1
|
Zhao H, Cao N, Liu Q, Zhang Y, Jin R, Lai H, Zheng L, Zhang H, Zhu Y, Ma Y, Yang Z, Wu Z, Li W, Liu Y, Cheng L, Chen Y. Inhibition of the E3 ligase UBR5 stabilizes TERT and protects vascular organoids from oxidative stress. J Transl Med 2024; 22:1080. [PMID: 39609696 PMCID: PMC11605888 DOI: 10.1186/s12967-024-05887-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 11/14/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND Excessive oxidative stress is known to cause endothelial dysfunction and drive cardiovascular diseases (CVD). While telomerase reverse transcriptase (TERT) shows protective effects against oxidative stress in rodents and is associated to human flow-mediated dilation in CVD, its regulatory mechanisms in human vascular systems under pathological oxidative stress require further investigation. METHODS Human induced pluripotent stem cells (hiPSCs) were used to create vascular organoids (VOs). These VOs and human umbilical vein endothelial cells (HUVECs) were subjected to oxidative stress through both hydrogen peroxide (H2O2) and oxidized low-density lipoprotein (oxLDL) models. The effects of TERT overexpression by inhibition of the ubiquitin protein ligase E3 component N-recognin 5 (UBR5) on reactive oxygen species (ROS)-induced vascular injury and cellular senescence were assessed using neovascular sprouting assays, senescence-associated β-galactosidase (SA-β-Gal) staining, and senescence-associated secretory phenotype (SASP) assays. RESULTS ROS significantly impaired VO development and endothelial progenitor cell (EPC) angiogenesis, evidenced by reduced neovascular sprouting and increased senescence markers, including elevated SA-β-Gal activity and SASP-related cytokine levels. Overexpression of TERT counteracted these effects, restoring VO development and EPC function. Immunoprecipitation-mass spectrometry identified UBR5 as a critical TERT regulator, facilitating its degradation. Inhibition of UBR5 stabilized TERT, improving VO angiogenic capacity, and reducing SA-β-Gal activity and SASP cytokine levels. CONCLUSIONS Inhibiting UBR5 stabilizes TERT, which preserves EPC angiogenic capacity, reduces VO impairment, and delays endothelial cell senescence under oxidative stress. These findings highlight the potential of targeting UBR5 to enhance vascular health in oxidative stress-related conditions.
Collapse
Affiliation(s)
- Haijing Zhao
- Department of Cardiology, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing, 100037, People's Republic of China
- Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, 100853, People's Republic of China
| | - Nian Cao
- Department of Cardiology, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing, 100037, People's Republic of China
| | - Qi Liu
- Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, 100853, People's Republic of China
| | - Yingyue Zhang
- Department of Cardiology, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing, 100037, People's Republic of China
- Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, 100853, People's Republic of China
| | - Rui Jin
- Beijing Institute of Biotechnology, Beijing, 100850, People's Republic of China
| | - Huiying Lai
- Department of Clinical Laboratory, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, People's Republic of China
| | - Li Zheng
- School of Medicine, Nankai University, Tianjin, 300071, People's Republic of China
| | - Honghong Zhang
- Department of Cardiology, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing, 100037, People's Republic of China
- Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, 100853, People's Republic of China
| | - Yue Zhu
- Department of Cardiology, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing, 100037, People's Republic of China
- Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, 100853, People's Republic of China
| | - Yuhan Ma
- School of Medicine, Nankai University, Tianjin, 300071, People's Republic of China
| | - Zengao Yang
- School of Medicine, South China University of Technology, Guangzhou, 510006, People's Republic of China
| | - Zhengfeng Wu
- Department of Cardiology, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing, 100037, People's Republic of China
- Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, 100853, People's Republic of China
| | - Weini Li
- Department of Biomedical Science, Cedars-Sinai Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, USA
| | - Yuqi Liu
- Department of Cardiology, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing, 100037, People's Republic of China.
- National Key Laboratory of Kidney Diseases, Beijing, 100853, People's Republic of China.
- Department of Cardiology, National Clinical Research Center of Geriatric Disease, Beijing, 100853, People's Republic of China.
- Beijing Key Laboratory of Chronic Heart Failure Precision Medicine, Beijing, 100853, People's Republic of China.
| | - Long Cheng
- The Key Laboratory of Geriatrics, Institute of Geriatric Medicine, Beijing Institute of Geriatrics, Chinese Academy of Medical Sciences, Beijing Hospital/National Centre of Gerontology of National Health Commission, Beijing, 100730, People's Republic of China.
- Beijing Institute of Biotechnology, Beijing, 100850, People's Republic of China.
| | - Yundai Chen
- Department of Cardiology, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing, 100037, People's Republic of China.
| |
Collapse
|
2
|
Cohen A, Jeng EE, Voorhies M, Symington J, Ali N, Rodriguez RA, Bassik MC, Sil A. Genome-scale CRISPR screening reveals that C3aR signaling is critical for rapid capture of fungi by macrophages. PLoS Pathog 2022; 18:e1010237. [PMID: 36174103 PMCID: PMC9578593 DOI: 10.1371/journal.ppat.1010237] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 10/18/2022] [Accepted: 07/13/2022] [Indexed: 11/18/2022] Open
Abstract
The fungal pathogen Histoplasma capsulatum (Hc) invades, replicates within, and destroys macrophages. To interrogate the molecular mechanisms underlying this interaction, we conducted a host-directed CRISPR-Cas9 screen and identified 361 genes that modify macrophage susceptibility to Hc infection, greatly expanding our understanding of host gene networks targeted by Hc. We identified pathways that have not been previously implicated in Hc interaction with macrophages, including the ragulator complex (involved in nutrient stress sensing), glycosylation enzymes, protein degradation machinery, mitochondrial respiration genes, solute transporters, and the ER membrane complex (EMC). The highest scoring protective hits included the complement C3a receptor (C3aR), a G-protein coupled receptor (GPCR) that recognizes the complement fragment C3a. Although it is known that complement components react with the fungal surface, leading to opsonization and release of small peptide fragments such as C3a, a role for C3aR in macrophage interactions with fungi has not been elucidated. We demonstrated that whereas C3aR is dispensable for macrophage phagocytosis of bacteria and latex beads, it is critical for optimal macrophage capture of pathogenic fungi, including Hc, the ubiquitous fungal pathogen Candida albicans, and the causative agent of Valley Fever Coccidioides posadasii. We showed that C3aR localizes to the early phagosome during Hc infection where it coordinates the formation of actin-rich membrane protrusions that promote Hc capture. We also showed that the EMC promotes surface expression of C3aR, likely explaining its identification in our screen. Taken together, our results provide new insight into host processes that affect Hc-macrophage interactions and uncover a novel and specific role for C3aR in macrophage recognition of fungi.
Collapse
Affiliation(s)
- Allison Cohen
- University of California San Francisco, Department of Microbiology and Immunology, San Francisco, California, United States of America
| | - Edwin E. Jeng
- Stanford University, Department of Genetics, Palo Alto, California, United States of America
| | - Mark Voorhies
- University of California San Francisco, Department of Microbiology and Immunology, San Francisco, California, United States of America
| | - Jane Symington
- University of California San Francisco, Department of Microbiology and Immunology, San Francisco, California, United States of America
| | - Nebat Ali
- University of California San Francisco, Department of Microbiology and Immunology, San Francisco, California, United States of America
| | - Rosa A. Rodriguez
- University of California San Francisco, Department of Microbiology and Immunology, San Francisco, California, United States of America
| | - Michael C. Bassik
- Stanford University, Department of Genetics, Palo Alto, California, United States of America
| | - Anita Sil
- University of California San Francisco, Department of Microbiology and Immunology, San Francisco, California, United States of America
| |
Collapse
|
3
|
Jung JU, Ghosh A, Earnest S, Deaton SL, Cobb MH. UBR5 is a novel regulator of WNK1 stability. Am J Physiol Cell Physiol 2022; 322:C1176-C1186. [PMID: 35442829 DOI: 10.1152/ajpcell.00417.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The with no lysine (K) 1 (WNK1) protein kinase maintains cellular ion homeostasis in many tissues through actions on ion cotransporters and channels. Increased accumulation of WNK1 protein leads to pseudohypoaldosteronism type II (PHAII), a form of familial hypertension. WNK1 can be degraded via its adaptor-dependent recruitment to the Cullin3-RBX1 E3 ligase complex by the ubiquitin-proteasome system. Disruption of this process also leads to disease. To determine if this is the primary mechanism of WNK1 turnover, we examined WNK1 protein stability and degradation by measuring its rate of decay after blockade of translation. Here, we show that WNK1 protein degradation exhibits atypical kinetics in Hela cells. Consistent with this apparent complexity, we found that multiple degradative pathways can modulate cellular WNK1 protein amount. WNK1 protein is degraded not only by the proteasome, but also by the lysosome. Non-lysosomal cysteine proteases calpain and caspases also influence WNK1 degradation, as inhibitors of these proteases modestly increased WNK1 protein expression. Importantly, we discovered that the E3 ubiquitin ligase UBR5 interacts with WNK1 and its deficiency results in increased WNK1 protein. Our results further demonstrate that increased WNK1 in UBR5-depleted cells is attributable to reduced lysosomal degradation of WNK1 protein. Taken together, our findings provide insights into the multiplicity of degradative pathways involved in WNK1 turnover and uncover UBR5 as a previously unknown regulator of WNK1 protein stability that leads to lysosomal degradation of WNK1 protein.
Collapse
Affiliation(s)
- Ji-Ung Jung
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX, United States
| | - Anwesha Ghosh
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX, United States
| | - Svetlana Earnest
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX, United States
| | - Staci L Deaton
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX, United States
| | - Melanie H Cobb
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
4
|
Kim SH, Cho JH, Park BO, Park BC, Kim JH, Park SG, Kim S. Phosphorylation of REPS1 at Ser709 by RSK attenuates the recycling of transferrin receptor. BMB Rep 2021. [PMID: 33407999 PMCID: PMC8167248 DOI: 10.5483/bmbrep.2021.54.5.266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
RalBP1 associated EPS domain containing 1 (REPS1) is conserved from Drosophila to humans and implicated in the endocytic system. However, an exact role of REPS1 remains largely unknown. Here, we demonstrated that mitogen activated protein kinase kinase (MEK)-p90 ribosomal S6 Kinase (RSK) signaling pathway directly phosphorylated REPS1 at Ser709 upon stimulation by epidermal growth factor (EGF) and amino acid. While REPS2 is known to be involved in the endocytosis of EGF receptor (EGFR), REPS1 knockout (KO) cells did not show any defect in the endocytosis of EGFR. However, in the REPS1 KO cells and the KO cells reconstituted with a non-phosphorylatable REPS1 (REPS1 S709A), the recycling of transferrin receptor (TfR) was attenuated compared to the cells reconstituted with wild type REPS1. Collectively, we suggested that the phosphorylation of REPS1 at S709 by RSK may have a role of the trafficking of TfR.
Collapse
Affiliation(s)
- Seong Heon Kim
- Department of Functional Genomics, KRIBB School of Biological Science, Korea University of Science and Technology, Daejeon 34113, Korea
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
| | - Jin-hwa Cho
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
| | - Bi-Oh Park
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Korea
| | - Byoung Chul Park
- Department of Functional Genomics, KRIBB School of Biological Science, Korea University of Science and Technology, Daejeon 34113, Korea
- Department of Proteome Structural Biology, KRIBB School of Biological Science, Korea University of Science and Technology, Daejeon 34113, Korea
| | - Jeong-Hoon Kim
- Department of Functional Genomics, KRIBB School of Biological Science, Korea University of Science and Technology, Daejeon 34113, Korea
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
| | - Sung Goo Park
- Department of Functional Genomics, KRIBB School of Biological Science, Korea University of Science and Technology, Daejeon 34113, Korea
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
| | - Sunhong Kim
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
- Department of Biomolecular Science, KRIBB School of Biological Science, Korea University of Science and Technology, Daejeon 34113, Korea
| |
Collapse
|
5
|
Kim SH, Park BO, Kim K, Park BC, Park SG, Kim JH, Kim S. Sjögren Syndrome antigen B regulates LIN28-let-7 axis in Caenorhabditis elegans and human. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2021; 1864:194684. [PMID: 33484878 DOI: 10.1016/j.bbagrm.2021.194684] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 01/05/2021] [Accepted: 01/13/2021] [Indexed: 02/06/2023]
Abstract
LIN28 protein and let-7 family micro RNAs (miRNAs) that are an evolutionarily conserved from nematodes to humans are the important regulators of developmental timing by dynamically interacting with each other. However, regulators of LIN28 remain largely elusive. Here, we show the evidences that Sjögren Syndrome antigen B (SSB) protein associates and cooperates with LIN28A and LIN28B, mammalian orthologues of Caenorhabditis elegans lin-28, proteins in the nucleus. Knockdown of SSB in HEK293 cell line resulted in the decrease of the amount of LIN28B mRNAs and proteins, and the increase of the level of mature let-7 miRNAs. Furthermore, RNA interference of ssb-1 gene, a worm SSB orthologue, was sufficient to cause a heterochronic defect in seam cells of C. elegans, recapitulating the phenotype of lin-28 downregulation. Collectively, we suggest that SSB is an important regulator for the LIN28-let-7 axis.
Collapse
Affiliation(s)
- Seong Heon Kim
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea; Department of Biomolecular Science, KRIBB School of Biological Science, Korea University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Bi-Oh Park
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea; College of Pharmacy, Chungbuk National University, Cheongju 34113, Republic of Korea
| | - Kidae Kim
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea; Department of Proteome Structural Biology, KRIBB School of Biological Science, Korea University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Byoung Chul Park
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea; Department of Proteome Structural Biology, KRIBB School of Biological Science, Korea University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Sung Goo Park
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea; Department of Functional Genomics, KRIBB School of Biological Science, Korea University of Science and Technology, Daejeon 34113, Republic of Korea.
| | - Jeong-Hoon Kim
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea; Department of Functional Genomics, KRIBB School of Biological Science, Korea University of Science and Technology, Daejeon 34113, Republic of Korea.
| | - Sunhong Kim
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea; Department of Biomolecular Science, KRIBB School of Biological Science, Korea University of Science and Technology, Daejeon 34113, Republic of Korea.
| |
Collapse
|
6
|
Hughes DC, Turner DC, Baehr LM, Seaborne RA, Viggars M, Jarvis JC, Gorski PP, Stewart CE, Owens DJ, Bodine SC, Sharples AP. Knockdown of the E3 ubiquitin ligase UBR5 and its role in skeletal muscle anabolism. Am J Physiol Cell Physiol 2021; 320:C45-C56. [PMID: 33052072 DOI: 10.1152/ajpcell.00432.2020] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
UBR5 is an E3 ubiquitin ligase positively associated with anabolism, hypertrophy, and recovery from atrophy in skeletal muscle. The precise mechanisms underpinning UBR5's role in the regulation of skeletal muscle mass remain unknown. The present study aimed to elucidate these mechanisms by silencing the UBR5 gene in vivo. To achieve this aim, we electroporated a UBR5-RNAi plasmid into mouse tibialis anterior muscle to investigate the impact of reduced UBR5 on anabolic signaling MEK/ERK/p90RSK and Akt/GSK3β/p70S6K/4E-BP1/rpS6 pathways. Seven days after UBR5 RNAi electroporation, although reductions in overall muscle mass were not detected, the mean cross-sectional area (CSA) of green fluorescent protein (GFP)-positive fibers were reduced (-9.5%) and the number of large fibers were lower versus the control. Importantly, UBR5-RNAi significantly reduced total RNA, muscle protein synthesis, ERK1/2, Akt, and GSK3β activity. Although p90RSK phosphorylation significantly increased, total p90RSK protein levels demonstrated a 45% reduction with UBR5-RNAi. Finally, these early events after 7 days of UBR5 knockdown culminated in significant reductions in muscle mass (-4.6%) and larger reductions in fiber CSA (-18.5%) after 30 days. This was associated with increased levels of phosphatase PP2Ac and inappropriate chronic elevation of p70S6K and rpS6 between 7 and 30 days, as well as corresponding reductions in eIF4e. This study demonstrates that UBR5 plays an important role in anabolism/hypertrophy, whereby knockdown of UBR5 culminates in skeletal muscle atrophy.
Collapse
Affiliation(s)
- David C Hughes
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Daniel C Turner
- Norwegian School of Sport Sciences (NiH), Institute for Physical Performance, Oslo, Norway
- School of Pharmacy and Bioengineering, Institute for Science & Technology in Medicine (ISTM), Keele University, Staffordshire, United Kingdom
- Stem Cells, Ageing and Molecular Physiology Unit (SCAMP), Research Institute for Sport & Exercise Sciences (RISES), Liverpool John Moores University, Liverpool, United Kingdom
| | - Leslie M Baehr
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Robert A Seaborne
- Stem Cells, Ageing and Molecular Physiology Unit (SCAMP), Research Institute for Sport & Exercise Sciences (RISES), Liverpool John Moores University, Liverpool, United Kingdom
- Centre for Genomics and Child Health, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Mark Viggars
- Stem Cells, Ageing and Molecular Physiology Unit (SCAMP), Research Institute for Sport & Exercise Sciences (RISES), Liverpool John Moores University, Liverpool, United Kingdom
| | - Jonathan C Jarvis
- Stem Cells, Ageing and Molecular Physiology Unit (SCAMP), Research Institute for Sport & Exercise Sciences (RISES), Liverpool John Moores University, Liverpool, United Kingdom
| | - Piotr P Gorski
- Norwegian School of Sport Sciences (NiH), Institute for Physical Performance, Oslo, Norway
- School of Pharmacy and Bioengineering, Institute for Science & Technology in Medicine (ISTM), Keele University, Staffordshire, United Kingdom
| | - Claire E Stewart
- Stem Cells, Ageing and Molecular Physiology Unit (SCAMP), Research Institute for Sport & Exercise Sciences (RISES), Liverpool John Moores University, Liverpool, United Kingdom
| | - Daniel J Owens
- Stem Cells, Ageing and Molecular Physiology Unit (SCAMP), Research Institute for Sport & Exercise Sciences (RISES), Liverpool John Moores University, Liverpool, United Kingdom
| | - Sue C Bodine
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Adam P Sharples
- Norwegian School of Sport Sciences (NiH), Institute for Physical Performance, Oslo, Norway
- School of Pharmacy and Bioengineering, Institute for Science & Technology in Medicine (ISTM), Keele University, Staffordshire, United Kingdom
- Stem Cells, Ageing and Molecular Physiology Unit (SCAMP), Research Institute for Sport & Exercise Sciences (RISES), Liverpool John Moores University, Liverpool, United Kingdom
| |
Collapse
|
7
|
Deubiquitinase OTUD5 is a positive regulator of mTORC1 and mTORC2 signaling pathways. Cell Death Differ 2020; 28:900-914. [PMID: 33110214 DOI: 10.1038/s41418-020-00649-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 10/12/2020] [Accepted: 10/14/2020] [Indexed: 12/11/2022] Open
Abstract
The mammalian Target of Rapamycin (mTOR) pathway regulates a variety of physiological processes, including cell growth and cancer progression. The regulatory mechanisms of these signals are extremely complex and comprise many feedback loops. Here, we identified the deubiquitinating enzyme ovarian tumor domain-containing protein 5 (OTUD5) as a novel positive regulator of the mTOR complex (mTORC) 1 and 2 signaling pathways. We demonstrated that OTUD5 stabilized β-transducin repeat-containing protein 1 (βTrCP1) proteins via its deubiquitinase (DUB) activity, leading to the degradation of Disheveled, Egl-10, and pleckstrin domain-containing mTOR-interacting protein (DEPTOR), which is an inhibitory protein of mTORC1 and 2. We also showed that mTOR directly phosphorylated OTUD5 and activated its DUB activity. RNA sequencing analysis revealed that OTUD5 regulates the downstream gene expression of mTOR. Additionally, OTUD5 depletion elicited several mTOR-related phenotypes such as decreased cell size and increased autophagy in mammalian cells as well as the suppression of a dRheb-induced curled wing phenotype by RNA interference of Duba, a fly ortholog of OTUD5, in Drosophila melanogaster. Furthermore, OTUD5 knockdown inhibited the proliferation of the cancer cell lines with mutations activating mTOR pathway. Our results suggested a positive feedback loop between OTUD5 and mTOR signaling pathway.
Collapse
|
8
|
Cammarata-Mouchtouris A, Nguyen XH, Acker A, Bonnay F, Goto A, Orian A, Fauvarque MO, Boutros M, Reichhart JM, Matt N. Hyd ubiquitinates the NF-κB co-factor Akirin to operate an effective immune response in Drosophila. PLoS Pathog 2020; 16:e1008458. [PMID: 32339205 PMCID: PMC7205318 DOI: 10.1371/journal.ppat.1008458] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 05/07/2020] [Accepted: 03/04/2020] [Indexed: 02/07/2023] Open
Abstract
The Immune Deficiency (IMD) pathway in Drosophila melanogaster is activated upon microbial challenge with Gram-negative bacteria to trigger the innate immune response. In order to decipher this nuclear factor κB (NF-κB) signaling pathway, we undertook an in vitro RNAi screen targeting E3 ubiquitin ligases specifically and identified the HECT-type E3 ubiquitin ligase Hyperplastic discs (Hyd) as a new actor in the IMD pathway. Hyd mediated Lys63 (K63)-linked polyubiquitination of the NF-κB cofactor Akirin was required for efficient binding of Akirin to the NF-κB transcription factor Relish. We showed that this Hyd-dependent interaction was required for the transcription of immunity-related genes that are activated by both Relish and Akirin but was dispensable for the transcription of genes that depend solely on Relish. Therefore Hyd is key in NF-κB transcriptional selectivity downstream of the IMD pathway. Drosophila depleted of Akirin or Hyd failed to express the full set of genes encoding immune-induced anti-microbial peptides and succumbed to immune challenges. We showed further that UBR5, the mammalian homolog of Hyd, was also required downstream of the NF-κB pathway for the activation of Interleukin 6 (IL6) transcription by LPS or IL-1β in cultured human cells. Our findings link the action of an E3 ubiquitin ligase to the activation of immune effector genes, deepening our understanding of the involvement of ubiquitination in inflammation and identifying a potential target for the control of inflammatory diseases. Ubiquitination has been recently identified in pathogenesis and progression of various diseases where inflammation is critical. NF-κB transcription factors are key actors in the transcriptional cascade leading to inflammation as they activate genes with pro- or anti-inflammatory activities. The similarity between the immune pathways in flies and mammals makes Drosophila melanogaster an excellent model to study the innate response. Accordingly, we decided to identify E3 ubiquitin-ligases involved in the regulation of NF-κB pathway, using Drosophila as a model system. A RNAi based screen in immortalized embryonic macrophage-like Drosophila cells points to the HECT-E3 ubiquitin ligase Hyd as a new regulator of the Immune-deficiency (IMD) NF-κB pathway, activated after Gram-negative immune challenge. More precisely, we showed that Hyd acts at the level of Akirin, an evolutionarily conserved player in the NF-κB pathway, required for the transcription of pro-inflammatory genes, but not for the NF-κB-dependent genes contributing to the down-regulation of inflammation. In addition, we could show that the human homologue of Hyd (UBR5) acts genetically at the level of human AKIRIN2, pointing to a unique dichotomy between Hyd/Akirin-dependent and -independent gene activation, allowing for the decoupling activation and resolution of inflammation. These results identified UBR5 as a putative target for anti-inflammatory compounds.
Collapse
Affiliation(s)
| | - Xuan-Hung Nguyen
- Vinmec Research Institute of Stem Cell and Gene Technology (VRISG) and College of Health Sciences, VinUniversity Hanoi, Vietnam
| | - Adrian Acker
- Université de Strasbourg, CNRS, M3I UPR 9022, Strasbourg, France
| | - François Bonnay
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna, Austria
| | - Akira Goto
- Université de Strasbourg, CNRS, M3I UPR 9022, Strasbourg, France
| | - Amir Orian
- Rappaport Research Institute and Rappaport Faculty of Medicine, Technion Integrated Cancer Center, Technion—Israel Institute of Technology, Haifa, Israel
| | | | - Michael Boutros
- Division of Signaling and Functional Genomics, German Cancer Research Center (DKFZ), and Department for Cell and Molecular Biology, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | | | - Nicolas Matt
- Université de Strasbourg, CNRS, M3I UPR 9022, Strasbourg, France
- * E-mail:
| |
Collapse
|
9
|
Ji H, Yi Q, Chen L, Wong L, Liu Y, Xu G, Zhao J, Huang T, Li B, Yang Y, Li W, Han L, Duan S. Circulating miR-3197 and miR-2116-5p as novel biomarkers for diabetic retinopathy. Clin Chim Acta 2019; 501:147-153. [PMID: 31678272 DOI: 10.1016/j.cca.2019.10.036] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 10/21/2019] [Accepted: 10/24/2019] [Indexed: 12/13/2022]
Abstract
Diabetic retinopathy (DR) is the leading cause of vision loss among older adults. The goal of this case-control study was to identify circulating miRNAs for the diagnosis of DR. The miRNeasy Serum/Plasma Kit was used to extract serum miRNAs. The μParaflo™ MicroRNA microarray was used to detect the expression levels of the miRNAs. The miRWalk algorithm was applied to predict the target genes of the miRNAs, which were further confirmed by the dual luciferase reporter gene system in HEK293T cells. A microarray was performed between 5 DR cases and 5 age-, sex-, body mass index-, and duration of diabetes-matched type 2 diabetic (T2DM) controls. The quantitative reverse transcription polymerase chain reaction technique was used to validate the differentially expressed circulating miRNAs in 45 DR cases and 45 well-matched controls. Receiver operating characteristic (ROC) curve analysis was used to evaluate the performance of the circulating miRNAs as diagnostic biomarkers for DR. Our microarray analysis screened out miR-2116-5p and miR-3197 as significantly up-regulated in DR cases compared with the controls. Furthermore, two miRNAs were validated in the 45 DR cases and 45 controls. The ROC analysis suggested that both miR-3197 and miR-2116-5p distinguished DR cases from controls. An additional dual-luciferase reporter gene assay confirmed that notch homolog 2 (NOTCH2) was the target gene of miR-2116-5p. Both miR-3197 and miR-2116-5p were identified as promising diagnostic biomarkers for DR. Future research is still needed to explore the molecular mechanisms of miR-3197 and miR-2116-5p in the pathogenesis of DR.
Collapse
Affiliation(s)
- Huihui Ji
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, China; Departmant of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Quanyong Yi
- Ningbo Eye Hospital, Minan Road 855, Ningbo, Zhejiang, China
| | - Lishuang Chen
- Ningbo Eye Hospital, Minan Road 855, Ningbo, Zhejiang, China
| | - Liping Wong
- Department of Social and Preventive Medicine, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Yanfen Liu
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Guodong Xu
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Jun Zhao
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Tianyi Huang
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Bin Li
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Yong Yang
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Wenxia Li
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Liyuan Han
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, China.
| | - Shiwei Duan
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang, China.
| |
Collapse
|
10
|
Saez I, Gerbracht JV, Koyuncu S, Lee HJ, Horn M, Kroef V, Denzel MS, Dieterich C, Gehring NH, Vilchez D. The E3 ubiquitin ligase UBR5 interacts with the H/ACA ribonucleoprotein complex and regulates ribosomal RNA biogenesis in embryonic stem cells. FEBS Lett 2019; 594:175-188. [PMID: 31365120 DOI: 10.1002/1873-3468.13559] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 07/24/2019] [Accepted: 07/26/2019] [Indexed: 12/27/2022]
Abstract
UBR5 is an E3 ubiquitin ligase involved in distinct processes such as transcriptional regulation and development. UBR5 is highly upregulated in embryonic stem cells (ESCs), whereas its expression decreases with differentiation, suggesting a role for UBR5 in ESC function. However, little is known about how UBR5 regulates ESC identity. Here, we define the protein interactome of UBR5 in ESCs and find interactions with distinct components of the H/ACA ribonucleoprotein complex, which is required for proper maturation of ribosomal RNA (rRNA). Notably, loss of UBR5 induces an abnormal accumulation of rRNA processing intermediates, resulting in diminished ribosomal levels. Consequently, lack of UBR5 triggers an increase in p53 levels and a concomitant decrease in cellular proliferation rates. Thus, our results indicate a link between UBR5 and rRNA maturation.
Collapse
Affiliation(s)
- Isabel Saez
- Institute for Genetics and Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Germany
| | | | - Seda Koyuncu
- Institute for Genetics and Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Germany
| | - Hyun Ju Lee
- Institute for Genetics and Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Germany
| | - Moritz Horn
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Virginia Kroef
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Martin S Denzel
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Christoph Dieterich
- Section of Bioinformatics and Systems Cardiology, Department of Internal Medicine III and Klaus, Tschira Institute for Computational Cardiology, University Hospital, Heidelberg, Germany
| | - Niels H Gehring
- Institute for Genetics, Department of Biology, University of Cologne, Germany
| | - David Vilchez
- Institute for Genetics and Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Germany
| |
Collapse
|
11
|
Seaborne RA, Hughes DC, Turner DC, Owens DJ, Baehr LM, Gorski P, Semenova EA, Borisov OV, Larin AK, Popov DV, Generozov EV, Sutherland H, Ahmetov II, Jarvis JC, Bodine SC, Sharples AP. UBR5 is a novel E3 ubiquitin ligase involved in skeletal muscle hypertrophy and recovery from atrophy. J Physiol 2019; 597:3727-3749. [PMID: 31093990 DOI: 10.1113/jp278073] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 05/10/2019] [Indexed: 01/03/2023] Open
Abstract
KEY POINTS We have recently identified that a HECT domain E3 ubiquitin ligase, named UBR5, is altered epigenetically (via DNA methylation) after human skeletal muscle hypertrophy, where its gene expression is positively correlated with increasing lean leg mass after training and retraining. In the present study we extensively investigate this novel and uncharacterised E3 ubiquitin ligase (UBR5) in skeletal muscle atrophy, recovery from atrophy and injury, anabolism and hypertrophy. We demonstrated that UBR5 was epigenetically altered via DNA methylation during recovery from atrophy. We also determined that UBR5 was alternatively regulated versus well characterised E3 ligases, MuRF1/MAFbx, at the gene expression level during atrophy, recovery from atrophy and hypertrophy. UBR5 also increased at the protein level during recovery from atrophy and injury, hypertrophy and during human muscle cell differentiation. Finally, in humans, genetic variations of the UBR5 gene were strongly associated with larger fast-twitch muscle fibres and strength/power performance versus endurance/untrained phenotypes. ABSTRACT We aimed to investigate a novel and uncharacterized E3 ubiquitin ligase in skeletal muscle atrophy, recovery from atrophy/injury, anabolism and hypertrophy. We demonstrated an alternate gene expression profile for UBR5 vs. well characterized E3-ligases, MuRF1/MAFbx, where, after atrophy evoked by continuous-low-frequency electrical-stimulation in rats, MuRF1/MAFbx were both elevated, yet UBR5 was unchanged. Furthermore, after recovery of muscle mass post TTX-induced atrophy in rats, UBR5 was hypomethylated and increased at the gene expression level, whereas a suppression of MuRF1/MAFbx was observed. At the protein level, we also demonstrated a significant increase in UBR5 after recovery of muscle mass from hindlimb unloading in both adult and aged rats, as well as after recovery from atrophy evoked by nerve crush injury in mice. During anabolism and hypertrophy, UBR5 gene expression increased following acute loading in three-dimensional bioengineered mouse muscle in vitro, and after chronic electrical stimulation-induced hypertrophy in rats in vivo, without increases in MuRF1/MAFbx. Additionally, UBR5 protein abundance increased following functional overload-induced hypertrophy of the plantaris muscle in mice and during differentiation of primary human muscle cells. Finally, in humans, genetic association studies (>700,000 single nucleotide polymorphisms) demonstrated that the A alleles of rs10505025 and rs4734621 single nucleotide polymorphisms in the UBR5 gene were strongly associated with larger cross-sectional area of fast-twitch muscle fibres and favoured strength/power vs. endurance/untrained phenotypes. Overall, we suggest that: (i) UBR5 comprises a novel E3 ubiquitin ligase that is inversely regulated to MuRF1/MAFbx; (ii) UBR5 is epigenetically regulated; and (iii) UBR5 is elevated at both the gene expression and protein level during recovery from skeletal muscle atrophy and hypertrophy.
Collapse
Affiliation(s)
- Robert A Seaborne
- Stem Cells, Ageing and Molecular Physiology Unit, Exercise Metabolism and Adaptation Research Group, Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, UK.,Institute for Science and Technology in Medicine (ISTM), School of Medicine, Keele University, Keele, UK.,Centre for Genomics and Child Health, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - David C Hughes
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Daniel C Turner
- Stem Cells, Ageing and Molecular Physiology Unit, Exercise Metabolism and Adaptation Research Group, Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, UK.,Institute for Science and Technology in Medicine (ISTM), School of Medicine, Keele University, Keele, UK
| | - Daniel J Owens
- Stem Cells, Ageing and Molecular Physiology Unit, Exercise Metabolism and Adaptation Research Group, Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, UK
| | - Leslie M Baehr
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Piotr Gorski
- Stem Cells, Ageing and Molecular Physiology Unit, Exercise Metabolism and Adaptation Research Group, Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, UK.,Institute for Science and Technology in Medicine (ISTM), School of Medicine, Keele University, Keele, UK
| | - Ekaterina A Semenova
- Department of Molecular Biology and Genetics, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Oleg V Borisov
- Department of Molecular Biology and Genetics, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia.,Institute for Genomic Statistics and Bioinformatics, University Hospital Bonn, Bonn, Germany
| | - Andrey K Larin
- Department of Molecular Biology and Genetics, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Daniil V Popov
- Laboratory of Exercise Physiology, Institute of Biomedical Problems of the Russian Academy of Sciences, Moscow, Russia
| | - Edward V Generozov
- Department of Molecular Biology and Genetics, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Hazel Sutherland
- Exercise Metabolism and Adaptation Research Group, Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, UK
| | - Ildus I Ahmetov
- Department of Molecular Biology and Genetics, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia.,Laboratory of Molecular Genetics, Kazan State Medical University, Kazan, Russia.,Department of Physical Education, Plekhanov Russian University of Economics, Moscow, Russia.,Exercise Metabolism and Adaptation Research Group, Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, UK
| | - Jonathan C Jarvis
- Exercise Metabolism and Adaptation Research Group, Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, UK
| | - Sue C Bodine
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Adam P Sharples
- Stem Cells, Ageing and Molecular Physiology Unit, Exercise Metabolism and Adaptation Research Group, Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, UK.,Institute for Science and Technology in Medicine (ISTM), School of Medicine, Keele University, Keele, UK
| |
Collapse
|