151
|
Ascenção K, Szabo C. Emerging roles of cystathionine β-synthase in various forms of cancer. Redox Biol 2022; 53:102331. [PMID: 35618601 PMCID: PMC9168780 DOI: 10.1016/j.redox.2022.102331] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 04/29/2022] [Accepted: 05/04/2022] [Indexed: 12/12/2022] Open
Abstract
The expression of the reverse transsulfuration enzyme cystathionine-β-synthase (CBS) is markedly increased in many forms of cancer, including colorectal, ovarian, lung, breast and kidney, while in other cancers (liver cancer and glioma) it becomes downregulated. According to the clinical database data in high-CBS-expressor cancers (e.g. colon or ovarian cancer), high CBS expression typically predicts lower survival, while in the low-CBS-expressor cancers (e.g. liver cancer), low CBS expression is associated with lower survival. In the high-CBS expressing tumor cells, CBS, and its product hydrogen sulfide (H2S) serves as a bioenergetic, proliferative, cytoprotective and stemness factor; it also supports angiogenesis and epithelial-to-mesenchymal transition in the cancer microenvironment. The current article reviews the various tumor-cell-supporting roles of the CBS/H2S axis in high-CBS expressor cancers and overviews the anticancer effects of CBS silencing and pharmacological CBS inhibition in various cancer models in vitro and in vivo; it also outlines potential approaches for biomarker identification, to support future targeted cancer therapies based on pharmacological CBS inhibition.
Collapse
|
152
|
Daks A, Fedorova O, Parfenyev S, Nevzorov I, Shuvalov O, Barlev NA. The Role of E3 Ligase Pirh2 in Disease. Cells 2022; 11:1515. [PMID: 35563824 PMCID: PMC9101203 DOI: 10.3390/cells11091515] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 02/04/2023] Open
Abstract
The p53-dependent ubiquitin ligase Pirh2 regulates a number of proteins involved in different cancer-associated processes. Targeting the p53 family proteins, Chk2, p27Kip1, Twist1 and others, Pirh2 participates in such cellular processes as proliferation, cell cycle regulation, apoptosis and cellular migration. Thus, it is not surprising that Pirh2 takes part in the initiation and progression of different diseases and pathologies including but not limited to cancer. In this review, we aimed to summarize the available data on Pirh2 regulation, its protein targets and its role in various diseases and pathological processes, thus making the Pirh2 protein a promising therapeutic target.
Collapse
Affiliation(s)
- Alexandra Daks
- Institute of Cytology RAS, 194064 St. Petersburg, Russia; (O.F.); (S.P.); (I.N.); (O.S.)
| | | | | | | | | | - Nickolai A. Barlev
- Institute of Cytology RAS, 194064 St. Petersburg, Russia; (O.F.); (S.P.); (I.N.); (O.S.)
| |
Collapse
|
153
|
Tu M, Saputo S. From Beginning to End: Expanding the SERINC3 Interactome Through an in silico Analysis. Bioinform Biol Insights 2022; 16:11779322221092944. [PMID: 35494555 PMCID: PMC9052817 DOI: 10.1177/11779322221092944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 03/07/2022] [Indexed: 11/15/2022] Open
Abstract
The serine incorporator (SERINC) family of proteins are a family of multipass transmembrane proteins associated with biosynthesis of serine-containing phospholipids and sphingolipids. Humans have 5 paralogs, SERINC1-5, which have been linked to disease including variable expression in tumor lines and possessing activity as restriction factors against HIV-1. Despite recent studies, the cellular function of SERINC proteins have yet to be fully elucidated. The goal of this study as to investigate the role of SERINC3 by expanding upon its interactome. We used a variety of bioinformatic tools to identify cellular factors that interact with SERINC3 and assessed how sequence variation might alter these interactions. Analysis of the promoter region indicates that SERINC3 is putatively regulated by transcription factors involved in tissue-specific development. Analysis of the unique 3′-untranslated region of one variant of HsSERINC3 revealed that this region serves as a conserved site of regulation by both RNA binding proteins and miRNA. In addition, SERINC3 is putatively regulated at the protein level by several posttranslational modifications. Our results show that extra-membrane portions of SERINC3 are subject to variation in the coding sequence as well as areas of relatively low conservation. Overall, our data suggest that regions of low homology as well as presence of variations in the nucleotide and protein sequences of HsSERINC3 suggest that these variations may lead to aberrant function and alternative regulatory mechanisms in homologs. The functional consequences of these sequence and structural variations need to be explored systematically to fully appreciate the role of SERINC3 in both health and disease.
Collapse
Affiliation(s)
- Mckenzie Tu
- Department of Chemistry and Biochemistry, SUNY Brockport, Brockport, NY, USA
| | - Sarah Saputo
- Department of Chemistry and Biochemistry, SUNY Brockport, Brockport, NY, USA
| |
Collapse
|
154
|
Zhao X, Wang J, Wang Y, Zhang M, Zhao W, Zhang H, Zhao L. Interferon‑stimulated gene 15 promotes progression of endometrial carcinoma and weakens antitumor immune response. Oncol Rep 2022; 47:110. [PMID: 35445736 PMCID: PMC9073416 DOI: 10.3892/or.2022.8321] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 12/14/2021] [Indexed: 11/24/2022] Open
Abstract
Endometrial carcinoma (EC) is one of the most common gynecological cancers with a poor prognosis. Therefore, clarifying the details of the molecular mechanisms is of great importance for EC diagnosis and clinical management. Interferon-stimulated gene 15 (ISG15) plays an important role in the development of various cancers. However, its role in EC remains unclear. High ISG15 expression was observed in EC, which was associated with poor clinical outcomes and pathological stage of patients with EC, thus representing a promising marker for EC progression. Further exploratory analysis revealed that the elevated ISG15 levels in EC were driven by aberrant DNA methylation, independent of copy number variation and specific transcription factor aberrations. Accordingly, knockdown of ISG15 by small interfering RNA attenuated the malignant cellular phenotype of EC cell lines, including proliferation and colony formation in vitro. Finally, investigation of the molecular mechanisms indicated that ISG15 promoted the cell cycle G1/S transition in EC. Furthermore, ISG15 promoted EC progression by activating the MYC proto-oncogene protein signaling pathway. Moreover, ECs with high levels of ISG15 harbored a more vital immune escape ability, evidenced not only by significantly less invasive CD8+ T cells, but also higher expression of T cell inhibitory factors, such as programmed death-ligand 1. These results suggest a tumor-promoting role of ISG15 in EC, which may be a promising marker for diagnosis, prognosis and therapeutic immunity.
Collapse
Affiliation(s)
- Xiwa Zhao
- Department of Obstetrics and Gynecology, The Fourth Hospital, Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Jingjing Wang
- The Research Center, The Fourth Hospital, Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Yaojie Wang
- The Research Center, The Fourth Hospital, Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Mengmeng Zhang
- Department of Obstetrics and Gynecology, The Fourth Hospital, Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Wei Zhao
- Department of Obstetrics and Gynecology, The Fourth Hospital, Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Hui Zhang
- Department of Obstetrics and Gynecology, The Fourth Hospital, Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Lianmei Zhao
- The Research Center, The Fourth Hospital, Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| |
Collapse
|
155
|
Abstract
Proteins are the molecular effectors of the information encoded in the genome. Proteomics aims at understanding the molecular functions of proteins in their biological context. In contrast to transcriptomics and genomics, the study of proteomes provides deeper insight into the dynamic regulatory layers encoded at the protein level, such as posttranslational modifications, subcellular localization, cell signaling, and protein-protein interactions. Currently, mass spectrometry (MS)-based proteomics is the technology of choice for studying proteomes at a system-wide scale, contributing to clinical biomarker discovery and fundamental molecular biology. MS technologies are continuously being developed to fulfill the requirements of speed, resolution, and quantitative accuracy, enabling the acquisition of comprehensive proteomes. In this review, we present how MS technology and acquisition methods have evolved to meet the requirements of cutting-edge proteomics research, which is describing the human proteome and its dynamic posttranslational modifications with unprecedented depth. Finally, we provide a perspective on studying proteomes at single-cell resolution. Expected final online publication date for the Annual Review of Genomics and Human Genetics, Volume 23 is October 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Ana Martinez-Val
- Novo Nordisk Foundation Center for Protein Research, Proteomics Program, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark;
| | - Ulises H Guzmán
- Novo Nordisk Foundation Center for Protein Research, Proteomics Program, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark;
| | - Jesper V Olsen
- Novo Nordisk Foundation Center for Protein Research, Proteomics Program, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark;
| |
Collapse
|
156
|
Tomczyk MM, Cheung KG, Xiang B, Tamanna N, Fonseca Teixeira AL, Agarwal P, Kereliuk SM, Spicer V, Lin L, Treberg J, Tong Q, Dolinsky VW. Mitochondrial Sirtuin-3 (SIRT3) Prevents Doxorubicin-Induced Dilated Cardiomyopathy by Modulating Protein Acetylation and Oxidative Stress. Circ Heart Fail 2022; 15:e008547. [PMID: 35418250 PMCID: PMC9117478 DOI: 10.1161/circheartfailure.121.008547] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND High doses of doxorubicin put cancer patients at risk for developing dilated cardiomyopathy. Previously, we showed that doxorubicin treatment decreases SIRT3 (sirtuin 3), the main mitochondrial deacetylase and increases protein acetylation in rat cardiomyocytes. Here, we hypothesize that SIRT3 expression can attenuate doxorubicin induced dilated cardiomyopathy in vivo by preventing the acetylation of mitochondrial proteins. METHODS Nontransgenic, M3-SIRT3 (truncated SIRT3; short isoform), and M1-SIRT3 (full-length SIRT3; mitochondrial localized) transgenic mice were treated with doxorubicin for 4 weeks (8 mg/kg body weight per week). Echocardiography was performed to assess cardiac structure and function and validated by immunohistochemistry and immunofluorescence (n=4-10). Mass spectrometry was performed on cardiac mitochondrial peptides in saline (n=6) and doxorubicin (n=5) treated hearts. Validation was performed in doxorubicin treated primary rat and human induced stem cell derived cardiomyocytes transduced with adenoviruses for M3-SIRT3 and M1-SIRT3 and deacetylase deficient mutants (n=4-10). RESULTS Echocardiography revealed that M3-SIRT3 transgenic mice were partially resistant to doxorubicin induced changes to cardiac structure and function whereas M1-SIRT3 expression prevented cardiac remodeling and dysfunction. In doxorubicin hearts, 37 unique acetylation sites on mitochondrial proteins were altered. Pathway analysis revealed these proteins are involved in energy production, fatty acid metabolism, and oxidative stress resistance. Increased M1-SIRT3 expression in primary rat and human cardiomyocytes attenuated doxorubicin-induced superoxide formation, whereas deacetylase deficient mutants were unable to prevent oxidative stress. CONCLUSIONS Doxorubicin reduced SIRT3 expression and markedly affected the cardiac mitochondrial acetylome. Increased M1-SIRT3 expression in vivo prevented doxorubicin-induced cardiac dysfunction, suggesting that SIRT3 could be a potential therapeutic target for mitigating doxorubicin-induced dilated cardiomyopathy.
Collapse
Affiliation(s)
- Mateusz M Tomczyk
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme of the Children's Hospital Research Institute of Manitoba' Winnipeg' Canada (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.).,Department of Pharmacology and Therapeutics (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.), University of Manitoba, Winnipeg, Canada.,Rady Faculty of Health Science, College of Medicine (M.M.T., K.G.C., B.X., P.A., S.M.K., V.S., V.W.D.), University of Manitoba, Winnipeg, Canada
| | - Kyle G Cheung
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme of the Children's Hospital Research Institute of Manitoba' Winnipeg' Canada (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.).,Department of Pharmacology and Therapeutics (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.), University of Manitoba, Winnipeg, Canada.,Rady Faculty of Health Science, College of Medicine (M.M.T., K.G.C., B.X., P.A., S.M.K., V.S., V.W.D.), University of Manitoba, Winnipeg, Canada
| | - Bo Xiang
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme of the Children's Hospital Research Institute of Manitoba' Winnipeg' Canada (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.).,Department of Pharmacology and Therapeutics (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.), University of Manitoba, Winnipeg, Canada.,Rady Faculty of Health Science, College of Medicine (M.M.T., K.G.C., B.X., P.A., S.M.K., V.S., V.W.D.), University of Manitoba, Winnipeg, Canada
| | - Nahid Tamanna
- Department of Biological Sciences (N.T., A.L.F.T., J.T.), University of Manitoba, Winnipeg, Canada
| | - Ana L Fonseca Teixeira
- Department of Biological Sciences (N.T., A.L.F.T., J.T.), University of Manitoba, Winnipeg, Canada
| | - Prasoon Agarwal
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme of the Children's Hospital Research Institute of Manitoba' Winnipeg' Canada (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.).,Department of Pharmacology and Therapeutics (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.), University of Manitoba, Winnipeg, Canada.,Rady Faculty of Health Science, College of Medicine (M.M.T., K.G.C., B.X., P.A., S.M.K., V.S., V.W.D.), University of Manitoba, Winnipeg, Canada.,KTH Royal Institute of Technology, School of Electrical Engineering and Computer Science, Stockholm, Sweden (P.A.).,Science for Life Laboratory, Solna, Sweden (P.A.)
| | - Stephanie M Kereliuk
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme of the Children's Hospital Research Institute of Manitoba' Winnipeg' Canada (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.).,Department of Pharmacology and Therapeutics (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.), University of Manitoba, Winnipeg, Canada.,Rady Faculty of Health Science, College of Medicine (M.M.T., K.G.C., B.X., P.A., S.M.K., V.S., V.W.D.), University of Manitoba, Winnipeg, Canada
| | - Victor Spicer
- Department of Internal Medicine (V.S.), University of Manitoba, Winnipeg, Canada.,Rady Faculty of Health Science, College of Medicine (M.M.T., K.G.C., B.X., P.A., S.M.K., V.S., V.W.D.), University of Manitoba, Winnipeg, Canada.,Manitoba Center for Proteomics and Systems Biology, Winnipeg, Canada (V.S.)
| | - Ligen Lin
- Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX (L.L., Q.T.).,Institute of Chinese Medical Sciences, University of Macau, China (L.L.)
| | - Jason Treberg
- Department of Biological Sciences (N.T., A.L.F.T., J.T.), University of Manitoba, Winnipeg, Canada
| | - Qiang Tong
- Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX (L.L., Q.T.)
| | - Vernon W Dolinsky
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme of the Children's Hospital Research Institute of Manitoba' Winnipeg' Canada (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.).,Department of Pharmacology and Therapeutics (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.), University of Manitoba, Winnipeg, Canada.,Rady Faculty of Health Science, College of Medicine (M.M.T., K.G.C., B.X., P.A., S.M.K., V.S., V.W.D.), University of Manitoba, Winnipeg, Canada
| |
Collapse
|
157
|
Rinaldi L, Fettweis G, Kim S, Garcia DA, Fujiwara S, Johnson TA, Tettey TT, Ozbun L, Pegoraro G, Puglia M, Blagoev B, Upadhyaya A, Stavreva DA, Hager GL. The glucocorticoid receptor associates with the cohesin loader NIPBL to promote long-range gene regulation. SCIENCE ADVANCES 2022; 8:eabj8360. [PMID: 35353576 PMCID: PMC8967222 DOI: 10.1126/sciadv.abj8360] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 02/07/2022] [Indexed: 05/13/2023]
Abstract
The cohesin complex is central to chromatin looping, but mechanisms by which these long-range chromatin interactions are formed and persist remain unclear. We demonstrate that interactions between a transcription factor (TF) and the cohesin loader NIPBL regulate enhancer-dependent gene activity. Using mass spectrometry, genome mapping, and single-molecule tracking methods, we demonstrate that the glucocorticoid (GC) receptor (GR) interacts with NIPBL and the cohesin complex at the chromatin level, promoting loop extrusion and long-range gene regulation. Real-time single-molecule experiments show that loss of cohesin markedly diminishes the concentration of TF molecules at specific nuclear confinement sites, increasing TF local concentration and promoting gene regulation. Last, patient-derived acute myeloid leukemia cells harboring cohesin mutations exhibit a reduced response to GCs, suggesting that the GR-NIPBL-cohesin interaction is defective in these patients, resulting in poor response to GC treatment.
Collapse
Affiliation(s)
- Lorenzo Rinaldi
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Gregory Fettweis
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sohyoung Kim
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - David A. Garcia
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
- Department of Physics, University of Maryland, College Park, MD 20742, USA
| | - Saori Fujiwara
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Thomas A. Johnson
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Theophilus T. Tettey
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Laurent Ozbun
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
- High-Throughput Imaging Facility (HiTIF), Center for Cancer Research (CCR), NCI/NIH, Bethesda, MD 20892, USA
| | - Gianluca Pegoraro
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
- High-Throughput Imaging Facility (HiTIF), Center for Cancer Research (CCR), NCI/NIH, Bethesda, MD 20892, USA
| | - Michele Puglia
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Blagoy Blagoev
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Arpita Upadhyaya
- Department of Physics, University of Maryland, College Park, MD 20742, USA
- Institute for Physical Science and Technology, University of Maryland, College Park, MD 20742, USA
| | - Diana A. Stavreva
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Gordon L. Hager
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
158
|
Zhou Q, Li S, Li M, Ke D, Wang Q, Yang Y, Liu GP, Wang XC, Liu E, Wang JZ. Human tau accumulation promotes glycogen synthase kinase-3β acetylation and thus upregulates the kinase: A vicious cycle in Alzheimer neurodegeneration. EBioMedicine 2022; 78:103970. [PMID: 35339896 PMCID: PMC8956943 DOI: 10.1016/j.ebiom.2022.103970] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 03/12/2022] [Accepted: 03/13/2022] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Glycogen synthase kinase-3β (GSK-3β) is one of the most effective kinases in promoting tau hyperphosphorylation and accumulation in Alzheimer's disease (AD). However, it is not clear how GSK-3β activity is regulated during AD progression. METHODS We firstly used mass spectrometry to identify the acetylation site of GSK-3β, and then established the cell and animal models of GSK-3β acetylation. Next, we conducted molecular, cell biological and behavioral tests. Finally, we designed a peptide to test whether blocking tau-mediated GSK-3β acetylation could be beneficial to AD. FINDINGS We found that GSK-3β protein levels increased in the brains of AD patients and the transgenic mice. Overexpressing tau increased GSK-3β protein level with increased acetylation and decreased ubiquitination-related proteolysis. Tau could directly acetylate GSK-3β at K15 both in vitro and in vivo. K15-acetylation inhibited ubiquitination-associated proteolysis of GSK-3β and changed its activity-dependent phosphorylation, leading to over-activation of the kinase. GSK-3β activation by K15-acetylation in turn exacerbated the AD-like pathologies. Importantly, competitively inhibiting GSK-3β K15-acetylation by a novel-designed peptide remarkably improved cognitive impairment and the AD-like pathologies in 3xTg-AD mice. INTERPRETATION Tau can directly acetylate GSK-3β at K15 which reveals a vicious cycle between tau hyperphosphorylation and GSK-3β activation. FUNDING This study was supported in parts by grants from Science and Technology Committee of China (2016YFC1305800), Hubei Province (2018ACA142), Natural Science Foundation of China (91949205, 82001134, 31730035, 81721005), Guangdong Provincial Key S&T Program (018B030336001).
Collapse
Affiliation(s)
- Qiuzhi Zhou
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shihong Li
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Mengzhu Li
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Dan Ke
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qun Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ying Yang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Gong-Ping Liu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiao-Chuan Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Enjie Liu
- Department of Pathology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Jian-Zhi Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226000, China.
| |
Collapse
|
159
|
Basello DA, Matera AG, Staněk D. A point mutation in human coilin prevents Cajal body formation. J Cell Sci 2022; 135:274900. [PMID: 35356988 PMCID: PMC9080554 DOI: 10.1242/jcs.259587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 03/20/2022] [Indexed: 11/20/2022] Open
Abstract
Coilin is a conserved protein essential for integrity of nuclear membrane-less inclusions called Cajal bodies. Here, we report an amino-acid substitution (p.K496E) found in a widely-used human EGFP-coilin construct that has a dominant negative effect on Cajal body formation. We show that this coilin-K496E variant fails to rescue Cajal bodies in cells lacking endogenous coilin, whereas the wild-type construct restores Cajal bodies in mouse and human coilin-knockout cells. In cells containing endogenous coilin, both the wild-type and K496E variant proteins accumulate in Cajal bodies. However, high-level overexpression of coilin-K496E causes Cajal body disintegration. Thus, a mutation in the C-terminal region of human coilin can disrupt Cajal body assembly. Caution should be used when interpreting data from coilin plasmids that are derived from this variant (currently deposited at Addgene).
Collapse
Affiliation(s)
- Davide A Basello
- Institute of Molecular Genetics, Czech Academy of Science, Prague, Czech Republic.,Faculty of Science, Charles University, Prague, Czech Republic
| | - A Gregory Matera
- Integrative Program for Biological and Genome Sciences, and Department of Biology, University of North Carolina, Chapel Hill, USA
| | - David Staněk
- Institute of Molecular Genetics, Czech Academy of Science, Prague, Czech Republic
| |
Collapse
|
160
|
Steger M, Karayel Ö, Demichev V. Ubiquitinomics: history, methods and applications in basic research and drug discovery. Proteomics 2022; 22:e2200074. [PMID: 35353442 DOI: 10.1002/pmic.202200074] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/21/2022] [Accepted: 03/23/2022] [Indexed: 11/08/2022]
Abstract
The ubiquitin-proteasome system (UPS) was discovered about 40 years ago and is known to regulate a multitude of cellular processes including protein homeostasis. ubiquitylated proteins are recognized by downstream effectors, resulting in alterations of protein abundance, activity, or localization. Not surprisingly, the ubiquitylation machinery is dysregulated in numerous diseases, including cancers and neurodegeneration. Mass spectrometry (MS)-based proteomics has emerged as a transformative technology for characterizing protein ubiquitylation in an unbiased fashion. Here, we provide an overview of the different MS-based approaches for studying protein ubiquitylation. We review various methods for enriching and quantifying ubiquitin modifications at the peptide or protein level, outline MS acquisition and data processing approaches and discuss key challenges. Finally, we examine how MS-based ubiquitinomics can aid both basic biology and drug discovery research. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Martin Steger
- Evotec München GmbH, Martinsried, 82152, Germany.,Present address: Max Planck Institute of Biochemistry, Martinsried, 82152, Germany
| | - Özge Karayel
- Max Planck Institute of Biochemistry, Martinsried, 82152, Germany.,Current address: Department of Physiological Chemistry, Genentech, South San Francisco, CA, 94080, USA
| | - Vadim Demichev
- Charité - Universitätsmedizin Berlin, Department of Biochemistry, Berlin, Germany
| |
Collapse
|
161
|
Lacoursiere RE, Hadi D, Shaw GS. Acetylation, Phosphorylation, Ubiquitination (Oh My!): Following Post-Translational Modifications on the Ubiquitin Road. Biomolecules 2022; 12:biom12030467. [PMID: 35327659 PMCID: PMC8946176 DOI: 10.3390/biom12030467] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/11/2022] [Accepted: 03/14/2022] [Indexed: 02/06/2023] Open
Abstract
Ubiquitination is controlled by a series of E1, E2, and E3 enzymes that can ligate ubiquitin to cellular proteins and dictate the turnover of a substrate and the outcome of signalling events such as DNA damage repair and cell cycle. This process is complex due to the combinatorial power of ~35 E2 and ~1000 E3 enzymes involved and the multiple lysine residues on ubiquitin that can be used to assemble polyubiquitin chains. Recently, mass spectrometric methods have identified that most enzymes in the ubiquitination cascade can be further modified through acetylation or phosphorylation under particular cellular conditions and altered modifications have been noted in different cancers and neurodegenerative diseases. This review provides a cohesive summary of ubiquitination, acetylation, and phosphorylation sites in ubiquitin, the human E1 enzyme UBA1, all E2 enzymes, and some representative E3 enzymes. The potential impacts these post-translational modifications might have on each protein function are highlighted, as well as the observations from human disease.
Collapse
|
162
|
Insights in Post-Translational Modifications: Ubiquitin and SUMO. Int J Mol Sci 2022; 23:ijms23063281. [PMID: 35328702 PMCID: PMC8952880 DOI: 10.3390/ijms23063281] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/15/2022] [Accepted: 03/16/2022] [Indexed: 12/23/2022] Open
Abstract
Both ubiquitination and SUMOylation are dynamic post-translational modifications that regulate thousands of target proteins to control virtually every cellular process. Unfortunately, the detailed mechanisms of how all these cellular processes are regulated by both modifications remain unclear. Target proteins can be modified by one or several moieties, giving rise to polymers of different morphology. The conjugation cascades of both modifications comprise a few activating and conjugating enzymes but close to thousands of ligating enzymes (E3s) in the case of ubiquitination. As a result, these E3s give substrate specificity and can form polymers on a target protein. Polymers can be quickly modified forming branches or cleaving chains leading the target protein to its cellular fate. The recent development of mass spectrometry(MS) -based approaches has increased the understanding of ubiquitination and SUMOylation by finding essential modified targets in particular signaling pathways. Here, we perform a concise overview comprising from the basic mechanisms of both ubiquitination and SUMOylation to recent MS-based approaches aimed to find specific targets for particular E3 enzymes.
Collapse
|
163
|
Gabriele RMC, Abel E, Fox NC, Wray S, Arber C. Knockdown of Amyloid Precursor Protein: Biological Consequences and Clinical Opportunities. Front Neurosci 2022; 16:835645. [PMID: 35360155 PMCID: PMC8964081 DOI: 10.3389/fnins.2022.835645] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 02/18/2022] [Indexed: 12/29/2022] Open
Abstract
Amyloid precursor protein (APP) and its cleavage fragment Amyloid-β (Aβ) have fundamental roles in Alzheimer's disease (AD). Genetic alterations that either increase the overall dosage of APP or alter its processing to favour the generation of longer, more aggregation prone Aβ species, are directly causative of the disease. People living with one copy of APP are asymptomatic and reducing APP has been shown to lower the relative production of aggregation-prone Aβ species in vitro. For these reasons, reducing APP expression is an attractive approach for AD treatment and prevention. In this review, we will describe the structure and the known functions of APP and go on to discuss the biological consequences of APP knockdown and knockout in model systems. We highlight progress in therapeutic strategies to reverse AD pathology via reducing APP expression. We conclude that new technologies that reduce the dosage of APP expression may allow disease modification and slow clinical progression, delaying or even preventing onset.
Collapse
Affiliation(s)
- Rebecca M. C. Gabriele
- Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Emily Abel
- Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, London, United Kingdom,UK Dementia Research Institute at University College London (UCL), Queen Square Institute of Neurology, London, United Kingdom
| | - Nick C. Fox
- Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, London, United Kingdom,UK Dementia Research Institute at University College London (UCL), Queen Square Institute of Neurology, London, United Kingdom
| | - Selina Wray
- Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Charles Arber
- Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, London, United Kingdom,*Correspondence: Charles Arber,
| |
Collapse
|
164
|
Makamte S, Thureau A, Jabrani A, Paquelin A, Plessis A, Sanial M, Rudenko O, Oteri F, Baaden M, Biou V. A large disordered region confers a wide spanning volume to vertebrate Suppressor of Fused as shown in a trans-species solution study. J Struct Biol 2022; 214:107853. [DOI: 10.1016/j.jsb.2022.107853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 03/10/2022] [Accepted: 03/24/2022] [Indexed: 10/18/2022]
|
165
|
Haitani T, Kobayashi M, Koyasu S, Akamatsu S, Suwa T, Onodera Y, Nam JM, Nguyen PTL, Menju T, Date H, Ogawa O, Harada H. Proteolysis of a histone acetyl reader, ATAD2, induces chemoresistance of cancer cells under severe hypoxia by inhibiting cell cycle progression in S phase. Cancer Lett 2022; 528:76-84. [PMID: 34973392 DOI: 10.1016/j.canlet.2021.12.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 12/01/2021] [Accepted: 12/26/2021] [Indexed: 11/27/2022]
Abstract
Cancer cells acquire chemoresistance in hypoxic regions of solid tumors, which is suggested to be at least partly due to reduction of their proliferative activity. However, molecular mechanisms behind it have not been fully elucidated. Here, we revealed the importance of active proteolysis of a histone acetylation reader, ATPase family AAA domain containing 2 (ATAD2), under hypoxia. We found that inactivation of an O2/Fe2+/α-ketoglutarate-dependent dioxygenase triggered ATAD2 proteolysis by the proteasome system upon severe hypoxia in a hypoxia-inducible factors (HIFs)-independent manner. Consistently, ATAD2 expression levels were markedly lower in perinecrotic hypoxic regions in both xenografted and clinical tumor tissues. The ATAD2 proteolysis was accompanied by a decrease in the amount of acetylated histone H3 lysine 27 and inhibited cell cycle progression from the early to late S phase under severe hypoxia. The retardation of S phase progression induced chemoresistance, which was blocked by overexpression of ATAD2. Together, these results indicate that ATAD2 proteolysis upon severe hypoxia induces chemoresistance of cancer cells through heterochromatinization and the subsequent retardation of S phase progression; therefore, inhibition of ATAD2 proteolysis is expected to be a strategy to overcome chemoresistance of hypoxic tumor cells.
Collapse
Affiliation(s)
- Takao Haitani
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan; Department of Genome Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan; Department of Urology, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Minoru Kobayashi
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan; Department of Genome Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Sho Koyasu
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan; Department of Genome Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan; Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8904, Japan
| | - Shusuke Akamatsu
- Department of Urology, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Tatsuya Suwa
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan; Department of Genome Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan; Department of Radiation Oncology and Image-applied Therapy, Graduate School of Medicine, Kyoto University, Kyoto, 606-8507, Japan
| | - Yasuhito Onodera
- Global Center for Biomedical Science and Engineering, Faculty of Medicine, Hokkaido University, Sapporo, 060-8638, Japan
| | - Jin-Min Nam
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan; Department of Genome Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Phuong Thi Lien Nguyen
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan; Department of Genome Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Toshi Menju
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, 606-8507, Japan
| | - Hiroshi Date
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, 606-8507, Japan
| | - Osamu Ogawa
- Department of Urology, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Hiroshi Harada
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan; Department of Genome Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan.
| |
Collapse
|
166
|
do Amaral MJ, Freire MHO, Almeida MS, Pinheiro AS, Cordeiro Y. Phase separation of the mammalian prion protein: physiological and pathological perspectives. J Neurochem 2022. [PMID: 35149997 DOI: 10.1111/jnc.15586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 01/24/2022] [Accepted: 01/31/2022] [Indexed: 11/27/2022]
Abstract
Abnormal phase transitions have been implicated in the occurrence of proteinopathies. Disordered proteins with nucleic acid binding ability drive the formation of reversible micron-sized condensates capable of controlling nucleic acid processing/transport. This mechanism, achieved via liquid-liquid phase separation (LLPS), underlies the formation of long-studied membraneless organelles (e.g., nucleolus) and various transient condensates formed by driver proteins. The prion protein (PrP) is not a classical nucleic acid-binding protein. However, it binds nucleic acids with high affinity, undergoes nucleocytoplasmic shuttling, contains a long intrinsically disordered region rich in glycines and evenly spaced aromatic residues, among other biochemical/biophysical properties of bona fide drivers of phase transitions. Because of this, our group and others have characterized LLPS of recombinant PrP. In vitro phase separation of PrP is modulated by nucleic acid aptamers, and, depending on the aptamer conformation, the liquid droplets evolve to solid-like species. Herein we discuss recent studies and previous evidence supporting PrP phase transitions. We focus on the central role of LLPS related to PrP physiology and pathology, with a special emphasis on the interaction of PrP with different ligands, such as proteins and nucleic acids, which can play a role in prion disease pathogenesis. Finally, we comment on therapeutic strategies directed at the nonfunctional phase separation that could potentially tackle prion diseases or other protein misfolding disorders.
Collapse
Affiliation(s)
- Mariana J do Amaral
- Faculty of Pharmacy, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | | | - Anderson S Pinheiro
- Department of Biochemistry, Institute of Chemistry, Rio de Janeiro, RJ, Brazil
| | - Yraima Cordeiro
- Faculty of Pharmacy, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
167
|
Wang W, Gong YN. MLKL ubiquitylation: more than a makeover. Cell Death Differ 2022; 29:269-271. [PMID: 35022572 PMCID: PMC8816916 DOI: 10.1038/s41418-022-00934-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 12/30/2021] [Accepted: 01/04/2022] [Indexed: 02/03/2023] Open
Affiliation(s)
- Weihong Wang
- grid.478063.e0000 0004 0456 9819Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15232 USA ,grid.21925.3d0000 0004 1936 9000Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261 USA
| | - Yi-Nan Gong
- grid.478063.e0000 0004 0456 9819Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15232 USA ,grid.21925.3d0000 0004 1936 9000Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261 USA
| |
Collapse
|
168
|
Zhou Z, Li Y, Xu H, Xie X, He Z, Lin S, Li R, Jin S, Cui J, Hu H, Liu F, Wu S, Ma W, Songyang Z. An inducible CRISPR/Cas9 screen identifies DTX2 as a transcriptional regulator of human telomerase. iScience 2022; 25:103813. [PMID: 35198878 PMCID: PMC8844827 DOI: 10.1016/j.isci.2022.103813] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 12/07/2021] [Accepted: 01/20/2022] [Indexed: 01/12/2023] Open
Abstract
Most tumor cells reactivate telomerase to ensure unlimited proliferation, whereas the expression of human telomerase reverse transcriptase (hTERT) is tightly regulated and rate-limiting for telomerase activity maintenance. Several general transcription factors (TFs) have been found in regulating hTERT transcription; however, a systematic study is lacking. Here we performed an inducible CRISPR/Cas9 KO screen using an hTERT core promoter-driven reporter. We identified numerous positive regulators including an E3 ligase DTX2. In telomerase-positive cancer cells, DTX2 depletion downregulated hTERT transcription and telomerase activity, contributing to progressive telomere shortening, growth arrest, and increased apoptosis. Utilizing BioID, we characterized multiple TFs as DTX2 proximal proteins, among which NFIC functioned corporately with DTX2 in promoting hTERT transcription. Further analysis demonstrated that DTX2 mediated K63-linked ubiquitination of NFIC, which facilitated NFIC binding to the hTERT promoter and enhanced hTERT expression. These findings highlight a new hTERT regulatory pathway that may be exploited for potential cancer therapeutics. An inducible CRISPR/Cas9 screen identifies regulators for hTERT transcription DTX2 deficiency leads to telomere shortening and cell growth arrest DTX2 mediates ubiquitination on NFIC, stabilizing NFIC binding on hTERT promoter DTX2-NFIC functions corporately to promote hTERT transcription and tumorigenesis
Collapse
Affiliation(s)
- Zhifen Zhou
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Guangzhou Key Laboratory of Healthy Aging Research, Institute of Healthy Aging Research, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Yujing Li
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Guangzhou Key Laboratory of Healthy Aging Research, Institute of Healthy Aging Research, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Huimin Xu
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Guangzhou Key Laboratory of Healthy Aging Research, Institute of Healthy Aging Research, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Xiaowei Xie
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Guangzhou Key Laboratory of Healthy Aging Research, Institute of Healthy Aging Research, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Zibin He
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Guangzhou Key Laboratory of Healthy Aging Research, Institute of Healthy Aging Research, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Song Lin
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Guangzhou Key Laboratory of Healthy Aging Research, Institute of Healthy Aging Research, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Ruofei Li
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Guangzhou Key Laboratory of Healthy Aging Research, Institute of Healthy Aging Research, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Shouheng Jin
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Guangzhou Key Laboratory of Healthy Aging Research, Institute of Healthy Aging Research, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Jun Cui
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Guangzhou Key Laboratory of Healthy Aging Research, Institute of Healthy Aging Research, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Hai Hu
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Feng Liu
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Guangzhou Key Laboratory of Healthy Aging Research, Institute of Healthy Aging Research, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Su Wu
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Guangzhou Key Laboratory of Healthy Aging Research, Institute of Healthy Aging Research, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
- Corresponding author
| | - Wenbin Ma
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Guangzhou Key Laboratory of Healthy Aging Research, Institute of Healthy Aging Research, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
- Corresponding author
| | - Zhou Songyang
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, Guangzhou Key Laboratory of Healthy Aging Research, Institute of Healthy Aging Research, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510005, China
- Corresponding author
| |
Collapse
|
169
|
Ren C, Han X, Lu C, Yang T, Qiao P, Sun Y, Yu Z. Ubiquitination of NF-κB p65 by FBXW2 suppresses breast cancer stemness, tumorigenesis, and paclitaxel resistance. Cell Death Differ 2022; 29:381-392. [PMID: 34465889 PMCID: PMC8816940 DOI: 10.1038/s41418-021-00862-4] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 08/22/2021] [Accepted: 08/23/2021] [Indexed: 02/07/2023] Open
Abstract
The F-box and WD-repeat-containing protein 2 (FBXW2) plays a crucial role as an E3 ligase in regulating tumorigenesis. However, the functions of FBXW2 in breast cancer are still unknown. Here, we find that nuclear factor-kB (NF-κB) p65 is a new substrate of FBXW2. FBXW2 directly binds to p65, leading to its ubiquitination and degradation. Interestingly, p300 acetylation of p65 blocks FBXW2 induced p65 ubiquitination. FBXW2-p65 axis is a crucial regulator of SOX2-induced stemness in breast cancer. Moreover, FBXW2 inhibits breast tumor growth by regulating p65 degradation in vitro and in vivo. FBXW2 overexpression abrogates the effects of p65 on paclitaxel resistance in vitro and in vivo. Furthermore, FBXW2 induced p65 degradation is also confirmed in FBXW2-knockout mice. Our results identify FBXW2 as an important E3 ligase for p65 degradation, which provide insights into the tumor suppressor functions of FBXW2 in breast cancer.
Collapse
Affiliation(s)
- Chune Ren
- grid.268079.20000 0004 1790 6079Department of Reproductive Medicine, Affiliated Hospital of Weifang Medical University, Weifang, PR China
| | - Xue Han
- grid.268079.20000 0004 1790 6079Department of Reproductive Medicine, Affiliated Hospital of Weifang Medical University, Weifang, PR China
| | - Chao Lu
- grid.268079.20000 0004 1790 6079Department of Reproductive Medicine, Affiliated Hospital of Weifang Medical University, Weifang, PR China
| | - Tingting Yang
- grid.268079.20000 0004 1790 6079Department of Reproductive Medicine, Affiliated Hospital of Weifang Medical University, Weifang, PR China
| | - Pengyun Qiao
- grid.268079.20000 0004 1790 6079Department of Reproductive Medicine, Affiliated Hospital of Weifang Medical University, Weifang, PR China
| | - Yonghong Sun
- grid.268079.20000 0004 1790 6079Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, PR China
| | - Zhenhai Yu
- grid.268079.20000 0004 1790 6079Department of Reproductive Medicine, Affiliated Hospital of Weifang Medical University, Weifang, PR China
| |
Collapse
|
170
|
From Drosophila to Human: Biological Function of E3 Ligase Godzilla and Its Role in Disease. Cells 2022; 11:cells11030380. [PMID: 35159190 PMCID: PMC8834447 DOI: 10.3390/cells11030380] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 12/29/2022] Open
Abstract
The ubiquitin–proteasome system is of fundamental importance in all fields of biology due to its impact on proteostasis and in regulating cellular processes. Ubiquitination, a type of protein post-translational modification, involves complex enzymatic machinery, such as E3 ubiquitin ligases. The E3 ligases regulate the covalent attachment of ubiquitin to a target protein and are involved in various cellular mechanisms, including the cell cycle, cell division, endoplasmic reticulum stress, and neurotransmission. Because the E3 ligases regulate so many physiological events, they are also associated with pathologic conditions, such as cancer, neurological disorders, and immune-related diseases. This review focuses specifically on the protease-associated transmembrane-containing the Really Interesting New Gene (RING) subset of E3 ligases. We describe the structure, partners, and physiological functions of the Drosophila Godzilla E3 ligase and its human homologues, RNF13, RNF167, and ZNRF4. Also, we summarize the information that has emerged during the last decade regarding the association of these E3 ligases with pathophysiological conditions, such as cancer, asthma, and rare genetic disorders. We conclude by highlighting the limitations of the current knowledge and pinpointing the unresolved questions relevant to RNF13, RNF167, and ZNRF4 ubiquitin ligases.
Collapse
|
171
|
Symmetry of Post-Translational Modifications in a Human Enzyme. Symmetry (Basel) 2022. [DOI: 10.3390/sym14020212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Paraoxonase 2 (PON2) is a member of a small family of human lactonases. Recently, post-translational modifications (PTMs) of PON2 were highlighted, one of which involved the modulation of the enzyme activity. Furthermore, two important single nucleotide polymorphisms (SNPs) involved in type 2 diabetes and its consequences, were found to modulate the enzyme activity as well. The position on the PON2 structural model of both residues corresponding to SNPs and PTMs suggested a symmetry of the molecule. By sequence and structure superposition we were able to confirm this finding. The result will be discussed in light of the evolution of symmetry in biological molecules and their function.
Collapse
|
172
|
Hwang JY, Wang H, Lu Y, Ikawa M, Chung JJ. C2cd6-encoded CatSperτ targets sperm calcium channel to Ca 2+ signaling domains in the flagellar membrane. Cell Rep 2022; 38:110226. [PMID: 34998468 PMCID: PMC8857959 DOI: 10.1016/j.celrep.2021.110226] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/29/2021] [Accepted: 12/10/2021] [Indexed: 12/14/2022] Open
Abstract
In mammalian sperm cells, regulation of spatiotemporal Ca2+ signaling relies on the quadrilinear Ca2+ signaling nanodomains in the flagellar membrane. The sperm-specific, multi-subunit CatSper Ca2+ channel, which is crucial for sperm hyperactivated motility and male fertility, organizes the nanodomains. Here, we report CatSperτ, the C2cd6-encoded membrane-associating C2 domain protein, can independently migrate to the flagella and serve as a major targeting component of the CatSper channel complex. CatSperτ loss of function in mice demonstrates that it is essential for sperm hyperactivated motility and male fertility. CatSperτ targets the CatSper channel into the quadrilinear nanodomains in the flagella of developing spermatids, whereas it is dispensable for functional channel assembly. CatSperτ interacts with ciliary trafficking machinery in a C2-dependent manner. These findings provide insights into the CatSper channel trafficking to the Ca2+ signaling nanodomains and the shared molecular mechanisms of ciliary and flagellar membrane targeting.
Collapse
Affiliation(s)
- Jae Yeon Hwang
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Huafeng Wang
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Yonggang Lu
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 5650871, Japan
| | - Masahito Ikawa
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 5650871, Japan
| | - Jean-Ju Chung
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT 06510, USA; Department of Gynecology and Obstetrics, Yale School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
173
|
Iannetta AA, Hicks LM. Maximizing Depth of PTM Coverage: Generating Robust MS Datasets for Computational Prediction Modeling. Methods Mol Biol 2022; 2499:1-41. [PMID: 35696073 DOI: 10.1007/978-1-0716-2317-6_1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Post-translational modifications (PTMs) regulate complex biological processes through the modulation of protein activity, stability, and localization. Insights into the specific modification type and localization within a protein sequence can help ascertain functional significance. Computational models are increasingly demonstrated to offer a low-cost, high-throughput method for comprehensive PTM predictions. Algorithms are optimized using existing experimental PTM data, thus accurate prediction performance relies on the creation of robust datasets. Herein, advancements in mass spectrometry-based proteomics technologies to maximize PTM coverage are reviewed. Further, requisite experimental validation approaches for PTM predictions are explored to ensure that follow-up mechanistic studies are focused on accurate modification sites.
Collapse
Affiliation(s)
- Anthony A Iannetta
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Leslie M Hicks
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
174
|
Paredes R, Doleschall N, Connors K, Geary B, Meyer S. EVI1 protein interaction dynamics: targetable for therapeutic intervention? Exp Hematol 2021; 107:1-8. [PMID: 34958895 DOI: 10.1016/j.exphem.2021.12.398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 12/18/2021] [Accepted: 12/20/2021] [Indexed: 11/04/2022]
Abstract
High expression of the transcriptional regulator EVI1 encoded at the MECOM locus at 3q26 is one of the most aggressive oncogenic drivers in acute myeloid leukaemia (AML) and carries a very poor prognosis. How EVI1 confers leukaemic transformation and chemotherapy resistance in AML is subject to important ongoing clinical and experimental studies. Recent discoveries have revealed critical details about genetic mechanisms of the activation of EVI1 overexpression and downstream events of aberrantly high EVI1 expression. Here we review and discuss aspects concerning the protein interactions of EVI1 and the related proteins MDS-EVI1 and ΔEVI1 from the perspective of their potential for therapeutic intervention.
Collapse
Affiliation(s)
- Roberto Paredes
- Stem Cell and Leukaemia Proteomics Laboratory, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK; Manchester Academic Health Science Centre, National Institute for Health Research Biomedical Research Centre, Manchester
| | - Nora Doleschall
- Stem Cell and Leukaemia Proteomics Laboratory, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK; Manchester Academic Health Science Centre, National Institute for Health Research Biomedical Research Centre, Manchester
| | - Kathleen Connors
- Stem Cell and Leukaemia Proteomics Laboratory, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK; Manchester Academic Health Science Centre, National Institute for Health Research Biomedical Research Centre, Manchester
| | - Bethany Geary
- Stem Cell and Leukaemia Proteomics Laboratory, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK; Manchester Academic Health Science Centre, National Institute for Health Research Biomedical Research Centre, Manchester
| | - Stefan Meyer
- Stem Cell and Leukaemia Proteomics Laboratory, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK; Manchester Academic Health Science Centre, National Institute for Health Research Biomedical Research Centre, Manchester; Department of Paediatric Haematology and Oncology, Royal Manchester Children's Hospital; Young Oncology Unit, The Christie NHS Foundation Trust.
| |
Collapse
|
175
|
Enkhbat G, Nakanishi A, Miki Y. The BRCA2 missense mutation K2497R suppressed self-degradation and increased ATP production and cell proliferation. Biochem Biophys Res Commun 2021; 590:27-33. [PMID: 34968781 DOI: 10.1016/j.bbrc.2021.12.073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 12/20/2021] [Indexed: 11/29/2022]
Abstract
Breast cancer susceptibility gene 2 (BRCA2) mediates genome maintenance during the S phase of the cell cycle, with important roles in replication stress, centrosome replication, and cytokinesis. In this study, we showed that a small heat shock protein, HSP27, interacted with and participated in the degradation of BRCA2 in estrogen-treated MCF-7 cells. BRCA2 degradation reportedly requires ubiquitination of the C-terminal region; thus, fragments of amino acid (aa) residues 2241-2940 were produced and assayed for their degradation following cycloheximide (CHX) treatment. The results showed that aa 2491-2580 affected the degradation of BRCA2, especially lysine (Lys) 2497. Furthermore, the K2497 A/R mutation increased ATP production and the proliferation of DLD-1 (BRCA2 knockout) cells compared to the cells expressing wild-type BRCA2-FLAG. Notably, a single residue, Lys2497, affected BRCA2 degradation, and K2497R is reportedly a missense mutation in hereditary breast cancer.
Collapse
Affiliation(s)
- Gerelmaa Enkhbat
- Department of Specialized Surgeries, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan; Department of Molecular Genetics, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Akira Nakanishi
- Department of Molecular Genetics, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan.
| | - Yoshio Miki
- Department of Molecular Genetics, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan; Department of Genetic Diagnosis, The Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan.
| |
Collapse
|
176
|
Arsenault HE, Ghizzoni JM, Leech CM, Diers AR, Gesta S, Vishnudas VK, Narain NR, Sarangarajan R, Benanti JA. Ubc1 turnover contributes to the spindle assembly checkpoint in Saccharomyces cerevisiae. G3 (BETHESDA, MD.) 2021; 11:jkab346. [PMID: 34586382 PMCID: PMC8664427 DOI: 10.1093/g3journal/jkab346] [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] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 09/20/2021] [Indexed: 11/21/2022]
Abstract
The spindle assembly checkpoint protects the integrity of the genome by ensuring that chromosomes are properly attached to the mitotic spindle before they are segregated during anaphase. Activation of the spindle checkpoint results in inhibition of the Anaphase-Promoting Complex (APC), an E3 ubiquitin ligase that triggers the metaphase-anaphase transition. Here, we show that levels of Ubc1, an E2 enzyme that functions in complex with the APC, modulate the response to spindle checkpoint activation in Saccharomyces cerevisiae. Overexpression of Ubc1 increased resistance to microtubule poisons, whereas Ubc1 shut-off sensitized cells. We also found that Ubc1 levels are regulated by the spindle checkpoint. Checkpoint activation or direct APC inhibition led to a decrease in Ubc1 levels, charging, and half-life. Additionally, stabilization of Ubc1 prevented its down-regulation by the spindle checkpoint and increased resistance to checkpoint-activating drugs. These results suggest that down-regulation of Ubc1 in response to spindle checkpoint signaling is necessary for a robust cell cycle arrest.
Collapse
Affiliation(s)
- Heather E Arsenault
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Julie M Ghizzoni
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Cassandra M Leech
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | | | | | | | | | | - Jennifer A Benanti
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| |
Collapse
|
177
|
Foster B, Attwood M, Gibbs-Seymour I. Tools for Decoding Ubiquitin Signaling in DNA Repair. Front Cell Dev Biol 2021; 9:760226. [PMID: 34950659 PMCID: PMC8690248 DOI: 10.3389/fcell.2021.760226] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 11/09/2021] [Indexed: 12/21/2022] Open
Abstract
The maintenance of genome stability requires dedicated DNA repair processes and pathways that are essential for the faithful duplication and propagation of chromosomes. These DNA repair mechanisms counteract the potentially deleterious impact of the frequent genotoxic challenges faced by cells from both exogenous and endogenous agents. Intrinsic to these mechanisms, cells have an arsenal of protein factors that can be utilised to promote repair processes in response to DNA lesions. Orchestration of the protein factors within the various cellular DNA repair pathways is performed, in part, by post-translational modifications, such as phosphorylation, ubiquitin, SUMO and other ubiquitin-like modifiers (UBLs). In this review, we firstly explore recent advances in the tools for identifying factors involved in both DNA repair and ubiquitin signaling pathways. We then expand on this by evaluating the growing repertoire of proteomic, biochemical and structural techniques available to further understand the mechanistic basis by which these complex modifications regulate DNA repair. Together, we provide a snapshot of the range of methods now available to investigate and decode how ubiquitin signaling can promote DNA repair and maintain genome stability in mammalian cells.
Collapse
Affiliation(s)
| | | | - Ian Gibbs-Seymour
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
178
|
Huntingtin Ubiquitination Mechanisms and Novel Possible Therapies to Decrease the Toxic Effects of Mutated Huntingtin. J Pers Med 2021; 11:jpm11121309. [PMID: 34945781 PMCID: PMC8709430 DOI: 10.3390/jpm11121309] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/19/2021] [Accepted: 11/21/2021] [Indexed: 12/24/2022] Open
Abstract
Huntington Disease (HD) is a dominant, lethal neurodegenerative disorder caused by the abnormal expansion (>35 copies) of a CAG triplet located in exon 1 of the HTT gene encoding the huntingtin protein (Htt). Mutated Htt (mHtt) easily aggregates, thereby inducing ER stress that in turn leads to neuronal injury and apoptosis. Therefore, both the inhibition of mHtt aggregate formation and the acceleration of mHtt degradation represent attractive strategies to delay HD progression, and even for HD treatment. Here, we describe the mechanism underlying mHtt degradation by the ubiquitin–proteasome system (UPS), which has been shown to play a more important role than the autophagy–lysosomal pathway. In particular, we focus on E3 ligase proteins involved in the UPS and detail their structure–function relationships. In this framework, we discuss the possible exploitation of PROteolysis TArgeting Chimeras (PROTACs) for HD therapy. PROTACs are heterobifunctional small molecules that comprise two different ligands joined by an appropriate linker; one of the ligands is specific for a selected E3 ubiquitin ligase, the other ligand is able to recruit a target protein of interest, in this case mHtt. As a consequence of PROTAC binding, mHtt and the E3 ubiquitin ligase can be brought to a relative position that allows mHtt to be ubiquitinated and, ultimately, allows a reduction in the amount of mHtt in the cell.
Collapse
|
179
|
Activation of homologous recombination in G1 preserves centromeric integrity. Nature 2021; 600:748-753. [PMID: 34853474 DOI: 10.1038/s41586-021-04200-z] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 11/02/2021] [Indexed: 01/01/2023]
Abstract
Centromeric integrity is key for proper chromosome segregation during cell division1. Centromeres have unique chromatin features that are essential for centromere maintenance2. Although they are intrinsically fragile and represent hotspots for chromosomal rearrangements3, little is known about how centromere integrity in response to DNA damage is preserved. DNA repair by homologous recombination requires the presence of the sister chromatid and is suppressed in the G1 phase of the cell cycle4. Here we demonstrate that DNA breaks that occur at centromeres in G1 recruit the homologous recombination machinery, despite the absence of a sister chromatid. Mechanistically, we show that the centromere-specific histone H3 variant CENP-A and its chaperone HJURP, together with dimethylation of lysine 4 in histone 3 (H3K4me2), enable a succession of events leading to the licensing of homologous recombination in G1. H3K4me2 promotes DNA-end resection by allowing DNA damage-induced centromeric transcription and increased formation of DNA-RNA hybrids. CENP-A and HJURP interact with the deubiquitinase USP11, enabling formation of the RAD51-BRCA1-BRCA2 complex5 and rendering the centromeres accessible to RAD51 recruitment and homologous recombination in G1. Finally, we show that inhibition of homologous recombination in G1 leads to centromeric instability and chromosomal translocations. Our results support a model in which licensing of homologous recombination at centromeric breaks occurs throughout the cell cycle to prevent the activation of mutagenic DNA repair pathways and preserve centromeric integrity.
Collapse
|
180
|
Gladyshev VN, Kritchevsky SB, Clarke SG, Cuervo AM, Fiehn O, de Magalhães JP, Mau T, Maes M, Moritz R, Niedernhofer LJ, Van Schaftingen E, Tranah GJ, Walsh K, Yura Y, Zhang B, Cummings SR. Molecular Damage in Aging. NATURE AGING 2021; 1:1096-1106. [PMID: 36846190 PMCID: PMC9957516 DOI: 10.1038/s43587-021-00150-3] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 11/04/2021] [Indexed: 11/09/2022]
Abstract
Cellular metabolism generates molecular damage affecting all levels of biological organization. Accumulation of this damage over time is thought to play a central role in the aging process, but damage manifests in diverse molecular forms complicating its assessment. Insufficient attention has been paid to date to the role of molecular damage in aging-related phenotypes, particularly in humans, in part because of the difficulty in measuring its various forms. Recently, omics approaches have been developed that begin to address this challenge, because they are able to assess a sizeable proportion of age-related damage at the level of small molecules, proteins, RNA, DNA, organelles and cells. This review describes the concept of molecular damage in aging and discusses its diverse aspects from theoretical models to experimental approaches. Measurement of multiple types of damage enables studies of the role of damage in human aging outcomes and lays a foundation for testing interventions to reduce the burden of molecular damage, opening new approaches to slowing aging and reducing its consequences.
Collapse
Affiliation(s)
- Vadim N. Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Stephen B. Kritchevsky
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - Steven G. Clarke
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Ana Maria Cuervo
- Department of Development and Molecular Biology, Albert Einstein College of Medicine, New York, NY 10461, USA
- Institute for Aging Studies, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Oliver Fiehn
- West Coast Metabolomics Center, University of California Davis, Davis, CA 95616, USA
| | - João Pedro de Magalhães
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L7 8TX, UK
| | - Theresa Mau
- San Francisco Coordinating Center, California Pacific Medical Center, Research Institute, San Francisco, CA 94143, USA
| | - Michal Maes
- Institute for Systems Biology, Seattle, WA 98109, USA
| | - Robert Moritz
- Institute for Systems Biology, Seattle, WA 98109, USA
| | - Laura J. Niedernhofer
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Emile Van Schaftingen
- De Duve Institute, Université catholique de Louvain, Bruxelles, Belgium
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Université catholique de Louvain, Bruxelles, Belgium
| | - Gregory J. Tranah
- San Francisco Coordinating Center, California Pacific Medical Center, Research Institute, San Francisco, CA 94143, USA
| | - Kenneth Walsh
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia-School of Medicine, Charlottesville, VA 22908, USA
| | - Yoshimitsu Yura
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia-School of Medicine, Charlottesville, VA 22908, USA
| | - Bohan Zhang
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Steven R. Cummings
- San Francisco Coordinating Center, California Pacific Medical Center, Research Institute, San Francisco, CA 94143, USA
| |
Collapse
|
181
|
Delgado ILS, Tavares A, Francisco S, Santos D, Coelho J, Basto AP, Zúquete S, Müller J, Hemphill A, Meissner M, Soares H, Leitão A, Nolasco S. Characterization of a MOB1 Homolog in the Apicomplexan Parasite Toxoplasma gondii. BIOLOGY 2021; 10:biology10121233. [PMID: 34943148 PMCID: PMC8698288 DOI: 10.3390/biology10121233] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/19/2021] [Accepted: 11/23/2021] [Indexed: 01/11/2023]
Abstract
Simple Summary Monopolar spindle One Binder1 (MOB1) proteins regulate key cellular functions, namely cell multiplication and cell division. The unicellular parasite Toxoplasma gondii transitions between several morphological stages, with the need to control the number of parasites in its cellular environment. We hypothesized that MOB1 proteins could participate in the regulation of the T. gondii life cycle, having identified one MOB1 protein (TgMOB1) coded in its genome. However, this study shows that TgMOB1 presents divergent features. While in organisms studied to date the lack of MOB1 has led to cell division defects, this did not occur in T. gondii in vitro cultures where mob1 was not an essential gene. Additionally, the identification of TgMOB1 proximity interacting partners detected novel MOB1 interactors. Still, TgMOB1 localizes to the region between the new-forming nuclei during cell division, and T. gondii parasites multiply slower with TgMOB1 overexpression and faster when there is a lack of TgMOB1, indicating an intricate role for TgMOB1 in T. gondii. This study uncovers new features of the T. gondii biology, a zoonotic parasite and model organism for the phylum Apicomplexa, and highlights the complex roles MOB1 proteins may assume, with possible implications for disease processes. Abstract Monopolar spindle One Binder1 (MOB1) proteins are conserved components of the tumor-suppressing Hippo pathway, regulating cellular processes such as cytokinesis. Apicomplexan parasites present a life cycle that relies on the parasites’ ability to differentiate between stages and regulate their proliferation; thus, Hippo signaling pathways could play an important role in the regulation of the apicomplexan life cycle. Here, we report the identification of one MOB1 protein in the apicomplexan Toxoplasma gondii. To characterize the function of MOB1, we generated gain-of-function transgenic lines with a ligand-controlled destabilization domain, and loss-of-function clonal lines obtained through CRISPR/Cas9 technology. Contrary to what has been characterized in other eukaryotes, MOB1 is not essential for cytokinesis in T. gondii. However, this picture is complex since we found MOB1 localized between the newly individualized daughter nuclei at the end of mitosis. Moreover, we detected a significant delay in the replication of overexpressing tachyzoites, contrasting with increased replication rates in knockout tachyzoites. Finally, using the proximity-biotinylation method, BioID, we identified novel members of the MOB1 interactome, a probable consequence of the observed lack of conservation of some key amino acid residues. Altogether, the results point to a complex evolutionary history of MOB1 roles in apicomplexans, sharing properties with other eukaryotes but also with divergent features, possibly associated with their complex life cycle.
Collapse
Affiliation(s)
- Inês L. S. Delgado
- CIISA—Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisboa, Portugal or (I.L.S.D.); (A.T.); (S.F.); (D.S.); (J.C.); (A.P.B.); (S.Z.); (A.L.)
- Faculdade de Medicina Veterinária, Universidade Lusófona, 1749-024 Lisboa, Portugal
| | - Alexandra Tavares
- CIISA—Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisboa, Portugal or (I.L.S.D.); (A.T.); (S.F.); (D.S.); (J.C.); (A.P.B.); (S.Z.); (A.L.)
| | - Samuel Francisco
- CIISA—Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisboa, Portugal or (I.L.S.D.); (A.T.); (S.F.); (D.S.); (J.C.); (A.P.B.); (S.Z.); (A.L.)
| | - Dulce Santos
- CIISA—Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisboa, Portugal or (I.L.S.D.); (A.T.); (S.F.); (D.S.); (J.C.); (A.P.B.); (S.Z.); (A.L.)
| | - João Coelho
- CIISA—Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisboa, Portugal or (I.L.S.D.); (A.T.); (S.F.); (D.S.); (J.C.); (A.P.B.); (S.Z.); (A.L.)
| | - Afonso P. Basto
- CIISA—Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisboa, Portugal or (I.L.S.D.); (A.T.); (S.F.); (D.S.); (J.C.); (A.P.B.); (S.Z.); (A.L.)
| | - Sara Zúquete
- CIISA—Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisboa, Portugal or (I.L.S.D.); (A.T.); (S.F.); (D.S.); (J.C.); (A.P.B.); (S.Z.); (A.L.)
| | - Joachim Müller
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Länggass-Strasse 122, CH-3012 Bern, Switzerland; (J.M.); (A.H.)
| | - Andrew Hemphill
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Länggass-Strasse 122, CH-3012 Bern, Switzerland; (J.M.); (A.H.)
| | - Markus Meissner
- Institute for Experimental Parasitology, Faculty of Veterinary Medicine, Ludwig-Maximilians-Universität Munich, D-82152 Munich, Germany;
| | - Helena Soares
- Escola Superior de Tecnologia da Saúde de Lisboa, Instituto Politécnico de Lisboa, 1990-096 Lisboa, Portugal; or
- Centro de Química Estrutural–Faculdade de Ciências da Universidade de Lisboa, 1749-016 Lisboa, Portugal
| | - Alexandre Leitão
- CIISA—Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisboa, Portugal or (I.L.S.D.); (A.T.); (S.F.); (D.S.); (J.C.); (A.P.B.); (S.Z.); (A.L.)
| | - Sofia Nolasco
- CIISA—Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisboa, Portugal or (I.L.S.D.); (A.T.); (S.F.); (D.S.); (J.C.); (A.P.B.); (S.Z.); (A.L.)
- Escola Superior de Tecnologia da Saúde de Lisboa, Instituto Politécnico de Lisboa, 1990-096 Lisboa, Portugal; or
- Correspondence: or
| |
Collapse
|
182
|
Kmiecik SW, Mayer MP. Molecular mechanisms of heat shock factor 1 regulation. Trends Biochem Sci 2021; 47:218-234. [PMID: 34810080 DOI: 10.1016/j.tibs.2021.10.004] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 10/08/2021] [Accepted: 10/22/2021] [Indexed: 02/06/2023]
Abstract
To thrive and to fulfill their functions, cells need to maintain proteome homeostasis even in the face of adverse environmental conditions or radical restructuring of the proteome during differentiation. At the center of the regulation of proteome homeostasis is an ancient transcriptional mechanism, the so-called heat shock response (HSR), orchestrated in all eukaryotic cells by heat shock transcription factor 1 (Hsf1). As Hsf1 is implicated in aging and several pathologies like cancer and neurodegenerative disorders, understanding the regulation of Hsf1 could open novel therapeutic opportunities. In this review, we discuss the regulation of Hsf1's transcriptional activity by multiple layers of control circuits involving Hsf1 synthesis and degradation, conformational rearrangements and post-translational modifications (PTMs), and molecular chaperones in negative feedback loops.
Collapse
Affiliation(s)
- Szymon W Kmiecik
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH-Alliance, Im Neuenheimer Feld 282, D-69120 Heidelberg, Germany
| | - Matthias P Mayer
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH-Alliance, Im Neuenheimer Feld 282, D-69120 Heidelberg, Germany.
| |
Collapse
|
183
|
Abstract
The intimate association between obesity and type II diabetes urges for a deeper understanding of adipocyte function. We and others have previously delineated a role for the tumor suppressor p53 in adipocyte biology. Here, we show that mice haploinsufficient for MDM2, a key regulator of p53, in their adipose stores suffer from overt obesity, glucose intolerance, and hepatic steatosis. These mice had decreased levels of circulating palmitoleic acid [non-esterified fatty acid (NEFA) 16:1] concomitant with impaired visceral adipose tissue expression of Scd1 and Ffar4. A similar decrease in Scd and Ffar4 expression was found in in vitro differentiated adipocytes with perturbed MDM2 expression. Lowered MDM2 levels led to nuclear exclusion of the transcriptional cofactors, MORC2 and LIPIN1, and thereby possibly hampered adipocyte function by antagonizing LIPIN1-mediated PPARγ coactivation. Collectively, these data argue for a hitherto unknown interplay between MDM2 and MORC2/LIPIN1 involved in balancing adipocyte function.
Collapse
|
184
|
Laigle V, Dingli F, Amhaz S, Perron T, Chouchène M, Colasse S, Petit I, Poullet P, Loew D, Prunier C, Levy L. Quantitative ubiquitylome analysis reveals specificity of RNF111/Arkadia E3 ubiquitin ligase for its degradative substrates SKI and SKIL/SnoN in TGF-β signaling pathway. Mol Cell Proteomics 2021; 20:100173. [PMID: 34740826 PMCID: PMC8665411 DOI: 10.1016/j.mcpro.2021.100173] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 10/06/2021] [Accepted: 11/01/2021] [Indexed: 11/16/2022] Open
Abstract
RNF111/Arkadia is an E3 ubiquitin ligase that activates the TGF-β pathway by degrading transcriptional repressors SKIL/SnoN and SKI, and truncations of the RING C-terminal domain of RNF111 that abolish its E3 function and subsequently TGF-β signaling are observed in some cancers. In the present study, we sought to perform a comprehensive analysis of RNF111 endogenous substrates upon TGF-β signaling activation using an integrative proteomic approach. In that aim we carried out label free quantitative proteomics after enrichment of ubiquitylated proteins (ubiquitylome) in parental U2OS cell line compared to U2OS CRISPR engineered clones expressing a truncated form of RNF111 devoid of its C-terminal RING domain. We compared two methods of enrichment for ubiquitylated proteins prior to proteomics analysis by mass spectrometry, the diGly remnant peptide immunoprecipitation with a K-ε-GG antibody (diGly) and a novel approach using protein immunoprecipitation with a ubiquitin pan nanobody (pan UB) that recognizes all ubiquitin chains and monoubiquitylation. While we detected SKIL ubiquitylation among 108 potential RNF111 substrates with the diGly method, we found that the pan UB method also constitutes a powerful approach since it enabled detection of 52 potential RNF111 substrates including SKI, SKIL and RNF111. Integrative comparison of the RNF111-dependent proteome and ubiquitylomes enabled identification of SKI and SKIL as the only targets ubiquitylated and degraded by RNF111 E3 ligase function in presence of TGF-β. Our results indicate that lysine 343 localized in the SAND domain of SKIL constitutes a target for RNF111 ubiquitylation and demonstrate that RNF111 E3 ubiquitin ligase function specifically targets SKI and SKIL ubiquitylation and degradation upon TGF-β pathway activation.
Collapse
Affiliation(s)
- Victor Laigle
- Institut Curie, PSL Research University, Laboratoire de Spectrométrie de Masse Protéomique, 75005 Paris, France
| | - Florent Dingli
- Institut Curie, PSL Research University, Laboratoire de Spectrométrie de Masse Protéomique, 75005 Paris, France
| | - Sadek Amhaz
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, F-75012, Paris, France
| | - Tiphaine Perron
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, F-75012, Paris, France
| | - Mouna Chouchène
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, F-75012, Paris, France
| | - Sabrina Colasse
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, F-75012, Paris, France
| | - Isabelle Petit
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, F-75012, Paris, France
| | | | - Damarys Loew
- Institut Curie, PSL Research University, Laboratoire de Spectrométrie de Masse Protéomique, 75005 Paris, France
| | - Céline Prunier
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, F-75012, Paris, France
| | - Laurence Levy
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, F-75012, Paris, France.
| |
Collapse
|
185
|
Lu X, Xuan W, Li J, Yao H, Huang C, Li J. AMPK protects against alcohol-induced liver injury through UQCRC2 to up-regulate mitophagy. Autophagy 2021; 17:3622-3643. [PMID: 33719895 PMCID: PMC8632272 DOI: 10.1080/15548627.2021.1886829] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 01/22/2021] [Accepted: 02/03/2021] [Indexed: 02/06/2023] Open
Abstract
Recent reports indicated that mitophagy protects against alcohol-induced liver injury, which helps remove damaged mitochondria to reduce the accumulation of reactive oxygen species (ROS). AMP-activated protein kinase (AMPK) has been recently used in ALD (alcoholic liver disease) and mitochondrial dysfunction research. However, the inner mechanism, whether AMPK can regulate mitophagy in ALD, remains unknown. Here we found that AMPK can significantly reduce alcohol-induced liver injury and enhances hepatocytes' mitophagy level. Next, we identified that AMPK rescued alcohol-induced low expression of UQCRC2 (ubiquinol-cytochrome c reductase core protein 2). Interestingly, UQCRC2 knockdown (KD) treatment causes impaired mitophagy, whereas UQCRC2 overexpression (OE) can significantly increase mitophagy to attenuate liver injury. Also, we identified that AMPK indirectly upregulates UQCRC2 protein level, and RNA-seq, chromatin immunoprecipitation (ChIP) assay, bioinformatics, and luciferase assays helped us understand that AMPK enhanced UQCRC2 gene transcription through activating NFE2L2/NRF2 (nuclear factor, erythroid 2 like 2). Our results demonstrate that AMPK regulating UQCRC2 is a significant mitochondrial event in mitophagy. It identifies a new signaling axis, AMPK-NFE2L2-UQCRC2, in the regulation of mitophagy levels in the liver, suggesting a possible therapeutic strategy to treat ALD.Abbreviations: AAV: AENO-associated virus; ALD: alcoholic liver disease; AMPK: AMP-activated protein kinase; BUN: blood urea nitrogen; H&E: hematoxylin and eosin; CCCP: carbonyl cyanide 3-chlorophenylhydrazone; ChIP: chromatin immunoprecipitation assay; CO-IP: co-immunoprecipitation; COPD: chronic obstructive pulmonary disease; EM: electron microscope; GOT1/AST: glutamic-oxaloacetic transaminase 1; GPT/ALT: glutamic-pyruvic transaminase; IF: immunofluorescence; IHC: immunohistochemistry; KD: knockdown; MAP1LC3/LC3: microtubule associated protein 1 light chain protein 3; MTDR: MitoTracker Deep Red; NFE2L2/NRF2: nuclear factor, erythroid 2 like 2; mtDNA: mitochondrial DNA; MTRC: MitoTracker Red CMXRos; OCR: Oxygen consumption rate; OE: overexpress; PINK1: PTEN induced kinase 1; qRT-PCR: quantitative real-time PCR; ROS: reactive oxygen species; SD: standard deviation; SOD2: superoxide dismutase 2; UQCRC2: ubiquinol-cytochrome c reductase core protein 2; WB: western blot; ΔΨ: mitochondrial membrane potential.
Collapse
Affiliation(s)
- Xinyi Lu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Wenting Xuan
- Department of Anesthesiology, Drum Tower Hospital, Medical College of Nanjing University, Nanjing, China
| | - Juanjuan Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Hongwei Yao
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Cheng Huang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Jun Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
186
|
Hayashi T, Matsushita T, Hisahara S, Iwahara N, Kuno A, Kunimoto R, Hosoda R, Tanno M, Shimohama S, Horio Y. Ubiquitin-dependent rapid degradation conceals a cell-protective function of cytoplasmic SIRT3 against oxidative stress. J Biochem 2021; 171:201-213. [PMID: 34718606 DOI: 10.1093/jb/mvab119] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 10/24/2021] [Indexed: 11/12/2022] Open
Abstract
SIRT3 is an NAD+-dependent protein deacetylase localized in mitochondria. Several studies reported localization of SIRT3 in the cytoplasm or nucleus, but data of these studies were not consistent. We detected expression of mitochondrial (SIRT3mt) and cytoplasmic (SIRT3ct) Sirt3 mRNAs in the mouse brain, and we also found SIRT3 immunostaining of mitochondria and cytoplasm in the brain and cultured neural cells. However, expression levels of SIRT3ct in COS cells transfected with SIRT3ct cDNA were much lower than those of SIRT3mt. We found that SIRT3ct but not SIRT3mt was promptly degraded by ubiquitin-dependent degradation, in which SIRT3ct degradation was mediated mainly by ubiquitination of NH2-terminal methionine and partly by that of lysine residues of SIRT3ct. SIRT3ct expression level was significantly enhanced by treatment of cells with staurosporine or H2O2. H2O2 treatment promoted nuclear translocation of SIRT3ct and induced histone H3 deacetylation and superoxide dismutase 2 expression. Overexpression of SIRT3ct decreased cell death caused by H2O2 at levels similar to those achieved by overexpression of SIRT3mt. Knockdown of Sirt3 mRNA increased cell death caused by amyloid-β (Aβ), and overexpression of SIRT3ct suppressed the toxic function of Aβ in PC12 cells. These results indicate that SIRT3ct promotes cell survival under physiological and pathological conditions.
Collapse
Affiliation(s)
- Takashi Hayashi
- Departments of Pharmacology, Neurology and Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, S 1, W 17, Chu-ouku, Sapporo 060-8556, Japan.,Department of Pharmacology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Takashi Matsushita
- Departments of Pharmacology, Neurology and Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, S 1, W 17, Chu-ouku, Sapporo 060-8556, Japan.,Department of Pharmacology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Shin Hisahara
- Department of Pharmacology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Naotoshi Iwahara
- Departments of Pharmacology, Neurology and Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, S 1, W 17, Chu-ouku, Sapporo 060-8556, Japan.,Department of Pharmacology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Atsushi Kuno
- Departments of Pharmacology, Neurology and Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, S 1, W 17, Chu-ouku, Sapporo 060-8556, Japan
| | - Risa Kunimoto
- Departments of Pharmacology, Neurology and Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, S 1, W 17, Chu-ouku, Sapporo 060-8556, Japan
| | - Ryusuke Hosoda
- Departments of Pharmacology, Neurology and Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, S 1, W 17, Chu-ouku, Sapporo 060-8556, Japan
| | - Masaya Tanno
- Departments of Pharmacology, Neurology and Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, S 1, W 17, Chu-ouku, Sapporo 060-8556, Japan
| | - Shun Shimohama
- Department of Pharmacology, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Yoshiyuki Horio
- Departments of Pharmacology, Neurology and Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, S 1, W 17, Chu-ouku, Sapporo 060-8556, Japan
| |
Collapse
|
187
|
Wilson MM, Callens C, Le Gallo M, Mironov S, Ding Q, Salamagnon A, Chavarria TE, Viel R, Peasah AD, Bhutkar A, Martin S, Godey F, Tas P, Kang HS, Juin PP, Jetten AM, Visvader JE, Weinberg RA, Attanasio M, Prigent C, Lees JA, Guen VJ. An EMT-primary cilium-GLIS2 signaling axis regulates mammogenesis and claudin-low breast tumorigenesis. SCIENCE ADVANCES 2021; 7:eabf6063. [PMID: 34705506 PMCID: PMC8550236 DOI: 10.1126/sciadv.abf6063] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 09/08/2021] [Indexed: 05/14/2023]
Abstract
The epithelial-mesenchymal transition (EMT) and primary ciliogenesis induce stem cell properties in basal mammary stem cells (MaSCs) to promote mammogenesis, but the underlying mechanisms remain incompletely understood. Here, we show that EMT transcription factors promote ciliogenesis upon entry into intermediate EMT states by activating ciliogenesis inducers, including FGFR1. The resulting primary cilia promote ubiquitination and inactivation of a transcriptional repressor, GLIS2, which localizes to the ciliary base. We show that GLIS2 inactivation promotes MaSC stemness, and GLIS2 is required for normal mammary gland development. Moreover, GLIS2 inactivation is required to induce the proliferative and tumorigenic capacities of the mammary tumor–initiating cells (MaTICs) of claudin-low breast cancers. Claudin-low breast tumors can be segregated from other breast tumor subtypes based on a GLIS2-dependent gene expression signature. Collectively, our findings establish molecular mechanisms by which EMT programs induce ciliogenesis to control MaSC and MaTIC stemness, mammary gland development, and claudin-low breast cancer formation.
Collapse
Affiliation(s)
- Molly M. Wilson
- Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Céline Callens
- Institut de Génétique et Développement de Rennes, Centre National de la Recherche Scientifique, Rennes, France
| | - Matthieu Le Gallo
- INSERM U1242, Rennes 1 University, Rennes, France
- Centre de Lutte Contre le Cancer Eugène Marquis, Rennes, France
| | - Svetlana Mironov
- Institut de Génétique et Développement de Rennes, Centre National de la Recherche Scientifique, Rennes, France
| | - Qiong Ding
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Amandine Salamagnon
- Institut de Génétique et Développement de Rennes, Centre National de la Recherche Scientifique, Rennes, France
| | - Tony E. Chavarria
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Roselyne Viel
- Plateforme d’Histopathologie de Haute Précision (H2P2), Rennes, France
| | - Abena D. Peasah
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Arjun Bhutkar
- Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA, USA
| | - Sophie Martin
- INSERM U1242, Rennes 1 University, Rennes, France
- Centre de Lutte Contre le Cancer Eugène Marquis, Rennes, France
| | - Florence Godey
- INSERM U1242, Rennes 1 University, Rennes, France
- Centre de Lutte Contre le Cancer Eugène Marquis, Rennes, France
| | - Patrick Tas
- INSERM U1242, Rennes 1 University, Rennes, France
- Centre de Lutte Contre le Cancer Eugène Marquis, Rennes, France
| | - Hong Soon Kang
- Cell Biology Section, Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | | | - Anton M. Jetten
- Cell Biology Section, Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Jane E. Visvader
- Stem Cells and Cancer Division, The Walter and Eliza Hall Institute of Medical Research and Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Robert A. Weinberg
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- MIT Department of Biology and the Whitehead Institute, Cambridge, MA, USA
| | - Massimo Attanasio
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Claude Prigent
- Institut de Génétique et Développement de Rennes, Centre National de la Recherche Scientifique, Rennes, France
- CRBM, CNRS, Université de Montpellier, Montpellier, France
| | - Jacqueline A. Lees
- Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Vincent J. Guen
- Institut de Génétique et Développement de Rennes, Centre National de la Recherche Scientifique, Rennes, France
- CRCINA, INSERM, Université de Nantes, Nantes, France
| |
Collapse
|
188
|
Akimov V, Fehling-Kaschek M, Barrio-Hernandez I, Puglia M, Bunkenborg J, Nielsen MM, Timmer J, Dengjel J, Blagoev B. Magnitude of Ubiquitination Determines the Fate of Epidermal Growth Factor Receptor Upon Ligand Stimulation. J Mol Biol 2021; 433:167240. [PMID: 34508725 DOI: 10.1016/j.jmb.2021.167240] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 08/17/2021] [Accepted: 09/01/2021] [Indexed: 12/23/2022]
Abstract
Receptor tyrosine kinases (RTK) bind growth factors and are critical for cell proliferation and differentiation. Their dysregulation leads to a loss of growth control, often resulting in cancer. Epidermal growth factor receptor (EGFR) is the prototypic RTK and can bind several ligands exhibiting distinct mitogenic potentials. Whereas the phosphorylation on individual EGFR sites and their roles for downstream signaling have been extensively studied, less is known about ligand-specific ubiquitination events on EGFR, which are crucial for signal attenuation and termination. We used a proteomics-based workflow for absolute quantitation combined with mathematical modeling to unveil potentially decisive ubiquitination events on EGFR from the first 30 seconds to 15 minutes of stimulation. Four ligands were used for stimulation: epidermal growth factor (EGF), heparin-binding-EGF like growth factor, transforming growth factor-α and epiregulin. Whereas only little differences in the order of individual ubiquitination sites were observed, the overall amount of modified receptor differed depending on the used ligand, indicating that absolute magnitude of EGFR ubiquitination, and not distinctly regulated ubiquitination sites, is a major determinant for signal attenuation and the subsequent cellular outcomes.
Collapse
Affiliation(s)
- Vyacheslav Akimov
- Center for Experimental BioInformatics, Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Mirjam Fehling-Kaschek
- Institut of Physics, University of Freiburg, Hermann-Herder-Str. 3, 79104 Freiburg, Germany
| | - Inigo Barrio-Hernandez
- Center for Experimental BioInformatics, Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Michele Puglia
- Center for Experimental BioInformatics, Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Jakob Bunkenborg
- Center for Experimental BioInformatics, Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Mogens M Nielsen
- Center for Experimental BioInformatics, Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Jens Timmer
- Institut of Physics, University of Freiburg, Hermann-Herder-Str. 3, 79104 Freiburg, Germany; Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestr. 18, 79104 Freiburg, Germany
| | - Jörn Dengjel
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland.
| | - Blagoy Blagoev
- Center for Experimental BioInformatics, Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark.
| |
Collapse
|
189
|
Arenas V, Castaño JL, Domínguez-García JJ, Yáñez L, Pipaón C. A Different View for an Old Disease: NEDDylation and Other Ubiquitin-Like Post-Translational Modifications in Chronic Lymphocytic Leukemia. Front Oncol 2021; 11:729550. [PMID: 34631557 PMCID: PMC8495217 DOI: 10.3389/fonc.2021.729550] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 09/07/2021] [Indexed: 12/03/2022] Open
Abstract
Despite the enormous amount of molecular data obtained over the years, the molecular etiology of chronic lymphocytic leukemia (CLL) is still largely unknown. All that information has enabled the development of new therapeutic approaches that have improved life expectancy of the patients but are still not curative. We must increase our knowledge of the molecular alterations responsible for the characteristics common to all CLL patients. One of such characteristics is the poor correlation between mRNA and protein expression, that suggests a role of post-translational mechanisms in CLL physiopathology. Drugs targeting these processes have indeed demonstrated an effect either alone or in combination with other aimed at specific pathways. A recent article unveiled an increment in ubiquitin-like modifications in CLL, with many protein members of relevant pathways affected. Interestingly, the inhibition of the NEDD8-activating protein NAE reverted a substantial number of those modifications. The present review gets the scarce data published about the role of NEDDylation in CLL together and establishes connections to what is known from other neoplasias, thus providing a new perspective to the underlying mechanisms in CLL.
Collapse
Affiliation(s)
- Víctor Arenas
- Laboratorio de Hematología Molecular, Servicio de Hematología, Hospital Universitario Marqués de Valdecilla-Instituto de Investigación Marqués de Valdecilla (IDIVAL), Santander, Spain
| | - Jose Luis Castaño
- Laboratorio de Hematología Molecular, Servicio de Hematología, Hospital Universitario Marqués de Valdecilla-Instituto de Investigación Marqués de Valdecilla (IDIVAL), Santander, Spain
| | - Juan José Domínguez-García
- Laboratorio de Hematología Molecular, Servicio de Hematología, Hospital Universitario Marqués de Valdecilla-Instituto de Investigación Marqués de Valdecilla (IDIVAL), Santander, Spain
| | - Lucrecia Yáñez
- Laboratorio de Hematología Molecular, Servicio de Hematología, Hospital Universitario Marqués de Valdecilla-Instituto de Investigación Marqués de Valdecilla (IDIVAL), Santander, Spain
| | - Carlos Pipaón
- Laboratorio de Hematología Molecular, Servicio de Hematología, Hospital Universitario Marqués de Valdecilla-Instituto de Investigación Marqués de Valdecilla (IDIVAL), Santander, Spain
| |
Collapse
|
190
|
Giordano I, Pirone L, Muratore V, Landaluze E, Pérez C, Lang V, Garde-Lapido E, Gonzalez-Lopez M, Barroso-Gomila O, Vertegaal ACO, Aransay AM, Rodriguez JA, Rodriguez MS, Sutherland JD, Barrio R. SALL1 Modulates CBX4 Stability, Nuclear Bodies, and Regulation of Target Genes. Front Cell Dev Biol 2021; 9:715868. [PMID: 34621739 PMCID: PMC8490708 DOI: 10.3389/fcell.2021.715868] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 08/30/2021] [Indexed: 11/16/2022] Open
Abstract
Development is orchestrated through a complex interplay of multiple transcription factors. The comprehension of this interplay will help us to understand developmental processes. Here we analyze the relationship between two key transcription factors: CBX4, a member of the Polycomb Repressive Complex 1 (PRC1), and SALL1, a member of the Spalt-like family with important roles in embryogenesis and limb development. Both proteins localize to nuclear bodies and are modified by the small ubiquitin-like modifier (SUMO). Our results show that CBX4 and SALL1 interact in the nucleoplasm and that increased SALL1 expression reduces ubiquitination of CBX4, enhancing its stability. This is accompanied by an increase in the number and size of CBX4-containing Polycomb bodies, and by a greater repression of CBX4 target genes. Thus, our findings uncover a new way of SALL1-mediated regulation of Polycomb bodies through modulation of CBX4 stability, with consequences in the regulation of its target genes, which could have an impact in cell differentiation and development.
Collapse
Affiliation(s)
- Immacolata Giordano
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance, Derio, Spain
| | - Lucia Pirone
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance, Derio, Spain
| | - Veronica Muratore
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance, Derio, Spain
| | - Eukene Landaluze
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance, Derio, Spain
| | - Coralia Pérez
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance, Derio, Spain
| | - Valerie Lang
- Viralgen Vector Core, Parque Científico y Tecnológico de Guipúzcoa, San Sebastián, Spain
| | - Elisa Garde-Lapido
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance, Derio, Spain
| | - Monika Gonzalez-Lopez
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance, Derio, Spain
| | - Orhi Barroso-Gomila
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance, Derio, Spain
| | - Alfred C O Vertegaal
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Ana M Aransay
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance, Derio, Spain.,Centro de Investigación Biomédica en Red. Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Jose Antonio Rodriguez
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country, Leioa, Spain
| | - Manuel S Rodriguez
- Laboratoire de Chimie de Coordination-CNRS, Paul Sabatier: Université Toulouse III, Toulouse, France
| | - James D Sutherland
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance, Derio, Spain
| | - Rosa Barrio
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance, Derio, Spain
| |
Collapse
|
191
|
Marsden AJ, Riley DRJ, Barry A, Khalil JS, Guinn BA, Kemp NT, Rivero F, Beltran-Alvarez P. Inhibition of Arginine Methylation Impairs Platelet Function. ACS Pharmacol Transl Sci 2021; 4:1567-1577. [PMID: 34661075 DOI: 10.1021/acsptsci.1c00135] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Indexed: 11/28/2022]
Abstract
Protein arginine methyltransferases (PRMTs) catalyze the transfer of methyl groups to arginine residues in proteins. PRMT inhibitors are novel, promising drugs against cancer that are currently in clinical trials, which include oral administration of the drugs. However, off-target activities of systemically available PRMT inhibitors have not yet been investigated. In this work, we study the relevance of arginine methylation in platelets and investigate the effect of PRMT inhibitors on platelet function and on the expression of relevant platelet receptors. We show that (1) key platelet proteins are modified by arginine methylation; (2) incubation of human platelets with PRMT inhibitors for 4 h results in impaired capacity of platelets to aggregate in response to thrombin and collagen, with IC50 values in the μM range; and (3) treatment with PRMT inhibitors leads to decreased membrane expression and reduced activation of the critical platelet integrin αIIbβ3. Our contribution opens new avenues for research on arginine methylation in platelets, including the repurposing of arginine methylation inhibitors as novel antiplatelet drugs. We also recommend that current and future clinical trials with PRMT inhibitors consider any adverse effects associated with platelet inhibition of these emerging anticancer drugs.
Collapse
Affiliation(s)
| | - David R J Riley
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, Hull HU6 7RX, U.K
| | - Antonia Barry
- Department of Biomedical Sciences, University of Hull, Hull HU6 7RX, U.K
| | - Jawad S Khalil
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, Hull HU6 7RX, U.K
| | - Barbara-Ann Guinn
- Department of Biomedical Sciences, University of Hull, Hull HU6 7RX, U.K
| | - Neil T Kemp
- Department of Physics and Mathematics, University of Hull, Hull HU6 7RX, U.K
| | - Francisco Rivero
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, Hull HU6 7RX, U.K
| | | |
Collapse
|
192
|
Rivera KD, Olive ME, Bergstrom EJ, Nelson AJ, Lee KA, Satpathy S, Carr SA, Udeshi ND. Automating UbiFast for High-throughput and Multiplexed Ubiquitin Enrichment. Mol Cell Proteomics 2021; 20:100154. [PMID: 34592423 PMCID: PMC9357436 DOI: 10.1016/j.mcpro.2021.100154] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 08/23/2021] [Accepted: 09/22/2021] [Indexed: 01/11/2023] Open
Abstract
Robust methods for deep-scale enrichment and site-specific identification of ubiquitylation sites are necessary for characterizing the myriad roles of protein ubiquitylation. To this end we previously developed UbiFast, a sensitive method for highly multiplexed ubiquitylation profiling where K-ϵ-GG peptides are enriched with anti-K-ε-GG antibody and labeled on-antibody with isobaric labeling reagents for sample multiplexing. Here, we present robotic automation of the UbiFast method using a magnetic bead-conjugated K-ε-GG antibody (mK-ε-GG) and a magnetic particle processor. We report the identification of ∼20,000 ubiquitylation sites from a TMT10-plex with 500 μg input per sample processed in ∼2 h. Automation of the UbiFast method greatly increased the number of identified and quantified ubiquitylation sites, improved reproducibility, and significantly reduced processing time. The automated method also significantly reduced variability across process replicates compared with the manual method. The workflow enables processing of up to 96 samples in a single day making it suitable to study ubiquitylation in large sample sets. Here we demonstrate the applicability of the method to profile small amounts of tissue using breast cancer patient–derived xenograft (PDX) tissue samples. HS mag anti-K-ε-GG antibody increases sensitivity of ubiquitylation site detection. Automated UbiFast increases reproducibility and sample processing throughput. The automated UbiFast workflow enables processing of up to 96 samples in one day. UbiFast can be employed to profile ubiquitylomes from small amounts of tumor tissue.
Collapse
Affiliation(s)
- Keith D Rivera
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Meagan E Olive
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Erik J Bergstrom
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | | | | | - Shankha Satpathy
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Steven A Carr
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.
| | - Namrata D Udeshi
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.
| |
Collapse
|
193
|
Schweiggert J, Habeck G, Hess S, Mikus F, Beloshistov R, Meese K, Hata S, Knobeloch K, Melchior F. SCF Fbxw5 targets kinesin-13 proteins to facilitate ciliogenesis. EMBO J 2021; 40:e107735. [PMID: 34368969 PMCID: PMC8441365 DOI: 10.15252/embj.2021107735] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 06/29/2021] [Accepted: 07/12/2021] [Indexed: 11/23/2022] Open
Abstract
Microtubule depolymerases of the kinesin-13 family play important roles in various cellular processes and are frequently overexpressed in different cancer types. Despite the importance of their correct abundance, remarkably little is known about how their levels are regulated in cells. Using comprehensive screening on protein microarrays, we identified 161 candidate substrates of the multi-subunit ubiquitin E3 ligase SCFFbxw5 , including the kinesin-13 member Kif2c/MCAK. In vitro reconstitution assays demonstrate that MCAK and its closely related orthologs Kif2a and Kif2b become efficiently polyubiquitylated by neddylated SCFFbxw5 and Cdc34, without requiring preceding modifications. In cells, SCFFbxw5 targets MCAK for proteasomal degradation predominantly during G2 . While this seems largely dispensable for mitotic progression, loss of Fbxw5 leads to increased MCAK levels at basal bodies and impairs ciliogenesis in the following G1 /G0 , which can be rescued by concomitant knockdown of MCAK, Kif2a or Kif2b. We thus propose a novel regulatory event of ciliogenesis that begins already within the G2 phase of the preceding cell cycle.
Collapse
Affiliation(s)
- Jörg Schweiggert
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH)University of HeidelbergDKFZ ‐ ZMBH AllianceHeidelbergGermany
| | - Gregor Habeck
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH)University of HeidelbergDKFZ ‐ ZMBH AllianceHeidelbergGermany
| | - Sandra Hess
- Institute of NeuropathologyFaculty of MedicineUniversity of FreiburgFreiburgGermany
- Faculty of BiologyUniversity of FreiburgFreiburgGermany
| | - Felix Mikus
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH)University of HeidelbergDKFZ ‐ ZMBH AllianceHeidelbergGermany
| | - Roman Beloshistov
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH)University of HeidelbergDKFZ ‐ ZMBH AllianceHeidelbergGermany
| | - Klaus Meese
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH)University of HeidelbergDKFZ ‐ ZMBH AllianceHeidelbergGermany
| | - Shoji Hata
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH)University of HeidelbergDKFZ ‐ ZMBH AllianceHeidelbergGermany
| | | | - Frauke Melchior
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH)University of HeidelbergDKFZ ‐ ZMBH AllianceHeidelbergGermany
| |
Collapse
|
194
|
Time-resolved in vivo ubiquitinome profiling by DIA-MS reveals USP7 targets on a proteome-wide scale. Nat Commun 2021; 12:5399. [PMID: 34518535 PMCID: PMC8438043 DOI: 10.1038/s41467-021-25454-1] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 08/11/2021] [Indexed: 11/08/2022] Open
Abstract
Mass spectrometry (MS)-based ubiquitinomics provides system-level understanding of ubiquitin signaling. Here we present a scalable workflow for deep and precise in vivo ubiquitinome profiling, coupling an improved sample preparation protocol with data-independent acquisition (DIA)-MS and neural network-based data processing specifically optimized for ubiquitinomics. Compared to data-dependent acquisition (DDA), our method more than triples identification numbers to 70,000 ubiquitinated peptides in single MS runs, while significantly improving robustness and quantification precision. Upon inhibition of the oncology target USP7, we simultaneously record ubiquitination and consequent changes in abundance of more than 8,000 proteins at high temporal resolution. While ubiquitination of hundreds of proteins increases within minutes of USP7 inhibition, we find that only a small fraction of those are ever degraded, thereby dissecting the scope of USP7 action. Our method enables rapid mode-of-action profiling of candidate drugs targeting DUBs or ubiquitin ligases at high precision and throughput. Combining improved sample preparation, data-independent acquisition mass spectrometry and deep learning, the authors develop a workflow for more robust and precise quantitative ubiquitinome profiling. They use this method to characterize targets of the deubiquitinase USP7 and effects of USP7 inhibitors.
Collapse
|
195
|
Yan Y, Gu T, Christensen SDK, Su J, Lassen TR, Hjortbak MV, Lo IJ, Venø ST, Tóth AE, Song P, Nielsen MS, Bøtker HE, Blagoev B, Drasbek KR, Kjems J. Cyclic Hypoxia Conditioning Alters the Content of Myoblast-Derived Extracellular Vesicles and Enhances Their Cell-Protective Functions. Biomedicines 2021; 9:biomedicines9091211. [PMID: 34572398 PMCID: PMC8471008 DOI: 10.3390/biomedicines9091211] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/06/2021] [Accepted: 09/08/2021] [Indexed: 11/16/2022] Open
Abstract
Remote ischemic conditioning (RIC) is a procedure that can attenuate ischemic-reperfusion injury by conducting brief cycles of ischemia and reperfusion in the arm or leg. Extracellular vesicles (EVs) circulating in the bloodstream can release their content into recipient cells to confer protective function on ischemia-reperfusion injured (IRI) organs. Skeletal muscle cells are potential candidates to release EVs as a protective signal during RIC. In this study, we used C2C12 cells as a model system and performed cyclic hypoxia-reoxygenation (HR) to mimic RIC. EVs were collected and subjected to small RNA profiling and proteomics. HR induced a distinct shift in the miRNA profile and protein content in EVs. HR EV treatment restored cell viability, dampened inflammation, and enhanced tube formation in in vitro assays. In vivo, HR EVs showed increased accumulation in the ischemic brain compared to EVs secreted from normoxic culture (N EVs) in a mouse undergoing transient middle cerebral artery occlusion (tMCAO). We conclude that HR conditioning changes the miRNA and protein profile in EVs released by C2C12 cells and enhances the protective signal in the EVs to recipient cells in vitro.
Collapse
Affiliation(s)
- Yan Yan
- Interdisciplinary Nanoscience Center, Aarhus University, 8000 Aarhus, Denmark; (Y.Y.); (J.S.); (I.L.); (P.S.)
- Omiics ApS, 8200 Aarhus, Denmark;
| | - Tingting Gu
- Center of Functionally Integrative Neuroscience, Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark; (T.G.); (K.R.D.)
| | - Stine Duelund Kaas Christensen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark; (S.D.K.C.); (B.B.)
| | - Junyi Su
- Interdisciplinary Nanoscience Center, Aarhus University, 8000 Aarhus, Denmark; (Y.Y.); (J.S.); (I.L.); (P.S.)
| | - Thomas Ravn Lassen
- Department of Cardiology, Aarhus University Hospital, Skejby, 8200 Aarhus, Denmark; (T.R.L.); (M.V.H.); (H.E.B.)
| | - Marie Vognstoft Hjortbak
- Department of Cardiology, Aarhus University Hospital, Skejby, 8200 Aarhus, Denmark; (T.R.L.); (M.V.H.); (H.E.B.)
| | - IJu Lo
- Interdisciplinary Nanoscience Center, Aarhus University, 8000 Aarhus, Denmark; (Y.Y.); (J.S.); (I.L.); (P.S.)
| | | | - Andrea Erzsebet Tóth
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark; (A.E.T.); (M.S.N.)
| | - Ping Song
- Interdisciplinary Nanoscience Center, Aarhus University, 8000 Aarhus, Denmark; (Y.Y.); (J.S.); (I.L.); (P.S.)
| | | | - Hans Erik Bøtker
- Department of Cardiology, Aarhus University Hospital, Skejby, 8200 Aarhus, Denmark; (T.R.L.); (M.V.H.); (H.E.B.)
| | - Blagoy Blagoev
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark; (S.D.K.C.); (B.B.)
| | - Kim Ryun Drasbek
- Center of Functionally Integrative Neuroscience, Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark; (T.G.); (K.R.D.)
| | - Jørgen Kjems
- Interdisciplinary Nanoscience Center, Aarhus University, 8000 Aarhus, Denmark; (Y.Y.); (J.S.); (I.L.); (P.S.)
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark
- Correspondence: ; Tel.: +45-289-920-86
| |
Collapse
|
196
|
Ramesh ST, Navyasree KV, Sah S, Ashok AB, Qathoon N, Mohanty S, Swain RK, Umasankar PK. BMP2K phosphorylates AP-2 and regulates clathrin-mediated endocytosis. Traffic 2021; 22:377-396. [PMID: 34480404 DOI: 10.1111/tra.12814] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/31/2021] [Accepted: 09/01/2021] [Indexed: 01/29/2023]
Abstract
Phosphorylation of the central adaptor protein complex, AP-2 is pivotal for clathrin-mediated endocytosis (CME). Here, we uncover the role of an uncharacterized kinase (BMP-2 inducible kinase-BMP2K) in AP-2 phosphorylation. We demonstrate that BMP2K can phosphorylate AP-2 in vitro and in vivo. Functional impairment of BMP2K impedes AP-2 phosphorylation leading to defects in clathrin-coated pit (CCP) morphology and cargo internalization. BMP2K engages AP-2 via its extended C-terminus and this interaction is important for its CCP localization and function. Notably, endogenous BMP2K levels decline upon functional impairment of AP-2 indicating AP-2 dependent BMP2K stabilization in cells. Further, functional inactivation of BMP2K in zebrafish embryos yields gastrulation phenotypes which mirror AP-2 loss-of-function suggesting physiological relevance of BMP2K in vertebrates. Together, our findings propose involvement of a novel kinase in AP-2 phosphorylation and in the operation of CME.
Collapse
Affiliation(s)
- Shikha T Ramesh
- Intracellular Trafficking Laboratory, Transdisciplinary Biology Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India.,Centre for Doctoral Studies, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Kolaparamba V Navyasree
- Intracellular Trafficking Laboratory, Transdisciplinary Biology Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India.,Centre for Doctoral Studies, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Sneha Sah
- Intracellular Trafficking Laboratory, Transdisciplinary Biology Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Anjitha B Ashok
- Intracellular Trafficking Laboratory, Transdisciplinary Biology Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Nishada Qathoon
- Intracellular Trafficking Laboratory, Transdisciplinary Biology Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | | | | | - Perunthottathu K Umasankar
- Intracellular Trafficking Laboratory, Transdisciplinary Biology Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| |
Collapse
|
197
|
Snyder NA, Silva GM. Deubiquitinating enzymes (DUBs): Regulation, homeostasis, and oxidative stress response. J Biol Chem 2021; 297:101077. [PMID: 34391779 PMCID: PMC8424594 DOI: 10.1016/j.jbc.2021.101077] [Citation(s) in RCA: 110] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 08/10/2021] [Accepted: 08/11/2021] [Indexed: 12/17/2022] Open
Abstract
Ubiquitin signaling is a conserved, widespread, and dynamic process in which protein substrates are rapidly modified by ubiquitin to impact protein activity, localization, or stability. To regulate this process, deubiquitinating enzymes (DUBs) counter the signal induced by ubiquitin conjugases and ligases by removing ubiquitin from these substrates. Many DUBs selectively regulate physiological pathways employing conserved mechanisms of ubiquitin bond cleavage. DUB activity is highly regulated in dynamic environments through protein-protein interaction, posttranslational modification, and relocalization. The largest family of DUBs, cysteine proteases, are also sensitive to regulation by oxidative stress, as reactive oxygen species (ROS) directly modify the catalytic cysteine required for their enzymatic activity. Current research has implicated DUB activity in human diseases, including various cancers and neurodegenerative disorders. Due to their selectivity and functional roles, DUBs have become important targets for therapeutic development to treat these conditions. This review will discuss the main classes of DUBs and their regulatory mechanisms with a particular focus on DUB redox regulation and its physiological impact during oxidative stress.
Collapse
Affiliation(s)
- Nathan A Snyder
- Department of Biology, Duke University, Durham, North Carolina, USA
| | - Gustavo M Silva
- Department of Biology, Duke University, Durham, North Carolina, USA.
| |
Collapse
|
198
|
Park HA, Stumpf A, Broman K, Jansen J, Dunn T, Scott M, Crowe-White KM. Role of lycopene in mitochondrial protection during differential levels of oxidative stress in primary cortical neurons. BRAIN DISORDERS 2021. [DOI: 10.1016/j.dscb.2021.100016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
|
199
|
Roy M, Singh R. TRIMs: selective recruitment at different steps of the NF-κB pathway-determinant of activation or resolution of inflammation. Cell Mol Life Sci 2021; 78:6069-6086. [PMID: 34283248 PMCID: PMC11072854 DOI: 10.1007/s00018-021-03900-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 07/04/2021] [Accepted: 07/13/2021] [Indexed: 12/25/2022]
Abstract
TNF-α-induced NF-κB pathway is an essential component of innate and adaptive immune pathway, and it is tightly regulated by various post-translational modifications including ubiquitination. Oscillations in NF-κB activation and temporal gene expression are emerging as critical determinants of inflammatory response, however, the regulators of unique outcomes in different patho-physiological conditions are not well understood. Tripartite Motif-containing proteins (TRIMs) are RING domain-containing E3 ligases involved in the regulation of cellular homeostasis, metabolism, cell death, inflammation, and host defence. Emerging reports suggest that TRIMs are recruited at different steps of TNF-α-induced NF-κB pathway and modulate via their E3 ligase activity. TRIMs show synergy and antagonism in the regulation of the NF-κB pathway and also regulate it in a feedback manner. TRIMs also regulate pattern recognition receptors (PRRs) mediated inflammatory pathways and may have evolved to directly regulate a specific arm of immune signalling. The review emphasizes TRIM-mediated ubiquitination and modulation of TNF-α-regulated temporal and NF-κB signaling and its possible impact on unique transcriptional and functional outcomes.
Collapse
Affiliation(s)
- Milton Roy
- Department of Biochemistry, Faculty of Science, The MS University of Baroda, Vadodara, Gujarat, 390002, India
- Institute for Cell Engineering, The Johns Hopkins University School of Medicine, 733 North Broadway, MRB 731, Baltimore, MD, 21205, USA
| | - Rajesh Singh
- Department of Biochemistry, Faculty of Science, The MS University of Baroda, Vadodara, Gujarat, 390002, India.
| |
Collapse
|
200
|
Thery F, Eggermont D, Impens F. Proteomics Mapping of the ISGylation Landscape in Innate Immunity. Front Immunol 2021; 12:720765. [PMID: 34447387 PMCID: PMC8383068 DOI: 10.3389/fimmu.2021.720765] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 07/16/2021] [Indexed: 01/12/2023] Open
Abstract
During infection, pathogen sensing and cytokine signaling by the host induce expression of antimicrobial proteins and specialized post-translational modifications. One such protein is ISG15, a ubiquitin-like protein (UBL) conserved among vertebrates. Similar to ubiquitin, ISG15 covalently conjugates to lysine residues in substrate proteins in a process called ISGylation. Mice deficient for ISGylation or lacking ISG15 are strongly susceptible to many viral pathogens and several intracellular bacterial pathogens. Although ISG15 was the first UBL discovered after ubiquitin, the mechanisms behind its protective activity are poorly understood. Largely, this stems from a lack of knowledge on the ISG15 substrate repertoire. To unravel the antiviral activity of ISG15, early studies used mass spectrometry-based proteomics in combination with ISG15 pulldown. Despite reporting hundreds of ISG15 substrates, these studies were unable to identify the exact sites of modification, impeding a clear understanding of the molecular consequences of protein ISGylation. More recently, a peptide-based enrichment approach revolutionized the study of ubiquitin allowing untargeted discovery of ubiquitin substrates, including knowledge of their exact modification sites. Shared molecular determinants between ISG15 and ubiquitin allowed to take advantage of this technology for proteome-wide mapping of ISG15 substrates and modification sites. In this review, we provide a comprehensive overview of mass spectrometry-based proteomics studies on protein ISGylation. We critically discuss the relevant literature, compare reported substrates and sites and make suggestions for future research.
Collapse
Affiliation(s)
- Fabien Thery
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium.,Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Denzel Eggermont
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium.,Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Francis Impens
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium.,Department of Biomolecular Medicine, Ghent University, Ghent, Belgium.,VIB Proteomics Core, VIB, Ghent, Belgium
| |
Collapse
|