51
|
Xu K, Chu Y, Liu Q, Fan W, He H, Huang F. NEDD4 E3 Ligases: Functions and Mechanisms in Bone and Tooth. Int J Mol Sci 2022; 23:ijms23179937. [PMID: 36077334 PMCID: PMC9455957 DOI: 10.3390/ijms23179937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/26/2022] [Accepted: 08/29/2022] [Indexed: 11/29/2022] Open
Abstract
Protein ubiquitination is a precisely controlled enzymatic cascade reaction belonging to the post-translational modification of proteins. In this process, E3 ligases catalyze the binding of ubiquitin (Ub) to protein substrates and define specificity. The neuronally expressed developmentally down-regulated 4 (NEDD4) subfamily, belonging to the homology to E6APC terminus (HECT) class of E3 ligases, has recently emerged as an essential determinant of multiple cellular processes in different tissues, including bone and tooth. Here, we place special emphasis on the regulatory role of the NEDD4 subfamily in the molecular and cell biology of osteogenesis. We elucidate in detail the specific roles, downstream substrates, and upstream regulatory mechanisms of the NEDD4 subfamily. Further, we provide an overview of the involvement of E3 ligases and deubiquitinases in the development, repair, and regeneration of another mineralized tissue—tooth.
Collapse
Affiliation(s)
- Ke Xu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510008, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510008, China
| | - Yanhao Chu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510008, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510008, China
| | - Qin Liu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510008, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510008, China
| | - Wenguo Fan
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510008, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510008, China
| | - Hongwen He
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510008, China
- Correspondence: (H.H.); (F.H.)
| | - Fang Huang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510008, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510008, China
- Correspondence: (H.H.); (F.H.)
| |
Collapse
|
52
|
Zou J, Zhou X, Chen X, Ma Y, Yu R. Shenkang Injection for Treating Renal Fibrosis-Metabonomics and Regulation of E3 Ubiquitin Ligase Smurfs on TGF-β/Smads Signal Transduction. Front Pharmacol 2022; 13:849832. [PMID: 35721120 PMCID: PMC9201572 DOI: 10.3389/fphar.2022.849832] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 04/25/2022] [Indexed: 12/31/2022] Open
Abstract
At present, TGF-β is the most critical fibrogenic factor known. Smad ubiquitin ligase Smurfs play an important role in the regulation of the TGF-/Smads signaling pathway, which is linked to metabolite changes in renal fibrosis. Previous studies have shown that Shenkang injection can prevent and treat chronic kidney disease through multiple channels of action. However, the precise relationship between Shenkang injection and the regulation of the TGF-/Smads signaling pathway in the treatment of chronic kidney disease is unknown. Here, we evaluated the pharmacological effects of Shenkang injection on ubiquitination and metabolic changes of the TGF-β/Smads signaling pathway in UUO mice using pathology-related indicators, immunoprecipitation, subcellular co-location, and metabonomics analysis. Our findings indicate that Shenkang injection can promote nuclear translocation of Smurf1 and Smurf2 to TGF- membrane receptors TR-I and Smad2 and ubiquitinated degradation of these proteins. Furthermore, the formation of TβR-I/TβR-II, TβR-I/Smad2, and TβR-I/Smad3 complexes was inhibited to negatively regulate the TGF-β/Smad signaling pathway induced renal tubular epithelial transdifferentiation (EMT). The EMT process is not very relevant in vivo, although it is clear that TGF-β induces EMT in cultured cells, which has been demonstrated by numerous teams around the world. However, this is not the case with the in vivo models of kidney fibrosis, especially UUO. In addition, Shenkang injection can improve amino acid metabolism, purine metabolism, and fatty acid metabolism disorders.
Collapse
Affiliation(s)
- Junju Zou
- Hunan Provincial Key Laboratory of Translational Research in TCM Prescriptions and Zheng, Hunan University of Chinese Medicine, Changsha, China
| | - Xiaotao Zhou
- School of Basic Medicine, Chengdu University of Chinese Medicine, Chengdu, China
| | - Xian Chen
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuerong Ma
- School of Basic Medicine, Chengdu University of Chinese Medicine, Chengdu, China
| | - Rong Yu
- Hunan Provincial Key Laboratory of Translational Research in TCM Prescriptions and Zheng, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
53
|
Vicidomini R, Serpe M. Local BMP signaling: A sensor for synaptic activity that balances synapse growth and function. Curr Top Dev Biol 2022; 150:211-254. [PMID: 35817503 PMCID: PMC11102767 DOI: 10.1016/bs.ctdb.2022.04.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Synapse development is coordinated by intercellular communication between the pre- and postsynaptic compartments, and by neuronal activity itself. In flies as in vertebrates, neuronal activity induces input-specific changes in the synaptic strength so that the entire circuit maintains stable function in the face of many challenges, including changes in synapse number and strength. But how do neurons sense synapse activity? In several studies carried out using the Drosophila neuromuscular junction (NMJ), we demonstrated that local BMP signaling provides an exquisite sensor for synapse activity. Here we review the main features of this exquisite sensor and discuss its functioning beyond monitoring the synapse activity but rather as a key controller that operates in coordination with other BMP signaling pathways to balance synapse growth, maturation and function.
Collapse
Affiliation(s)
- Rosario Vicidomini
- Neurosciences and Cellular and Structural Biology Division, Eunice Kennedy Shiver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Mihaela Serpe
- Neurosciences and Cellular and Structural Biology Division, Eunice Kennedy Shiver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States.
| |
Collapse
|
54
|
Haouari S, Vourc’h P, Jeanne M, Marouillat S, Veyrat-Durebex C, Lanznaster D, Laumonnier F, Corcia P, Blasco H, Andres CR. The Roles of NEDD4 Subfamily of HECT E3 Ubiquitin Ligases in Neurodevelopment and Neurodegeneration. Int J Mol Sci 2022; 23:ijms23073882. [PMID: 35409239 PMCID: PMC8999422 DOI: 10.3390/ijms23073882] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 02/06/2023] Open
Abstract
The ubiquitin pathway regulates the function of many proteins and controls cellular protein homeostasis. In recent years, it has attracted great interest in neurodevelopmental and neurodegenerative diseases. Here, we have presented the first review on the roles of the 9 proteins of the HECT E3 ligase NEDD4 subfamily in the development and function of neurons in the central nervous system (CNS). We discussed their regulation and their direct or indirect involvement in neurodevelopmental diseases, such as intellectual disability, and neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease or Amyotrophic Lateral Sclerosis. Further studies on the roles of these proteins, their regulation and their targets in neurons will certainly contribute to a better understanding of neuronal function and dysfunction, and will also provide interesting information for the development of therapeutics targeting them.
Collapse
Affiliation(s)
- Shanez Haouari
- UMR 1253, iBrain, Université de Tours, Inserm, 37044 Tours, France; (S.H.); (M.J.); (S.M.); (C.V.-D.); (D.L.); (F.L.); (P.C.); (H.B.); (C.R.A.)
| | - Patrick Vourc’h
- UMR 1253, iBrain, Université de Tours, Inserm, 37044 Tours, France; (S.H.); (M.J.); (S.M.); (C.V.-D.); (D.L.); (F.L.); (P.C.); (H.B.); (C.R.A.)
- CHRU de Tours, Service de Biochimie et Biologie Moléculaire, 37044 Tours, France
- Correspondence: ; Tel.: +33-(0)2-34-37-89-10; Fax: +33-(0)2-47-36-61-85
| | - Médéric Jeanne
- UMR 1253, iBrain, Université de Tours, Inserm, 37044 Tours, France; (S.H.); (M.J.); (S.M.); (C.V.-D.); (D.L.); (F.L.); (P.C.); (H.B.); (C.R.A.)
- CHRU de Tours, Service de Génétique, 37044 Tours, France
| | - Sylviane Marouillat
- UMR 1253, iBrain, Université de Tours, Inserm, 37044 Tours, France; (S.H.); (M.J.); (S.M.); (C.V.-D.); (D.L.); (F.L.); (P.C.); (H.B.); (C.R.A.)
| | - Charlotte Veyrat-Durebex
- UMR 1253, iBrain, Université de Tours, Inserm, 37044 Tours, France; (S.H.); (M.J.); (S.M.); (C.V.-D.); (D.L.); (F.L.); (P.C.); (H.B.); (C.R.A.)
- CHRU de Tours, Service de Biochimie et Biologie Moléculaire, 37044 Tours, France
| | - Débora Lanznaster
- UMR 1253, iBrain, Université de Tours, Inserm, 37044 Tours, France; (S.H.); (M.J.); (S.M.); (C.V.-D.); (D.L.); (F.L.); (P.C.); (H.B.); (C.R.A.)
| | - Frédéric Laumonnier
- UMR 1253, iBrain, Université de Tours, Inserm, 37044 Tours, France; (S.H.); (M.J.); (S.M.); (C.V.-D.); (D.L.); (F.L.); (P.C.); (H.B.); (C.R.A.)
| | - Philippe Corcia
- UMR 1253, iBrain, Université de Tours, Inserm, 37044 Tours, France; (S.H.); (M.J.); (S.M.); (C.V.-D.); (D.L.); (F.L.); (P.C.); (H.B.); (C.R.A.)
- CHRU de Tours, Service de Neurologie, 37044 Tours, France
| | - Hélène Blasco
- UMR 1253, iBrain, Université de Tours, Inserm, 37044 Tours, France; (S.H.); (M.J.); (S.M.); (C.V.-D.); (D.L.); (F.L.); (P.C.); (H.B.); (C.R.A.)
- CHRU de Tours, Service de Biochimie et Biologie Moléculaire, 37044 Tours, France
| | - Christian R. Andres
- UMR 1253, iBrain, Université de Tours, Inserm, 37044 Tours, France; (S.H.); (M.J.); (S.M.); (C.V.-D.); (D.L.); (F.L.); (P.C.); (H.B.); (C.R.A.)
- CHRU de Tours, Service de Biochimie et Biologie Moléculaire, 37044 Tours, France
| |
Collapse
|
55
|
Kirkpatrick BW, Cooke ME, Frie M, Sporer KRB, Lett B, Wells SJ, Coussens PM. Genome-wide association analysis for susceptibility to infection by Mycobacterium avium ssp. paratuberculosis in US Holsteins. J Dairy Sci 2022; 105:4301-4313. [PMID: 35307176 DOI: 10.3168/jds.2021-21276] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 01/27/2022] [Indexed: 12/19/2022]
Abstract
Paratuberculosis, or Johne's disease, is a chronic, granulomatous, gastrointestinal tract disease of cattle and other ruminants caused by the bacterium Mycobacterium avium subspecies paratuberculosis (MAP). Control of Johne's disease is based on programs of testing and culling animals positive for infection with MAP and concurrently modifying management to reduce the likelihood of infection. The current study was motivated by the hypothesis that genetic variation in host susceptibility to MAP infection can be dissected and quantifiable associations with genetic markers identified. Two separate GWAS analyses were conducted, the first using 897 genotyped Holstein artificial insemination sires with phenotypes derived from incidence of MAP infection among daughters based on milk ELISA testing records. The second GWAS analysis was a case-control design using US Holstein cows phenotyped for MAP infection by serum ELISA or fecal culture tests. Cases included cows positive for either serum ELISA, fecal culture, or both. Controls consisted of animals negative for all tests conducted. A total of 376 samples (70 cases and 306 controls) from a University of Minnesota Johne's management demonstration project and 184 samples (76 cases and 108 controls) from a Michigan State University study were used. Medium-density (sires) and high-density (cows) genotype data were imputed to full genome sequence for the analyses. Marker-trait associations were analyzed using the single-step (ss)GWAS procedure implemented in the BLUPF90 suite of programs. Evidence of significant genomic contributions for susceptibility to MAP infection were observed on multiple chromosomes. Results were combined across studies in a meta-analysis, and increased support for genomic regions on BTA7 and BTA21 were observed. Gene set enrichment analysis suggested pathways for antigen processing and presentation, antimicrobial peptides and natural killer cell-mediated cytotoxicity are relevant to variation in host susceptibility to MAP infection, among others. Genomic prediction was evaluated using a 5-fold cross-validation, and moderate correlations were observed between genomic breeding value predictions and daughter averages (∼0.43 to 0.53) for MAP infection in testing data sets. These results suggest that genomic selection against susceptibility to MAP infection is feasible in Holstein cattle.
Collapse
Affiliation(s)
- B W Kirkpatrick
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, 1675 Observatory Drive, Madison 53706.
| | - M E Cooke
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, 1675 Observatory Drive, Madison 53706
| | - M Frie
- Department of Animal Science, Michigan State University, 474 S Shaw Ln, East Lansing 48824
| | - K R B Sporer
- Department of Animal Science, Michigan State University, 474 S Shaw Ln, East Lansing 48824
| | - B Lett
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, 1675 Observatory Drive, Madison 53706
| | - S J Wells
- Department of Veterinary Population Medicine, University of Minnesota, 1365 Gortner Avenue, St. Paul 55108
| | - P M Coussens
- Department of Animal Science, Michigan State University, 474 S Shaw Ln, East Lansing 48824
| |
Collapse
|
56
|
Yan Y, Wang Q. BMP Signaling: Lighting up the Way for Embryonic Dorsoventral Patterning. Front Cell Dev Biol 2022; 9:799772. [PMID: 35036406 PMCID: PMC8753366 DOI: 10.3389/fcell.2021.799772] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/06/2021] [Indexed: 11/13/2022] Open
Abstract
One of the most significant events during early embryonic development is the establishment of a basic embryonic body plan, which is defined by anteroposterior, dorsoventral (DV), and left-right axes. It is well-known that the morphogen gradient created by BMP signaling activity is crucial for DV axis patterning across a diverse set of vertebrates. The regulation of BMP signaling during DV patterning has been strongly conserved across evolution. This is a remarkable regulatory and evolutionary feat, as the BMP gradient has been maintained despite the tremendous variation in embryonic size and shape across species. Interestingly, the embryonic DV axis exhibits robust stability, even in face of variations in BMP signaling. Multiple lines of genetic, molecular, and embryological evidence have suggested that numerous BMP signaling components and their attendant regulators act in concert to shape the developing DV axis. In this review, we summarize the current knowledge of the function and regulation of BMP signaling in DV patterning. Throughout, we focus specifically on popular model animals, such as Xenopus and zebrafish, highlighting the similarities and differences of the regulatory networks between species. We also review recent advances regarding the molecular nature of DV patterning, including the initiation of the DV axis, the formation of the BMP gradient, and the regulatory molecular mechanisms behind BMP signaling during the establishment of the DV axis. Collectively, this review will help clarify our current understanding of the molecular nature of DV axis formation.
Collapse
Affiliation(s)
- Yifang Yan
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.,National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China.,Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China.,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Qiang Wang
- State Key Laboratory of Membrane Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
57
|
Dong L, Liu L, Li Y, Li W, Zhou L, Xia Q. E3 ligase Smurf1 protects against misfolded SOD1 in neuronal cells by promoting its K63 ubiquitylation and aggresome formation. Hum Mol Genet 2022; 31:2035-2048. [PMID: 35022748 DOI: 10.1093/hmg/ddac008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 12/15/2021] [Accepted: 01/07/2022] [Indexed: 11/15/2022] Open
Abstract
K63-linked polyubiquitination of the neurodegenerative disease-associated misfolded protein copper-zinc superoxide dismutase (SOD1) is associated with the formation of inclusion bodies. Highly expressed E3 ligase Smurf1 promotes cellular homostasis through the enhanced capability of aggregate degradation. However, it is not well explored the role of Smurf1 in the dynamics of SOD1 aggresomes. In this study, we report that Smurf1 promotes the recruitment of SOD1 to form aggresomes. Mechanistically, Smurf1 interacts with mutant SOD1 to promote aggresome formation by modification of its K63-linked polyubiquitination. Moreover, overexpressed Smurf1 enhances mutant SOD1 aggresome formation and autophagic degradation to prevent cell death. Thus, our data suggest that Smurf1 plays an important role in attenuating protein misfolding-induced cell toxicity by both driving the sequestration of misfolded SOD1 into aggresomes and autophagic degradation.
Collapse
Affiliation(s)
- Lei Dong
- School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Liqun Liu
- School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Yang Li
- School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Wenxuan Li
- School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Liying Zhou
- Beijing Tide Pharmaceutical CO., LTD, Beijing, 100000, China
| | - Qin Xia
- School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| |
Collapse
|
58
|
Han D, Li S, Xia Q, Meng X, Dong L. Overexpressed Smurf1 is degraded in glioblastoma cells through autophagy in a p62-dependent manner. FEBS Open Bio 2022; 12:118-129. [PMID: 34614303 PMCID: PMC8727935 DOI: 10.1002/2211-5463.13310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 10/05/2021] [Accepted: 10/05/2021] [Indexed: 11/07/2022] Open
Abstract
Homologous to E6AP C-terminus (HECT)-type E3 ubiquitin ligase SMAD-specific E3 ubiquitin protein ligase 1 (Smurf1) was originally identified to ubiquitinate Smad protein in the TGF-β/BMP signaling pathway. Recently, Smurf1 has been reported to promote tumorigenesis by regulating multiple biological processes. High expression of Smurf1 plays a vital role in brain tumor progression by mediating aberrant cell signaling pathways. Previous reports have shown that Smurf1 is degraded mainly through the ubiquitin-proteasome system, but it remains unclear whether Smurf1 is degraded by autophagy in tumor cells. In this study, we show that autophagy activators promote Smurf1 degradation in glioblastoma (GB) cells. The autophagy receptor p62 colocalizes with ubiquitinated substrates to promote sequestration of cytoplasm cargo into the autophagosome. We report that autophagic degradation of Smurf1 is dependent on p62. Moreover, the autophagic degradation of Smurf1 is prevented in the absence of the HECT domain or E3 ubiquitin ligase activity. We further proved that activation of autophagy leads to a decrease of Smurf1 and the inhibition of the phosphoinositide 3-kinase/protein kinase B signaling pathway in GB cells. Our results suggest that enhancement of autophagic degradation of Smurf1 may be a potential approach to treating GB.
Collapse
Affiliation(s)
- Da Han
- School of Life ScienceBeijing Institute of TechnologyChina
| | - Shengzhen Li
- School of Life ScienceBeijing Institute of TechnologyChina
| | - Qin Xia
- School of Life ScienceBeijing Institute of TechnologyChina
| | - Xinyi Meng
- School of Life ScienceBeijing Institute of TechnologyChina
| | - Lei Dong
- School of Life ScienceBeijing Institute of TechnologyChina
| |
Collapse
|
59
|
Schminke B, Kauffmann P, Schubert A, Altherr M, Gelis T, Miosge N. SMURF1 and SMURF2 in Progenitor Cells from Articular Cartilage and Meniscus during Late-Stage Osteoarthritis. Cartilage 2021; 13:117S-128S. [PMID: 33090007 PMCID: PMC8721605 DOI: 10.1177/1947603520967069] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
OBJECTIVE The aim of this study was to investigate the roles of SMURF1 and SMURF2 in progenitor cells from the human knee in late-stage osteoarthritis (OA). DESIGN We applied immunohistochemistry, immunocytochemistry, RNAi, lentiviral transfection, and Western blot analysis. We obtained chondrogenic progenitor cells (CPCs) from the articular cartilage and meniscus progenitor cells (MPCs) from the nonvascularized part of the meniscus. RESULTS SMURF1 and SMURF2 appeared in both osteoarthritic tissues. CPCs and MPCs exhibited comparable amounts of these proteins, which influence the balance between RUNX2 and SOX9. The overexpression of SMURF1 reduced the levels of RUNX2, SOX9, and TGFBR1. The overexpression of SMURF2 also reduced the levels of RUNX2 and TGFBR1, while SOX9 levels were not affected. The knockdown of SMURF1 had no effect on RUNX2, SOX9, or TGFBR1. The knockdown of SMURF2 enhanced RUNX2 and SOX9 levels in CPCs. The respective protein levels in MPCs were not affected. CONCLUSIONS This study shows that SMURF1 and SMURF2 are regulatory players for the expression of the major regulator transcription factors RUNX2 and SOX9 in CPCs and MPCs. Our novel findings may help elucidate new treatment strategies for cartilage regeneration.
Collapse
Affiliation(s)
- Boris Schminke
- Department of Oral and Maxillofacial
Surgery, University Medical Center Goettingen, Göttingen, Germany,Oral Biology and Tissue Regeneration
Work Group, University Medical Center Goettingen, Göttingen, Germany
| | - Philipp Kauffmann
- Department of Oral and Maxillofacial
Surgery, University Medical Center Goettingen, Göttingen, Germany
| | - Andrea Schubert
- Oral Biology and Tissue Regeneration
Work Group, University Medical Center Goettingen, Göttingen, Germany
| | - Manuel Altherr
- Oral Biology and Tissue Regeneration
Work Group, University Medical Center Goettingen, Göttingen, Germany
| | - Thomas Gelis
- Oral Biology and Tissue Regeneration
Work Group, University Medical Center Goettingen, Göttingen, Germany
| | - Nicolai Miosge
- Oral Biology and Tissue Regeneration
Work Group, University Medical Center Goettingen, Göttingen, Germany,Nicolai Miosge, Oral Biology and Tissue
Regeneration Work Group, University Medical Center Göttingen,
Robert-Koch-Strasse 40, Göttingen, 37075, Germany.
| |
Collapse
|
60
|
Min KD, Asakura M, Shirai M, Yamazaki S, Ito S, Fu HY, Asanuma H, Asano Y, Minamino T, Takashima S, Kitakaze M. ASB2 is a novel E3 ligase of SMAD9 required for cardiogenesis. Sci Rep 2021; 11:23056. [PMID: 34845242 PMCID: PMC8630118 DOI: 10.1038/s41598-021-02390-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 11/15/2021] [Indexed: 12/27/2022] Open
Abstract
Cardiogenesis requires the orchestrated spatiotemporal tuning of BMP signalling upon the balance between induction and counter-acting suppression of the differentiation of the cardiac tissue. SMADs are key intracellular transducers and the selective degradation of SMADs by the ubiquitin-proteasome system is pivotal in the spatiotemporal tuning of BMP signalling. However, among three SMADs for BMP signalling, SMAD1/5/9, only the specific E3 ligase of SMAD9 remains poorly investigated. Here, we report for the first time that SMAD9, but not the other SMADs, is ubiquitylated by the E3 ligase ASB2 and targeted for proteasomal degradation. ASB2, as well as Smad9, is conserved among vertebrates. ASB2 expression was specific to the cardiac region from the very early stage of cardiac differentiation in embryogenesis of mouse. Knockdown of Asb2 in zebrafish resulted in a thinned ventricular wall and dilated ventricle, which were rescued by simultaneous knockdown of Smad9. Abundant Smad9 protein leads to dysregulated cardiac differentiation through a mechanism involving Tbx2, and the BMP signal conducted by Smad9 was downregulated under quantitative suppression of Smad9 by Asb2. Our findings demonstrate that ASB2 is the E3 ligase of SMAD9 and plays a pivotal role in cardiogenesis through regulating BMP signalling.
Collapse
Affiliation(s)
- Kyung-Duk Min
- Department of Clinical Research and Development, National Cerebral and Cardiovascular Center, 6-1 Kishibe- Shimmachi, Suita, Osaka, 564-8565, Japan
| | - Masanori Asakura
- Department of Clinical Research and Development, National Cerebral and Cardiovascular Center, 6-1 Kishibe- Shimmachi, Suita, Osaka, 564-8565, Japan
- Department of Cardiovascular and Renal Medicine, Hyogo College of Medicine, Hyogo, Japan
| | - Manabu Shirai
- Department of Bioscience, National Cerebral and Cardiovascular Center, Osaka, Japan
- Omics Research Center, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Satoru Yamazaki
- Department of Cell Biology, National Cerebral and Cardiovascular Center, Osaka, Japan
- Department of Molecular Pharmacology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Shin Ito
- Department of Clinical Research and Development, National Cerebral and Cardiovascular Center, 6-1 Kishibe- Shimmachi, Suita, Osaka, 564-8565, Japan
| | - Hai Ying Fu
- Department of Clinical Research and Development, National Cerebral and Cardiovascular Center, 6-1 Kishibe- Shimmachi, Suita, Osaka, 564-8565, Japan
- Department of Cardiorenal and Cerebrovascular Medicine, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Hiroshi Asanuma
- Department of Internal Medicine, Meiji University of Integrative Medicine, Kyoto, Japan
| | - Yoshihiro Asano
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tetsuo Minamino
- Department of Cardiorenal and Cerebrovascular Medicine, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Seiji Takashima
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Masafumi Kitakaze
- Department of Clinical Research and Development, National Cerebral and Cardiovascular Center, 6-1 Kishibe- Shimmachi, Suita, Osaka, 564-8565, Japan.
- Hanwa Daini Senboku Hospital, Sakai, Osaka, Japan.
| |
Collapse
|
61
|
Du MG, Liu F, Chang Y, Tong S, Liu W, Chen YJ, Xie P. Neddylation modification of the U3 snoRNA-binding protein RRP9 by Smurf1 promotes tumorigenesis. J Biol Chem 2021; 297:101307. [PMID: 34662580 PMCID: PMC8569593 DOI: 10.1016/j.jbc.2021.101307] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 09/29/2021] [Accepted: 10/13/2021] [Indexed: 11/16/2022] Open
Abstract
Neddylation is a posttranslational modification that attaches ubiquitin-like protein Nedd8 to protein targets via Nedd8-specific E1-E2-E3 enzymes and modulates many important biological processes. Nedd8 attaches to a lysine residue of a substrate, not for degradation, but for modulation of substrate activity. We previously identified the HECT-type ubiquitin ligase Smurf1, which controls diverse cellular processes, is activated by Nedd8 through covalent neddylation. Smurf1 functions as a thioester bond-type Nedd8 ligase to catalyze its own neddylation. Numerous ubiquitination substrates of Smurf1 have been identified, but the neddylation substrates of Smurf1 remain unknown. Here, we show that Smurf1 interacts with RRP9, a core component of the U3 snoRNP complex, which is involved in pre-rRNA processing. Our in vivo and in vitro neddylation modification assays show that RRP9 is conjugated with Nedd8. RRP9 neddylation is catalyzed by Smurf1 and removed by the NEDP1 deneddylase. We identified Lys221 as a major neddylation site on RRP9. Deficiency of RRP9 neddylation inhibits pre-rRNA processing and leads to downregulation of ribosomal biogenesis. Consequently, functional studies suggest that ectopic expression of RRP9 promotes tumor cell proliferation, colony formation, and cell migration, whereas unneddylated RRP9, K221R mutant has no such effect. Furthermore, in human colorectal cancer, elevated expression of RRP9 and Smurf1 correlates with cancer progression. These results reveal that Smurf1 plays a multifaceted role in pre-rRNA processing by catalyzing RRP9 neddylation and shed new light on the oncogenic role of RRP9.
Collapse
Affiliation(s)
- Meng-Ge Du
- Department of Cell Biology, The Municipal Key Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing, China
| | - Fan Liu
- Department of Cell Biology, The Municipal Key Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing, China
| | - Yan Chang
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Shuai Tong
- Department of Cell Biology, The Municipal Key Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing, China
| | - Wei Liu
- Department of Cell Biology, The Municipal Key Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing, China
| | - Yu-Jiao Chen
- Department of Cell Biology, The Municipal Key Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing, China
| | - Ping Xie
- Department of Cell Biology, The Municipal Key Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing, China.
| |
Collapse
|
62
|
Shen J, Fu B, Li Y, Wu Y, Sang H, Zhang H, Lin H, Liu H, Huang W. E3 Ubiquitin Ligase-Mediated Regulation of Osteoblast Differentiation and Bone Formation. Front Cell Dev Biol 2021; 9:706395. [PMID: 34513836 PMCID: PMC8430030 DOI: 10.3389/fcell.2021.706395] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 08/03/2021] [Indexed: 12/13/2022] Open
Abstract
The ubiquitin–proteasome system (UPS) is an essential pathway that regulates the homeostasis and function of intracellular proteins and is a crucial protein-degradation system in osteoblast differentiation and bone formation. Abnormal regulation of ubiquitination leads to osteoblast differentiation disorders, interfering with bone formation and ultimately leading to osteoporosis. E3 ubiquitin ligases (E3) promote addition of a ubiquitin moiety to substrate proteins, specifically recognizing the substrate and modulating tyrosine kinase receptors, signaling proteins, and transcription factors involved in the regulation of osteoblast proliferation, differentiation, survival, and bone formation. In this review, we summarize current progress in the understanding of the function and regulatory effects of E3 ligases on the transcription factors and signaling pathways that regulate osteoblast differentiation and bone formation. A deep understanding of E3 ligase-mediated regulation of osteoblast differentiation provides a scientific rationale for the discovery and development of novel E3-targeting therapeutic strategies for osteoporosis.
Collapse
Affiliation(s)
- Jianlin Shen
- Guangdong Innovation Platform for Translation of 3D Printing Application, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,Department of Orthopedics, Affiliated Hospital of Putian University, Putian, China
| | - Bowen Fu
- Guangdong Innovation Platform for Translation of 3D Printing Application, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Yanfang Li
- Department of Pediatric Surgery, Affiliated Hospital of Putian University, Putian, China
| | - Yanjiao Wu
- Department of Orthopedics, Shunde Hospital of Southern Medical University, Guangzhou, China
| | - Hongxun Sang
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Heshi Zhang
- Department of Vessel and Breast, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Haibin Lin
- Department of Orthopedics, Affiliated Hospital of Putian University, Putian, China
| | - Huan Liu
- Department of Orthopedics, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Wenhua Huang
- Guangdong Innovation Platform for Translation of 3D Printing Application, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Medical Biomechanics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Orthopedic Center, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
63
|
Tril dampens Nodal signaling through Pellino2- and Traf6-mediated activation of Nedd4l. Proc Natl Acad Sci U S A 2021; 118:2104661118. [PMID: 34475212 DOI: 10.1073/pnas.2104661118] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 07/27/2021] [Indexed: 11/18/2022] Open
Abstract
Toll-like receptor 4 (Tlr) interactor with leucine-rich repeats (Tril) functions as a Tlr coreceptor to mediate innate immunity in adults. In Xenopus embryos, Tril triggers degradation of the transforming growth factor β (Tgf-ß) family inhibitor, Smad7. This enhances bone morphogenetic protein (Bmp) signaling to enable ventral mesoderm to commit to a blood fate. Here, we show that Tril simultaneously dampens Nodal signaling by catalytically activating the ubiquitin ligase NEDD4 Like (Nedd4l). Nedd4l then targets Nodal receptors for degradation. How Tril signals are transduced in a nonimmune context is unknown. We identify the ubiquitin ligase Pellino2 as a protein that binds to the cytoplasmic tail of Tril and subsequently forms a complex with Nedd4l and another E3 ligase, TNF-receptor associated factor 6 (Traf6). Pellino2 and Traf6 are essential for catalytic activation of Nedd4l, both in Xenopus and in mammalian cells. Traf6 ubiquitinates Nedd4l, which is then recruited to membrane compartments where activation occurs. Collectively, our findings reveal that Tril initiates a noncanonical Tlr-like signaling cascade to activate Nedd4l, thereby coordinately regulating the Bmp and Nodal arms of the Tgf-ß superfamily during vertebrate development.
Collapse
|
64
|
Zhang F, Yang Y, Chen X, Liu Y, Hu Q, Huang B, Liu Y, Pan Y, Zhang Y, Liu D, Liang R, Li G, Wei Q, Li L, Jin L. The long non-coding RNA βFaar regulates islet β-cell function and survival during obesity in mice. Nat Commun 2021; 12:3997. [PMID: 34183666 PMCID: PMC8238983 DOI: 10.1038/s41467-021-24302-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 06/07/2021] [Indexed: 02/08/2023] Open
Abstract
Despite obesity being a predisposing factor for pancreatic β-cell dysfunction and loss, the mechanisms underlying its negative effect on insulin-secreting cells remain poorly understood. In this study, we identify an islet-enriched long non-coding RNA (lncRNA), which we name β-cell function and apoptosis regulator (βFaar). βFaar is dramatically downregulated in the islets of the obese mice, and a low level of βFaar is necessary for the development of obesity-associated β-cell dysfunction and apoptosis. Mechanistically, βFaar promote the synthesis and secretion of insulin by upregulating islet-specific genes Ins2, NeuroD1, and Creb1 through sponging miR-138-5p. In addition, using quantitative mass spectrometry, we identify TRAF3IP2 and SMURF1 as interacting proteins that are specifically associated with βFaar. We demonstrate that SMURF1 ubiquitin ligase activity is essential for TRAF3IP2 ubiquitination and activation of NF-κB-mediate β-cell apoptosis. Our experiments provide direct evidence that dysregulated βFaar contributes to the development of obesity-induced β-cell injury and apoptosis. Beta-cell function is often impaired in obesity through incompletely understood mechanisms. Here the authors show that the long noncoding RNA βFaar is reduced by diet-induced obesity in mice, which leads to impaired beta-cell function via miR-138-5p and survival via TRAF3 Interacting Protein 2.
Collapse
Affiliation(s)
- Fangfang Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu Province, China
| | - Yue Yang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu Province, China
| | - Xi Chen
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu Province, China
| | - Yue Liu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu Province, China
| | - Qianxing Hu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu Province, China
| | - Bin Huang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu Province, China
| | - Yuhong Liu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu Province, China
| | - Yi Pan
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu Province, China
| | - Yanfeng Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu Province, China
| | - Dechen Liu
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
| | - Rui Liang
- Organ Transplant Center, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Guoqing Li
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China.,Pancreatic Research Institute, Southeast University, Nanjing, China
| | - Qiong Wei
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China. .,Pancreatic Research Institute, Southeast University, Nanjing, China.
| | - Ling Li
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China. .,Pancreatic Research Institute, Southeast University, Nanjing, China.
| | - Liang Jin
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu Province, China.
| |
Collapse
|
65
|
Han O, Pak B, Jin SW. The Role of BMP Signaling in Endothelial Heterogeneity. Front Cell Dev Biol 2021; 9:673396. [PMID: 34235147 PMCID: PMC8255612 DOI: 10.3389/fcell.2021.673396] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 05/21/2021] [Indexed: 01/07/2023] Open
Abstract
Bone morphogenetic proteins (BMPs), which compose the largest group of the transforming growth factor-β (TGF-ß) superfamily, have been implied to play a crucial role in diverse physiological processes. The most intriguing feature of BMP signaling is that it elicits heterogeneous responses from cells with equivalent identity, thus permitting highly context-dependent signaling outcomes. In endothelial cells (ECs), which are increasingly perceived as a highly heterogeneous population of cells with respect to their morphology, function, as well as molecular characteristics, BMP signaling has shown to elicit diverse and often opposite effects, illustrating the innate complexity of signaling responses. In this review, we provide a concise yet comprehensive overview of how outcomes of BMP signaling are modulated in a context-dependent manner with an emphasis on the underlying molecular mechanisms and summarize how these regulations of the BMP signaling promote endothelial heterogeneity.
Collapse
Affiliation(s)
- Orjin Han
- Cell Logistics Research Center, School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, South Korea
| | - Boryeong Pak
- Cell Logistics Research Center, School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, South Korea
| | - Suk-Won Jin
- Cell Logistics Research Center, School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, South Korea
| |
Collapse
|
66
|
The Role of HECT-Type E3 Ligase in the Development of Cardiac Disease. Int J Mol Sci 2021; 22:ijms22116065. [PMID: 34199773 PMCID: PMC8199989 DOI: 10.3390/ijms22116065] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 05/26/2021] [Accepted: 06/01/2021] [Indexed: 12/12/2022] Open
Abstract
Despite advances in medicine, cardiac disease remains an increasing health problem associated with a high mortality rate. Maladaptive cardiac remodeling, such as cardiac hypertrophy and fibrosis, is a risk factor for heart failure; therefore, it is critical to identify new therapeutic targets. Failing heart is reported to be associated with hyper-ubiquitylation and impairment of the ubiquitin–proteasome system, indicating an importance of ubiquitylation in the development of cardiac disease. Ubiquitylation is a post-translational modification that plays a pivotal role in protein function and degradation. In 1995, homologous to E6AP C-terminus (HECT) type E3 ligases were discovered. E3 ligases are key enzymes in ubiquitylation and are classified into three families: really interesting new genes (RING), HECT, and RING-between-RINGs (RBRs). Moreover, 28 HECT-type E3 ligases have been identified in human beings. It is well conserved in evolution and is characterized by the direct attachment of ubiquitin to substrates. HECT-type E3 ligase is reported to be involved in a wide range of human diseases and health. The role of HECT-type E3 ligases in the development of cardiac diseases has been uncovered in the last decade. There are only a few review articles summarizing recent advancements regarding HECT-type E3 ligase in the field of cardiac disease. This study focused on cardiac remodeling and described the role of HECT-type E3 ligases in the development of cardiac disease. Moreover, this study revealed that the current knowledge could be exploited for the development of new clinical therapies.
Collapse
|
67
|
Mark KG, Rape M. Ubiquitin-dependent regulation of transcription in development and disease. EMBO Rep 2021; 22:e51078. [PMID: 33779035 DOI: 10.15252/embr.202051078] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 11/29/2020] [Accepted: 03/01/2021] [Indexed: 12/19/2022] Open
Abstract
Transcription is an elaborate process that is required to establish and maintain the identity of the more than two hundred cell types of a metazoan organism. Strict regulation of gene expression is therefore vital for tissue formation and homeostasis. An accumulating body of work found that ubiquitylation of histones, transcription factors, or RNA polymerase II is crucial for ensuring that transcription occurs at the right time and place during development. Here, we will review principles of ubiquitin-dependent control of gene expression and discuss how breakdown of these regulatory circuits leads to a wide array of human diseases.
Collapse
Affiliation(s)
- Kevin G Mark
- Department of Molecular Cell Biology, University of California at Berkeley, Berkeley, CA, USA
| | - Michael Rape
- Department of Molecular Cell Biology, University of California at Berkeley, Berkeley, CA, USA.,Howard Hughes Medical Institute, University of California at Berkeley, Berkeley, CA, USA
| |
Collapse
|
68
|
Li Y, Cui C, Xie F, Kiełbasa S, Mei H, van Dinther M, van Dam H, Bauer A, Zhang L, Ten Dijke P. VprBP mitigates TGF-β and Activin signaling by promoting Smurf1-mediated type I receptor degradation. J Mol Cell Biol 2021; 12:138-151. [PMID: 31291647 PMCID: PMC7109606 DOI: 10.1093/jmcb/mjz057] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 04/16/2019] [Accepted: 06/06/2019] [Indexed: 12/13/2022] Open
Abstract
The transforming growth factor-β (TGF-β) family controls embryogenesis, stem cell differentiation, and tissue homeostasis. However, how post-translation modifications contribute to fine-tuning of TGF-β family signaling responses is not well understood. Inhibitory (I)-Smads can antagonize TGF-β/Smad signaling by recruiting Smurf E3 ubiquitin ligases to target the active TGF-β receptor for proteasomal degradation. A proteomic interaction screen identified Vpr binding protein (VprBP) as novel binding partner of Smad7. Mis-expression studies revealed that VprBP negatively controls Smad2 phosphorylation, Smad2-Smad4 interaction, as well as TGF-β target gene expression. VprBP was found to promote Smad7-Smurf1-TβRI complex formation and induce proteasomal degradation of TGF-β type I receptor (TβRI). Moreover, VprBP appears to stabilize Smurf1 by suppressing Smurf1 poly-ubiquitination. In multiple adult and mouse embryonic stem cells, depletion of VprBP promotes TGF-β or Activin-induced responses. In the mouse embryo VprBP expression negatively correlates with mesoderm marker expression, and VprBP attenuated mesoderm induction during zebrafish embryogenesis. Our findings thereby uncover a novel regulatory mechanism by which Smurf1 controls the TGF-β and Activin cascade and identify VprBP as a critical determinant of embryonic mesoderm induction.
Collapse
Affiliation(s)
- Yihao Li
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, 2300RC Leiden, The Netherlands
| | - Chao Cui
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, 2300RC Leiden, The Netherlands
| | - Feng Xie
- MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Szymon Kiełbasa
- Department of Human Genetics, Leiden Genome Technology Centre, Leiden University Medical Center, 2300RC Leiden, The Netherlands
| | - Hailiang Mei
- Sequence Analysis Support Core, Leiden University Medical Center, 2300RC Leiden, The Netherlands
| | - Maarten van Dinther
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, 2300RC Leiden, The Netherlands
| | - Hans van Dam
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, 2300RC Leiden, The Netherlands
| | - Andreas Bauer
- Novartis Institutes for BioMedical Research, Inc., Novartis Campus, Forum 2.5.01.30, CH-4056, Basel, Switzerland
| | - Long Zhang
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, 2300RC Leiden, The Netherlands.,MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Peter Ten Dijke
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, 2300RC Leiden, The Netherlands.,MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
69
|
A comprehensive phenotypic CRISPR-Cas9 screen of the ubiquitin pathway uncovers roles of ubiquitin ligases in mitosis. Mol Cell 2021; 81:1319-1336.e9. [PMID: 33539788 DOI: 10.1016/j.molcel.2021.01.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 10/20/2020] [Accepted: 01/11/2021] [Indexed: 12/13/2022]
Abstract
The human ubiquitin proteasome system, composed of over 700 ubiquitin ligases (E3s) and deubiquitinases (DUBs), has been difficult to characterize systematically and phenotypically. We performed chemical-genetic CRISPR-Cas9 screens to identify E3s/DUBs whose loss renders cells sensitive or resistant to 41 compounds targeting a broad range of biological processes, including cell cycle progression, genome stability, metabolism, and vesicular transport. Genes and compounds clustered functionally, with inhibitors of related pathways interacting similarly with E3s/DUBs. Some genes, such as FBXW7, showed interactions with many of the compounds. Others, such as RNF25 and FBXO42, showed interactions primarily with a single compound (methyl methanesulfonate for RNF25) or a set of related compounds (the mitotic cluster for FBXO42). Mutation of several E3s with sensitivity to mitotic inhibitors led to increased aberrant mitoses, suggesting a role for these genes in cell cycle regulation. Our comprehensive CRISPR-Cas9 screen uncovered 466 gene-compound interactions covering 25% of the interrogated E3s/DUBs.
Collapse
|
70
|
Sinha A, Iyengar PV, ten Dijke P. E3 Ubiquitin Ligases: Key Regulators of TGFβ Signaling in Cancer Progression. Int J Mol Sci 2021; 22:E476. [PMID: 33418880 PMCID: PMC7825147 DOI: 10.3390/ijms22020476] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 12/25/2020] [Accepted: 12/29/2020] [Indexed: 02/07/2023] Open
Abstract
Transforming growth factor β (TGFβ) is a secreted growth and differentiation factor that influences vital cellular processes like proliferation, adhesion, motility, and apoptosis. Regulation of the TGFβ signaling pathway is of key importance to maintain tissue homeostasis. Perturbation of this signaling pathway has been implicated in a plethora of diseases, including cancer. The effect of TGFβ is dependent on cellular context, and TGFβ can perform both anti- and pro-oncogenic roles. TGFβ acts by binding to specific cell surface TGFβ type I and type II transmembrane receptors that are endowed with serine/threonine kinase activity. Upon ligand-induced receptor phosphorylation, SMAD proteins and other intracellular effectors become activated and mediate biological responses. The levels, localization, and function of TGFβ signaling mediators, regulators, and effectors are highly dynamic and regulated by a myriad of post-translational modifications. One such crucial modification is ubiquitination. The ubiquitin modification is also a mechanism by which crosstalk with other signaling pathways is achieved. Crucial effector components of the ubiquitination cascade include the very diverse family of E3 ubiquitin ligases. This review summarizes the diverse roles of E3 ligases that act on TGFβ receptor and intracellular signaling components. E3 ligases regulate TGFβ signaling both positively and negatively by regulating degradation of receptors and various signaling intermediates. We also highlight the function of E3 ligases in connection with TGFβ's dual role during tumorigenesis. We conclude with a perspective on the emerging possibility of defining E3 ligases as drug targets and how they may be used to selectively target TGFβ-induced pro-oncogenic responses.
Collapse
Affiliation(s)
| | | | - Peter ten Dijke
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (A.S.); (P.V.I.)
| |
Collapse
|
71
|
Dunmore BJ, Jones RJ, Toshner MR, Upton PD, Morrell NW. Approaches to treat pulmonary arterial hypertension by targeting bmpr2 - from cell membrane to nucleus. Cardiovasc Res 2021; 117:2309-2325. [PMID: 33399862 DOI: 10.1093/cvr/cvaa350] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 11/06/2020] [Accepted: 12/15/2020] [Indexed: 12/12/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is estimated to affect between 10-50 people per million worldwide. The lack of cure and devastating nature of the disease means that treatment is crucial to arrest rapid clinical worsening. Current therapies are limited by their focus on inhibiting residual vasoconstriction rather than targeting key regulators of the cellular pathology. Potential disease-modifying therapies may come from research directed towards causal pathways involved in the cellular and molecular mechanisms of disease. It is widely acknowledged, that targeting reduced expression of the critical bone morphogenetic protein type-2 receptor (BMPR2) and its associated signalling pathways is a compelling therapeutic avenue to explore. In this review we highlight the advances that have been made in understanding this pathway and the therapeutics that are being tested in clinical trials and the clinic to treat PAH.
Collapse
Affiliation(s)
- Benjamin J Dunmore
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's and Royal Papworth Hospitals, Cambridge, UK
| | - Rowena J Jones
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's and Royal Papworth Hospitals, Cambridge, UK
| | - Mark R Toshner
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's and Royal Papworth Hospitals, Cambridge, UK
| | - Paul D Upton
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's and Royal Papworth Hospitals, Cambridge, UK
| | - Nicholas W Morrell
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's and Royal Papworth Hospitals, Cambridge, UK
| |
Collapse
|
72
|
Hagg A, Kharoud S, Goodchild G, Goodman CA, Chen JL, Thomson RE, Qian H, Gregorevic P, Harrison CA, Walton KL. TMEPAI/PMEPA1 Is a Positive Regulator of Skeletal Muscle Mass. Front Physiol 2020; 11:560225. [PMID: 33250771 PMCID: PMC7672205 DOI: 10.3389/fphys.2020.560225] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 10/12/2020] [Indexed: 12/20/2022] Open
Abstract
Inhibition of myostatin- and activin-mediated SMAD2/3 signaling using ligand traps, such as soluble receptors, ligand-targeting propeptides and antibodies, or follistatin can increase skeletal muscle mass in healthy mice and ameliorate wasting in models of cancer cachexia and muscular dystrophy. However, clinical translation of these extracellular approaches targeting myostatin and activin has been hindered by the challenges of achieving efficacy without potential effects in other tissues. Toward the goal of developing tissue-specific myostatin/activin interventions, we explored the ability of transmembrane prostate androgen-induced (TMEPAI), an inhibitor of transforming growth factor-β (TGF-β1)-mediated SMAD2/3 signaling, to promote growth, and counter atrophy, in skeletal muscle. In this study, we show that TMEPAI can block activin A, activin B, myostatin and GDF-11 activity in vitro. To determine the physiological significance of TMEPAI, we employed Adeno-associated viral vector (AAV) delivery of a TMEPAI expression cassette to the muscles of healthy mice, which increased mass by as much as 30%, due to hypertrophy of muscle fibers. To demonstrate that TMEPAI mediates its effects via inhibition of the SMAD2/3 pathway, tibialis anterior (TA) muscles of mice were co-injected with AAV vectors expressing activin A and TMEPAI. In this setting, TMEPAI blocked skeletal muscle wasting driven by activin-induced phosphorylation of SMAD3. In a model of cancer cachexia associated with elevated circulating activin A, delivery of AAV:TMEPAI into TA muscles of mice bearing C26 colon tumors ameliorated the muscle atrophy normally associated with cancer progression. Collectively, the findings indicate that muscle-directed TMEPAI gene delivery can inactivate the activin/myostatin-SMAD3 pathway to positively regulate muscle mass in healthy settings and models of disease.
Collapse
Affiliation(s)
- Adam Hagg
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.,Centre for Muscle Research, Department of Physiology, The University of Melbourne, Melbourne, VIC, Australia.,Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Swati Kharoud
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.,Faculty of Science, Engineering and Technology, Swinburne University of Technology, Melbourne, VIC, Australia
| | - Georgia Goodchild
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Craig A Goodman
- Centre for Muscle Research, Department of Physiology, The University of Melbourne, Melbourne, VIC, Australia.,Australian Institute for Musculoskeletal Science, Sunshine Hospital, The University of Melbourne, St Albans, VIC, Australia
| | - Justin L Chen
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.,Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Rachel E Thomson
- Centre for Muscle Research, Department of Physiology, The University of Melbourne, Melbourne, VIC, Australia.,Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Hongwei Qian
- Centre for Muscle Research, Department of Physiology, The University of Melbourne, Melbourne, VIC, Australia.,Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Paul Gregorevic
- Centre for Muscle Research, Department of Physiology, The University of Melbourne, Melbourne, VIC, Australia.,Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia.,Department of Neurology, The University of Washington School of Medicine, Seattle, WA, United States
| | - Craig A Harrison
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.,Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Kelly L Walton
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.,Hudson Institute of Medical Research, Clayton, VIC, Australia
| |
Collapse
|
73
|
A novel negative regulatory mechanism of Smurf2 in BMP/Smad signaling in bone. Bone Res 2020; 8:41. [PMID: 33298874 PMCID: PMC7680794 DOI: 10.1038/s41413-020-00115-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 08/08/2020] [Accepted: 08/18/2020] [Indexed: 12/19/2022] Open
Abstract
Transforming growth factor-β (TGF-β) and bone morphogenetic protein (BMP) play important roles in bone metabolism. Smad ubiquitination regulatory factors (Smurfs) regulate TGF-β/BMP signaling via ubiquitination, resulting in degradation of signaling molecules to prevent excessive activation of TGF-β/BMP signaling. Though Smurf2 has been shown to negatively regulate TGF-β/Smad signaling, its involvement in BMP/Smad signaling in bone metabolism has not been thoroughly investigated. In the present study, we sought to evaluate the role of Smurf2 in BMP/Smad signaling in bone metabolism. Absorbable collagen sponges containing 3 μg of recombinant human BMP2 (rhBMP2) were implanted in the dorsal muscle pouches of wild type (WT) and Smurf2−/− mice. The rhBMP2-induced ectopic bone in Smurf2−/− mice showed greater bone mass, higher mineral apposition and bone formation rates, and greater osteoblast numbers than the ectopic bone in WT mice. In WT mice, the ectopic bone consisted of a thin discontinuous outer cortical shell and scant inner trabecular bone. In contrast, in Smurf2−/− mice, the induced bone consisted of a thick, continuous outer cortical shell and abundant inner trabecular bone. Additionally, rhBMP2-stimulated bone marrow stromal cells (BMSCs) from Smurf2−/− mice showed increased osteogenic differentiation. Smurf2 induced the ubiquitination of Smad1/5. BMP/Smad signaling was enhanced in Smurf2−/− BMSCs stimulated with rhBMP2, and the inhibition of BMP/Smad signaling suppressed osteogenic differentiation of these BMSCs. These findings demonstrate that Smurf2 negatively regulates BMP/Smad signaling, thereby identifying a new regulatory mechanism in bone metabolism.
Collapse
|
74
|
Xia Q, Li Y, Han D, Dong L. SMURF1, a promoter of tumor cell progression? Cancer Gene Ther 2020; 28:551-565. [PMID: 33204002 DOI: 10.1038/s41417-020-00255-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 10/14/2020] [Accepted: 10/29/2020] [Indexed: 12/20/2022]
Abstract
Overexpression of HECT-type E3 ubiquitin ligase SMURF1 is correlated with poor prognosis in patients with various cancers, such as glioblastoma, colon cancer, and clear cell renal cell carcinoma. SMURF1 acts as a tumor promoter by ubiquitination modification and/or degradation of tumor-suppressing proteins. Combined treatment of Smurf1 knockdown with rapamycin showed collaborative antitumor effects in mice. This review described the role of HECT, WW, and C2 domains in regulating SMURF1 substrate selection. We summarized up to date SMURF1 substrates regulating different type cell signaling, thus, accelerating tumor progression, invasion, and metastasis. Furthermore, the downregulation of SMURF1 expression, inhibition of its E3 activity and regulation of its specificity to substrates prevent tumor progression. The potential application of SMURF1 regulators, specifically, wisely choose certain drugs by blocking SMURF1 selectivity in tumor suppressors, to develop novel anticancer treatments.
Collapse
Affiliation(s)
- Qin Xia
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Yang Li
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Da Han
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Lei Dong
- School of Life Science, Beijing Institute of Technology, Beijing, China.
| |
Collapse
|
75
|
Chen J, A. Gingold J. Dysregulated PJA1-TGF-β signaling in cancer stem cell-associated liver cancers. Oncoscience 2020; 7:88-95. [PMID: 33457451 PMCID: PMC7781490 DOI: 10.18632/oncoscience.522] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 11/18/2020] [Indexed: 12/12/2022] Open
Abstract
The transforming growth factor beta (TGF-β) signaling pathway plays important roles in cell differentiation, stem cell modulation, organ lineage, and immune suppression. TGF-β signaling is negatively regulated by the ubiquitin-proteasome pathway. Although mouse models of cancer arising from a defective TGF-β pathway clearly demonstrate the tumor-suppressive role of TGF-β, the underlying mechanism by which a defective TGF-β pathway triggers liver cancer development is poorly understood. This review summarizes key findings from our recent studies connecting TGF-β to hepatic oncogenesis and highlights the vulnerability of TGF-β signaling to PJA1-mediated ubiquitination. TGF-β, together with the chromatin insulator CCCTC-binding factor (CTCF), epigenetically and transcriptionally regulate tumor promoter genes, including IGF2 and TERT, in TGF-β-defective mice and in human liver cancers. Dysfunction of the TGF-β-regulated SPTBN1/SMAD3/CTCF complex increases stem cell-like properties in hepatocellular carcinoma (HCC) cells and enhances tumorigenesis in tumor-initiating cells in a mouse model. PJA1, a novel E3 ubiquitin ligase, is a key negative regulator of TGF-β signaling. PJA1 overexpression is detected in HCCs and is sufficient to suppress SMAD3- and SPTBN1-mediated TGF-β tumor suppressor signaling, promoting HCC proliferation. Dysregulated PJA1-TGF-β signaling activates oncogenic genes and promotes tumorigenesis in human liver cancers. In addition, inhibition of PJA1 by treatment with E3 ligase inhibitors restores TGF-β tumor-suppressor function and suppresses liver cancer progression. These new findings suggest potential therapeutic avenues for targeting dysregulated PJA1-TGF-β signaling via cancer stem cells in liver cancers.
Collapse
Affiliation(s)
- Jian Chen
- Department of Gastroenterology, Hepatology, & Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Sandhill Therapeutics, Inc., Dallas, TX, USA
| | - Julian A. Gingold
- Montefiore Medical Center, OB/GYN and Women’s Health, Bronx, NY, USA
| |
Collapse
|
76
|
Halloran D, Durbano HW, Nohe A. Bone Morphogenetic Protein-2 in Development and Bone Homeostasis. J Dev Biol 2020; 8:E19. [PMID: 32933207 PMCID: PMC7557435 DOI: 10.3390/jdb8030019] [Citation(s) in RCA: 176] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/01/2020] [Accepted: 09/11/2020] [Indexed: 12/11/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) are multi-functional growth factors belonging to the Transforming Growth Factor-Beta (TGF-β) superfamily. These proteins are essential to many developmental processes, including cardiogenesis, neurogenesis, and osteogenesis. Specifically, within the BMP family, Bone Morphogenetic Protein-2 (BMP-2) was the first BMP to be characterized and has been well-studied. BMP-2 has important roles during embryonic development, as well as bone remodeling and homeostasis in adulthood. Some of its specific functions include digit formation and activating osteogenic genes, such as Runt-Related Transcription Factor 2 (RUNX2). Because of its diverse functions and osteogenic potential, the Food and Drug Administration (FDA) approved usage of recombinant human BMP-2 (rhBMP-2) during spinal fusion surgery, tibial shaft repair, and maxillary sinus reconstructive surgery. However, shortly after initial injections of rhBMP-2, several adverse complications were reported, and alternative therapeutics have been developed to limit these side-effects. As the clinical application of BMP-2 is largely implicated in bone, we focus primarily on its role in bone. However, we also describe briefly the role of BMP-2 in development. We then focus on the structure of BMP-2, its activation and regulation signaling pathways, BMP-2 clinical applications, and limitations of using BMP-2 as a therapeutic. Further, this review explores other potential treatments that may be useful in treating bone disorders.
Collapse
Affiliation(s)
| | | | - Anja Nohe
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA; (D.H.); (H.W.D.)
| |
Collapse
|
77
|
Nguyen TH, Han TH, Newfeld SJ, Serpe M. Selective Disruption of Synaptic BMP Signaling by a Smad Mutation Adjacent to the Highly Conserved H2 Helix. Genetics 2020; 216:159-175. [PMID: 32737119 PMCID: PMC7463279 DOI: 10.1534/genetics.120.303484] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 07/16/2020] [Indexed: 01/08/2023] Open
Abstract
Bone morphogenetic proteins (BMPs) shape normal development and function via canonical and noncanonical signaling pathways. BMPs initiate canonical signaling by binding to transmembrane receptors that phosphorylate Smad proteins and induce their translocation into the nucleus and regulation of target genes. Phosphorylated Smads also accumulate at cellular junctions, but this noncanonical, local BMP signaling modality remains less defined. We have recently reported that phosphorylated Smad (pMad in Drosophila) accumulates at synaptic junctions in protein complexes with genetically distinct composition and regulation. Here, we examined a wide collection of DrosophilaMad alleles and searched for molecular features relevant to pMad accumulation at synaptic junctions. We found that strong Mad alleles generally disrupt both synaptic and nuclear pMad, whereas moderate Mad alleles have a wider range of phenotypes and can selectively impact different BMP signaling pathways. Interestingly, regulatory Mad mutations reveal that synaptic pMad appears to be more sensitive to a net reduction in Mad levels than nuclear pMad. Importantly, a previously uncharacterized allele, Mad8 , showed markedly reduced synaptic pMad but only moderately diminished nuclear pMad. The postsynaptic composition and electrophysiological properties of Mad8 neuromuscular junctions (NMJs) were also altered. Using biochemical approaches, we examined how a single point mutation in Mad8 could influence the Mad-receptor interface and identified a key motif, the H2 helix. Our study highlights the biological relevance of Smad-dependent, synaptic BMP signaling and uncovers a highly conserved structural feature of Smads, critical for normal development and function.
Collapse
Affiliation(s)
- Tho Huu Nguyen
- Section on Cellular Communication, Eunice Kennedy Shiver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892
| | - Tae Hee Han
- Section on Cellular Communication, Eunice Kennedy Shiver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892
| | - Stuart J Newfeld
- School of Life Sciences, Arizona State University, Tempe, Arizona 85287-4501
| | - Mihaela Serpe
- Section on Cellular Communication, Eunice Kennedy Shiver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892
| |
Collapse
|
78
|
Zhang Y, Qian H, Wu B, You S, Wu S, Lu S, Wang P, Cao L, Zhang N, Sun Y. E3 Ubiquitin ligase NEDD4 family‑regulatory network in cardiovascular disease. Int J Biol Sci 2020; 16:2727-2740. [PMID: 33110392 PMCID: PMC7586430 DOI: 10.7150/ijbs.48437] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 08/06/2020] [Indexed: 12/17/2022] Open
Abstract
Protein ubiquitination represents a critical modification occurring after translation. E3 ligase catalyzes the covalent binding of ubiquitin to the protein substrate, which could be degraded. Ubiquitination as an important protein post-translational modification is closely related to cardiovascular disease. The NEDD4 family, belonging to HECT class of E3 ubiquitin ligases can recognize different substrate proteins, including PTEN, ENaC, Nav1.5, SMAD2, PARP1, Septin4, ALK1, SERCA2a, TGFβR3 and so on, via the WW domain to catalyze ubiquitination, thus participating in multiple cardiovascular-related disease such as hypertension, arrhythmia, myocardial infarction, heart failure, cardiotoxicity, cardiac hypertrophy, myocardial fibrosis, cardiac remodeling, atherosclerosis, pulmonary hypertension and heart valve disease. However, there is currently no review comprehensively clarifying the important role of NEDD4 family proteins in the cardiovascular system. Therefore, the present review summarized recent studies about NEDD4 family members in cardiovascular disease, providing novel insights into the prevention and treatment of cardiovascular disease. In addition, assessing transgenic animals and performing gene silencing would further identify the ubiquitination targets of NEDD4. NEDD4 quantification in clinical samples would also constitute an important method for determining NEDD4 significance in cardiovascular disease.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Hao Qian
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Boquan Wu
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Shilong You
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Shaojun Wu
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Saien Lu
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Pingyuan Wang
- Staff scientist, Center for Molecular Medicine National Heart Lung and Blood Institute, National Institutes of Health, the United States
| | - Liu Cao
- Key Laboratory of Medical Cell Biology, Ministry of Education; Institute of Translational Medicine, China Medical University; Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, Shenyang, Liaoning, China
| | - Naijin Zhang
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Yingxian Sun
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| |
Collapse
|
79
|
Xia Q, Zhang H, Zhang P, Li Y, Xu M, Li X, Li X, Dong L. Oncogenic Smurf1 promotes PTEN wild-type glioblastoma growth by mediating PTEN ubiquitylation. Oncogene 2020; 39:5902-5915. [PMID: 32737433 DOI: 10.1038/s41388-020-01400-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 07/01/2020] [Accepted: 07/20/2020] [Indexed: 02/08/2023]
Abstract
PI3K/Akt/mTOR signaling pathway activity is highly elevated in glioblastoma (GBM). Although rapamycin is known to inhibit this pathway, GBM patients are resistant to rapamycin monotherapy. This may be related to mutations of tumor suppressor phosphatase and tensin homolog (PTEN). Here, we show that higher expression of E3 ligase Smad ubiquitylation regulatory factor 1 (Smurf1) in GBM is correlated with poor prognosis. Smurf1 promotes cell growth and colony formation by accelerating cell cycle and aberrant signaling pathways. In addition, we show that Smurf1 ubiquitylates and degrades PTEN. We further demonstrate that the oncogenic role of Smurf1 is dependent on PTEN. Upregulated Smurf1 impairs PTEN activity, leading to consistent activation of PI3K/Akt/mTOR signaling pathway; and depletion of Smurf1 dramatically inhibits cell proliferation and tumor growth. Moreover, loss of Smurf1 abolishes the aberrant regulation of PTEN, causing negative feedback on PI3K/Akt/mTOR signaling pathway, and thus leading to rescue of tumor sensitivity to rapamycin in an orthotopic GBM model. Taken together, we show that Smurf1 promotes tumor progression via PTEN, and combined treatment of Smurf1 knockdown with mammalian target of rapamycin (mTOR) inhibition reduces tumor progression. These results identify a unique role of Smurf1 in mTOR inhibitor resistance and provide a strong rationale for combined therapy targeting GBM.
Collapse
Affiliation(s)
- Qin Xia
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Hanwen Zhang
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Pei Zhang
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Yang Li
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Mengchuan Xu
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Xiaobo Li
- Tianjin Key Laboratory of Medical Epigenetics, Department of Immunology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Xuejun Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lei Dong
- School of Life Science, Beijing Institute of Technology, Beijing, China.
| |
Collapse
|
80
|
Wang M, Dai W, Ke Z, Li Y. Functional roles of E3 ubiquitin ligases in gastric cancer. Oncol Lett 2020; 20:22. [PMID: 32774495 PMCID: PMC7405480 DOI: 10.3892/ol.2020.11883] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 04/29/2020] [Indexed: 12/15/2022] Open
Abstract
To date, >650 E3 ubiquitin ligases have been described in humans, including >600 really interesting new genes (RINGs), 28 homologous to E6-associated protein C-terminus (HECTs) and several RING-in-between-RINGs. They are considered key regulators and therapeutic targets of many types of human cancers, including gastric cancer (GC). Among them, some RING and HECT E3 ligases are closely related to the proliferation, infiltration and prognosis of GC. During the past few years, abnormal expressions and functions of many E3 ligases have been identified in GC. However, the functional roles of E3 ligases in GC have not been fully elucidated. The present article focuses on the functional roles of E3 ligases related to the proteasome in GC. In this comprehensive review, the latest research progress on E3 ligases involved in GC and elaborate their structure, classification, functional roles and therapeutic value in GC was summarized. Finally, 30 E3 ligases that serve essential roles in regulating the development of GC were described. Some of these ligases may serve as oncogenes or tumor suppressors in GC, whereas the pathological mechanism of others needs further study; for example, constitutive photomorphogenic 1. In conclusion, the present review demonstrated that E3 ligases are crucial tumor regulatory factors and potential therapeutic targets in GC. Therefore, more studies should focus on the therapeutic targeting of E3 ligases in GC.
Collapse
Affiliation(s)
- Mingliang Wang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Wei Dai
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Zhangyan Ke
- Department of Geriatric Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Yongxiang Li
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| |
Collapse
|
81
|
Bai Y, Ying Y. The Post-translational Modifications of Smurf2 in TGF-β Signaling. Front Mol Biosci 2020; 7:128. [PMID: 32733916 PMCID: PMC7358609 DOI: 10.3389/fmolb.2020.00128] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 06/02/2020] [Indexed: 12/18/2022] Open
Abstract
Smad ubiquitin regulatory factor 2 (Smurf2), an essential negative regulator of TGF-β signaling, ubiquitinates TGF-β receptors (TβRs) and Smad proteins, inducing their proteasomal degradation. Smurf2 plays crucial roles in regulating TGF-β signaling and maintaining normal cellular functions and tissue homeostasis; dysfunction of Smurf2 triggers abnormal TGF-β signaling in pathological states. Smurf2 has been reported as a potentially strong candidate for targeting therapies for related diseases. Recent work has begun to focus on the regulation of Smurf2 itself, and emerging evidence indicates that Smurf2 is regulated by post-translational modifications (PTMs) mechanisms. These mechanisms predominantly regulate the expression level and E3 ligase activity of Smurf2, strongly suggesting that this protein contributes to complicated roles under multiple pathophysiological conditions. In this review, we cover some significant and novel mechanisms of the PTMs that potentially control Smurf2 participation in TGF-β signaling, including ubiquitylation, SUMOylation, neddylation, phosphorylation, and methylation in order to provide a broad view of the depth and sophistication of Smurf2 function in TGF-β regulation, as well as perspectives for future therapeutic directions for its associated diseases.
Collapse
Affiliation(s)
- Yangjinming Bai
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology and Department of Pathophysiology, Schools of Basic Medical Sciences, Nanchang University, Nanchang, China.,Nanchang Joint Program, Queen Mary School, Nanchang University, Nanchang, China
| | - Ying Ying
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology and Department of Pathophysiology, Schools of Basic Medical Sciences, Nanchang University, Nanchang, China
| |
Collapse
|
82
|
Current potential therapeutic strategies targeting the TGF-β/Smad signaling pathway to attenuate keloid and hypertrophic scar formation. Biomed Pharmacother 2020; 129:110287. [PMID: 32540643 DOI: 10.1016/j.biopha.2020.110287] [Citation(s) in RCA: 227] [Impact Index Per Article: 45.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 05/08/2020] [Accepted: 05/16/2020] [Indexed: 12/12/2022] Open
Abstract
Aberrant scar formation, which includes keloid and hypertrophic scars, is associated with a pathological disorganized wound healing process with chronic inflammation. The TGF-β/Smad signaling pathway is the most canonical pathway through which the formation of collagen in the fibroblasts and myofibroblasts is regulated. Sustained activation of the TGF-β/Smad signaling pathway results in the long-term overactivation of fibroblasts and myofibroblasts, which is necessary for the excessive collagen formation in aberrant scars. There are two categories of therapeutic strategies that aim to target the TGF-β/Smad signaling pathway in fibroblasts and myofibroblasts to interfere with their cellular functions and reduce cell proliferation. The first therapeutic strategy includes medications, and the second strategy is composed of genetic and cellular therapeutics. Therefore, the focus of this review is to critically evaluate these two main therapeutic strategies that target the TGF-β/Smad pathway to attenuate abnormal skin scar formation.
Collapse
|
83
|
Activity of Smurf2 Ubiquitin Ligase Is Regulated by the Wnt Pathway Protein Dishevelled. Cells 2020; 9:cells9051147. [PMID: 32392721 PMCID: PMC7290506 DOI: 10.3390/cells9051147] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/01/2020] [Accepted: 05/04/2020] [Indexed: 01/17/2023] Open
Abstract
Wnt and BMP signaling pathways are two key molecular machineries regulating development and homeostasis. The efficient coordination of Wnt and BMP is essential in many developmental processes such as establishment of antero-posterior and dorso-ventral body axis, regulation of convergent extension, or development of various organ systems. SMAD ubiquitination regulatory factor (Smurf) family of E3 ubiquitin ligases are important and evolutionary conserved regulators of TGF-β/BMP signaling pathways. Smurf2 has been previously shown to regulate Wnt/planar cell polarity (PCP) signaling pathway by ubiquitinating Prickle1, one of the key components of PCP. We explored the role of Smurf2 in Wnt pathways in further detail and identified that Smurf2 is also a ubiquitin ligase of Dishevelled (DVL), the key cytoplasmic signal transducer in the Wnt pathway. Interestingly, the Smurf2 and DVL relationship expands beyond substrate-E3 ligase. We can show that DVL activates Smurf2, which allows Smurf2 to ubiquitinate its substrates from Wnt/PCP (Prickle1) as well as TGF-β/BMP (Smad2) pathways more efficiently. Using SMAD7 as an example of Smurf2 activator we show that DVL and SMAD7 both activates Smurf2 activity. In HEK293 cells the deficiency of DVL phenocopies absence of Smurf2 and leads to the increased phosphorylation of R-Smads. Smurf2-DVL connection provides a novel and intriguing point of crosstalk for Wnt and BMP pathways.
Collapse
|
84
|
Al-Rawi R, Al-Beshri A, Mikhail FM, McCormick K. Fragile Bones Secondary to SMURF1 Gene Duplication. Calcif Tissue Int 2020; 106:567-573. [PMID: 32040594 DOI: 10.1007/s00223-020-00668-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 01/28/2020] [Indexed: 10/25/2022]
Abstract
Studies on mice have shown that the Smad Ubiquitin Regulatory Factor-1 (SMURF1) gene negatively regulates osteoblast function and the response to bone morphogenetic protein in a dose-dependent fashion (Chan et al. in Mol Cell Biol 27(16):5776-5789, https://doi.org/10.1128/MCB.00218-07, 2007; Yamashita et al. in Cell 121(1):101-113, https://doi.org/10.1016/j.cell.2005.01.035, 2005). In addition, a tumorigenic role for SMURF1 has been implicated due to the interference with apoptosis signals (Nie et al. in J Biol Chem 285(30):22818-22830, https://doi.org/10.1074/jbc.M110.126920, 2010; Wang et al. in Nat Commun 5:4901, https://doi.org/10.1038/ncomms5901, 2014). A 10-year-old girl with a history of severe developmental delay, infantile seizures, and B-cell lymphoma, in remission for approximately 3.5 years, was referred to the metabolic bone clinic for fractures and low bone mineral density. Array comparative genomic hybridization revealed a pathogenic microduplication in chromosome 7 at bands 7q21.3q22.1 that encompasses the SMURF1 gene. The clinical features of this child are congruous with the phenotype as ascribed excess Smurf1 mutations in mice. This is the first case description of osteoporosis in a child secondary to a microduplication involving SMURF1 gene.
Collapse
Affiliation(s)
- Rawan Al-Rawi
- Department of Pediatrics, University of Alabama at Birmingham, 1601 4th Avenue South, 1600 7th AVE S, Birmingham, AL, 35233-1711, USA.
| | - Ali Al-Beshri
- Department of Internal Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Fady M Mikhail
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kenneth McCormick
- Department of Pediatrics, University of Alabama at Birmingham, 1601 4th Avenue South, 1600 7th AVE S, Birmingham, AL, 35233-1711, USA
| |
Collapse
|
85
|
Zhang X, Zhan Y, Lin W, Zhao F, Guo C, Chen Y, Du M, Li D, Zhang L, An W, Wang H, Xie P. Smurf1 aggravates non‐alcoholic fatty liver disease by stabilizing SREBP‐1c in an E3 activity‐independent manner. FASEB J 2020; 34:7631-7643. [DOI: 10.1096/fj.201902952rr] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 03/19/2020] [Accepted: 03/24/2020] [Indexed: 01/23/2023]
Affiliation(s)
- Xin Zhang
- State Key Laboratory of Cellular Stress Biology School of Life Sciences Innovation Center for Cell Biology Xiamen University Xiamen China
| | - Yutao Zhan
- Municipal Laboratory for Liver Protection and Regulation of Regeneration Department of Cell Biology Capital Medical University Beijing China
- Department of Gastroenterology Beijing Tongren Hospital Capital Medical University Beijing China
| | - Wenjun Lin
- Municipal Laboratory for Liver Protection and Regulation of Regeneration Department of Cell Biology Capital Medical University Beijing China
- Department of Gastroenterology Beijing Tongren Hospital Capital Medical University Beijing China
| | - Fei Zhao
- Department of Gastroenterology Beijing Tongren Hospital Capital Medical University Beijing China
| | - Chaojing Guo
- State Key Laboratory of Proteomics National Center of Protein Sciences (Beijing) Beijing Institute of Lifeomics Beijing China
| | - Yujiao Chen
- Municipal Laboratory for Liver Protection and Regulation of Regeneration Department of Cell Biology Capital Medical University Beijing China
| | - Mengge Du
- Municipal Laboratory for Liver Protection and Regulation of Regeneration Department of Cell Biology Capital Medical University Beijing China
| | - Dongnian Li
- Department of Gastroenterology Beijing Tongren Hospital Capital Medical University Beijing China
| | - Lingqiang Zhang
- State Key Laboratory of Proteomics National Center of Protein Sciences (Beijing) Beijing Institute of Lifeomics Beijing China
| | - Wei An
- Municipal Laboratory for Liver Protection and Regulation of Regeneration Department of Cell Biology Capital Medical University Beijing China
| | - Hong‐Rui Wang
- State Key Laboratory of Cellular Stress Biology School of Life Sciences Innovation Center for Cell Biology Xiamen University Xiamen China
| | - Ping Xie
- Municipal Laboratory for Liver Protection and Regulation of Regeneration Department of Cell Biology Capital Medical University Beijing China
| |
Collapse
|
86
|
Sun Z, Cai S, Zabkiewicz C, Liu C, Ye L. Bone morphogenetic proteins mediate crosstalk between cancer cells and the tumour microenvironment at primary tumours and metastases (Review). Int J Oncol 2020; 56:1335-1351. [PMID: 32236571 DOI: 10.3892/ijo.2020.5030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 02/28/2020] [Indexed: 11/05/2022] Open
Abstract
Bone morphogenetic proteins (BMP) are pluripotent molecules, co‑ordinating cellular functions from early embryonic and postnatal development to tissue repair, regeneration and homeostasis. They are also involved in tumourigenesis, disease progression and the metastasis of various solid tumours. Emerging evidence has indicated that BMPs are able to promote disease progression and metastasis by orchestrating communication between cancer cells and the surrounding microenvironment. The interactions occur between BMPs and epidermal growth factor receptor, hepatocyte growth factor, fibroblast growth factor, vascular endothelial growth factor and extracellular matrix components. Overall, these interactions co‑ordinate the cellular functions of tumour cells and other types of cell in the tumour to promote the growth of the primary tumour, local invasion, angiogenesis and metastasis, and the establishment and survival of cancer cells in the metastatic niche. Therefore, the present study aimed to provide an informative summary of the involvement of BMPs in the tumour microenvironment.
Collapse
Affiliation(s)
- Zhiwei Sun
- VIP‑II Division of Medical Department, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Shuo Cai
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom
| | - Catherine Zabkiewicz
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom
| | - Chang Liu
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom
| | - Lin Ye
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom
| |
Collapse
|
87
|
Takebayashi-Suzuki K, Suzuki A. Intracellular Communication among Morphogen Signaling Pathways during Vertebrate Body Plan Formation. Genes (Basel) 2020; 11:E341. [PMID: 32213808 PMCID: PMC7141137 DOI: 10.3390/genes11030341] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 03/18/2020] [Accepted: 03/19/2020] [Indexed: 12/25/2022] Open
Abstract
During embryonic development in vertebrates, morphogens play an important role in cell fate determination and morphogenesis. Bone morphogenetic proteins (BMPs) belonging to the transforming growth factor-β (TGF-β) family control the dorsal-ventral (DV) patterning of embryos, whereas other morphogens such as fibroblast growth factor (FGF), Wnt family members, and retinoic acid (RA) regulate the formation of the anterior-posterior (AP) axis. Activation of morphogen signaling results in changes in the expression of target genes including transcription factors that direct cell fate along the body axes. To ensure the correct establishment of the body plan, the processes of DV and AP axis formation must be linked and coordinately regulated by a fine-tuning of morphogen signaling. In this review, we focus on the interplay of various intracellular regulatory mechanisms and discuss how communication among morphogen signaling pathways modulates body axis formation in vertebrate embryos.
Collapse
Affiliation(s)
- Kimiko Takebayashi-Suzuki
- Amphibian Research Center, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima, Hiroshima 739-8526, Japan
| | - Atsushi Suzuki
- Graduate School of Integrated Sciences for Life, Amphibian Research Center, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-8526, Japan
| |
Collapse
|
88
|
Maintenance of the Undifferentiated State in Myogenic Progenitor Cells by TGFβ Signaling is Smad Independent and Requires MEK Activation. Int J Mol Sci 2020; 21:ijms21031057. [PMID: 32033454 PMCID: PMC7038076 DOI: 10.3390/ijms21031057] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/28/2020] [Accepted: 02/04/2020] [Indexed: 12/12/2022] Open
Abstract
Transforming growth factor β (TGFβ) is a pluripotent cytokine and regulates a myriad of biological processes. It has been established that TGFβ potently inhibits skeletal muscle differentiation; however, the molecular mechanism is not clearly defined. Previously, we reported that inhibition of the TGFβ canonical pathway by an inhibitory Smad, Smad7, does not reverse this effect on differentiation, suggesting that activation of receptor Smads (R-Smads) by TGFβ is not responsible for repression of myogenesis. In addition, pharmacological blockade of Smad3 activation by TGFβ did not reverse TGFβ's inhibitory effect on myogenesis. In considering other pathways, we observed that TGFβ potently activates MEK/ERK, and a pharmacological inhibitor of MEK reversed TGFβ's inhibitory effect on myogenesis, as indicated by a myogenin promoter-reporter gene, sarcomeric myosin heavy chain accumulation, and phenotypic myotube formation. Furthermore, we found that c-Jun, a known potent repressor of myogenesis, which is coincidently also a down-stream target of MEK/ERK signaling, was phosphorylated and accumulates in the nucleus in response to TGFβ activation. Taken together, these observations support a model in which TGFβ activates a MEK/ERK/c-Jun pathway to repress skeletal myogenesis, maintaining the pluripotent undifferentiated state in myogenic progenitors.
Collapse
|
89
|
Totland MZ, Rasmussen NL, Knudsen LM, Leithe E. Regulation of gap junction intercellular communication by connexin ubiquitination: physiological and pathophysiological implications. Cell Mol Life Sci 2020; 77:573-591. [PMID: 31501970 PMCID: PMC7040059 DOI: 10.1007/s00018-019-03285-0] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 08/10/2019] [Accepted: 08/16/2019] [Indexed: 12/15/2022]
Abstract
Gap junctions consist of arrays of intercellular channels that enable adjacent cells to communicate both electrically and metabolically. Gap junctions have a wide diversity of physiological functions, playing critical roles in both excitable and non-excitable tissues. Gap junction channels are formed by integral membrane proteins called connexins. Inherited or acquired alterations in connexins are associated with numerous diseases, including heart failure, neuropathologies, deafness, skin disorders, cataracts and cancer. Gap junctions are highly dynamic structures and by modulating the turnover rate of connexins, cells can rapidly alter the number of gap junction channels at the plasma membrane in response to extracellular or intracellular cues. Increasing evidence suggests that ubiquitination has important roles in the regulation of endoplasmic reticulum-associated degradation of connexins as well as in the modulation of gap junction endocytosis and post-endocytic sorting of connexins to lysosomes. In recent years, researchers have also started to provide insights into the physiological roles of connexin ubiquitination in specific tissue types. This review provides an overview of the advances made in understanding the roles of connexin ubiquitination in the regulation of gap junction intercellular communication and discusses the emerging physiological and pathophysiological implications of these processes.
Collapse
Affiliation(s)
- Max Zachrisson Totland
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, 0424, Oslo, Norway
- K.G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Oslo, Norway
| | - Nikoline Lander Rasmussen
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, 0424, Oslo, Norway
- K.G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Oslo, Norway
- Department of Medical Biology, University of Tromsø, Tromsø, Norway
| | - Lars Mørland Knudsen
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, 0424, Oslo, Norway
- K.G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Oslo, Norway
| | - Edward Leithe
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, 0424, Oslo, Norway.
- K.G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
90
|
Fu L, Cui CP, Zhang X, Zhang L. The functions and regulation of Smurfs in cancers. Semin Cancer Biol 2019; 67:102-116. [PMID: 31899247 DOI: 10.1016/j.semcancer.2019.12.023] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 11/10/2019] [Accepted: 12/26/2019] [Indexed: 02/06/2023]
Abstract
Smad ubiquitination regulatory factor 1 (Smurf1) and Smurf2 are HECT-type E3 ubiquitin ligases, and both Smurfs were initially identified to regulate Smad protein stability in the TGF-β/BMP signaling pathway. In recent years, Smurfs have exhibited E3 ligase-dependent and -independent activities in various kinds of cells. Smurfs act as either potent tumor promoters or tumor suppressors in different tumors by regulating biological processes, including metastasis, apoptosis, cell cycle, senescence and genomic stability. The regulation of Smurfs activity and expression has therefore emerged as a hot spot in tumor biology research. Further, the Smurf1- or Smurf2-deficient mice provide more in vivo clues for the functional study of Smurfs in tumorigenesis and development. In this review, we summarize these milestone findings and, in turn, reveal new avenues for the prevention and treatment of cancer by regulating Smurfs.
Collapse
Affiliation(s)
- Lin Fu
- Institute of Chronic Disease, Qingdao Municipal Hospital, Qingdao University, Qingdao 266000, China
| | - Chun-Ping Cui
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 100850, China
| | - Xueli Zhang
- Department of General Surgery, Shanghai Fengxian Central Hospital Graduate Training Base, Fengxian Hospital, Southern Medical University, Shanghai, China.
| | - Lingqiang Zhang
- Institute of Chronic Disease, Qingdao Municipal Hospital, Qingdao University, Qingdao 266000, China; State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 100850, China; Peixian People's Hospital, Jiangsu Province 221600, China.
| |
Collapse
|
91
|
García‐García P, Ruiz M, Reyes R, Delgado A, Évora C, Riancho JA, Rodríguez‐Rey JC, Pérez‐Campo FM. Smurf1 Silencing Using a LNA-ASOs/Lipid Nanoparticle System to Promote Bone Regeneration. Stem Cells Transl Med 2019; 8:1306-1317. [PMID: 31631568 PMCID: PMC6877774 DOI: 10.1002/sctm.19-0145] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 09/17/2019] [Indexed: 12/19/2022] Open
Abstract
Despite the great advance of bone tissue engineering in the last few years, repair of bone defects remains a major problem. Low cell engraftment and dose-dependent side effects linked to the concomitant administration of bone morphogenetic proteins (BMPs) are the main problems currently hindering the clinical use of mesenchymal stem cell (MSC)-based therapies in this field. We have managed to bypass these drawbacks by combining the silencing the Smurf1 ubiquitin ligase in MSCs with the use of a scaffold that sustainably releases low doses of BMP-2. In this system, Smurf1 silencing is achieved by using GapmeRs, a clinically safe method that avoids the use of viral vectors, facilitating its translation to the clinic. Here, we show that a single transient transfection with a small quantity of a Smurf1-specific GapmeR is able to induce a significant level of silencing of the target gene, enough to prime MSCs for osteogenic differentiation. Smurf1 silencing highly increases MSCs responsiveness to BMP-2, allowing a dramatic reduction of the dose needed to achieve the desired therapeutic effect. The combination of these primed cells with alginate scaffolds designed to sustainably and locally release low doses of BMP-2 to the defect microenvironment is able to induce the formation of a mature bone matrix both in an osteoporotic rat calvaria system and in a mouse ectopic model. Importantly, this approach also enhances osteogenic differentiation in MSCs from osteoporotic patients, characterized by a reduced bone-forming potential, even at low BMP doses, underscoring the regenerative potential of this system. Stem Cells Translational Medicine 2019;8:1306&1317.
Collapse
Affiliation(s)
- Patricia García‐García
- Department of Chemical Engineering and Pharmaceutical TechnologyInstitute of Biomedical Technologies (ITB), University of La LagunaLa LagunaSpain
| | - Mario Ruiz
- Department of Molecular Biology, Faculty of MedicineUniversity of Cantabria, IDIVALSantanderSpain
| | - Ricardo Reyes
- Department of Biochemistry, Microbiology, Cellular Biology and GeneticsInstitute of Biomedical Technologies (ITB), University of La LagunaLa LagunaSpain
| | - Araceli Delgado
- Department of Chemical Engineering and Pharmaceutical TechnologyInstitute of Biomedical Technologies (ITB), University of La LagunaLa LagunaSpain
| | - Carmen Évora
- Department of Chemical Engineering and Pharmaceutical TechnologyInstitute of Biomedical Technologies (ITB), University of La LagunaLa LagunaSpain
| | - José Antonio Riancho
- Department of Internal Medicine, Hospital U M ValdecillaUniversity of Cantabria, IDIVALSantanderSpain
| | | | - Flor María Pérez‐Campo
- Department of Molecular Biology, Faculty of MedicineUniversity of Cantabria, IDIVALSantanderSpain
| |
Collapse
|
92
|
Bernassola F, Chillemi G, Melino G. HECT-Type E3 Ubiquitin Ligases in Cancer. Trends Biochem Sci 2019; 44:1057-1075. [DOI: 10.1016/j.tibs.2019.08.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 08/13/2019] [Accepted: 08/23/2019] [Indexed: 12/30/2022]
|
93
|
Seo D, Jung SM, Park JS, Lee J, Ha J, Kim M, Park SH. The deubiquitinating enzyme PSMD14 facilitates tumor growth and chemoresistance through stabilizing the ALK2 receptor in the initiation of BMP6 signaling pathway. EBioMedicine 2019; 49:55-71. [PMID: 31685442 PMCID: PMC7113187 DOI: 10.1016/j.ebiom.2019.10.039] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 09/23/2019] [Accepted: 10/21/2019] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Although bone morphogenetic protein 6 (BMP6) signaling pathway has been implicated in many types of cancer, its role of tumorigenesis seems to be controversial and its ubiquitin-modifying mechanisms have not been fully addressed. Our study was designed to investigate how BMP6 signaling pathway is regulated by ubiquitin-modifying systems and to address molecular and clinical significance in colorectal cancers. METHODS Human deubiquitnase (DUB) siRNA library was used to screen the specific DUB, named PSMD14, involved in BMP6 signaling pathway. Immunoblot, immunoprecipitation and ubiquitination assays were used to analyze targets of the PSMD14. A role of PSMD14-mediated BMP6 signaling pathway for malignant cancer progression was investigated using in vitro and in vivo model of colorectal cancers as well as clinical samples of colorectal cancer patients. FINDINGS The deubiquitinase PSMD14 acts as a positive regulator for the initiation of the BMP6 signaling pathway through deubiquitinating K48-linked ALK2 type I receptor ubiquitination mediated by Smurf1 E3 ligase, resulting in increased stability of the ALK2. This role of PSMD14 is independent of its intrinsic role in the 26S proteasome system. Furthermore, either PSMD14 or ALK2 depletion significantly decreases tumorigenesis of HCT116 colorectal cancer cells in a xenograft model as well as cancer stemness/chemoresistance, and expression of the PSMD14 and ALK2 gene are correlated with malignant progression and the survival of colorectal cancer patients. INTERPRETATION These findings suggest that the PSMD14-ALK2 axis plays an essential role in initiation of the BMP6 signaling pathway and contributes to tumorigenesis and chemoresistance of colorectal cancers.
Collapse
Affiliation(s)
- Dongyeob Seo
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Su Myung Jung
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jin Seok Park
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jaewon Lee
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jihoon Ha
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Minbeom Kim
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Seok Hee Park
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| |
Collapse
|
94
|
Valer JA, Sánchez-de-Diego C, Pimenta-Lopes C, Rosa JL, Ventura F. ACVR1 Function in Health and Disease. Cells 2019; 8:cells8111366. [PMID: 31683698 PMCID: PMC6912516 DOI: 10.3390/cells8111366] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 10/28/2019] [Accepted: 10/30/2019] [Indexed: 12/12/2022] Open
Abstract
Activin A receptor type I (ACVR1) encodes for a bone morphogenetic protein type I receptor of the TGFβ receptor superfamily. It is involved in a wide variety of biological processes, including bone, heart, cartilage, nervous, and reproductive system development and regulation. Moreover, ACVR1 has been extensively studied for its causal role in fibrodysplasia ossificans progressiva (FOP), a rare genetic disorder characterised by progressive heterotopic ossification. ACVR1 is linked to different pathologies, including cardiac malformations and alterations in the reproductive system. More recently, ACVR1 has been experimentally validated as a cancer driver gene in diffuse intrinsic pontine glioma (DIPG), a malignant childhood brainstem glioma, and its function is being studied in other cancer types. Here, we review ACVR1 receptor function and signalling in physiological and pathological processes and its regulation according to cell type and mutational status. Learning from different functions and alterations linked to ACVR1 is a key step in the development of interdisciplinary research towards the identification of novel treatments for these pathologies.
Collapse
Affiliation(s)
- José Antonio Valer
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, 08907 Barcelona, Spain.
| | - Cristina Sánchez-de-Diego
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, 08907 Barcelona, Spain.
| | - Carolina Pimenta-Lopes
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, 08907 Barcelona, Spain.
| | - Jose Luis Rosa
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, 08907 Barcelona, Spain.
| | - Francesc Ventura
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, 08907 Barcelona, Spain.
| |
Collapse
|
95
|
Cooke M, Baker MJ, Kazanietz MG, Casado-Medrano V. PKCε regulates Rho GTPases and actin cytoskeleton reorganization in non-small cell lung cancer cells. Small GTPases 2019; 12:202-208. [PMID: 31648598 DOI: 10.1080/21541248.2019.1684785] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Oncogenic protein kinase C epsilon (PKCε) promotes the formation of membrane ruffles and motility in non-small cell lung cancer (NSCLC) cells. We found that PKCε is down-regulated when NSCLC cells undergo epithelial-to-mesenchymal transition (EMT) in response to TGF-β, thus becoming dispensable for migration and invasion in the mesenchymal state. PKCε silencing or inhibition leads to stress fibre formation, suggesting that this kinase negatively regulates RhoA activity. Ruffle formation induced by PKCε activation in the epithelial state is dependent on PI3K, but does not involve the PI3K-dependent Rac-GEFs Ect2, Trio, Vav2 or Tiam1, suggesting alternative Rac-GEFs as mediators of this response. In the proposed model, PKCε acts as a rheostat for Rho GTPases that differs in the epithelial and mesenchymal states.
Collapse
Affiliation(s)
- Mariana Cooke
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Martin J Baker
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Marcelo G Kazanietz
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Victoria Casado-Medrano
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
96
|
Ruetalo N, Anders S, Stollmaier C, Jäckl M, Schütz-Stoffregen MC, Stefan N, Wolf C, Wiesner S. The WW1 Domain Enhances Autoinhibition in Smurf Ubiquitin Ligases. J Mol Biol 2019; 431:4834-4847. [PMID: 31628949 DOI: 10.1016/j.jmb.2019.09.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 09/23/2019] [Accepted: 09/26/2019] [Indexed: 10/25/2022]
Abstract
Downregulation of ubiquitin (Ub) ligase activity prevents premature ubiquitination and is critical for cellular homeostasis. Nedd4 Ub ligases share a common domain architecture and yet are regulated in distinct ways through interactions of the catalytic HECT domain with the N-terminal C2 domain or the central WW domain region. Smurf1 and Smurf2 are two highly related Nedd4 ligases with ~70% overall sequence identity. Here, we show that the Smurf1 C2 domain interacts with the HECT domain and inhibits ligase activity in trans. However, in contrast to Smurf2, we find that full-length Smurf1 is a highly active Ub ligase, and we can attribute this striking difference in regulation to the lack of one WW domain (WW1) in Smurf1. Using NMR spectroscopy and biochemical assays, we identified the WW1 region as an additional inhibitory element in Smurf2 that cooperates with the C2 domain to enhance HECT domain binding and Smurf2 inhibition. Our work provides important insights into Smurf regulation and highlights that the activities of highly related proteins can be controlled in distinct ways.
Collapse
Affiliation(s)
- Natalia Ruetalo
- Max Planck Institute for Developmental Biology, Max-Planck-Ring 5, 72076 Tübingen, Germany
| | - Samira Anders
- Max Planck Institute for Developmental Biology, Max-Planck-Ring 5, 72076 Tübingen, Germany
| | - Carsten Stollmaier
- Max Planck Institute for Developmental Biology, Max-Planck-Ring 5, 72076 Tübingen, Germany; Institute of Biophysics and Physical Biochemistry, University of Regensburg, Universitätsstr. 31, 93040 Regensburg, Germany
| | - Magnus Jäckl
- Max Planck Institute for Developmental Biology, Max-Planck-Ring 5, 72076 Tübingen, Germany
| | - Mira C Schütz-Stoffregen
- Max Planck Institute for Developmental Biology, Max-Planck-Ring 5, 72076 Tübingen, Germany; Institute of Biophysics and Physical Biochemistry, University of Regensburg, Universitätsstr. 31, 93040 Regensburg, Germany
| | - Nadine Stefan
- Institute of Biophysics and Physical Biochemistry, University of Regensburg, Universitätsstr. 31, 93040 Regensburg, Germany
| | - Christine Wolf
- Max Planck Institute for Developmental Biology, Max-Planck-Ring 5, 72076 Tübingen, Germany
| | - Silke Wiesner
- Max Planck Institute for Developmental Biology, Max-Planck-Ring 5, 72076 Tübingen, Germany; Institute of Biophysics and Physical Biochemistry, University of Regensburg, Universitätsstr. 31, 93040 Regensburg, Germany.
| |
Collapse
|
97
|
Lear TB, McKelvey AC, Evankovich JW, Rajbhandari S, Coon TA, Dunn SR, Londino JD, McVerry BJ, Zhang Y, Valenzi E, Burton CL, Gordon R, Gingras S, Lockwood KC, Jurczak MJ, Lafyatis R, Shlomchik MJ, Liu Y, Chen BB. KIAA0317 regulates pulmonary inflammation through SOCS2 degradation. JCI Insight 2019; 4:129110. [PMID: 31578312 DOI: 10.1172/jci.insight.129110] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 09/04/2019] [Indexed: 01/08/2023] Open
Abstract
Dysregulated proinflammatory cytokine release has been implicated in the pathogenesis of several life-threatening acute lung illnesses such as pneumonia, sepsis, and acute respiratory distress syndrome. Suppressors of cytokine signaling proteins, particularly SOCS2, have recently been described as antiinflammatory mediators. However, the regulation of SOCS2 protein has not been described. Here we describe a mechanism of SOCS2 regulation by the action of the ubiquitin E3 ligase KIAA0317. KIAA0317-mediated degradation of SOCS2 exacerbated inflammation in vitro, and depletion of KIAA0317 in vivo ameliorated pulmonary inflammation. KIAA0317-knockout mice exhibited resistance to LPS-induced pulmonary inflammation, while KIAA03017 reexpression mitigated this effect. We uncovered a small molecule inhibitor of KIAA0317 protein (BC-1365) that prevented SOCS2 degradation and attenuated LPS- and P. aeruginosa-induced lung inflammation in vivo. These studies show KIAA0317 to be a critical mediator of pulmonary inflammation through its degradation of SOCS2 and a potential candidate target for therapeutic inhibition.
Collapse
Affiliation(s)
- Travis B Lear
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, School of Medicine.,Department of Environmental and Occupational Health, School of Public Health
| | - Alison C McKelvey
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, School of Medicine
| | - John W Evankovich
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, School of Medicine
| | - Shristi Rajbhandari
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, School of Medicine
| | - Tiffany A Coon
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, School of Medicine
| | - Sarah R Dunn
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, School of Medicine
| | - James D Londino
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, School of Medicine
| | - Bryan J McVerry
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, School of Medicine.,Department of Environmental and Occupational Health, School of Public Health
| | - Yingze Zhang
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, School of Medicine
| | - Eleanor Valenzi
- Division of Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine
| | - Christine L Burton
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, School of Medicine
| | | | | | | | - Michael J Jurczak
- Division of Endocrinology and Metabolism, Department of Medicine, School of Medicine
| | - Robert Lafyatis
- Division of Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine
| | | | - Yuan Liu
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, School of Medicine.,Aging Institute and.,McGowan Institute for Regenerative Medicine
| | - Bill B Chen
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, School of Medicine.,Aging Institute and.,Vascular Medicine Institute, Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
98
|
Guo L, Wang R, Zhang K, Yuan J, Wang J, Wang X, Ma J, Wu C. A PINCH-1-Smurf1 signaling axis mediates mechano-regulation of BMPR2 and stem cell differentiation. J Cell Biol 2019; 218:3773-3794. [PMID: 31578224 PMCID: PMC6829670 DOI: 10.1083/jcb.201902022] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 06/30/2019] [Accepted: 08/08/2019] [Indexed: 12/13/2022] Open
Abstract
Mechanical cues from extracellular matrix exert strong effects on stem cell differentiation. This study finds that a signaling axis consisting of PINCH-1, Smurf1, and BMPR2 senses mechanical signals from extracellular matrix and regulates BMP signaling and mesenchymal stem cell differentiation. Mechano-environment plays multiple critical roles in the control of mesenchymal stem cell (MSC) fate decision, but the underlying signaling mechanisms remain undefined. We report here a signaling axis consisting of PINCH-1, SMAD specific E3 ubiquitin protein ligase 1 (Smurf1), and bone morphogenetic protein type 2 receptor (BMPR2) that links mechano-environment to MSC fate decision. PINCH-1 interacts with Smurf1, which inhibits the latter from interacting with BMPR2 and consequently suppresses BMPR2 degradation, resulting in augmented BMP signaling and MSC osteogenic differentiation (OD). Extracellular matrix (ECM) stiffening increases PINCH-1 level and consequently activates this signaling axis. Depletion of PINCH-1 blocks stiff ECM-induced BMP signaling and OD, whereas overexpression of PINCH-1 overrides signals from soft ECM and promotes OD. Finally, perturbation of either Smurf1 or BMPR2 expression is sufficient to block the effects of PINCH-1 on BMP signaling and MSC fate decision. Our findings delineate a key signaling mechanism through which mechano-environment controls BMPR2 level and MSC fate decision.
Collapse
Affiliation(s)
- Ling Guo
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Academy for Advanced Interdisciplinary Studies and Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Rong Wang
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Academy for Advanced Interdisciplinary Studies and Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Kuo Zhang
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Academy for Advanced Interdisciplinary Studies and Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Jifan Yuan
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Academy for Advanced Interdisciplinary Studies and Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Jiaxin Wang
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Academy for Advanced Interdisciplinary Studies and Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Xiaoxia Wang
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Academy for Advanced Interdisciplinary Studies and Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Jianfei Ma
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Academy for Advanced Interdisciplinary Studies and Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Chuanyue Wu
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
99
|
Groppe JC. Induced degradation of protein kinases by bifunctional small molecules: a next-generation strategy. Expert Opin Drug Discov 2019; 14:1237-1253. [DOI: 10.1080/17460441.2019.1660641] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Jay C. Groppe
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, USA
| |
Collapse
|
100
|
Zhu B, Xue F, Zhang C, Li G. LMCD1 promotes osteogenic differentiation of human bone marrow stem cells by regulating BMP signaling. Cell Death Dis 2019; 10:647. [PMID: 31501411 PMCID: PMC6733937 DOI: 10.1038/s41419-019-1876-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 07/24/2019] [Accepted: 08/01/2019] [Indexed: 12/16/2022]
Abstract
Human bone marrow stem cells (BMSCs) are heterogeneous progenitor cells with two defining features, self-renew and multi-lineage differentiation. As one of the differentiation directions, osteogenesis is vital for bone homeostasis. A growing body of evidences show that ubiquitin-dependent protein degradation plays an essential role in the osteogenic differentiation of BMSCs. In this study, we found that LMCD1 was upregulated during osteogenic differentiation process of BMSCs by analyzing GSE80614. In vitro and in vivo functional studies confirmed that LMCD1 was critical to the osteogenic commitment of BMSCs. Compared to those of the controls, downregulation of LMCD1 significantly restrained osteogenic differentiation and enhanced adipogenic differentiation, while upregulation of LMCD1 increased the osteogenic differentiation and suppressed adipogenic differentiation. Mechanically, we found that LMCD1 could protect RUNX2 and Smad1 protein from Smurf1-induced ubiquitination degradation thereby regulating BMP signaling. In conclusion, our findings suggest that LMCD1 is a novel regulator of osteogenic differentiation and may be a potential therapeutic target for bone metabolism related diseases.
Collapse
Affiliation(s)
- Bin Zhu
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, NO.600 Yishan Road, 200233, Shanghai, China
| | - Feng Xue
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, NO.600 Yishan Road, 200233, Shanghai, China
| | - Changqing Zhang
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, NO.600 Yishan Road, 200233, Shanghai, China.
| | - Guangyi Li
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, NO.600 Yishan Road, 200233, Shanghai, China.
| |
Collapse
|