiseasesChina‐Canada Center of Research for Digestive Diseases (ccCRDD)Shanghai University of Traditional Chinese MedicineShanghaiChina
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine)ShanghaiChina
| | - Wenjun Zhou
- Institute of Digestive DiseasesChina‐Canada Center of Research for Digestive Diseases (ccCRDD)Shanghai University of Traditional Chinese MedicineShanghaiChina
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine)ShanghaiChina
| | - Guang Ji
- Institute of Digestive DiseasesChina‐Canada Center of Research for Digestive Diseases (ccCRDD)Shanghai University of Traditional Chinese MedicineShanghaiChina
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine)ShanghaiChina
| | - Yanqi Dang
- Institute of Digestive DiseasesChina‐Canada Center of Research for Digestive Diseases (ccCRDD)Shanghai University of Traditional Chinese MedicineShanghaiChina
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine)ShanghaiChina
| |
Collapse
|
4
|
Si Y, Ou H, Jin X, Gu M, Sheng S, Peng W, Yang D, Zhan X, Zhang L, Yu Q, Liu X, Liu Y. G protein pathway suppressor 2 suppresses aerobic glycolysis through RACK1-mediated HIF-1α degradation in breast cancer. Free Radic Biol Med 2024; 222:478-492. [PMID: 38942092 DOI: 10.1016/j.freeradbiomed.2024.06.021] [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: 03/25/2024] [Revised: 06/16/2024] [Accepted: 06/25/2024] [Indexed: 06/30/2024]
Abstract
Aerobic glycolysis has been recognized as a hallmark of human cancer. G protein pathway suppressor 2 (GPS2) is a negative regulator of the G protein-MAPK pathway and a core subunit of the NCoR/SMRT transcriptional co-repressor complex. However, how its biological properties intersect with cellular metabolism in breast cancer (BC) development remains poorly elucidated. Here, we report that GPS2 is low expressed in BC tissues and negatively correlated with poor prognosis. Both in vitro and in vivo studies demonstrate that GPS2 suppresses malignant progression of BC. Moreover, GPS2 suppresses aerobic glycolysis in BC cells. Mechanistically, GPS2 destabilizes HIF-1α to reduce the transcription of its downstream glycolytic regulators (PGK1, PGAM1, ENO1, PKM2, LDHA, PDK1, PDK2, and PDK4), and then suppresses cellular aerobic glycolysis. Notably, receptor for activated C kinase 1 (RACK1) is identified as a key ubiquitin ligase for GPS2 to promote HIF-1α degradation. GPS2 stabilizes the binding of HIF-1α to RACK1 by directly binding to RACK1, resulting in polyubiquitination and instability of HIF-1α. Amino acid residues 70-92 aa of the GPS2 N-terminus bind RACK1. A 23-amino-acid-long GPS2-derived peptide was developed based on this N-terminal region, which promotes the interaction of RACK1 with HIF-1α, downregulates HIF-1α expression and significantly suppresses BC tumorigenesis in vitro and in vivo. In conclusion, our findings indicate that GPS2 decreases the stability of HIF-1α, which in turn suppresses aerobic glycolysis and tumorigenesis in BC, suggesting that targeting HIF-1α degradation and treating with peptides may be a promising approach to treat BC.
Collapse
Affiliation(s)
- Yuan Si
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China.
| | - Hongling Ou
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xin Jin
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Institute of Modern Biology, Nanjing University, Nanjing, Jiangsu, China
| | - Manxiang Gu
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Songran Sheng
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Wenkang Peng
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Dan Yang
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xiangrong Zhan
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Liang Zhang
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Qingqing Yu
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xuewen Liu
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, China.
| | - Ying Liu
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, China.
| |
Collapse
|
5
|
Gao Y, Kwan J, Orofino J, Burrone G, Mitra S, Fan TY, English J, Hekman R, Emili A, Lyons SM, Cardamone MD, Perissi V. Inhibition of K63 ubiquitination by G-Protein pathway suppressor 2 (GPS2) regulates mitochondria-associated translation. Pharmacol Res 2024; 207:107336. [PMID: 39094987 PMCID: PMC11905147 DOI: 10.1016/j.phrs.2024.107336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 07/29/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024]
Abstract
G-Protein Pathway Suppressor 2 (GPS2) is an inhibitor of non-proteolytic K63 ubiquitination mediated by the E2 ubiquitin-conjugating enzyme Ubc13. Previous studies have associated GPS2-mediated restriction of ubiquitination with the regulation of insulin signaling, inflammatory responses and mitochondria-nuclear communication across different tissues and cell types. However, a detailed understanding of the targets of GPS2/Ubc13 activity is lacking. Here, we have dissected the GPS2-regulated K63 ubiquitome in mouse embryonic fibroblasts and human breast cancer cells, unexpectedly finding an enrichment for proteins involved in RNA binding and translation on the outer mitochondrial membrane. Validation of selected targets of GPS2-mediated regulation, including the RNA-binding protein PABPC1 and translation factors RPS1, RACK1 and eIF3M, revealed a mitochondrial-specific strategy for regulating the translation of nuclear-encoded mitochondrial proteins via non-proteolytic ubiquitination. Removal of GPS2-mediated inhibition, either via genetic deletion or stress-induced nuclear translocation, promotes the import-coupled translation of selected mRNAs leading to the increased expression of an adaptive antioxidant program. In light of GPS2 role in nuclear-mitochondria communication, these findings reveal an exquisite regulatory network for modulating mitochondrial gene expression through spatially coordinated transcription and translation.
Collapse
Affiliation(s)
- Yuan Gao
- Department of Biochemistry and Cell Biology, Chobanian&Avedisian School of Medicine, Boston University, Boston, MA 02115, United States
| | - Julian Kwan
- Department of Biochemistry and Cell Biology, Chobanian&Avedisian School of Medicine, Boston University, Boston, MA 02115, United States; Center for Network and Systems Biology, Boston University, Boston, MA 02115, United States
| | - Joseph Orofino
- Department of Biochemistry and Cell Biology, Chobanian&Avedisian School of Medicine, Boston University, Boston, MA 02115, United States
| | - Giulia Burrone
- Department of Biochemistry and Cell Biology, Chobanian&Avedisian School of Medicine, Boston University, Boston, MA 02115, United States; Department of Computer Science, University of Torino, Torino, Italy; Department of Clinical and Biological Science, University of Torino, Torino, Italy; Graduate Program in Complex Systems for Quantitative Biomedicine, University of Torino, Torino, Italy
| | - Sahana Mitra
- Department of Biochemistry and Cell Biology, Chobanian&Avedisian School of Medicine, Boston University, Boston, MA 02115, United States
| | - Ting-Yu Fan
- Department of Biochemistry and Cell Biology, Chobanian&Avedisian School of Medicine, Boston University, Boston, MA 02115, United States
| | - Justin English
- Department of Biochemistry and Cell Biology, Chobanian&Avedisian School of Medicine, Boston University, Boston, MA 02115, United States; Graduate Program in Pharmacology and Experimental Therapeutics, Chobanian&Avedisian School of Medicine, Boston University, Boston, MA 02115, United States
| | - Ryan Hekman
- Department of Biochemistry and Cell Biology, Chobanian&Avedisian School of Medicine, Boston University, Boston, MA 02115, United States; Center for Network and Systems Biology, Boston University, Boston, MA 02115, United States
| | - Andrew Emili
- Department of Biochemistry and Cell Biology, Chobanian&Avedisian School of Medicine, Boston University, Boston, MA 02115, United States; Center for Network and Systems Biology, Boston University, Boston, MA 02115, United States; Biology Department, Boston University, Boston, MA 02115, United States
| | - Shawn M Lyons
- Department of Biochemistry and Cell Biology, Chobanian&Avedisian School of Medicine, Boston University, Boston, MA 02115, United States
| | - Maria Dafne Cardamone
- Department of Biochemistry and Cell Biology, Chobanian&Avedisian School of Medicine, Boston University, Boston, MA 02115, United States
| | - Valentina Perissi
- Department of Biochemistry and Cell Biology, Chobanian&Avedisian School of Medicine, Boston University, Boston, MA 02115, United States.
| |
Collapse
|
6
|
Hu L, Liu H, Ma H, Zeng X, Cao Y, Liu B, Li H, Zhang X. TRAF6-mediated ubiquitination of AKT1 in the nucleus occurs in a β-arrestin2-dependent manner upon insulin stimulation. Biochem Pharmacol 2024; 226:116362. [PMID: 38871335 DOI: 10.1016/j.bcp.2024.116362] [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: 02/01/2024] [Revised: 05/25/2024] [Accepted: 06/10/2024] [Indexed: 06/15/2024]
Abstract
AKT, also known as protein kinase B (PKB), serves as a crucial regulator of numerous biological functions, including cell growth, metabolism, and tumorigenesis. Increasing evidence suggests that the kinase activity of AKT is regulated via ubiquitination by various E3 ligase enzymes in response to different stimuli. However, the molecular mechanisms underlying insulin-induced AKT ubiquitination are not yet fully understood. Here, we show that activation of the insulin receptor (IR) leads to enhanced ubiquitination of AKT1 at K8 and K14 residues, facilitated by the cytosolic E3 ubiquitin ligase enzyme, TRAF6. Further investigation using AKT1 mutants with modified nucleocytoplasmic shuttling properties reveals that TRAF6-mediated AKT1 ubiquitination occurs within the nucleus in a β-Arr2-dependent manner. The nuclear entry of TRAF6 depends on importin β1, while β-Arr2 regulates this process by facilitating the interaction between TRAF6 and importin β1. Additionally, the ubiquitination of AKT1 is essential for its translocation to the activated IR on the plasma membrane, where it plays a functional role in recruiting Glut4 and facilitating glucose uptake. This study uncovers the cellular components and processes involved in insulin-induced ubiquitination and activation of AKT1, providing insights and detailed strategies for manipulating AKT1.
Collapse
Affiliation(s)
- Li Hu
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, China
| | - Haiping Liu
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, China
| | - Haixiang Ma
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, China
| | - Xingyue Zeng
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, China
| | - Yongkai Cao
- Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen 518035, China
| | - Bing Liu
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, China
| | - Huijun Li
- Department of Pharmaceuticals, People's Hospital of Zunyi City Bo Zhou District, Zunyi 563000, China
| | - Xiaohan Zhang
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, China.
| |
Collapse
|
7
|
Lu Y, Ma WB, Ren GM, Li YT, Wang T, Zhan YQ, Xiang SS, Chen H, Gao HY, Zhao K, Yu M, Li CY, Yang XM, Yin RH. GPS2 promotes erythroid differentiation in K562 erythroleukemia cells primarily via NCOR1. Int J Hematol 2024; 120:157-166. [PMID: 38814500 DOI: 10.1007/s12185-024-03797-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 05/10/2024] [Accepted: 05/20/2024] [Indexed: 05/31/2024]
Abstract
G protein pathway suppressor 2 (GPS2) has been shown to play a pivotal role in human and mouse definitive erythropoiesis in an EKLF-dependent manner. However, whether GPS2 affects human primitive erythropoiesis is still unknown. This study demonstrated that GPS2 positively regulates erythroid differentiation in K562 cells, which have a primitive erythroid phenotype. Overexpression of GPS2 promoted hemin-induced hemoglobin synthesis in K562 cells as assessed by the increased percentage of benzidine-positive cells and the deeper red coloration of the cell pellets. In contrast, knockdown of GPS2 inhibited hemin-induced erythroid differentiation of K562 cells. GPS2 overexpression also enhanced erythroid differentiation of K562 cells induced by cytosine arabinoside (Ara-C). GPS2 induced hemoglobin synthesis by increasing the expression of globin and ALAS2 genes, either under steady state or upon hemin treatment. Promotion of erythroid differentiation of K562 cells by GPS2 mainly relies on NCOR1, as knockdown of NCOR1 or lack of the NCOR1-binding domain of GPS2 potently diminished the promotive effect. Thus, our study revealed a previously unknown role of GPS2 in regulating human primitive erythropoiesis in K562 cells.
Collapse
Affiliation(s)
- Ying Lu
- College of Life Science and Bioengineering, Faculty of Environmental and Life Sciences, Beijing University of Technology, Beijing, 100124, China
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Wen-Bing Ma
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, China
| | - Guang-Ming Ren
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Ya-Ting Li
- College of Life Science and Bioengineering, Faculty of Environmental and Life Sciences, Beijing University of Technology, Beijing, 100124, China
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Ting Wang
- College of Life Science and Bioengineering, Faculty of Environmental and Life Sciences, Beijing University of Technology, Beijing, 100124, China
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Yi-Qun Zhan
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Shen-Si Xiang
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Hui Chen
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Hui-Ying Gao
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Ke Zhao
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Miao Yu
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Chang-Yan Li
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Xiao-Ming Yang
- College of Life Science and Bioengineering, Faculty of Environmental and Life Sciences, Beijing University of Technology, Beijing, 100124, China.
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing, 100850, China.
| | - Rong-Hua Yin
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing, 100850, China.
| |
Collapse
|
8
|
Hu T, Mu C, Li Y, Hao W, Yu X, Wang Y, Han W, Li Q. GPS2 ameliorates cigarette smoking-induced pulmonary vascular remodeling by modulating the ras-Raf-ERK axis. Respir Res 2024; 25:210. [PMID: 38755610 PMCID: PMC11100185 DOI: 10.1186/s12931-024-02831-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 05/01/2024] [Indexed: 05/18/2024] Open
Abstract
BACKGROUND Mitogen-activated protein kinase (MAPK)signaling-mediated smoking-associated pulmonary vascular remodeling (PVR) plays an important role in the pathogenesis of group 3 pulmonary hypertension (PH). And G protein pathway suppressor 2 (GPS2) could suppress G-protein signaling such as Ras and MAPK, but its role in cigarette smoking -induced PVR (CS-PVR) is unclear. METHODS An in vivo model of smoke-exposed rats was constructed to assess the role of GPS2 in smoking-induced PH and PVR. In vitro, the effects of GPS2 overexpression and silencing on the function of human pulmonary arterial smooth cells (HPASMCs) and the underlying mechanisms were explored. RESULTS GPS2 expression was downregulated in rat pulmonary arteries (PAs) and HPASMCs after CS exposure. More importantly, CS-exposed rats with GPS2 overexpression had lower right ventricular systolic pressure (RVSP), right ventricular hypertrophy index (RVHI), and wall thickness (WT%) than those without. And enhanced proliferation and migration of HPASMCs induced by cigarette smoking extract (CSE) can be evidently inhibited by overexpressed GPS2. Besides, GPS2siRNA significantly enhanced the proliferation, and migration of HPASMCs as well as activated Ras and Raf/ERK signaling, while these effects were inhibited by zoledronic acid (ZOL). In addition, GPS2 promoter methylation level in rat PAs and HPASMCs was increased after CS exposure, and 5-aza-2-deoxycytidine (5-aza) inhibited CSE-induced GPS2 hypermethylation and downregulation in vitro. CONCLUSIONS GPS2 overexpression could improve the CS-PVR, suggesting that GPS2 might serve as a novel therapeutic target for PH-COPD in the future.
Collapse
Affiliation(s)
- Ting Hu
- Department of Respiratory and Critical Care Medicine, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
- Qingdao Key Lab of Common Diseases, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, 5 Donghai Middle Road, Qingdao, 266071, China
| | - Chaohui Mu
- Department of Respiratory and Critical Care Medicine, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
- Qingdao Key Lab of Common Diseases, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, 5 Donghai Middle Road, Qingdao, 266071, China
| | - Yanmiao Li
- Department of Respiratory and Critical Care Medicine, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
- Qingdao Key Lab of Common Diseases, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, 5 Donghai Middle Road, Qingdao, 266071, China
| | - Wanming Hao
- Qingdao Key Lab of Common Diseases, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, 5 Donghai Middle Road, Qingdao, 266071, China
- Department of Respiratory and Critical Care Medicine, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, 5 Donghai Middle Road, Qingdao, 266071, China
| | - Xinjuan Yu
- Qingdao Key Lab of Common Diseases, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, 5 Donghai Middle Road, Qingdao, 266071, China
- Department of Respiratory and Critical Care Medicine, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, 5 Donghai Middle Road, Qingdao, 266071, China
| | - Yixuan Wang
- Qingdao Key Lab of Common Diseases, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, 5 Donghai Middle Road, Qingdao, 266071, China
- Department of Respiratory and Critical Care Medicine, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, 5 Donghai Middle Road, Qingdao, 266071, China
| | - Wei Han
- Qingdao Key Lab of Common Diseases, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, 5 Donghai Middle Road, Qingdao, 266071, China.
- Department of Respiratory and Critical Care Medicine, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, 5 Donghai Middle Road, Qingdao, 266071, China.
| | - Qinghai Li
- Qingdao Key Lab of Common Diseases, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, 5 Donghai Middle Road, Qingdao, 266071, China.
- Department of Respiratory and Critical Care Medicine, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, 5 Donghai Middle Road, Qingdao, 266071, China.
| |
Collapse
|
9
|
Ma M, Cao R, Tian Y, Fu X. Ubiquitination and Metabolic Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1466:47-79. [PMID: 39546135 DOI: 10.1007/978-981-97-7288-9_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
The increasing incidence of metabolic diseases, including obesity, type 2 diabetes mellitus (T2DM), and non-alcoholic fatty liver disease (NAFLD), in the past decade is a serious concern worldwide. Disruption of cellular protein homeostasis has been considered as a crucial contributor to the pathogenesis of metabolic diseases. To maintain protein homeostasis, cells have evolved multiple dynamic and self-regulating quality control processes to adapt new environmental conditions and prevent prolonged damage. Among them, the ubiquitin proteasome system (UPS), the primary proteolytic pathway for degradation of aberrant proteins via ubiquitination, has an essential role in maintaining cellular homeostasis in response to intracellular stress. Correspondingly, accumulating evidences have shown that dysregulation of ubiquitination can aggravate various metabolic derangements in many tissues, including the liver, skeletal muscle, pancreas, and adipose tissue, and is involved in the initiation and progression of diverse metabolic diseases. In this part, we will summarize the role of ubiquitination in the pathogenesis of metabolic diseases, including obesity, T2DM and NAFLD, and discuss its potential as a therapeutic target.
Collapse
Affiliation(s)
- Meilin Ma
- Division of Endocrinology and Metabolism, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China
| | - Rong Cao
- Division of Endocrinology and Metabolism, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China
| | - Yan Tian
- Division of Endocrinology and Metabolism, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China
| | - Xianghui Fu
- State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
10
|
Jiang N, Li W, Jiang S, Xie M, Liu R. Acetylation in pathogenesis: Revealing emerging mechanisms and therapeutic prospects. Biomed Pharmacother 2023; 167:115519. [PMID: 37729729 DOI: 10.1016/j.biopha.2023.115519] [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: 07/18/2023] [Revised: 09/08/2023] [Accepted: 09/14/2023] [Indexed: 09/22/2023] Open
Abstract
Protein acetylation modifications play a central and pivotal role in a myriad of biological processes, spanning cellular metabolism, proliferation, differentiation, apoptosis, and beyond, by effectively reshaping protein structure and function. The metabolic state of cells is intricately connected to epigenetic modifications, which in turn influence chromatin status and gene expression patterns. Notably, pathological alterations in protein acetylation modifications are frequently observed in diseases such as metabolic syndrome, cardiovascular disorders, and cancer. Such abnormalities can result in altered protein properties and loss of function, which are closely associated with developing and progressing related diseases. In recent years, the advancement of precision medicine has highlighted the potential value of protein acetylation in disease diagnosis, treatment, and prevention. This review includes provocative and thought-provoking papers outlining recent breakthroughs in acetylation modifications as they relate to cardiovascular disease, mitochondrial metabolic regulation, liver health, neurological health, obesity, diabetes, and cancer. Additionally, it covers the molecular mechanisms and research challenges in understanding the role of acetylation in disease regulation. By summarizing novel targets and prognostic markers for the treatment of related diseases, we aim to contribute to the field. Furthermore, we discuss current hot topics in acetylation research related to health regulation, including N4-acetylcytidine and liquid-liquid phase separation. The primary objective of this review is to provide insights into the functional diversity and underlying mechanisms by which acetylation regulates proteins in disease contexts.
Collapse
Affiliation(s)
- Nan Jiang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Wenyong Li
- School of Biology and Food Engineering, Fuyang Normal University, Fuyang, Anhui 236037, China
| | - Shuanglin Jiang
- School of Biology and Food Engineering, Fuyang Normal University, Fuyang, Anhui 236037, China
| | - Ming Xie
- North China Petroleum Bureau General Hospital, Renqiu 062550, China.
| | - Ran Liu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China.
| |
Collapse
|
11
|
González-Arzola K, Díaz-Quintana A. Mitochondrial Factors in the Cell Nucleus. Int J Mol Sci 2023; 24:13656. [PMID: 37686461 PMCID: PMC10563088 DOI: 10.3390/ijms241713656] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/31/2023] [Accepted: 08/31/2023] [Indexed: 09/10/2023] Open
Abstract
The origin of eukaryotic organisms involved the integration of mitochondria into the ancestor cell, with a massive gene transfer from the original proteobacterium to the host nucleus. Thus, mitochondrial performance relies on a mosaic of nuclear gene products from a variety of genomes. The concerted regulation of their synthesis is necessary for metabolic housekeeping and stress response. This governance involves crosstalk between mitochondrial, cytoplasmic, and nuclear factors. While anterograde and retrograde regulation preserve mitochondrial homeostasis, the mitochondria can modulate a wide set of nuclear genes in response to an extensive variety of conditions, whose response mechanisms often merge. In this review, we summarise how mitochondrial metabolites and proteins-encoded either in the nucleus or in the organelle-target the cell nucleus and exert different actions modulating gene expression and the chromatin state, or even causing DNA fragmentation in response to common stress conditions, such as hypoxia, oxidative stress, unfolded protein stress, and DNA damage.
Collapse
Affiliation(s)
- Katiuska González-Arzola
- Centro Andaluz de Biología Molecular y Medicina Regenerativa—CABIMER, Consejo Superior de Investigaciones Científicas—Universidad de Sevilla—Universidad Pablo de Olavide, 41092 Seville, Spain
- Departamento de Bioquímica Vegetal y Biología Molecular, Universidad de Sevilla, 41012 Seville, Spain
| | - Antonio Díaz-Quintana
- Departamento de Bioquímica Vegetal y Biología Molecular, Universidad de Sevilla, 41012 Seville, Spain
- Instituto de Investigaciones Químicas—cicCartuja, Universidad de Sevilla—C.S.I.C, 41092 Seville, Spain
| |
Collapse
|
12
|
Grigorova N, Ivanova Z, Vachkova E, Petrova V, Penev T. DHA-Provoked Reduction in Adipogenesis and Glucose Uptake Could Be Mediated by Gps2 Upregulation in Immature 3T3-L1 Cells. Int J Mol Sci 2023; 24:13325. [PMID: 37686130 PMCID: PMC10487817 DOI: 10.3390/ijms241713325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 08/24/2023] [Accepted: 08/26/2023] [Indexed: 09/10/2023] Open
Abstract
The signaling pathway of fatty acids in the context of obesity is an extensively explored topic, yet their primary mechanism of action remains incompletely understood. This study aims to examine the effect of docosahexaenoic acid (DHA) on some crucial aspects of adipogenesis in differentiating 3T3-L1 cells, using palmitic acid-treated (PA), standard differentiated, and undifferentiated adipocytes as controls. Employing 60 µM DHA or PA, 3T3-L1 preadipocytes were treated from the onset of adipogenesis, with negative and positive controls included. After eight days, we performed microscopic observations, cell viability assays, the determination of adiponectin concentration, intracellular lipid accumulation, and gene expression analysis. Our findings demonstrated that DHA inhibits adipogenesis, lipolysis, and glucose uptake by suppressing peroxisome proliferator-activated receptor gamma (Pparg) and G-protein coupled receptor 120 (Gpr120) gene expression. Cell cytotoxicity was ruled out as a causative factor, and β-oxidation involvement was suspected. These results challenge the conventional belief that omega-3 fatty acids, acting as Pparg and Gpr120 agonists, promote adipogenesis and enhance insulin-dependent glucose cell flux. Moreover, we propose a novel hypothesis suggesting the key role of the co-repressor G protein pathway suppressor 2 in mediating this process. Additional investigations are required to elucidate the molecular mechanisms driving DHA's anti-adipogenic effect and its broader health implications.
Collapse
Affiliation(s)
- Natalia Grigorova
- Department of Pharmacology, Animal Physiology, Biochemistry and Chemistry, Faculty of Veterinary Medicine, Trakia University, 6000 Stara Zagora, Bulgaria; (Z.I.); (E.V.); (V.P.)
| | - Zhenya Ivanova
- Department of Pharmacology, Animal Physiology, Biochemistry and Chemistry, Faculty of Veterinary Medicine, Trakia University, 6000 Stara Zagora, Bulgaria; (Z.I.); (E.V.); (V.P.)
| | - Ekaterina Vachkova
- Department of Pharmacology, Animal Physiology, Biochemistry and Chemistry, Faculty of Veterinary Medicine, Trakia University, 6000 Stara Zagora, Bulgaria; (Z.I.); (E.V.); (V.P.)
| | - Valeria Petrova
- Department of Pharmacology, Animal Physiology, Biochemistry and Chemistry, Faculty of Veterinary Medicine, Trakia University, 6000 Stara Zagora, Bulgaria; (Z.I.); (E.V.); (V.P.)
| | - Toncho Penev
- Department of Ecology and Animal Hygiene, Faculty of Agriculture, Trakia University, 6000 Stara Zagora, Bulgaria;
| |
Collapse
|
13
|
English J, Orofino J, Cederquist CT, Paul I, Li H, Auwerx J, Emili A, Belkina A, Cardamone D, Perissi V. GPS2-mediated regulation of the adipocyte secretome modulates adipose tissue remodeling at the onset of diet-induced obesity. Mol Metab 2023; 69:101682. [PMID: 36731652 PMCID: PMC9922684 DOI: 10.1016/j.molmet.2023.101682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 01/22/2023] [Indexed: 02/01/2023] Open
Abstract
OBJECTIVE Dysfunctional, unhealthy expansion of white adipose tissue due to excess dietary intake is a process at the root of obesity and Type 2 Diabetes development. The objective of this study is to contribute to a better understanding of the underlying mechanism(s) regulating the early stages of adipose tissue expansion and adaptation to dietary stress due to an acute, high-fat diet (HFD) challenge, with a focus on the communication between adipocytes and other stromal cells. METHODS We profiled the early response to high-fat diet exposure in wildtype and adipocyte-specific GPS2-KO (GPS2-AKO) mice at the cellular, tissue and organismal level. A multi-pronged approach was employed to disentangle the complex cellular interactions dictating tissue remodeling, via single-cell RNA sequencing and FACS profiling of the stromal fraction, and semi-quantitative proteomics of the adipocyte-derived exosomal cargo after 5 weeks of HFD feeding. RESULTS Our results indicate that loss of GPS2 in mature adipocytes leads to impaired adaptation to the metabolic stress imposed by HFD feeding. GPS2-AKO mice are significantly more inflamed, insulin resistant, and obese, compared to the WT counterparts. At the cellular level, lack of GPS2 in adipocytes impacts upon other stromal populations, with both the eWAT and scWAT depots exhibiting changes in the immune and non-immune compartments that contribute to an increase in inflammatory and anti-adipogenic cell types. Our studies also revealed that adipocyte to stromal cell communication is facilitated by exosomes, and that transcriptional rewiring of the exosomal cargo is crucial for tissue remodeling. Loss of GPS2 results in increased expression of secreted factors promoting a TGFβ-driven fibrotic microenvironment favoring unhealthy tissue remodeling and expansion. CONCLUSIONS Adipocytes serve as an intercellular signaling hub, communicating with the stromal compartment via paracrine signaling. Our study highlights the importance of proper regulation of the 'secretome' released by energetically stressed adipocytes at the onset of obesity. Altered transcriptional regulation of factors secreted via adipocyte-derived exosomes (AdExos), in the absence of GPS2, contributes to the establishment of an anti-adipogenic, pro-fibrotic adipose tissue environment, and to hastened progression towards a metabolically dysfunctional phenotype.
Collapse
Affiliation(s)
- Justin English
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA; Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA.
| | - Joseph Orofino
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA.
| | - Carly T. Cederquist
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Indranil Paul
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA; Center for Network Systems Biology, Boston University, Boston, MA, USA.
| | - Hao Li
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland.
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland.
| | - Andrew Emili
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA; Center for Network Systems Biology, Boston University, Boston, MA, USA.
| | - Anna Belkina
- Flow Cytometry Core Facility, Boston University School of Medicine, Boston, MA, USA; Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA.
| | - Dafne Cardamone
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA.
| | - Valentina Perissi
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA; School of Life Science, Northwestern Polytechnical University, Xi'an 710072, China.
| |
Collapse
|
14
|
Richter HJ, Hauck AK, Batmanov K, Inoue SI, So BN, Kim M, Emmett MJ, Cohen RN, Lazar MA. Balanced control of thermogenesis by nuclear receptor corepressors in brown adipose tissue. Proc Natl Acad Sci U S A 2022; 119:e2205276119. [PMID: 35939699 PMCID: PMC9388101 DOI: 10.1073/pnas.2205276119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 07/07/2022] [Indexed: 11/18/2022] Open
Abstract
Brown adipose tissue (BAT) is a key thermogenic organ whose expression of uncoupling protein 1 (UCP1) and ability to maintain body temperature in response to acute cold exposure require histone deacetylase 3 (HDAC3). HDAC3 exists in tight association with nuclear receptor corepressors (NCoRs) NCoR1 and NCoR2 (also known as silencing mediator of retinoid and thyroid receptors [SMRT]), but the functions of NCoR1/2 in BAT have not been established. Here we report that as expected, genetic loss of NCoR1/2 in BAT (NCoR1/2 BAT-dKO) leads to loss of HDAC3 activity. In addition, HDAC3 is no longer bound at its physiological genomic sites in the absence of NCoR1/2, leading to a shared deregulation of BAT lipid metabolism between NCoR1/2 BAT-dKO and HDAC3 BAT-KO mice. Despite these commonalities, loss of NCoR1/2 in BAT does not phenocopy the cold sensitivity observed in HDAC3 BAT-KO, nor does loss of either corepressor alone. Instead, BAT lacking NCoR1/2 is inflamed, particularly with respect to the interleukin-17 axis that increases thermogenic capacity by enhancing innervation. Integration of BAT RNA sequencing and chromatin immunoprecipitation sequencing data revealed that NCoR1/2 directly regulate Mmp9, which integrates extracellular matrix remodeling and inflammation. These findings reveal pleiotropic functions of the NCoR/HDAC3 corepressor complex in BAT, such that HDAC3-independent suppression of BAT inflammation counterbalances stimulation of HDAC3 activity in the control of thermogenesis.
Collapse
Affiliation(s)
- Hannah J. Richter
- Biochemistry and Molecular Biophysics Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| | - Amy K. Hauck
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| | - Kirill Batmanov
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| | - Shin-Ichi Inoue
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| | - Bethany N. So
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| | - Mindy Kim
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| | - Matthew J. Emmett
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| | - Ronald N. Cohen
- Section of Endocrinology, Diabetes, and Metabolism, University of Chicago, Chicago, IL 60637
| | - Mitchell A. Lazar
- Biochemistry and Molecular Biophysics Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| |
Collapse
|
15
|
Non-proteolytic ubiquitylation in cellular signaling and human disease. Commun Biol 2022; 5:114. [PMID: 35136173 PMCID: PMC8826416 DOI: 10.1038/s42003-022-03060-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 01/18/2022] [Indexed: 12/18/2022] Open
Abstract
Ubiquitylation is one of the most common post-translational modifications (PTMs) of proteins that frequently targets substrates for proteasomal degradation. However it can also result in non-proteolytic events which play important functions in cellular processes such as intracellular signaling, membrane trafficking, DNA repair and cell cycle. Emerging evidence demonstrates that dysfunction of non-proteolytic ubiquitylation is associated with the development of multiple human diseases. In this review, we summarize the current knowledge and the latest concepts on how non-proteolytic ubiquitylation pathways are involved in cellular signaling and in disease-mediating processes. Our review, may advance our understanding of the non-degradative ubiquitylation process. Evanthia Pangou and co-authors review recent insights into the important roles of non-proteolytic ubiquitylation in cellular signaling as well as in physiology and disease.
Collapse
|
16
|
The Deubiquitinase OTUB1 Is a Key Regulator of Energy Metabolism. Int J Mol Sci 2022; 23:ijms23031536. [PMID: 35163456 PMCID: PMC8836018 DOI: 10.3390/ijms23031536] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 01/26/2022] [Accepted: 01/27/2022] [Indexed: 11/17/2022] Open
Abstract
Dysregulated energy metabolism is a major contributor to a multitude of pathologies, including obesity and diabetes. Understanding the regulation of metabolic homeostasis is of utmost importance for the identification of therapeutic targets for the treatment of metabolically driven diseases. We previously identified the deubiquitinase OTUB1 as substrate for the cellular oxygen sensor factor-inhibiting HIF (FIH) with regulatory effects on cellular energy metabolism, but the physiological relevance of OTUB1 is unclear. Here, we report that the induced global deletion of OTUB1 in adult mice (Otub1 iKO) elevated energy expenditure, reduced age-dependent body weight gain, facilitated blood glucose clearance and lowered basal plasma insulin levels. The respiratory exchange ratio was maintained, indicating an unaltered nutrient oxidation. In addition, Otub1 deletion in cells enhanced AKT activity, leading to a larger cell size, higher ATP levels and reduced AMPK phosphorylation. AKT is an integral part of insulin-mediated signaling and Otub1 iKO mice presented with increased AKT phosphorylation following acute insulin administration combined with insulin hypersensitivity. We conclude that OTUB1 is an important regulator of metabolic homeostasis.
Collapse
|
17
|
B Tóth B, Barta Z, Barta ÁB, Fésüs L. Regulatory modules of human thermogenic adipocytes: functional genomics of large cohort and Meta-analysis derived marker-genes. BMC Genomics 2021; 22:886. [PMID: 34895148 PMCID: PMC8665548 DOI: 10.1186/s12864-021-08126-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 10/27/2021] [Indexed: 11/18/2022] Open
Abstract
Background Recently, ProFAT and BATLAS studies identified brown and white adipocytes marker genes based on analysis of large databases. They offered scores to determine the thermogenic status of adipocytes using the gene-expression data of these markers. In this work, we investigated the functional context of these genes. Results Gene Set Enrichment Analyses (KEGG, Reactome) of the BATLAS and ProFAT marker-genes identified pathways deterministic in the formation of brown and white adipocytes. The collection of the annotated proteins of the defined pathways resulted in expanded white and brown characteristic protein-sets, which theoretically contain all functional proteins that could be involved in the formation of adipocytes. Based on our previously obtained RNA-seq data, we visualized the expression profile of these proteins coding genes and found patterns consistent with the two adipocyte phenotypes. The trajectory of the regulatory processes could be outlined by the transcriptional profile of progenitor and differentiated adipocytes, highlighting the importance of suppression processes in browning. Protein interaction network-based functional genomics by STRING, Cytoscape and R-Igraph platforms revealed that different biological processes shape the brown and white adipocytes and highlighted key regulatory elements and modules including GAPDH-CS, DECR1, SOD2, IL6, HRAS, MTOR, INS-AKT, ERBB2 and 4-NFKB, and SLIT-ROBO-MAPK. To assess the potential role of a particular protein in shaping adipocytes, we assigned interaction network location-based scores (betweenness centrality, number of bridges) to them and created a freely accessible platform, the AdipoNET (https//adiponet.com), to conveniently use these data. The Eukaryote Promoter Database predicted the response elements in the UCP1 promoter for the identified, potentially important transcription factors (HIF1A, MYC, REL, PPARG, TP53, AR, RUNX, and FoxO1). Conclusion Our integrative approach-based results allowed us to investigate potential regulatory elements of thermogenesis in adipose tissue. The analyses revealed that some unique biological processes form the brown and white adipocyte phenotypes, which presumes the existence of the transitional states. The data also suggests that the two phenotypes are not mutually exclusive, and differentiation of thermogenic adipocyte requires induction of browning as well as repressions of whitening. The recognition of these simultaneous actions and the identified regulatory modules can open new direction in obesity research. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-08126-8.
Collapse
Affiliation(s)
- Beáta B Tóth
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Egyetem Tér 1, Debrecen, H-4032, Hungary.
| | - Zoltán Barta
- MTA-DE Behavioural Ecology Research Group, Department of Evolutionary Zoology and Human Biology, University of Debrecen, Egyetem tér 1, Debrecen, H-4032, Hungary
| | - Ákos Barnabás Barta
- Vienna University of Economics and Business (WU), Welthandelspl. 1, 1020, Wien, Austria
| | - László Fésüs
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Egyetem Tér 1, Debrecen, H-4032, Hungary.
| |
Collapse
|
18
|
Barilla S, Treuter E, Venteclef N. Transcriptional and epigenetic control of adipocyte remodeling during obesity. Obesity (Silver Spring) 2021; 29:2013-2025. [PMID: 34813171 DOI: 10.1002/oby.23248] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 04/27/2021] [Accepted: 05/07/2021] [Indexed: 01/05/2023]
Abstract
The rising prevalence of obesity over the past decades coincides with the rising awareness that a detailed understanding of both adipose tissue biology and obesity-associated remodeling is crucial for developing therapeutic and preventive strategies. Substantial progress has been made in identifying the signaling pathways and transcriptional networks that orchestrate alterations of adipocyte gene expression linked to diverse phenotypes. Owing to recent advances in epigenomics, we also gained a better appreciation for the fact that different environmental cues can epigenetically reprogram adipocyte fate and function, mainly by altering DNA methylation and histone modification patterns. Intriguingly, it appears that transcription factors and chromatin-modifying coregulator complexes are the key regulatory components that coordinate both signaling-induced transcriptional and epigenetic alterations in adipocytes. In this review, we summarize and discuss current molecular insights into how these alterations and the involved regulatory components trigger adipogenesis and adipose tissue remodeling in response to energy surplus.
Collapse
Affiliation(s)
- Serena Barilla
- Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden
| | - Eckardt Treuter
- Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden
| | - Nicolas Venteclef
- Cordeliers Research Center, Inserm, University of Paris, IMMEDIAB Laboratory, Paris, France
- Inovarion, Paris, France
| |
Collapse
|
19
|
McKenna M, Balasuriya N, Zhong S, Li SSC, O'Donoghue P. Phospho-Form Specific Substrates of Protein Kinase B (AKT1). Front Bioeng Biotechnol 2021; 8:619252. [PMID: 33614606 PMCID: PMC7886700 DOI: 10.3389/fbioe.2020.619252] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 12/29/2020] [Indexed: 11/17/2022] Open
Abstract
Protein kinase B (AKT1) is hyper-activated in diverse human tumors. AKT1 is activated by phosphorylation at two key regulatory sites, Thr308 and Ser473. Active AKT1 phosphorylates many, perhaps hundreds, of downstream cellular targets in the cytosol and nucleus. AKT1 is well-known for phosphorylating proteins that regulate cell survival and apoptosis, however, the full catalog of AKT1 substrates remains unknown. Using peptide arrays, we recently discovered that each phosphorylated form of AKT1 (pAKT1S473, pAKT1T308, and ppAKT1S473,T308) has a distinct substrate specificity, and these data were used to predict potential new AKT1 substrates. To test the high-confidence predictions, we synthesized target peptides representing putative AKT1 substrates. Peptides substrates were synthesized by solid phase synthesis and their purity was confirmed by mass spectrometry. Most of the predicted peptides showed phosphate accepting activity similar to or greater than that observed with a peptide derived from a well-established AKT1 substrate, glycogen synthase kinase 3β (GSK-3β). Among the novel substrates, AKT1 was most active with peptides representing PIP3-binding protein Rab11 family-interacting protein 2 and cysteinyl leukotriene receptor 1, indicating their potential role in AKT1-dependent cellular signaling. The ppAKT1S473,T308 enzyme was highly selective for peptides containing a patch of basic residues at −5, −4, −3 and aromatic residues (Phe/Tyr) at +1 positions from the phosphorylation site. The pAKT1S473 variant preferred more acidic peptides, Ser or Pro at +4, and was agnostic to the residue at −5. The data further support our hypothesis that Ser473 phosphorylation plays a key role in modulating AKT1 substrate selectivity.
Collapse
Affiliation(s)
- McShane McKenna
- Department of Biochemistry, The University of Western Ontario, London, ON, Canada
| | - Nileeka Balasuriya
- Department of Biochemistry, The University of Western Ontario, London, ON, Canada
| | - Shanshan Zhong
- Department of Biochemistry, The University of Western Ontario, London, ON, Canada
| | - Shawn Shun-Cheng Li
- Department of Biochemistry, The University of Western Ontario, London, ON, Canada
| | - Patrick O'Donoghue
- Department of Biochemistry, The University of Western Ontario, London, ON, Canada.,Department of Chemistry, The University of Western Ontario, London, ON, Canada
| |
Collapse
|
20
|
GPS2 promotes erythroid differentiation by control of the stability of EKLF protein. Blood 2021; 135:2302-2315. [PMID: 32384137 DOI: 10.1182/blood.2019003867] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 03/05/2020] [Indexed: 02/08/2023] Open
Abstract
Erythropoiesis is a complex multistage process that involves differentiation of early erythroid progenitors to enucleated mature red blood cells, in which lineage-specific transcription factors play essential roles. Erythroid Krüppel-like factor (EKLF/KLF1) is a pleiotropic erythroid transcription factor that is required for the proper maturation of the erythroid cells, whose expression and activation are tightly controlled in a temporal and differentiation stage-specific manner. Here, we uncover a novel role of G-protein pathway suppressor 2 (GPS2), a subunit of the nuclear receptor corepressor/silencing mediator of retinoic acid and thyroid hormone receptor corepressor complex, in erythrocyte differentiation. Our study demonstrates that knockdown of GPS2 significantly suppresses erythroid differentiation of human CD34+ cells cultured in vitro and xenotransplanted in nonobese diabetic/severe combined immunodeficiency/interleukin-2 receptor γ-chain null mice. Moreover, global deletion of GPS2 in mice causes impaired erythropoiesis in the fetal liver and leads to severe anemia. Flow cytometric analysis and Wright-Giemsa staining show a defective differentiation at late stages of erythropoiesis in Gps2-/- embryos. Mechanistically, GPS2 interacts with EKLF and prevents proteasome-mediated degradation of EKLF, thereby increasing EKLF stability and transcriptional activity. Moreover, we identify the amino acids 191-230 region in EKLF protein, responsible for GPS2 binding, that is highly conserved in mammals and essential for EKLF protein stability. Collectively, our study uncovers a previously unknown role of GPS2 as a posttranslational regulator that enhances the stability of EKLF protein and thereby promotes erythroid differentiation.
Collapse
|
21
|
Blaustein M, Piegari E, Martínez Calejman C, Vila A, Amante A, Manese MV, Zeida A, Abrami L, Veggetti M, Guertin DA, van der Goot FG, Corvi MM, Colman-Lerner A. Akt Is S-Palmitoylated: A New Layer of Regulation for Akt. Front Cell Dev Biol 2021; 9:626404. [PMID: 33659252 PMCID: PMC7917195 DOI: 10.3389/fcell.2021.626404] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 01/25/2021] [Indexed: 11/13/2022] Open
Abstract
The protein kinase Akt/PKB participates in a great variety of processes, including translation, cell proliferation and survival, as well as malignant transformation and viral infection. In the last few years, novel Akt posttranslational modifications have been found. However, how these modification patterns affect Akt subcellular localization, target specificity and, in general, function is not thoroughly understood. Here, we postulate and experimentally demonstrate by acyl-biotin exchange (ABE) assay and 3H-palmitate metabolic labeling that Akt is S-palmitoylated, a modification related to protein sorting throughout subcellular membranes. Mutating cysteine 344 into serine blocked Akt S-palmitoylation and diminished its phosphorylation at two key sites, T308 and T450. Particularly, we show that palmitoylation-deficient Akt increases its recruitment to cytoplasmic structures that colocalize with lysosomes, a process stimulated during autophagy. Finally, we found that cysteine 344 in Akt1 is important for proper its function, since Akt1-C344S was unable to support adipocyte cell differentiation in vitro. These results add an unexpected new layer to the already complex Akt molecular code, improving our understanding of cell decision-making mechanisms such as cell survival, differentiation and death.
Collapse
Affiliation(s)
- Matías Blaustein
- Departamento de Fisiología, Biología Molecular y Celular (DFBMC), Facultad de Ciencias Exactas y Naturales (FCEN), Universidad de Buenos Aires (UBA), Buenos Aires, Argentina.,Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-UBA, Buenos Aires, Argentina.,Instituto de Biociencias, Biotecnología y Biología Traslacional (iB3), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Estefanía Piegari
- Departamento de Fisiología, Biología Molecular y Celular (DFBMC), Facultad de Ciencias Exactas y Naturales (FCEN), Universidad de Buenos Aires (UBA), Buenos Aires, Argentina.,Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-UBA, Buenos Aires, Argentina
| | - Camila Martínez Calejman
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, United States
| | - Antonella Vila
- Departamento de Fisiología, Biología Molecular y Celular (DFBMC), Facultad de Ciencias Exactas y Naturales (FCEN), Universidad de Buenos Aires (UBA), Buenos Aires, Argentina.,Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-UBA, Buenos Aires, Argentina.,Instituto de Biociencias, Biotecnología y Biología Traslacional (iB3), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Analía Amante
- Departamento de Fisiología, Biología Molecular y Celular (DFBMC), Facultad de Ciencias Exactas y Naturales (FCEN), Universidad de Buenos Aires (UBA), Buenos Aires, Argentina.,Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-UBA, Buenos Aires, Argentina.,Instituto de Biociencias, Biotecnología y Biología Traslacional (iB3), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María Victoria Manese
- Laboratorio de bioquímica y biología celular de parásitos, Instituto Tecnológico de Chascomús (IIB-INTECH), Universidad Nacional de San Martín (UNSAM) - CONICET, Chascomús, Argentina
| | - Ari Zeida
- Departamento de Bioquímica and Centro de Investigaciones Biomédicas (CEINBIO), Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Laurence Abrami
- Global Health Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Mariela Veggetti
- Departamento de Fisiología, Biología Molecular y Celular (DFBMC), Facultad de Ciencias Exactas y Naturales (FCEN), Universidad de Buenos Aires (UBA), Buenos Aires, Argentina.,Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-UBA, Buenos Aires, Argentina
| | - David A Guertin
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, United States.,Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, United States.,Lei Weibo Institute for Rare Diseases, University of Massachusetts Medical School, Worcester, MA, United States
| | - F Gisou van der Goot
- Global Health Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - María Martha Corvi
- Laboratorio de bioquímica y biología celular de parásitos, Instituto Tecnológico de Chascomús (IIB-INTECH), Universidad Nacional de San Martín (UNSAM) - CONICET, Chascomús, Argentina
| | - Alejandro Colman-Lerner
- Departamento de Fisiología, Biología Molecular y Celular (DFBMC), Facultad de Ciencias Exactas y Naturales (FCEN), Universidad de Buenos Aires (UBA), Buenos Aires, Argentina.,Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-UBA, Buenos Aires, Argentina
| |
Collapse
|
22
|
Chan S, Smith E, Gao Y, Kwan J, Blum BC, Tilston-Lunel AM, Turcinovic I, Varelas X, Cardamone MD, Monti S, Emili A, Perissi V. Loss of G-Protein Pathway Suppressor 2 Promotes Tumor Growth Through Activation of AKT Signaling. Front Cell Dev Biol 2021; 8:608044. [PMID: 33490071 PMCID: PMC7817781 DOI: 10.3389/fcell.2020.608044] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 12/04/2020] [Indexed: 02/06/2023] Open
Abstract
G Protein Suppressor 2 (GPS2) is a multifunctional protein that exerts important roles in inflammation and metabolism in adipose, liver, and immune cells. GPS2 has recently been identified as a significantly mutated gene in breast cancer and other malignancies and proposed to work as a putative tumor suppressor. However, molecular mechanisms by which GPS2 prevents cancer development and/or progression are largely unknown. Here, we have profiled the phenotypic changes induced by GPS2 depletion in MDA-MB-231 triple negative breast cancer cells and investigated the underlying molecular mechanisms. We found that GPS2-deleted MDA-MB-231 cells exhibited increased proliferative, migratory, and invasive properties in vitro, and conferred greater tumor burden in vivo in an orthotopic xenograft mouse model. Transcriptomic, proteomic and phospho-proteomic profiling of GPS2-deleted MBA-MB-231 revealed a network of altered signals that relate to cell growth and PI3K/AKT signaling. Overlay of GPS2-regulated gene expression with MDA-MB-231 cells modified to express constitutively active AKT showed significant overlap, suggesting that sustained AKT activation is associated with loss of GPS2. Accordingly, we demonstrate that the pro-oncogenic phenotypes associated with GPS2 deletion are rescued by pharmacological inhibition of AKT with MK2206. Collectively, these observations confirm a tumor suppressor role for GPS2 and reveal that loss of GPS2 promotes breast cancer cell proliferation and tumor growth through uncontrolled activation of AKT signaling. Moreover, our study points to GPS2 as a potential biomarker for a subclass of breast cancers that would be responsive to PI3K-class inhibitor drugs.
Collapse
Affiliation(s)
- Stefanie Chan
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, United States
| | - Emma Smith
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, United States
| | - Yuan Gao
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, United States
| | - Julian Kwan
- Center for Network Systems Biology, Boston University, Boston, MA, United States
| | - Benjamin C. Blum
- Center for Network Systems Biology, Boston University, Boston, MA, United States
| | | | - Isabella Turcinovic
- Center for Network Systems Biology, Boston University, Boston, MA, United States
| | - Xaralabos Varelas
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, United States
| | - Maria Dafne Cardamone
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, United States
| | - Stefano Monti
- Division of Computational Biology, Department of Medicine, Boston University School of Medicine, Boston, MA, United States
| | - Andrew Emili
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, United States
- Center for Network Systems Biology, Boston University, Boston, MA, United States
| | - Valentina Perissi
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
23
|
English J, Son JM, Cardamone MD, Lee C, Perissi V. Decoding the rosetta stone of mitonuclear communication. Pharmacol Res 2020; 161:105161. [PMID: 32846213 PMCID: PMC7755734 DOI: 10.1016/j.phrs.2020.105161] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/04/2020] [Accepted: 08/14/2020] [Indexed: 12/12/2022]
Abstract
Cellular homeostasis in eukaryotic cells requires synchronized coordination of multiple organelles. A key role in this stage is played by mitochondria, which have recently emerged as highly interconnected and multifunctional hubs that process and coordinate diverse cellular functions. Beyond producing ATP, mitochondria generate key metabolites and are central to apoptotic and metabolic signaling pathways. Because most mitochondrial proteins are encoded in the nuclear genome, the biogenesis of new mitochondria and the maintenance of mitochondrial functions and flexibility critically depend upon effective mitonuclear communication. This review addresses the complex network of signaling molecules and pathways allowing mitochondria-nuclear communication and coordinated regulation of their independent but interconnected genomes, and discusses the extent to which dynamic communication between the two organelles has evolved for mutual benefit and for the overall maintenance of cellular and organismal fitness.
Collapse
Affiliation(s)
- Justin English
- Department of Biochemistry, Boston University, Boston, MA, 02115, USA; Graduate Program in Biomolecular Pharmacology, Department of Pharmacology and Experimental Therapeutics, Boston University, Boston, MA, 02115, USA
| | - Jyung Mean Son
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | | | - Changhan Lee
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA; USC Norris Comprehensive Cancer Center, Los Angeles, CA, 90089, USA; Biomedical Sciences, Graduate School, Ajou University, Suwon, 16499, South Korea
| | - Valentina Perissi
- Department of Biochemistry, Boston University, Boston, MA, 02115, USA.
| |
Collapse
|
24
|
Drareni K, Ballaire R, Alzaid F, Goncalves A, Chollet C, Barilla S, Nguewa JL, Dias K, Lemoine S, Riveline JP, Roussel R, Dalmas E, Velho G, Treuter E, Gautier JF, Venteclef N. Adipocyte Reprogramming by the Transcriptional Coregulator GPS2 Impacts Beta Cell Insulin Secretion. Cell Rep 2020; 32:108141. [PMID: 32937117 PMCID: PMC7495095 DOI: 10.1016/j.celrep.2020.108141] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 07/03/2020] [Accepted: 08/21/2020] [Indexed: 01/10/2023] Open
Abstract
Glucose homeostasis is maintained through organ crosstalk that regulates secretion of insulin to keep blood glucose levels within a physiological range. In type 2 diabetes, this coordinated response is altered, leading to a deregulation of beta cell function and inadequate insulin secretion. Reprogramming of white adipose tissue has a central role in this deregulation, but the critical regulatory components remain unclear. Here, we demonstrate that expression of the transcriptional coregulator GPS2 in white adipose tissue is correlated with insulin secretion rate in humans. The causality of this relationship is confirmed using adipocyte-specific GPS2 knockout mice, in which inappropriate secretion of insulin promotes glucose intolerance. This phenotype is driven by adipose-tissue-secreted factors, which cause increased pancreatic islet inflammation and impaired beta cell function. Thus, our study suggests that, in mice and in humans, GPS2 controls the reprogramming of white adipocytes to influence pancreatic islet function and insulin secretion.
Collapse
Affiliation(s)
- Karima Drareni
- Cordeliers Research Centre, INSERM, Immunity and Metabolism in Diabetes Laboratory, Sorbonne Université, Université de Paris, 75006 Paris, France.
| | | | - Fawaz Alzaid
- Cordeliers Research Centre, INSERM, Immunity and Metabolism in Diabetes Laboratory, Sorbonne Université, Université de Paris, 75006 Paris, France
| | - Andreia Goncalves
- Cordeliers Research Centre, INSERM, Immunity and Metabolism in Diabetes Laboratory, Sorbonne Université, Université de Paris, 75006 Paris, France
| | - Catherine Chollet
- Cordeliers Research Centre, INSERM, Immunity and Metabolism in Diabetes Laboratory, Sorbonne Université, Université de Paris, 75006 Paris, France
| | - Serena Barilla
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge 14157, Sweden
| | - Jean-Louis Nguewa
- Department of Diabetes, Clinical Investigation Centre (CIC-9504), Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Karine Dias
- École Normale Supérieure, PSL Research University, Centre National de la Recherche Scientifique (CNRS), INSERM, Institut de Biologie de l'École Normale Supérieure (IBENS), Plateforme Génomique, Paris, France
| | - Sophie Lemoine
- École Normale Supérieure, PSL Research University, Centre National de la Recherche Scientifique (CNRS), INSERM, Institut de Biologie de l'École Normale Supérieure (IBENS), Plateforme Génomique, Paris, France
| | - Jean-Pierre Riveline
- Cordeliers Research Centre, INSERM, Immunity and Metabolism in Diabetes Laboratory, Sorbonne Université, Université de Paris, 75006 Paris, France; Department of Diabetes, Clinical Investigation Centre (CIC-9504), Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Ronan Roussel
- Cordeliers Research Centre, INSERM, Immunity and Metabolism in Diabetes Laboratory, Sorbonne Université, Université de Paris, 75006 Paris, France; Department of Diabetology, Endocrinology and Nutrition, DHU FIRE, Bichat Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Elise Dalmas
- Cordeliers Research Centre, INSERM, Immunity and Metabolism in Diabetes Laboratory, Sorbonne Université, Université de Paris, 75006 Paris, France
| | - Gilberto Velho
- Cordeliers Research Centre, INSERM, Immunity and Metabolism in Diabetes Laboratory, Sorbonne Université, Université de Paris, 75006 Paris, France
| | - Eckardt Treuter
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge 14157, Sweden
| | - Jean-François Gautier
- Cordeliers Research Centre, INSERM, Immunity and Metabolism in Diabetes Laboratory, Sorbonne Université, Université de Paris, 75006 Paris, France; Department of Diabetes, Clinical Investigation Centre (CIC-9504), Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Nicolas Venteclef
- Cordeliers Research Centre, INSERM, Immunity and Metabolism in Diabetes Laboratory, Sorbonne Université, Université de Paris, 75006 Paris, France.
| |
Collapse
|
25
|
Barilla S, Liang N, Mileti E, Ballaire R, Lhomme M, Ponnaiah M, Lemoine S, Soprani A, Gautier JF, Amri EZ, Le Goff W, Venteclef N, Treuter E. Loss of G protein pathway suppressor 2 in human adipocytes triggers lipid remodeling by upregulating ATP binding cassette subfamily G member 1. Mol Metab 2020; 42:101066. [PMID: 32798719 PMCID: PMC7509237 DOI: 10.1016/j.molmet.2020.101066] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/05/2020] [Accepted: 08/11/2020] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVE Adipogenesis is critical for adipose tissue remodeling during the development of obesity. While the role of transcription factors in the orchestration of adipogenic pathways is already established, the involvement of coregulators that transduce regulatory signals into epigenome alterations and transcriptional responses remains poorly understood. The aim of our study was to investigate which pathways are controlled by G protein pathway suppressor 2 (GPS2) during the differentiation of human adipocytes. METHODS We generated a unique loss-of-function model by RNAi depletion of GPS2 in human multipotent adipose-derived stem (hMADS) cells. We thoroughly characterized the coregulator depletion-dependent pathway alterations during adipocyte differentiation at the level of transcriptome (RNA-seq), epigenome (ChIP-seq H3K27ac), cistrome (ChIP-seq GPS2), and lipidome. We validated the in vivo relevance of the identified pathways in non-diabetic and diabetic obese patients. RESULTS The loss of GPS2 triggers the reprogramming of cellular processes related to adipocyte differentiation by increasing the responses to the adipogenic cocktail. In particular, GPS2 depletion increases the expression of BMP4, an important trigger for the commitment of fibroblast-like progenitors toward the adipogenic lineage and increases the expression of inflammatory and metabolic genes. GPS2-depleted human adipocytes are characterized by hypertrophy, triglyceride and phospholipid accumulation, and sphingomyelin depletion. These changes are likely a consequence of the increased expression of ATP-binding cassette subfamily G member 1 (ABCG1) that mediates sphingomyelin efflux from adipocytes and modulates lipoprotein lipase (LPL) activity. We identify ABCG1 as a direct transcriptional target, as GPS2 depletion leads to coordinated changes of transcription and H3K27 acetylation at promoters and enhancers that are occupied by GPS2 in wild-type adipocytes. We find that in omental adipose tissue of obese humans, GPS2 levels correlate with ABCG1 levels, type 2 diabetic status, and lipid metabolic status, supporting the in vivo relevance of the hMADS cell-derived in vitro data. CONCLUSION Our study reveals a dual regulatory role of GPS2 in epigenetically modulating the chromatin landscape and gene expression during human adipocyte differentiation and identifies a hitherto unknown GPS2-ABCG1 pathway potentially linked to adipocyte hypertrophy in humans.
Collapse
Affiliation(s)
- Serena Barilla
- Department of Biosciences and Nutrition, Karolinska Institute, 14183 Huddinge, Sweden.
| | - Ning Liang
- Department of Biosciences and Nutrition, Karolinska Institute, 14183 Huddinge, Sweden
| | - Enrichetta Mileti
- Department of Biosciences and Nutrition, Karolinska Institute, 14183 Huddinge, Sweden
| | - Raphaëlle Ballaire
- Centre de Recherche des Cordeliers, Inserm, University of Paris, IMMEDIAB Laboratory, F-75006, Paris, France; Inovarion, Paris, France
| | - Marie Lhomme
- ICANalytics Lipidomic, Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
| | - Maharajah Ponnaiah
- ICANalytics Lipidomic, Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
| | - Sophie Lemoine
- École Normale Supérieure, PSL Research University, Centre National de la Recherche Scientifique (CNRS), Inserm, Institut de Biologie de l'École Normale Supérieure (IBENS), Plateforme Génomique, Paris, France
| | - Antoine Soprani
- Centre de Recherche des Cordeliers, Inserm, University of Paris, IMMEDIAB Laboratory, F-75006, Paris, France; Department of Digestive Surgery, Générale de Santé (GDS), Geoffroy Saint Hilaire Clinic, 75005, Paris, France
| | - Jean-Francois Gautier
- Centre de Recherche des Cordeliers, Inserm, University of Paris, IMMEDIAB Laboratory, F-75006, Paris, France; Lariboisière Hospital, AP-HP, Diabetology Department, University of Paris, Paris, France
| | - Ez-Zoubir Amri
- University of Côte d'Azur, CNRS, Inserm, iBV, Nice, France
| | - Wilfried Le Goff
- Sorbonne University, Inserm, Institute of Cardiometabolism and Nutrition (ICAN), UMR_S1166, Hôpital de la Pitié, Paris, F-75013, France
| | - Nicolas Venteclef
- Centre de Recherche des Cordeliers, Inserm, University of Paris, IMMEDIAB Laboratory, F-75006, Paris, France; Lariboisière Hospital, AP-HP, Diabetology Department, University of Paris, Paris, France
| | - Eckardt Treuter
- Department of Biosciences and Nutrition, Karolinska Institute, 14183 Huddinge, Sweden.
| |
Collapse
|
26
|
Zhu Y, Jiang X, Ye P, Wang Z, Zheng Y, Liu Z, Chen S, Zhang D. Knockout of AKAP150 improves impaired BK channel-mediated vascular dysfunction through the Akt/GSK3β signalling pathway in diabetes mellitus. J Cell Mol Med 2020; 24:4716-4725. [PMID: 32163656 PMCID: PMC7176888 DOI: 10.1111/jcmm.15143] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 02/05/2020] [Accepted: 02/19/2020] [Indexed: 12/15/2022] Open
Abstract
Vascular dysfunction resulting from diabetes is an important factor in arteriosclerosis. Previous studies have shown that during hyperglycaemia and diabetes, AKAP150 promotes vascular tone enhancement by intensifying the remodelling of the BK channel. However, the interaction between AKAP150 and the BK channel remains open to discussion. In this study, we investigated the regulation of impaired BK channel-mediated vascular dysfunction in diabetes mellitus. Using AKAP150 null mice (AKAP150-/- ) and wild-type (WT) control mice (C57BL/6J), diabetes was induced by intraperitoneal injection of streptozotocin. We found that knockout of AKAP150 reversed vascular remodelling and fibrosis in mice with diabetes and in AKAP150-/- diabetic mice. Impaired Akt/GSK3β signalling contributed to decreased BK-β1 expression in aortas from diabetic mice, and the silencing of AKAP150 increased Akt phosphorylation and BK-β1 expression in MOVAS cells treated with HG medium. The inhibition of Akt activity caused a decrease in BK-β1 expression, and treatment with AKAP150 siRNA suppressed GSK3β expression in the nuclei of MOVAS cells treated with HG. Knockout of AKAP150 reverses impaired BK channel-mediated vascular dysfunction through the Akt/GSK3β signalling pathway in diabetes mellitus.
Collapse
Affiliation(s)
- Yan‐Rong Zhu
- Department of CardiologyNanjing First HospitalNanjing Medical UniversityNanjingChina
| | - Xiao‐Xin Jiang
- Department of CardiologyNanjing First HospitalNanjing Medical UniversityNanjingChina
| | - Peng Ye
- Department of CardiologyNanjing First HospitalNanjing Medical UniversityNanjingChina
| | - Zhi‐Mei Wang
- Department of CardiologyNanjing First HospitalNanjing Medical UniversityNanjingChina
| | - Yaguo Zheng
- Department of CardiologyNanjing First HospitalNanjing Medical UniversityNanjingChina
| | - Zhizhong Liu
- Department of CardiologyNanjing First HospitalNanjing Medical UniversityNanjingChina
| | - Shao‐Liang Chen
- Department of CardiologyNanjing First HospitalNanjing Medical UniversityNanjingChina
| | - Dai‐Min Zhang
- Department of CardiologyNanjing First HospitalNanjing Medical UniversityNanjingChina
| |
Collapse
|
27
|
Reynolds JC, Bwiza CP, Lee C. Mitonuclear genomics and aging. Hum Genet 2020; 139:381-399. [PMID: 31997134 PMCID: PMC7147958 DOI: 10.1007/s00439-020-02119-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Accepted: 01/17/2020] [Indexed: 12/25/2022]
Abstract
Our cells operate based on two distinct genomes that are enclosed in the nucleus and mitochondria. The mitochondrial genome presumably originates from endosymbiotic bacteria. With time, a large portion of the original genes in the bacterial genome is considered to have been lost or transferred to the nuclear genome, leaving a reduced 16.5 Kb circular mitochondrial DNA (mtDNA). Traditionally only 37 genes, including 13 proteins, were thought to be encoded within mtDNA, its genetic repertoire is expanding with the identification of mitochondrial-derived peptides (MDPs). The biology of aging has been largely unveiled to be regulated by genes that are encoded in the nuclear genome, whereas the mitochondrial genome remained more cryptic. However, recent studies position mitochondria and mtDNA as an important counterpart to the nuclear genome, whereby the two organelles constantly regulate each other. Thus, the genomic network that regulates lifespan and/or healthspan is likely constituted by two unique, yet co-evolved, genomes. Here, we will discuss aspects of mitochondrial biology, especially mitochondrial communication that may add substantial momentum to aging research by accounting for both mitonuclear genomes to more comprehensively and inclusively map the genetic and molecular networks that govern aging and age-related diseases.
Collapse
Affiliation(s)
- Joseph C Reynolds
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA
| | - Conscience P Bwiza
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA
| | - Changhan Lee
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA.
- USC Norris Comprehensive Cancer Center, Los Angeles, CA, 90089, USA.
- Biomedical Sciences, Graduate School, Ajou University, Suwon, 16499, South Korea.
| |
Collapse
|
28
|
Drareni K, Ballaire R, Barilla S, Mathew MJ, Toubal A, Fan R, Liang N, Chollet C, Huang Z, Kondili M, Foufelle F, Soprani A, Roussel R, Gautier JF, Alzaid F, Treuter E, Venteclef N. GPS2 Deficiency Triggers Maladaptive White Adipose Tissue Expansion in Obesity via HIF1A Activation. Cell Rep 2019; 24:2957-2971.e6. [PMID: 30208320 PMCID: PMC6153369 DOI: 10.1016/j.celrep.2018.08.032] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 07/27/2018] [Accepted: 08/10/2018] [Indexed: 12/12/2022] Open
Abstract
Hypertrophic white adipose tissue (WAT) represents a maladaptive mechanism linked to the risk for developing type 2 diabetes in humans. However, the molecular events that predispose WAT to hypertrophy are poorly defined. Here, we demonstrate that adipocyte hypertrophy is triggered by loss of the corepressor GPS2 during obesity. Adipocyte-specific GPS2 deficiency in mice (GPS2 AKO) causes adipocyte hypertrophy, inflammation, and mitochondrial dysfunction during surplus energy. This phenotype is driven by HIF1A activation that orchestrates inadequate WAT remodeling and disrupts mitochondrial activity, which can be reversed by pharmacological or genetic HIF1A inhibition. Correlation analysis of gene expression in human adipose tissue reveals a negative relationship between GPS2 and HIF1A, adipocyte hypertrophy, and insulin resistance. We propose therefore that the obesity-associated loss of GPS2 in adipocytes predisposes for a maladaptive WAT expansion and a pro-diabetic status in mice and humans. Adipose-specific GPS2 deficiency predisposes for adipocyte hypertrophy Loss of GPS2 triggers transcriptional activation of HIF1A pathways Deregulation of GPS2-HIF1A interplay provokes disrupted mitochondrial activity GPS2 and HIF1A levels are negatively correlated in human adipose tissue
Collapse
Affiliation(s)
- Karima Drareni
- INSERM, Cordeliers Research Centre, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Paris, France
| | - Raphaëlle Ballaire
- INSERM, Cordeliers Research Centre, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Paris, France; Inovarion, 75013 Paris, France
| | - Serena Barilla
- Karolinska Institutet, Department of Biosciences and Nutrition, Huddinge, Sweden
| | - Mano J Mathew
- INSERM, Cordeliers Research Centre, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Paris, France
| | - Amine Toubal
- INSERM, Cordeliers Research Centre, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Paris, France
| | - Rongrong Fan
- Karolinska Institutet, Department of Biosciences and Nutrition, Huddinge, Sweden
| | - Ning Liang
- Karolinska Institutet, Department of Biosciences and Nutrition, Huddinge, Sweden
| | - Catherine Chollet
- INSERM, Cordeliers Research Centre, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Paris, France
| | - Zhiqiang Huang
- Karolinska Institutet, Department of Biosciences and Nutrition, Huddinge, Sweden
| | - Maria Kondili
- INSERM, Cordeliers Research Centre, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Paris, France
| | - Fabienne Foufelle
- INSERM, Cordeliers Research Centre, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Paris, France
| | - Antoine Soprani
- INSERM, Cordeliers Research Centre, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Paris, France; Clinique Geoffroy Saint-Hilaire, Ramsey General de Santé, Paris, France
| | - Ronan Roussel
- INSERM, Cordeliers Research Centre, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Paris, France; Diabetology, Endocrinology and Nutrition Department, DHU FIRE, Bichat Hospital, AP-HP, Paris, France; Faculty of Medicine, University Paris-Diderot, Paris, France
| | - Jean-François Gautier
- INSERM, Cordeliers Research Centre, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Paris, France; Assistance Publique-Hôpitaux de Paris, Lariboisière Hospital, Department of Diabetes, Clinical Investigation Centre (CIC-9504), University Paris-Diderot, Paris, France; Faculty of Medicine, University Paris-Diderot, Paris, France
| | - Fawaz Alzaid
- INSERM, Cordeliers Research Centre, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Paris, France
| | - Eckardt Treuter
- Karolinska Institutet, Department of Biosciences and Nutrition, Huddinge, Sweden.
| | - Nicolas Venteclef
- INSERM, Cordeliers Research Centre, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Paris, France.
| |
Collapse
|
29
|
Soledad RB, Charles S, Samarjit D. The secret messages between mitochondria and nucleus in muscle cell biology. Arch Biochem Biophys 2019; 666:52-62. [PMID: 30935885 PMCID: PMC6538274 DOI: 10.1016/j.abb.2019.03.019] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 03/01/2019] [Accepted: 03/28/2019] [Indexed: 02/06/2023]
Abstract
Over two thousand proteins are found in the mitochondrial compartment but the mitochondrial genome codes for only 13 proteins. The majority of mitochondrial proteins are products of nuclear genes and are synthesized in the cytosol, then translocated into the mitochondria. Most of the subunits of the five respiratory chain complexes in the inner mitochondrial membrane, which generate a proton gradient across the membrane and produce ATP, are encoded by nuclear genes. Therefore, it is quite clear that import of nuclear-encoded proteins into the mitochondria is essential for mitochondrial function. Nuclear to mitochondrial communication is well studied. However, there is another arm to this communication, mitochondria to nucleus retrograde signaling. This plays an important role in the maintenance of cellular homeostasis, and is less well studied. Several transcription factors, including Sp1, SIRT3 and GSP2, are activated by altered mitochondrial function. These activated transcription factors then translocate to the nucleus. Based on the mitochondrially generated molecular signal, nuclear genes are targeted, which alters transcription of nuclear genes that code for mitochondrial proteins. This review article will mainly focus on this interactive and bi-directional communication between mitochondria and nucleus, and how this communication plays a significant role in muscle cell biology.
Collapse
Affiliation(s)
| | - Steenbergen Charles
- Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
| | - Das Samarjit
- Department of Pathology, Johns Hopkins University, Baltimore, MD, United States.
| |
Collapse
|
30
|
SETDB1-mediated methylation of Akt promotes its K63-linked ubiquitination and activation leading to tumorigenesis. Nat Cell Biol 2019; 21:214-225. [PMID: 30692626 PMCID: PMC6414065 DOI: 10.1038/s41556-018-0266-1] [Citation(s) in RCA: 142] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 12/12/2018] [Indexed: 11/12/2022]
Abstract
The serine/threonine kinase Akt plays a central role in cell proliferation, survival and metabolism and its hyperactivation is linked to cancer progression. Here we report that Akt undergoes K64 methylation by SETDB1, which is crucial for cell membrane recruitment, phosphorylation and activation of Akt upon growth factor stimulation. Furthermore, we reveal an adaptor function of histone demethylase JMJD2A, which recognizes Akt K64 methylation and recruits E3 ligase TRAF6 and Skp2-SCF to the Akt complex, independently of its demethylase activity, thereby initiating K63-linked ubiquitination, cell membrane recruitment and activation of Akt. Notably, cancer associated Akt mutant (E17K) displays enhanced K64 methylation, leading to its hyper-phosphorylation and activation. SETDB1-mediated Akt K64 methylation is upregulated and correlated with Akt hyperactivation in non-small-cell lung carcinoma (NSCLC), promotes tumor development and predicts poor outcome. Collectively, these findings reveal complicated layers of Akt activation regulation coordinated by SETDB1-mediated Akt K64 methylation to drive tumorigenesis.
Collapse
|
31
|
AKT methylation by SETDB1 promotes AKT kinase activity and oncogenic functions. Nat Cell Biol 2019; 21:226-237. [PMID: 30692625 PMCID: PMC6377565 DOI: 10.1038/s41556-018-0261-6] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 12/10/2018] [Indexed: 12/15/2022]
Abstract
Aberrant activation of Akt disturbs proliferation, survival and metabolic homeostasis of various human cancers. Thus, it is critical to understand upstream signaling pathways governing Akt activation. Here, we report that Akt undergoes SETDB1-mediated lysine-methylation to promote its activation, which is antagonized by the Jumonji-family demethylase, KDM4B. Notably, compared with wild-type mice, mice harboring non-methylated mutant Akt1 not only exhibited reduced body size, but also were less prone to carcinogen-induced skin tumors in part due to reduced Akt activation. Mechanistically, Phosphatidylinositol (3,4,5)-trisphosphate (PIP3) interaction with Akt facilitates its interaction with SETDB1 for subsequent Akt methylation, which in turn sustains Akt phosphorylation. Pathologically, genetic alterations including SETDB1 amplification aberrantly promote Akt methylation to facilitate its activation and oncogenic functions. Thus, Akt methylation is an important step synergizing with PI3K signaling to control Akt activation, suggesting that targeting the SETDB1 signaling could be a potential therapeutic strategy for combatting hyperactive Akt-driven cancers.
Collapse
|
32
|
Cardamone MD, Tanasa B, Cederquist CT, Huang J, Mahdaviani K, Li W, Rosenfeld MG, Liesa M, Perissi V. Mitochondrial Retrograde Signaling in Mammals Is Mediated by the Transcriptional Cofactor GPS2 via Direct Mitochondria-to-Nucleus Translocation. Mol Cell 2019; 69:757-772.e7. [PMID: 29499132 DOI: 10.1016/j.molcel.2018.01.037] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 12/15/2017] [Accepted: 01/29/2018] [Indexed: 12/24/2022]
Abstract
As most of the mitochondrial proteome is encoded in the nucleus, mitochondrial functions critically depend on nuclear gene expression and bidirectional mito-nuclear communication. However, mitochondria-to-nucleus communication pathways in mammals are incompletely understood. Here, we identify G-Protein Pathway Suppressor 2 (GPS2) as a mediator of mitochondrial retrograde signaling and a transcriptional activator of nuclear-encoded mitochondrial genes. GPS2-regulated translocation from mitochondria to nucleus is essential for the transcriptional activation of a nuclear stress response to mitochondrial depolarization and for supporting basal mitochondrial biogenesis in differentiating adipocytes and brown adipose tissue (BAT) from mice. In the nucleus, GPS2 recruitment to target gene promoters regulates histone H3K9 demethylation and RNA POL2 activation through inhibition of Ubc13-mediated ubiquitination. These findings, together, reveal an additional layer of regulation of mitochondrial gene transcription, uncover a direct mitochondria-nuclear communication pathway, and indicate that GPS2 retrograde signaling is a key component of the mitochondrial stress response in mammals.
Collapse
Affiliation(s)
- Maria Dafne Cardamone
- Biochemistry Department, Boston University School of Medicine, Boston, MA 02118, USA
| | - Bogdan Tanasa
- Howard Hughes Medical Institute, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Carly T Cederquist
- Biochemistry Department, Boston University School of Medicine, Boston, MA 02118, USA
| | - Jiawen Huang
- Biochemistry Department, Boston University School of Medicine, Boston, MA 02118, USA
| | - Kiana Mahdaviani
- Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Wenbo Li
- Howard Hughes Medical Institute, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Michael G Rosenfeld
- Howard Hughes Medical Institute, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Marc Liesa
- Department of Medicine, Division of Endocrinology and Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Valentina Perissi
- Biochemistry Department, Boston University School of Medicine, Boston, MA 02118, USA.
| |
Collapse
|
33
|
Liang N, Jakobsson T, Fan R, Treuter E. The Nuclear Receptor-Co-repressor Complex in Control of Liver Metabolism and Disease. Front Endocrinol (Lausanne) 2019; 10:411. [PMID: 31293521 PMCID: PMC6606711 DOI: 10.3389/fendo.2019.00411] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 06/07/2019] [Indexed: 12/21/2022] Open
Abstract
Hepatocytes are the major cell-type in the liver responsible for the coordination of metabolism in response to multiple signaling inputs. Coordination occurs primarily at the level of gene expression via transcriptional networks composed of transcription factors, in particular nuclear receptors (NRs), and associated co-regulators, including chromatin-modifying complexes. Disturbance of these networks by genetic, environmental or nutritional factors can lead to metabolic dysregulation and has been linked to the progression of non-alcoholic fatty liver disease (NAFLD) toward steatohepatitis and even liver cancer. Since there are currently no approved therapies, major efforts are dedicated to identify the critical factors that can be employed for drug development. Amongst the identified factors with clinical significance are currently lipid-sensing NRs including PPARs, LXRs, and FXR. However, major obstacles of NR-targeting are the undesired side effects associated with the genome-wide NR activities in multiple cell-types. Thus, of particular interest are co-regulators that determine NR activities, context-selectivity, and associated chromatin states. Current research on the role of co-regulators in hepatocytes is still premature due to the large number of candidates, the limited number of available mouse models, and the technical challenges in studying their chromatin occupancy. As a result, how NR-co-regulator networks in hepatocytes are coordinated by extracellular signals, and how NR-pathway selectivity is achieved, remains currently poorly understood. We will here review a notable exception, namely a fundamental transcriptional co-repressor complex that during the past decade has become the probably most-studied and best-understood physiological relevant co-regulator in hepatocytes. This multiprotein complex contains the core subunits HDAC3, NCOR, SMRT, TBL1, TBLR1, and GPS2 and is referred to as the "NR-co-repressor complex." We will particularly discuss recent advances in characterizing hepatocyte-specific loss-of-function mouse models and in applying genome-wide sequencing approaches including ChIP-seq. Both have been instrumental to uncover the role of each of the subunits under physiological conditions and in disease models, but they also revealed insights into the NR target range and genomic mechanisms of action of the co-repressor complex. We will integrate a discussion of translational aspects about the role of the complex in NAFLD pathways and in particular about the hypothesis that patient-specific alterations of specific subunits may determine NAFLD susceptibility and the therapeutic outcomes of NR-directed treatments.
Collapse
Affiliation(s)
- Ning Liang
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Tomas Jakobsson
- Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Rongrong Fan
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Eckardt Treuter
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
- *Correspondence: Eckardt Treuter
| |
Collapse
|
34
|
Huang Z, Liang N, Damdimopoulos A, Fan R, Treuter E. G protein pathway suppressor 2 (GPS2) links inflammation and cholesterol efflux by controlling lipopolysaccharide‐induced ATP‐binding cassette transporter A1 expression in macrophages. FASEB J 2018; 33:1631-1643. [DOI: 10.1096/fj.201801123r] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Zhiqiang Huang
- Department of Biosciences and NutritionCenter for Innovative Medicine (CIMED)Karolinska InstitutetHuddingeSweden
| | - Ning Liang
- Department of Biosciences and NutritionCenter for Innovative Medicine (CIMED)Karolinska InstitutetHuddingeSweden
| | - Anastasius Damdimopoulos
- Department of Biosciences and NutritionCenter for Innovative Medicine (CIMED)Karolinska InstitutetHuddingeSweden
| | - Rongrong Fan
- Department of Biosciences and NutritionCenter for Innovative Medicine (CIMED)Karolinska InstitutetHuddingeSweden
| | - Eckardt Treuter
- Department of Biosciences and NutritionCenter for Innovative Medicine (CIMED)Karolinska InstitutetHuddingeSweden
| |
Collapse
|
35
|
Vincent AE, Rosa HS, Pabis K, Lawless C, Chen C, Grünewald A, Rygiel KA, Rocha MC, Reeve AK, Falkous G, Perissi V, White K, Davey T, Petrof BJ, Sayer AA, Cooper C, Deehan D, Taylor RW, Turnbull DM, Picard M. Subcellular origin of mitochondrial DNA deletions in human skeletal muscle. Ann Neurol 2018; 84:289-301. [PMID: 30014514 PMCID: PMC6141001 DOI: 10.1002/ana.25288] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 07/02/2018] [Accepted: 07/02/2018] [Indexed: 01/07/2023]
Abstract
OBJECTIVE In patients with mitochondrial DNA (mtDNA) maintenance disorders and with aging, mtDNA deletions sporadically form and clonally expand within individual muscle fibers, causing respiratory chain deficiency. This study aimed to identify the sub-cellular origin and potential mechanisms underlying this process. METHODS Serial skeletal muscle cryosections from patients with multiple mtDNA deletions were subjected to subcellular immunofluorescent, histochemical, and genetic analysis. RESULTS We report respiratory chain-deficient perinuclear foci containing mtDNA deletions, which show local elevations of both mitochondrial mass and mtDNA copy number. These subcellular foci of respiratory chain deficiency are associated with a local increase in mitochondrial biogenesis and unfolded protein response signaling pathways. We also find that the commonly reported segmental pattern of mitochondrial deficiency is consistent with the three-dimensional organization of the human skeletal muscle mitochondrial network. INTERPRETATION We propose that mtDNA deletions first exceed the biochemical threshold causing biochemical deficiency in focal regions adjacent to the myonuclei, and induce mitochondrial biogenesis before spreading across the muscle fiber. These subcellular resolution data provide new insights into the possible origin of mitochondrial respiratory chain deficiency in mitochondrial myopathy. Ann Neurol 2018;84:289-301.
Collapse
Affiliation(s)
- Amy E. Vincent
- Wellcome Centre for Mitochondrial Research and Newcastle Centre for Ageing and VitalityInstitute of Neuroscience, Newcastle UniversityNewcastle upon TyneUnited Kingdom
| | - Hannah S. Rosa
- Wellcome Centre for Mitochondrial Research and Newcastle Centre for Ageing and VitalityInstitute of Neuroscience, Newcastle UniversityNewcastle upon TyneUnited Kingdom
| | - Kamil Pabis
- Wellcome Centre for Mitochondrial Research and Newcastle Centre for Ageing and VitalityInstitute of Neuroscience, Newcastle UniversityNewcastle upon TyneUnited Kingdom
| | - Conor Lawless
- Wellcome Centre for Mitochondrial Research and Newcastle Centre for Ageing and VitalityInstitute of Neuroscience, Newcastle UniversityNewcastle upon TyneUnited Kingdom
| | - Chun Chen
- Wellcome Centre for Mitochondrial Research and Newcastle Centre for Ageing and VitalityInstitute of Neuroscience, Newcastle UniversityNewcastle upon TyneUnited Kingdom
| | - Anne Grünewald
- Wellcome Centre for Mitochondrial Research and Newcastle Centre for Ageing and VitalityInstitute of Neuroscience, Newcastle UniversityNewcastle upon TyneUnited Kingdom
- Institute of NeurogeneticsUniversity of LübeckLübeckGermany
- Molecular and Functional Neurobiology GroupLuxembourg Center for Systems Biomedicine, University of LuxembourgEsch‐sur‐AlzetteLuxembourg
| | - Karolina A. Rygiel
- Wellcome Centre for Mitochondrial Research and Newcastle Centre for Ageing and VitalityInstitute of Neuroscience, Newcastle UniversityNewcastle upon TyneUnited Kingdom
| | - Mariana C. Rocha
- Wellcome Centre for Mitochondrial Research and Newcastle Centre for Ageing and VitalityInstitute of Neuroscience, Newcastle UniversityNewcastle upon TyneUnited Kingdom
| | - Amy K. Reeve
- Wellcome Centre for Mitochondrial Research and Newcastle Centre for Ageing and VitalityInstitute of Neuroscience, Newcastle UniversityNewcastle upon TyneUnited Kingdom
| | - Gavin Falkous
- Wellcome Centre for Mitochondrial Research and Newcastle Centre for Ageing and VitalityInstitute of Neuroscience, Newcastle UniversityNewcastle upon TyneUnited Kingdom
| | - Valentina Perissi
- Department of BiochemistryBoston University School of MedicineBostonMA
| | - Kathryn White
- Electron Microscopy Research ServicesNewcastle UniversityNewcastle upon TyneUnited Kingdom
| | - Tracey Davey
- Electron Microscopy Research ServicesNewcastle UniversityNewcastle upon TyneUnited Kingdom
| | - Basil J. Petrof
- Meakins‐Christie Laboratories, Department of MedicineMcGill University Health CentreMontrealQuebecCanada
| | - Avan A. Sayer
- National Institute for Health Research Newcastle Biomedical Research CentreNewcastle upon Tyne Hospitals National Health Service Foundation Trust and Newcastle UniversityNewcastle upon TyneUnited Kingdom
| | - Cyrus Cooper
- Medical Research Council Lifecourse Epidemiology UnitUniversity of SouthamptonSouthamptonUnited Kingdom
| | - David Deehan
- Institute of Cellular MedicineNewcastle UniversityNewcastle Upon TyneUnited Kingdom
| | - Robert W. Taylor
- Wellcome Centre for Mitochondrial Research and Newcastle Centre for Ageing and VitalityInstitute of Neuroscience, Newcastle UniversityNewcastle upon TyneUnited Kingdom
| | - Doug M. Turnbull
- Wellcome Centre for Mitochondrial Research and Newcastle Centre for Ageing and VitalityInstitute of Neuroscience, Newcastle UniversityNewcastle upon TyneUnited Kingdom
| | - Martin Picard
- Department of Psychiatry, Division of Behavioral MedicineColumbia University Medical CenterNew YorkNY
- Department of Neurology and Columbia Translational Neuroscience InitiativeH. Houston Merritt Center, Columbia University Medical CenterNew YorkNY
- Columbia University Aging CenterColumbia UniversityNew YorkNY
| |
Collapse
|
36
|
Zhang P, Xu L, Guan H, Liu L, Liu J, Huang Z, Cao X, Liao Z, Xiao H, Li Y. Beraprost sodium, a prostacyclin analogue, reduces fructose-induced hepatocellular steatosis in mice and in vitro via the microRNA-200a and SIRT1 signaling pathway. Metabolism 2017; 73:9-21. [PMID: 28732575 DOI: 10.1016/j.metabol.2017.05.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 04/24/2017] [Accepted: 05/06/2017] [Indexed: 12/30/2022]
Abstract
PURPOSE To determine whether beraprost sodium, a prostacyclin analogue, could reduce hepatic lipid accumulation induced by fructose in mice and cultured human hepatocytes, and to investigate the expression of microRNAs and the sirtuin 1 (SIRT1) pathway. METHODS Male C57BL/6JNju mice were divided into three groups and fed one of the following diets: a normal diet, a high fructose diet, or a high fructose diet with beraprost sodium treatment. In addition, human-derived HepG2 cells were cultured and treated with fructose (25mmol/L) with or without beraprost sodium (10μmol/L) for 24h, and transfected with small interfering RNA (siRNA) against SIRT1, miR-200a mimic, or miR-200a inhibitor for 48h. The miRNA microarray analysis was performed on the HepG2 cells, and the expression profiles of miRNAs were analyzed using Gene Cluster 3.0 and verified using qPCR. RESULTS Beraprost sodium treatment attenuated hepatic steatosis, induced the transcription of genes involved in lipid metabolism in C57BL/6 mice (P<0.05), and increased the expression of hepatic SIRT1 and peroxisome proliferator activated receptor α (PPARα) in the cells treated with fructose. These effects were blocked in HepG2 cells after transfection with siRNA against SIRT1. MiR-200a was highly expressed during fructose treatment and was down regulated by beraprost sodium (P<0.05). A luciferase assay showed that miR-200a regulated SIRT1 by binding to the 3' UTR. Overexpression of miR-200a inhibited expression of hepatic SIRT1. CONCLUSIONS Our study demonstrated that SIRT1 pathway mediated the effects of beraprost sodium on attenuation of hepatic lipid disorders induced by fructose and revealed the primary role of miR-200a in the regulation of hepatic SIRT1 by beraprost sodium. Our findings suggested that SIRT1 might be a therapeutic target of fructose-related metabolism disorders.
Collapse
Affiliation(s)
- Pengyuan Zhang
- The First Affiliated Hospital, Sun Yat-sen University, Department of Endocrinology, Guangzhou, 510080, China
| | - Lijuan Xu
- The First Affiliated Hospital, Sun Yat-sen University, Department of Endocrinology, Guangzhou, 510080, China
| | - Hongyu Guan
- The First Affiliated Hospital, Sun Yat-sen University, Department of Endocrinology, Guangzhou, 510080, China
| | - Liehua Liu
- The First Affiliated Hospital, Sun Yat-sen University, Department of Endocrinology, Guangzhou, 510080, China
| | - Juan Liu
- The First Affiliated Hospital, Sun Yat-sen University, Department of Endocrinology, Guangzhou, 510080, China
| | - Zhimin Huang
- The First Affiliated Hospital, Sun Yat-sen University, Department of Endocrinology, Guangzhou, 510080, China
| | - Xiaopei Cao
- The First Affiliated Hospital, Sun Yat-sen University, Department of Endocrinology, Guangzhou, 510080, China
| | - Zhihong Liao
- The First Affiliated Hospital, Sun Yat-sen University, Department of Endocrinology, Guangzhou, 510080, China
| | - Haipeng Xiao
- The First Affiliated Hospital, Sun Yat-sen University, Department of Endocrinology, Guangzhou, 510080, China
| | - Yanbing Li
- The First Affiliated Hospital, Sun Yat-sen University, Department of Endocrinology, Guangzhou, 510080, China.
| |
Collapse
|
37
|
Kenny TC, Manfredi G, Germain D. The Mitochondrial Unfolded Protein Response as a Non-Oncogene Addiction to Support Adaptation to Stress during Transformation in Cancer and Beyond. Front Oncol 2017; 7:159. [PMID: 28798902 PMCID: PMC5526845 DOI: 10.3389/fonc.2017.00159] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 07/10/2017] [Indexed: 12/27/2022] Open
Abstract
Upon accumulation of misfolded proteins in the mitochondria, the mitochondrial unfolded protein response (UPRmt) is activated. This review focuses on the role of this response in cancer. We discuss evidence that during transformation, the UPRmt may play an essential role in the maintenance of the integrity of the mitochondria in the face of increased oxidative stress. However, the role of the UPRmt in other diseases is also emerging and is therefore also briefly discussed.
Collapse
Affiliation(s)
- Timothy C Kenny
- Department of Medicine, Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY, Unites States
| | - Giovanni Manfredi
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, Unites States
| | - Doris Germain
- Department of Medicine, Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY, Unites States
| |
Collapse
|
38
|
Lentucci C, Belkina AC, Cederquist CT, Chan M, Johnson HE, Prasad S, Lopacinski A, Nikolajczyk BS, Monti S, Snyder-Cappione J, Tanasa B, Cardamone MD, Perissi V. Inhibition of Ubc13-mediated Ubiquitination by GPS2 Regulates Multiple Stages of B Cell Development. J Biol Chem 2016; 292:2754-2772. [PMID: 28039360 DOI: 10.1074/jbc.m116.755132] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 12/21/2016] [Indexed: 12/12/2022] Open
Abstract
Non-proteolytic ubiquitin signaling mediated by Lys63 ubiquitin chains plays a critical role in multiple pathways that are key to the development and activation of immune cells. Our previous work indicates that GPS2 (G-protein Pathway Suppressor 2) is a multifunctional protein regulating TNFα signaling and lipid metabolism in the adipose tissue through modulation of Lys63 ubiquitination events. However, the full extent of GPS2-mediated regulation of ubiquitination and the underlying molecular mechanisms are unknown. Here, we report that GPS2 is required for restricting the activation of TLR and BCR signaling pathways and the AKT/FOXO1 pathway in immune cells based on direct inhibition of Ubc13 enzymatic activity. Relevance of this regulatory strategy is confirmed in vivo by B cell-targeted deletion of GPS2, resulting in developmental defects at multiple stages of B cell differentiation. Together, these findings reveal that GPS2 genomic and non-genomic functions are critical for the development and cellular homeostasis of B cells.
Collapse
Affiliation(s)
| | - Anna C Belkina
- the Flow Cytometry Core Facility, Boston University School of Medicine, Boston, Massachusetts 02118 and.,Microbiology, and
| | | | | | | | | | | | | | | | - Jennifer Snyder-Cappione
- the Flow Cytometry Core Facility, Boston University School of Medicine, Boston, Massachusetts 02118 and.,Microbiology, and
| | - Bogdan Tanasa
- the Department of Pediatrics, Stanford University School of Medicine, Stanford, California 94305
| | | | | |
Collapse
|