1
|
Bian Q, Chen Y, Zhang J, Du X, Zhou Y, Zhang Q, Ding C, Wei H, Fu B. Maternal natural killer cells drive neuroimmune disorders in offspring through aberrant secretion of extracellular granzyme B. Immunity 2025:S1074-7613(25)00191-8. [PMID: 40381623 DOI: 10.1016/j.immuni.2025.04.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 05/28/2024] [Accepted: 04/22/2025] [Indexed: 05/20/2025]
Abstract
Prenatal viral infections can lead to neurodevelopmental disorders in offspring. However, how viral-induced maternal perturbations impact fetal brain macrophages remains insufficiently clear. Here, we demonstrated that inflammatory decidual natural killer (NK) cells, triggered by viral infection-induced maternal immune activation, drove macrophage activation, neurodevelopmental disorders, and behavioral deficits in offspring. Extracellular granzyme B (GzmB), predominantly released by the maternal CD49a+ tissue-resident NK subset under type I interferon stimulation, crossed the maternal-fetal barrier and promoted interferon-stimulated gene (ISG)-expressing fetal macrophage accumulation and microglial activation. Targeting extracellular GzmB through the systemic administration of the serine protease inhibitor Serpina3n or genetic ablation of Gzmb in maternal NK cells mitigated neuroimmune disorders in the fetal brain. These findings suggest that exposure to a perturbed maternal milieu reprograms decidual NK cell immunity, disrupting fetal neuroimmune homeostasis and increasing offspring susceptibility to neurodevelopmental disorders later in life.
Collapse
Affiliation(s)
- Qiwu Bian
- National Key Laboratory of Immune Response and Immunotherapy, Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science & Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, Anhui, China
| | - Yihang Chen
- National Key Laboratory of Immune Response and Immunotherapy, Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science & Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, Anhui, China
| | - Jinghe Zhang
- National Key Laboratory of Immune Response and Immunotherapy, Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science & Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, Anhui, China
| | - Xianghui Du
- National Key Laboratory of Immune Response and Immunotherapy, Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science & Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, Anhui, China
| | - Yonggang Zhou
- National Key Laboratory of Immune Response and Immunotherapy, Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science & Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, Anhui, China
| | - Qiao Zhang
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, Human Phenome Institute, Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai 200433, China
| | - Chen Ding
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, Human Phenome Institute, Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai 200433, China
| | - Haiming Wei
- National Key Laboratory of Immune Response and Immunotherapy, Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science & Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, Anhui, China.
| | - Binqing Fu
- National Key Laboratory of Immune Response and Immunotherapy, Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science & Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, Anhui, China.
| |
Collapse
|
2
|
Matsuhisa K, Sato S, Kaneko M. Identification of E3 Ubiquitin Ligase Substrates Using Biotin Ligase-Based Proximity Labeling Approaches. Biomedicines 2025; 13:854. [PMID: 40299435 PMCID: PMC12024899 DOI: 10.3390/biomedicines13040854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 04/30/2025] Open
Abstract
Ubiquitylation is a post-translational modification originally identified as the first step in protein degradation by the ubiquitin-proteasome system. Ubiquitylation is also known to regulate many cellular processes without degrading the ubiquitylated proteins. Substrate proteins are specifically recognized and ubiquitylated by ubiquitin ligases. It is necessary to identify the substrates for each ubiquitin ligase to understand the physiological and pathological roles of ubiquitylation. Recently, a promiscuous mutant of a biotin ligase derived from Escherichia coli, BioID, and its variants have been utilized to analyze protein-protein interaction. In this review, we summarize the current knowledge regarding the molecular mechanisms underlying ubiquitylation, BioID-based approaches for interactome studies, and the application of BirA and its variants for the identification of ubiquitin ligase substrates.
Collapse
Affiliation(s)
- Koji Matsuhisa
- Lee Kong Chian School of Medicine, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore;
| | - Shinya Sato
- Department of Pharmacology and Therapeutic Innovation, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8521, Japan;
| | - Masayuki Kaneko
- Department of Pharmacology and Therapeutic Innovation, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8521, Japan;
| |
Collapse
|
3
|
Ahammed MS, Wang X. Promoting proteostasis by cAMP/PKA and cGMP/PKG. Trends Mol Med 2025; 31:224-239. [PMID: 39477759 PMCID: PMC11908951 DOI: 10.1016/j.molmed.2024.10.006] [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: 07/19/2024] [Revised: 09/23/2024] [Accepted: 10/09/2024] [Indexed: 11/06/2024]
Abstract
Proteasome functional insufficiency (PFI) is implicated in neurodegeneration and heart failure, where aberrant protein aggregation is common and impairs the ubiquitin (Ub)-proteasome system (UPS), exacerbating increased proteotoxic stress (IPTS) and creating a vicious circle. Breaking this circle represents a key to treating these diseases. Protein kinase (PK)-A and PKG can activate the proteasome and promote proteasomal degradation of misfolded proteins. PKA does so by phosphorylating Ser14-RPN6/PSMD11, but how PKG activates the proteasome remains unknown. Emerging evidence supports a strategy to treat diseases with IPTS by augmenting cAMP/PKA and cGMP/PKG. Conceivably, targeted activation of PKA and PKG at proteasome nanodomains would minimize the undesired effects from their actions on other targets. In this review, we discuss PKA and PKG regulation of proteostasis via the UPS.
Collapse
Affiliation(s)
- Md Salim Ahammed
- Division of Basic Biomedical Sciences, University of South Dakota Sanford School of Medicine, Vermillion, SD 57069, USA
| | - Xuejun Wang
- Division of Basic Biomedical Sciences, University of South Dakota Sanford School of Medicine, Vermillion, SD 57069, USA.
| |
Collapse
|
4
|
Ahmad S, Christova T, Pye M, Narimatsu M, Song S, Wrana JL, Attisano L. Small Extracellular Vesicles Promote Axon Outgrowth by Engaging the Wnt-Planar Cell Polarity Pathway. Cells 2025; 14:56. [PMID: 39791757 PMCID: PMC11720052 DOI: 10.3390/cells14010056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 12/14/2024] [Accepted: 12/17/2024] [Indexed: 01/30/2025] Open
Abstract
In neurons, the acquisition of a polarized morphology is achieved upon the outgrowth of a single axon from one of several neurites. Small extracellular vesicles (sEVs), such as exosomes, from diverse sources are known to promote neurite outgrowth and thus may have therapeutic potential. However, the effect of fibroblast-derived exosomes on axon elongation in neurons of the central nervous system under growth-permissive conditions remains unclear. Here, we show that fibroblast-derived sEVs promote axon outgrowth and a polarized neuronal morphology in mouse primary embryonic cortical neurons. Mechanistically, we demonstrate that the sEV-induced increase in axon outgrowth requires endogenous Wnts and core PCP components including Prickle, Vangl, Frizzled, and Dishevelled. We demonstrate that sEVs are internalized by neurons, colocalize with Wnt7b, and induce relocalization of Vangl2 to the distal axon during axon outgrowth. In contrast, sEVs derived from neurons or astrocytes do not promote axon outgrowth, while sEVs from activated astrocytes inhibit elongation. Thus, our data reveal that fibroblast-derived sEVs promote axon elongation through the Wnt-PCP pathway in a manner that is dependent on endogenous Wnts.
Collapse
Affiliation(s)
- Samar Ahmad
- Department of Biochemistry, Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada; (S.A.); (T.C.); (S.S.)
| | - Tania Christova
- Department of Biochemistry, Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada; (S.A.); (T.C.); (S.S.)
| | - Melanie Pye
- Center for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, ON M5G 1X5, Canada; (M.P.); (M.N.); (J.L.W.)
| | - Masahiro Narimatsu
- Center for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, ON M5G 1X5, Canada; (M.P.); (M.N.); (J.L.W.)
| | - Siyuan Song
- Department of Biochemistry, Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada; (S.A.); (T.C.); (S.S.)
| | - Jeffrey L. Wrana
- Center for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, ON M5G 1X5, Canada; (M.P.); (M.N.); (J.L.W.)
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1X5, Canada
| | - Liliana Attisano
- Department of Biochemistry, Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada; (S.A.); (T.C.); (S.S.)
| |
Collapse
|
5
|
Ambrozkiewicz MC, Lorenz S. Understanding ubiquitination in neurodevelopment by integrating insights across space and time. Nat Struct Mol Biol 2025; 32:14-22. [PMID: 39633012 DOI: 10.1038/s41594-024-01422-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 10/08/2024] [Indexed: 12/07/2024]
Abstract
Ubiquitination regulates a myriad of eukaryotic signaling cascades by modifying substrate proteins, thereby determining their functions and fates. In this perspective, we discuss current challenges in investigating the ubiquitin system in the developing brain. We foster the concept that ubiquitination pathways are spatiotemporally regulated and tightly intertwined with molecular and cellular transitions during neurogenesis and neural circuit assembly. Focusing on the neurologically highly relevant class of homologous to E6AP C-terminus (HECT) ubiquitin ligases, we propose cross-disciplinary translational approaches bridging state-of-the-art cell biology, proteomics, biochemistry, structural biology and neuroscience to dissect ubiquitination in neurodevelopment and its specific perturbations in brain diseases. We highlight that a comprehensive understanding of ubiquitin signaling in the brain may reveal new horizons in basic neuroscience and clinical applications.
Collapse
Affiliation(s)
- Mateusz C Ambrozkiewicz
- Institute of Cell Biology and Neurobiology, Research Group 'Proteostasis', Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, Berlin, Germany.
| | - Sonja Lorenz
- Max Planck Institute for Multidisciplinary Sciences, Research Group 'Ubiquitin Signaling Specificity', Am Fassberg 11, Göttingen, Germany.
| |
Collapse
|
6
|
Chen J, Dang YM, Liu MC, Gao L, Guan T, Hu A, Xiong L, Lin H. AMPK induces PIAS3 mediated SUMOylation of E3 ubiquitin ligase Smurf1 impairing osteogenic differentiation and traumatic heterotopic ossification. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119771. [PMID: 38844181 DOI: 10.1016/j.bbamcr.2024.119771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 05/04/2024] [Accepted: 05/27/2024] [Indexed: 06/15/2024]
Abstract
AMP-activated protein kinase (AMPK) is a typical sensor of intracellular energy metabolism. Our previous study revealed the role of activated AMPK in the suppression of osteogenic differentiation and traumatic heterotopic ossification, but the underlying mechanism remains poorly understood. The E3 ubiquitin ligase Smurf1 is a crucial regulator of osteogenic differentiation and bone formation. We report here that Smurf1 is primarily SUMOylated at a C-terminal lysine residue (K324), which enhances its activity, facilitating ALK2 proteolysis and subsequent bone morphogenetic protein (BMP) signaling pathway inhibition. Furthermore, SUMOylation of the SUMO E3 ligase PIAS3 and Smurf1 SUMOylation was suppressed during the osteogenic differentiation and traumatic heterotopic ossification. More importantly, we found that AMPK activation enhances the SUMOylation of Smurf1, which is mediated by PIAS3 and increases the association between PIAS3 and AMPK. Overall, our study revealed that Smurf1 can be SUMOylated by PIAS3, Furthermore, Smurf1 SUMOylation mediates osteogenic differentiation and traumatic heterotopic ossification through suppression of the BMP signaling pathway. This study revealed that promotion of Smurf1 SUMOylation by AMPK activation may be implicated in traumatic heterotopic ossification treatment.
Collapse
Affiliation(s)
- Jie Chen
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Yan-Miao Dang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Meng-Chao Liu
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Linqing Gao
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Tianshu Guan
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Anxin Hu
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Lixia Xiong
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Hui Lin
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330006, China.
| |
Collapse
|
7
|
Yagishita H, Go Y, Okamoto K, Arimura N, Ikegaya Y, Sasaki T. A method to analyze gene expression profiles from hippocampal neurons electrophysiologically recorded in vivo. Front Neurosci 2024; 18:1360432. [PMID: 38694898 PMCID: PMC11061373 DOI: 10.3389/fnins.2024.1360432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 03/26/2024] [Indexed: 05/04/2024] Open
Abstract
Hippocampal pyramidal neurons exhibit diverse spike patterns and gene expression profiles. However, their relationships with single neurons are not fully understood. In this study, we designed an electrophysiology-based experimental procedure to identify gene expression profiles using RNA sequencing of single hippocampal pyramidal neurons whose spike patterns were recorded in living mice. This technique involves a sequence of experiments consisting of in vivo juxtacellular recording and labeling, brain slicing, cell collection, and transcriptome analysis. We demonstrated that the expression levels of a subset of genes in individual hippocampal pyramidal neurons were significantly correlated with their spike burstiness, submillisecond-level spike rise times or spike rates, directly measured by in vivo electrophysiological recordings. Because this methodological approach can be applied across a wide range of brain regions, it is expected to contribute to studies on various neuronal heterogeneities to understand how physiological spike patterns are associated with gene expression profiles.
Collapse
Affiliation(s)
- Haruya Yagishita
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, Japan
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yasuhiro Go
- Graduate School of Information Science, University of Hyogo, Hyogo, Japan
- Department of System Neuroscience, Division of Behavioral Development, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi, Japan
- Cognitive Genomics Research Group, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Aichi, Japan
| | - Kazuki Okamoto
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
- Department of Neuroanatomy, Graduate School of Medicine, Juntendo University, Tokyo, Japan
- Department of Cell Biology and Neuroscience, Graduate School of Medicine, Juntendo University, Bunkyo, Tokyo, Japan
| | - Nariko Arimura
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Yuji Ikegaya
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
- Center for Information and Neural Networks, National Institute of Information and Communications Technology, Osaka, Japan
- Institute for AI and Beyond, The University of Tokyo, Tokyo, Japan
| | - Takuya Sasaki
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, Japan
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
8
|
Huang Z, Zhou L, Duan J, Qin S, Jiang J, Chen H, Wang K, Liu R, Yuan M, Tang X, Nice EC, Wei Y, Zhang W, Huang C. Oxidative Stress Promotes Liver Cancer Metastasis via RNF25-Mediated E-Cadherin Protein Degradation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306929. [PMID: 38286671 PMCID: PMC10987140 DOI: 10.1002/advs.202306929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 12/25/2023] [Indexed: 01/31/2024]
Abstract
Loss of E-cadherin (ECAD) is required in tumor metastasis. Protein degradation of ECAD in response to oxidative stress is found in metastasis of hepatocellular carcinoma (HCC) and is independent of transcriptional repression as usually known. Mechanistically, protein kinase A (PKA) senses oxidative stress by redox modification in its β catalytic subunit (PRKACB) at Cys200 and Cys344. The activation of PKA kinase activity subsequently induces RNF25 phosphorylation at Ser450 to initiate RNF25-catalyzed degradation of ECAD. Functionally, RNF25 repression induces ECAD protein expression and inhibits HCC metastasis in vitro and in vivo. Altogether, these results indicate that RNF25 is a critical regulator of ECAD protein turnover, and PKA is a necessary redox sensor to enable this process. This study provides some mechanistic insight into how oxidative stress-induced ECAD degradation promotes tumor metastasis of HCC.
Collapse
Affiliation(s)
- Zhao Huang
- Department of BiotherapyCancer Center and State Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - Li Zhou
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education)Institute for Viral HepatitisDepartment of Infectious DiseasesThe Second Affiliated HospitalChongqing Medical UniversityChongqing400016China
| | - Jiufei Duan
- Department of BiotherapyCancer Center and State Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - Siyuan Qin
- Department of BiotherapyCancer Center and State Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - Jingwen Jiang
- West China School of Public Health and West China Fourth HospitalSichuan UniversityChengdu610041China
| | - Haining Chen
- Colorectal Cancer CenterDepartment of General SurgeryWest China HospitalSichuan UniversityChengdu610041China
| | - Kui Wang
- West China School of Basic Medical Sciences & Forensic MedicineState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - Rui Liu
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesResearch Unit of Oral Carcinogenesis and ManagementChinese Academy of Medical SciencesWest China Hospital of StomatologySichuan UniversityChengdu610041China
| | - Minlan Yuan
- Mental Health Center and Psychiatric LaboratoryThe State Key Laboratory of BiotherapyWest China Biomedical Big Data CenterWest China Hospital of Sichuan UniversityChengdu610041China
| | - Xiangdong Tang
- Sleep Medicine CenterDepartment of Respiratory and Critical Care MedicineMental Health CenterTranslational Neuroscience CenterState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - Edouard C. Nice
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonVIC3167Australia
| | - Yuquan Wei
- Department of BiotherapyCancer Center and State Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
| | - Wei Zhang
- Frontiers Medical CenterTianfu Jincheng LaboratoryChengdu610212China
- Medical Big Data CenterSichuan UniversityChengdu610041China
| | - Canhua Huang
- Department of BiotherapyCancer Center and State Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengdu610041China
- Frontiers Medical CenterTianfu Jincheng LaboratoryChengdu610212China
| |
Collapse
|
9
|
Hughes DC, Goodman CA, Baehr LM, Gregorevic P, Bodine SC. A critical discussion on the relationship between E3 ubiquitin ligases, protein degradation, and skeletal muscle wasting: it's not that simple. Am J Physiol Cell Physiol 2023; 325:C1567-C1582. [PMID: 37955121 PMCID: PMC10861180 DOI: 10.1152/ajpcell.00457.2023] [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: 09/18/2023] [Revised: 11/07/2023] [Accepted: 11/07/2023] [Indexed: 11/14/2023]
Abstract
Ubiquitination is an important post-translational modification (PTM) for protein substrates, whereby ubiquitin is added to proteins through the coordinated activity of activating (E1), ubiquitin-conjugating (E2), and ubiquitin ligase (E3) enzymes. The E3s provide key functions in the recognition of specific protein substrates to be ubiquitinated and aid in determining their proteolytic or nonproteolytic fates, which has led to their study as indicators of altered cellular processes. MuRF1 and MAFbx/Atrogin-1 were two of the first E3 ubiquitin ligases identified as being upregulated in a range of different skeletal muscle atrophy models. Since their discovery, the expression of these E3 ubiquitin ligases has often been studied as a surrogate measure of changes to bulk protein degradation rates. However, emerging evidence has highlighted the dynamic and complex regulation of the ubiquitin proteasome system (UPS) in skeletal muscle and demonstrated that protein ubiquitination is not necessarily equivalent to protein degradation. These observations highlight the potential challenges of quantifying E3 ubiquitin ligases as markers of protein degradation rates or ubiquitin proteasome system (UPS) activation. This perspective examines the usefulness of monitoring E3 ubiquitin ligases for determining specific or bulk protein degradation rates in the settings of skeletal muscle atrophy. Specific questions that remain unanswered within the skeletal muscle atrophy field are also identified, to encourage the pursuit of new research that will be critical in moving forward our understanding of the molecular mechanisms that govern protein function and degradation in muscle.
Collapse
Affiliation(s)
- David C Hughes
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
| | - Craig A Goodman
- Centre for Muscle Research (CMR), Department of Anatomy and Physiology, The University of Melbourne, Parkville, Victoria, Australia
| | - Leslie M Baehr
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
| | - Paul Gregorevic
- Centre for Muscle Research (CMR), Department of Anatomy and Physiology, The University of Melbourne, Parkville, Victoria, Australia
- Department of Neurology, The University of Washington School of Medicine, Seattle, Washington, United States
| | - Sue C Bodine
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
| |
Collapse
|
10
|
Xu Y, Qu M, He Y, He Q, Shen T, Luo J, Tan D, Bao H, Xu C, Ji X, Hu X, Barkat MQ, Zeng LH, Wu X. Smurf1 polyubiquitinates on K285/K282 of the kinases Mst1/2 to attenuate their tumor-suppressor functions. J Biol Chem 2023; 299:105395. [PMID: 37890777 PMCID: PMC10696403 DOI: 10.1016/j.jbc.2023.105395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 09/29/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Sterile 20-like kinases Mst1 and Mst2 (Mst1/2) and large tumor suppressor 1/2 are core kinases to mediate Hippo signaling in maintaining tissue homeostasis. We have previously demonstrated that Smad ubiquitin (Ub) regulatory factor 1 (Smurf1), a HECT-type E3 ligase, ubiquitinates and in turn destabilizes large tumor suppressor 1/2 to induce the transcriptional output of Hippo signaling. Here, we unexpectedly find that Smurf1 interacts with and polyubiquitinates Mst1/2 by virtue of K27- and K29-linked Ub chains, resulting in the proteasomal degradation of Mst1/2 and attenuation of their tumor-suppressor functions. Among the potential Ub acceptor sites on Mst1/2, K285/K282 are conserved and essential for Smurf1-induced polyubiquitination and degradation of Mst1/2 as well as transcriptional output of Hippo signaling. As a result, K285R/K282R mutation of Mst1/2 not only negates the transcriptional output of Hippo signaling but enhances the tumor-suppressor functions of Mst1/2. Together, we demonstrate that Smurf1-mediated polyubiquitination on K285/K282 of Mst1/2 destabilizes Mst1/2 to attenuate their tumor-suppressor functions. Thus, the present study identifies Smurf1-mediated ubiquitination of Mst1/2 as a hitherto uncharacterized mechanism fine-tuning the Hippo signaling pathway and may provide additional targets for therapeutic intervention of diseases associated with this important pathway.
Collapse
Affiliation(s)
- Yana Xu
- Department of Orthopaedics, The Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, China
| | - Meiyu Qu
- Department of Orthopaedics, The Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Hangzhou City University School of Medicine, Hangzhou, China
| | - Yangxun He
- Department of Orthopaedics, The Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiangqiang He
- Department of Orthopaedics, The Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, China
| | - Tingyu Shen
- Department of Orthopaedics, The Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiahao Luo
- Department of Orthopaedics, The Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dan Tan
- Department of Orthopaedics, The Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hangyang Bao
- Department of Orthopaedics, The Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chengyun Xu
- Department of Orthopaedics, The Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xing Ji
- Department of Orthopaedics, The Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Hangzhou City University School of Medicine, Hangzhou, China
| | - Xinhua Hu
- Department of Clinical Pharmacology, The Affiliated Second Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Muhammad Qasim Barkat
- Department of Orthopaedics, The Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ling-Hui Zeng
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Hangzhou City University School of Medicine, Hangzhou, China.
| | - Ximei Wu
- Department of Orthopaedics, The Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
11
|
Wan JX, Wang YQ, Lan SN, Chen L, Feng MQ, Chen X. Research Progress in Function and Regulation of E3 Ubiquitin Ligase SMURF1. Curr Med Sci 2023; 43:855-868. [PMID: 37558865 DOI: 10.1007/s11596-023-2774-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 06/08/2023] [Indexed: 08/11/2023]
Abstract
Smad ubiquitylation regulatory factor 1 (Smurf1) is an important homologous member of E6-AP C-terminus type E3 ubiquitin ligase. Initially, Smurf1 was reportedly involved in the negative regulation of the bone morphogenesis protein (BMP) pathway. After further research, several studies have confirmed that Smurf1 is widely involved in various biological processes, such as bone homeostasis regulation, cell migration, apoptosis, and planar cell polarity. At the same time, recent studies have provided a deeper understanding of the regulatory mechanisms of Smurf1's expression, activity, and substrate selectivity. In our review, a brief summary of recent important biological functions and regulatory mechanisms of E3 ubiquitin ligase Smurf1 is proposed.
Collapse
Affiliation(s)
- Ji-Xi Wan
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yu-Qi Wang
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Si-Na Lan
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Liu Chen
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Ming-Qian Feng
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
- College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, 430070, China
| | - Xin Chen
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
12
|
Souza-Costa LP, Andrade-Chaves JT, Andrade JM, Costa VV, Franco LH. Uncovering new insights into the role of the ubiquitin ligase Smurf1 on the regulation of innate immune signaling and resistance to infection. Front Immunol 2023; 14:1185741. [PMID: 37228615 PMCID: PMC10203584 DOI: 10.3389/fimmu.2023.1185741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 03/28/2023] [Indexed: 05/27/2023] Open
Abstract
Innate immunity is the body's first line of defense against infections. Innate immune cells express pattern recognition receptors in distinct cellular compartments that are responsible to detect either pathogens-associated molecules or cellular components derived from damaged cells, to trigger intracellular signaling pathways that lead to the activation of inflammatory responses. Inflammation is essential to coordinate immune cell recruitment, pathogen elimination and to keep normal tissue homeostasis. However, uncontrolled, misplaced or aberrant inflammatory responses could lead to tissue damage and drive chronic inflammatory diseases and autoimmunity. In this context, molecular mechanisms that tightly regulate the expression of molecules required for the signaling of innate immune receptors are crucial to prevent pathological immune responses. In this review, we discuss the ubiquitination process and its importance in the regulation of innate immune signaling and inflammation. Then, we summarize the roles of Smurf1, a protein that works on ubiquitination, on the regulation of innate immune signaling and antimicrobial mechanisms, emphasizing its substrates and highlighting its potential as a therapeutic target for infectious and inflammatory conditions.
Collapse
Affiliation(s)
- Luiz Pedro Souza-Costa
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Josiane Teixeira Andrade-Chaves
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Juvana Moreira Andrade
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Vivian Vasconcelos Costa
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Luis Henrique Franco
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
13
|
Gu X, Jia C, Wang J. Advances in Understanding the Molecular Mechanisms of Neuronal Polarity. Mol Neurobiol 2023; 60:2851-2870. [PMID: 36738353 DOI: 10.1007/s12035-023-03242-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 01/22/2023] [Indexed: 02/05/2023]
Abstract
The establishment and maintenance of neuronal polarity are important for neural development and function. Abnormal neuronal polarity establishment commonly leads to a variety of neurodevelopmental disorders. Over the past three decades, with the continuous development and improvement of biological research methods and techniques, we have made tremendous progress in the understanding of the molecular mechanisms of neuronal polarity establishment. The activity of positive and negative feedback signals and actin waves are both essential in this process. They drive the directional transport and aggregation of key molecules of neuronal polarity, promote the spatiotemporal regulation of ordered and coordinated interactions of actin filaments and microtubules, stimulate the specialization and growth of axons, and inhibit the formation of multiple axons. In this review, we focus on recent advances in these areas, in particular the important findings about neuronal polarity in two classical models, in vitro primary hippocampal/cortical neurons and in vivo cortical pyramidal neurons, and discuss our current understanding of neuronal polarity..
Collapse
Affiliation(s)
- Xi Gu
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.
| | - Chunhong Jia
- Department of Pediatrics, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Junhao Wang
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| |
Collapse
|
14
|
Urrutia PJ, González-Billault C. A Role for Second Messengers in Axodendritic Neuronal Polarity. J Neurosci 2023; 43:2037-2052. [PMID: 36948585 PMCID: PMC10039749 DOI: 10.1523/jneurosci.1065-19.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 02/07/2023] [Accepted: 02/10/2023] [Indexed: 03/24/2023] Open
Abstract
Neuronal polarization is a complex molecular process regulated by intrinsic and extrinsic mechanisms. Nerve cells integrate multiple extracellular cues to generate intracellular messengers that ultimately control cell morphology, metabolism, and gene expression. Therefore, second messengers' local concentration and temporal regulation are crucial elements for acquiring a polarized morphology in neurons. This review article summarizes the main findings and current understanding of how Ca2+, IP3, cAMP, cGMP, and hydrogen peroxide control different aspects of neuronal polarization, and highlights questions that still need to be resolved to fully understand the fascinating cellular processes involved in axodendritic polarization.
Collapse
Affiliation(s)
- Pamela J Urrutia
- Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile 7800003
- School of Medical Technology, Faculty of Medicine and Science, Universidad San Sebastián, Santiago, Chile 7510157
| | - Christian González-Billault
- Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile 7800003
- Department of Neurosciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile 8380453
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile 7800003
- Buck Institute for Research on Aging, Novato, California 94945
| |
Collapse
|
15
|
Bone Tissue and the Nervous System: What Do They Have in Common? Cells 2022; 12:cells12010051. [PMID: 36611845 PMCID: PMC9818711 DOI: 10.3390/cells12010051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/12/2022] [Accepted: 12/16/2022] [Indexed: 12/25/2022] Open
Abstract
Degenerative diseases affecting bone tissues and the brain represent important problems with high socio-economic impact. Certain bone diseases, such as osteoporosis, are considered risk factors for the progression of neurological disorders. Often, patients with neurodegenerative diseases have bone fractures or reduced mobility linked to osteoarthritis. The bone is a dynamic tissue involved not only in movement but also in the maintenance of mineral metabolism. Bone is also associated with the generation of both hematopoietic stem cells (HSCs), and thus the generation of the immune system, and mesenchymal stem cells (MSCs). Bone marrow is a lymphoid organ and contains MSCs and HSCs, both of which are involved in brain health via the production of cytokines with endocrine functions. Hence, it seems clear that bone is involved in the regulation of the neuronal system and vice versa. This review summarizes the recent knowledge on the interactions between the nervous system and bone and highlights the importance of the interaction between nerve and bone cells. In addition, experimental models that study the interaction between nerve and skeletal cells are discussed, and innovative models are suggested to better evaluate the molecular interactions between these two cell types.
Collapse
|
16
|
The Twofold Role of Osteogenic Small Molecules in Parkinson's Disease Therapeutics: Crosstalk of Osteogenesis and Neurogenesis. BIOMED RESEARCH INTERNATIONAL 2022; 2022:3813541. [PMID: 36545269 PMCID: PMC9763015 DOI: 10.1155/2022/3813541] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/17/2022] [Accepted: 11/23/2022] [Indexed: 12/14/2022]
Abstract
Deemed one of the most problematic neurodegenerative diseases in the elderly population, Parkinson's disease remains incurable to date. Ongoing diagnostic studies, however, have revealed that a large number of small molecule drugs that trigger the BMP2-Smad signaling pathway with an osteogenic nature may be effective in Parkinson's disease treatment. Although BMP2 and Smad1, 3, and 5 biomolecules promote neurite outgrowth and neuroprotection in dopaminergic cells as well, small molecules are quicker at crossing the BBB and reaching the damaged dopaminergic neurons located in the substantia nigra due to a molecular weight less than 500 Da. It is worth noting that osteogenic small molecules that inhibit Smurf1 phosphorylation do not offer therapeutic opportunities for Parkinson's disease; whereas, osteogenic small molecules that trigger Smad1, 3, and 5 phosphorylation may have strong therapeutic implications in Parkinson's disease by increasing the survival rate of dopaminergic cells and neuritogenesis. Notably, from a different perspective, it might be said that osteogenic small molecules can possibly put forth therapeutic options for Parkinson's disease by improving neuritogenesis and cell survival.
Collapse
|
17
|
Li H, Lu Q, Li Y, Yan Y, Yin Z, Guo J, Xu W. Smurf participates in Helicoverpa armigera diapause by regulating the transforming growth factor-β signaling pathway. INSECT SCIENCE 2022; 29:1251-1261. [PMID: 35064956 DOI: 10.1111/1744-7917.13007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/28/2021] [Accepted: 01/07/2022] [Indexed: 06/14/2023]
Abstract
Diapause, an important strategy used by insects to avoid adverse environments, is regulated by various cell signaling pathways. The results of our previous studies demonstrated that the transforming growth factor-β (TGF-β) signaling pathway regulated pupal diapause in Helicoverpa armigera, which was accompanied by downregulation of proteins in TGF-β signaling. However, to date the mechanism underlying this phenomenon remains unknown. Here, we cloned the E3 ubiquitin ligases gene Smurf. In vitro experiments showed that Smurf directly bound to TGF-β receptor type I (TGFβRI) and Smad2. Overexpressing Smurf promoted ubiquitination of TGFβRI and Smad2, thereby downregulating their protein levels. Conversely, silencing of the Smurf gene suppressed ubiquitination of TGFβRI and Smad2 thereby increasing their protein levels. Results from in vivo co-immunoprecipitation assays revealed that the binding of Smurf to TGFβRI or Smad2 was stronger in diapause pupae than in nondiapause pupae. Injection of Smurf inhibitor A01 into diapause pupae markedly upregulated expression of TGFβRI and Smad2 proteins, leading to resumption of development in diapause pupae. Taken together, these findings suggested that ubiquitin ligase E3 Smurf participated in H. armigera diapause by regulating TGF-β signaling, and thus could be playing a crucial role in insect diapause.
Collapse
Affiliation(s)
- Haiyin Li
- Guizhou Provincial Key Laboratory for Agricultural Pest Management of the Mountainous Region, Institute of Entomology, Guizhou University, Guiyang, China
| | - Qin Lu
- Guizhou Provincial Key Laboratory for Agricultural Pest Management of the Mountainous Region, Institute of Entomology, Guizhou University, Guiyang, China
| | - Yan Li
- Guizhou Provincial Key Laboratory for Agricultural Pest Management of the Mountainous Region, Institute of Entomology, Guizhou University, Guiyang, China
| | - Yufang Yan
- Guizhou Provincial Key Laboratory for Agricultural Pest Management of the Mountainous Region, Institute of Entomology, Guizhou University, Guiyang, China
| | - Zhiyong Yin
- Guizhou Provincial Key Laboratory for Agricultural Pest Management of the Mountainous Region, Institute of Entomology, Guizhou University, Guiyang, China
| | - Jianjun Guo
- Guizhou Provincial Key Laboratory for Agricultural Pest Management of the Mountainous Region, Institute of Entomology, Guizhou University, Guiyang, China
| | - Weihua Xu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
18
|
Yu Y, Portolés S, Ren Y, Sun G, Wang XF, Zhang H, Guo S. The key clock component ZEITLUPE (ZTL) negatively regulates ABA signaling by degradation of CHLH in Arabidopsis. FRONTIERS IN PLANT SCIENCE 2022; 13:995907. [PMID: 36176682 PMCID: PMC9513469 DOI: 10.3389/fpls.2022.995907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 08/18/2022] [Indexed: 06/16/2023]
Abstract
Ubiquitination-mediated protein degradation plays important roles in ABA signal transduction and delivering responses to chloroplast stress signals in plants, but additional E3 ligases of protein ubiquitination remain to be identified to understand the complex signaling network. Here we reported that ZEITLUPE (ZTL), an F-box protein, negatively regulates abscisic acid (ABA) signaling during ABA-inhibited early seedling growth and ABA-induced stomatal closure in Arabidopsis thaliana. Using molecular biology and biochemistry approaches, we demonstrated that ZTL interacts with and ubiquitinates its substrate, CHLH/ABAR (Mg-chelatase H subunit/putative ABA receptor), to modulate CHLH stability via the 26S proteasome pathway. CHLH acts genetically downstream of ZTL in ABA and drought stress signaling. Interestingly, ABA conversely induces ZTL phosphorylation, and high levels of ABA also induce CHLH proteasomal degradation, implying that phosphorylated ZTL protein may enhance the affinity to CHLH, leading to the increased degradation of CHLH after ABA treatment. Taken together, our results revealed a possible mechanism of reciprocal regulation between ABA signaling and the circadian clock, which is thought to be essential for plant fitness and survival.
Collapse
Affiliation(s)
- Yongtao Yu
- National Watermelon and Melon Improvement Center, Beijing Academy of Agriculture and Forestry Sciences, Key Laboratory of Biology and Genetic Improvement of Horticultural Crops (North China), Beijing Key Laboratory of Vegetable Germplasm Improvement, Beijing, China
| | - Sergi Portolés
- MOE Key Lab of Bioinformatics, Center for Plant Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Yi Ren
- National Watermelon and Melon Improvement Center, Beijing Academy of Agriculture and Forestry Sciences, Key Laboratory of Biology and Genetic Improvement of Horticultural Crops (North China), Beijing Key Laboratory of Vegetable Germplasm Improvement, Beijing, China
| | - Guangyu Sun
- Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, College of Life Sciences, Northeast Forestry University, Harbin, Heilongjiang, China
| | - Xiao-Fang Wang
- MOE Key Lab of Bioinformatics, Center for Plant Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Huihui Zhang
- Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, College of Life Sciences, Northeast Forestry University, Harbin, Heilongjiang, China
| | - Shaogui Guo
- National Watermelon and Melon Improvement Center, Beijing Academy of Agriculture and Forestry Sciences, Key Laboratory of Biology and Genetic Improvement of Horticultural Crops (North China), Beijing Key Laboratory of Vegetable Germplasm Improvement, Beijing, China
| |
Collapse
|
19
|
Khan TA, Guo A, Martin J, Te Chien C, Liu T, Szczurkowska J, Shelly M. Directed mechanisms for apical dendrite development during neuronal polarization. Dev Biol 2022; 490:110-116. [PMID: 35809631 DOI: 10.1016/j.ydbio.2022.07.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 06/09/2022] [Accepted: 07/01/2022] [Indexed: 12/18/2022]
Abstract
The development of the dendrite and the axon during neuronal polarization underlies the directed flow of information in the brain. Seminal studies on axon development have dominated the mechanistic analysis of neuronal polarization. These studies, many originating from examinations in cultured hippocampal and cortical neurons in vitro, have established a prevalent view that axon formation precedes and is necessary for neuronal polarization. There is also in vivo evidence supporting this view. Nevertheless, the establishment of bipolar polarity and the leading edge, and apical dendrite development in pyramidal neurons in vivo occur when axon formation is prevented. Furthermore, recent mounting evidence suggest that directed mechanisms might mediate bipolar polarity/leading process and subsequent apical dendrite development. In the presence of spatially directed extracellular cues in the developing brain, these events may operate independently of axon forming events. In this perspective we summarize evidence in support of these evolving views in neuronal polarization and highlight recent findings on dedicated mechanisms acting in apical dendrite development.
Collapse
Affiliation(s)
- Tamor A Khan
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794-5230, USA
| | - Alan Guo
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794-5230, USA
| | - Jacqueline Martin
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794-5230, USA
| | - Chia Te Chien
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794-5230, USA
| | - Tianrui Liu
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794-5230, USA
| | - Joanna Szczurkowska
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794-5230, USA
| | - Maya Shelly
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794-5230, USA.
| |
Collapse
|
20
|
Vignoli B, Canossa M. Perirhinal Cortex LTP Does Not Require Astrocyte BDNF-TrkB Signaling. Cells 2022; 11:1501. [PMID: 35563806 PMCID: PMC9103847 DOI: 10.3390/cells11091501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/21/2022] [Accepted: 04/26/2022] [Indexed: 11/16/2022] Open
Abstract
Neurons release and respond to brain-derived neurotrophic factor (BDNF) with bursts of brain activity. BDNF action is known to extend to peri-synaptic astrocytes, contributing to synaptic strengthening. This implies that astrocytes have a set of dynamic responses, some of which might be secondary to activation of the tropomyosin tyrosine kinase B (TrkB) receptor. Here, we assessed the contribution of BDNF to long-term synaptic potentiation (LTP), by specifically deleting TrkB in cortical astrocytes. TrkB deletion had no effect on LTP induction, stabilization and maintenance, indicating that TrkB signaling in astrocytes is extraneous to transducing BDNF activity for synaptic strengthening.
Collapse
Affiliation(s)
- Beatrice Vignoli
- Department of Physics, University of Trento, 38123 Povo (TN), Italy
| | - Marco Canossa
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123 Povo (TN), Italy;
| |
Collapse
|
21
|
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
|
22
|
Szczurkowska J, Guo A, Martin J, Lee SI, Martinez E, Chien CT, Khan TA, Singh R, Dadson D, Tran TS, Pautot S, Shelly M. Semaphorin3A/PlexinA3 association with the Scribble scaffold for cGMP increase is required for apical dendrite development. Cell Rep 2022; 38:110483. [PMID: 35294878 PMCID: PMC8994670 DOI: 10.1016/j.celrep.2022.110483] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 10/03/2021] [Accepted: 02/12/2022] [Indexed: 11/30/2022] Open
Abstract
The development of the apical dendrite from the leading process of the bipolar pyramidal neuron might be directed by spatially organized extrinsic cues acting on localized intrinsic determinants. The extracellular cues regulating apical dendrite polarization remain elusive. We show that leading process and apical dendrite development are directed by class III Semaphorins and mediated by a localized cGMP-synthesizing complex. The scaffolding protein Scribble that associates with the cGMP-synthesizing enzyme soluble guanylate cyclase (sGC) also associates with the Semaphorin3A (Sema3A) co-receptor PlexinA3. Deletion or knockdown of PlexinA3 and Sema3A or disruption of PlexinA3-Scribble association prevents Sema3A-mediated cGMP increase and causes defects in apical dendrite development. These manipulations also impair bipolar polarity and leading process establishment. Local cGMP elevation or sGC expression rescues the effects of PlexinA3 knockdown or PlexinA3-Scribble complex disruption. During neuronal polarization, leading process and apical dendrite development are directed by a scaffold that links Semaphorin cue to cGMP increase. Szczurkowska et al. show that spatially directed Sema3A may promote development of the leading process and the apical dendrite via the co-receptor PlexinA3 by orchestrating localized cGMP increase on the scaffold protein, Scribble, at the leading edge of developing pyramidal neurons.
Collapse
Affiliation(s)
- Joanna Szczurkowska
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY 11794, USA
| | - Alan Guo
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY 11794, USA
| | - Jacqueline Martin
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY 11794, USA
| | - Seong-Il Lee
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY 11794, USA
| | - Edward Martinez
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| | - Chia Te Chien
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY 11794, USA
| | - Tamor A Khan
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY 11794, USA
| | - Ravnit Singh
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY 11794, USA
| | - Doreen Dadson
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY 11794, USA
| | - Tracy S Tran
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| | | | - Maya Shelly
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY 11794, USA.
| |
Collapse
|
23
|
Chen JS, Jones CM, Igarashi MG, Ren L, Johnson AE, Gould KL. Localization of the ubiquitin ligase Dma1 to the fission yeast contractile ring is modulated by phosphorylation. FEBS Lett 2021; 595:2781-2792. [PMID: 34674264 PMCID: PMC8721890 DOI: 10.1002/1873-3468.14211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/13/2021] [Accepted: 10/14/2021] [Indexed: 11/06/2022]
Abstract
The timing of cytokinesis relative to other mitotic events in the fission yeast Schizosaccharomyces pombe is controlled by the septation initiation network (SIN). During a mitotic checkpoint, the SIN is inhibited by the E3 ubiquitin ligase Dma1 to prevent chromosome mis-segregation. Dma1 dynamically localizes to spindle pole bodies (SPBs) and the contractile ring (CR) during mitosis, though its role at the CR is unknown. Here, we examined whether Dma1 phosphorylation affects its localization or function. We found that preventing Dma1 phosphorylation by substituting the six phosphosites with alanines diminished its CR localization but did not affect its mitotic checkpoint function. These studies reinforce the conclusion that Dma1 localization to the SPB is key to its role in the mitotic checkpoint.
Collapse
Affiliation(s)
- Jun-Song Chen
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37240
| | | | - Maya G. Igarashi
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37240
| | - Liping Ren
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37240
| | | | - Kathleen L. Gould
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37240
| |
Collapse
|
24
|
The Moringin/α-CD Pretreatment Induces Neuroprotection in an In Vitro Model of Alzheimer's Disease: A Transcriptomic Study. Curr Issues Mol Biol 2021; 43:197-214. [PMID: 34073287 PMCID: PMC8929117 DOI: 10.3390/cimb43010017] [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: 04/06/2021] [Revised: 05/19/2021] [Accepted: 05/24/2021] [Indexed: 12/23/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder and represents the most common form of senile dementia. Autophagy and mitophagy are cellular processes that play a key role in the aggregation of β-amyloid (Aβ) and tau phosphorylation. As a consequence, impairment of these processes leads to the progression of AD. Thus, interest is growing in the search for new natural compounds, such as Moringin (MOR), with neuroprotective, anti-amyloidogenic, antioxidative, and anti-inflammatory properties that could be used for AD prevention. However, MOR appears to be poorly soluble and stable in water. To increase its solubility MOR was conjugated with α-cyclodextrin (MOR/α-CD). In this work, it was evaluated if MOR/α-CD pretreatment was able to exert neuroprotective effects in an AD in vitro model through the evaluation of the transcriptional profile by next-generation sequencing (NGS). To induce the AD model, retinoic acid-differentiated SH-SY5Y cells were exposed to Aβ1-42. The MOR/α-CD pretreatment reduced the expression of the genes which encode proteins involved in senescence, autophagy, and mitophagy processes. Additionally, MOR/α-CD was able to induce neuronal remodeling modulating the axon guidance, principally downregulating the Slit/Robo signaling pathway. Noteworthy, MOR/α-CD, modulating these important pathways, may induce neuronal protection against Aβ1-42 toxicity as demonstrated also by the reduction of cleaved caspase 3. These data indicated that MOR/α-CD could attenuate the progression of the disease and promote neuronal repair.
Collapse
|
25
|
Szczurkowska J, Lee SI, Guo A, Cwetsch AW, Khan T, Rao S, Walz G, Huber TB, Cancedda L, Pautot S, Shelly M. A Localized Scaffold for cGMP Increase Is Required for Apical Dendrite Development. Cell Rep 2021; 31:107519. [PMID: 32294442 PMCID: PMC7293895 DOI: 10.1016/j.celrep.2020.03.083] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 12/18/2019] [Accepted: 03/24/2020] [Indexed: 10/26/2022] Open
Abstract
Studies in cultured neurons have established that axon specification instructs neuronal polarization and is necessary for dendrite development. However, dendrite formation in vivo occurs when axon formation is prevented. The mechanisms promoting dendrite development remain elusive. We find that apical dendrite development is directed by a localized cyclic guanosine monophosphate (cGMP)-synthesizing complex. We show that the scaffolding protein Scribble associates with cGMP-synthesizing enzymes soluble-guanylate-cyclase (sGC) and neuronal nitric oxide synthase (nNOS). The Scribble scaffold is preferentially localized to and mediates cGMP increase in dendrites. These events are regulated by kinesin KifC2. Knockdown of Scribble, sGC-β1, or KifC2 or disrupting their associations prevents cGMP increase in dendrites and causes severe defects in apical dendrite development. Local cGMP elevation or sGC expression rescues the effects of Scribble knockdown on dendrite development, indicating that Scribble is an upstream regulator of cGMP. During neuronal polarization, dendrite development is directed by the Scribble scaffold that might link extracellular cues to localized cGMP increase.
Collapse
Affiliation(s)
- Joanna Szczurkowska
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY 11794-5230, USA
| | - Seong-Il Lee
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY 11794-5230, USA
| | - Alan Guo
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY 11794-5230, USA
| | - Andrzej W Cwetsch
- Local Micro-environment and Brain Development Laboratory, Istituto Italiano di Tecnologia, Genova, Italy; Università degli Studi di Genova, Genova, Italy
| | - Tanvir Khan
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY 11794-5230, USA
| | - Sneha Rao
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY 11794-5230, USA
| | - Gerd Walz
- Department of Medicine IV, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Tobias B Huber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Laura Cancedda
- Local Micro-environment and Brain Development Laboratory, Istituto Italiano di Tecnologia, Genova, Italy; Dulbecco Telethon Institute, Italy
| | | | - Maya Shelly
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY 11794-5230, USA.
| |
Collapse
|
26
|
Pinto MJ, Tomé D, Almeida RD. The Ubiquitinated Axon: Local Control of Axon Development and Function by Ubiquitin. J Neurosci 2021; 41:2796-2813. [PMID: 33789876 PMCID: PMC8018891 DOI: 10.1523/jneurosci.2251-20.2021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 01/15/2021] [Accepted: 01/22/2021] [Indexed: 02/01/2023] Open
Abstract
Ubiquitin tagging sets protein fate. With a wide range of possible patterns and reversibility, ubiquitination can assume many shapes to meet specific demands of a particular cell across time and space. In neurons, unique cells with functionally distinct axons and dendrites harboring dynamic synapses, the ubiquitin code is exploited at the height of its power. Indeed, wide expression of ubiquitination and proteasome machinery at synapses, a diverse brain ubiquitome, and the existence of ubiquitin-related neurodevelopmental diseases support a fundamental role of ubiquitin signaling in the developing and mature brain. While special attention has been given to dendritic ubiquitin-dependent control, how axonal biology is governed by this small but versatile molecule has been considerably less discussed. Herein, we set out to explore the ubiquitin-mediated spatiotemporal control of an axon's lifetime: from its differentiation and growth through presynaptic formation, function, and pruning.
Collapse
Affiliation(s)
- Maria J Pinto
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, 3004-504, Portugal
| | - Diogo Tomé
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, 3004-504, Portugal
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro, 3810-193, Portugal
| | - Ramiro D Almeida
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, 3004-504, Portugal
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro, 3810-193, Portugal
| |
Collapse
|
27
|
Yang L, Zhou W, Lin H. Posttranslational Modifications of Smurfs: Emerging Regulation in Cancer. Front Oncol 2021; 10:610663. [PMID: 33718111 PMCID: PMC7950759 DOI: 10.3389/fonc.2020.610663] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 12/30/2020] [Indexed: 12/14/2022] Open
Abstract
Smad ubiquitination regulatory factors (Smurfs) belong to the Nedd4 subfamily of HECT-type E3 ubiquitin ligases. Under normal situations, Smurfs are exactly managed by upstream regulators, and thereby strictly control tumor biological processes, including cell growth, differentiation, apoptosis, polarization, epithelial mesenchymal transition (EMT), and invasion. Disruption of Smurf activity has been implicated in cancer progression, and Smurf activity is controlled by a series of posttranslational modifications (PTMs), including phosphorylation, ubiquitination, neddylation, sumoylation, and methylation. The effect and function of Smurfs depend on PTMs and regulate biological processes. Specifically, these modifications regulate the functional expression of Smurfs by affecting protein degradation and protein interactions. In this review, we summarize the complexity and diversity of Smurf PTMs from biochemical and biological perspectives and highlight the understanding of their roles in cancer.
Collapse
Affiliation(s)
- Longtao Yang
- Second Clinical Medical School, Nanchang University, Nanchang, China
| | - Wenwen Zhou
- Second Clinical Medical School, Nanchang University, Nanchang, China
| | - Hui Lin
- Department of Pathophysiology, School of Basic Medical Sciences, Nanchang University, Nanchang, China
| |
Collapse
|
28
|
Asano K. Multipoint Recognition of Molecular Conformations with Organocatalysts for Asymmetric Synthetic Reactions. BULLETIN OF THE CHEMICAL SOCIETY OF JAPAN 2021. [DOI: 10.1246/bcsj.20200343] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Keisuke Asano
- Department of Material Chemistry, Graduate School of Engineering, Kyoto University, Kyotodaigaku-Katsura, Nishikyo, Kyoto 615-8510, Japan
| |
Collapse
|
29
|
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
|
30
|
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
|
31
|
TGFβ1 Induces Axonal Outgrowth via ALK5/PKA/SMURF1-Mediated Degradation of RhoA and Stabilization of PAR6. eNeuro 2020; 7:ENEURO.0104-20.2020. [PMID: 32887692 PMCID: PMC7540929 DOI: 10.1523/eneuro.0104-20.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 07/21/2020] [Accepted: 08/06/2020] [Indexed: 12/11/2022] Open
Abstract
Transforming growth factor (TGF)β1 has repeatedly been associated with axonal regeneration and recovery after injury to the CNS. We found TGFβ1 upregulated in the stroke-denervated mouse spinal cord after ischemic injury to the motor cortex as early as 4 d postinjury (dpi) and persisting up to 28 dpi. Given the potential role of TGFβ1 in structural plasticity and functional recovery after stroke highlighted in several published studies, we investigated its downstream signaling in an in vitro model of neurite outgrowth. We found that in this model, TGFβ1 rescues neurite outgrowth under growth inhibitory conditions via the canonical TGFβR2/ALK5 signaling axis. Thereby, protein kinase A (PKA)-mediated phosphorylation of the E3 ubiquitin ligase SMURF1 induces a switch of its substrate preference from PAR6 to the Ras homolog A (RhoA), in this way enhancing outgrowth on the level of the cytoskeleton. This proposed mechanism of TGFβ1 signaling could underly the observed increase in structural plasticity after stroke in vivo as suggested by the temporal and spatial expression of TGFβ1. In accordance with previous publications, this study corroborates the potential of TGFβ1 and associated signaling cascades as a target for future therapeutic interventions to enhance structural plasticity and functional recovery for stroke patients.
Collapse
|
32
|
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
|
33
|
Mechanisms of axon polarization in pyramidal neurons. Mol Cell Neurosci 2020; 107:103522. [PMID: 32653476 DOI: 10.1016/j.mcn.2020.103522] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 06/19/2020] [Accepted: 06/29/2020] [Indexed: 01/19/2023] Open
Abstract
Neurons are highly polarized cells that have specialized regions for synaptic input, the dendrites, and synaptic output, the axons. This polarity is critical for appropriate neural circuit formation and function. One of the central gaps in our knowledge is understanding how developing neurons initiate axon polarity. Given the critical nature of this polarity on neural circuit formation and function, neurons have evolved multiple mechanisms comprised of extracellular and intracellular cues that allow them to initiate and form axons. These mechanisms engage a variety of signaling cascades that provide positive and negative cues to ensure axon polarization. This review highlights our current knowledge of the molecular underpinnings of axon polarization in pyramidal neurons and their relevance to the development of the brain.
Collapse
|
34
|
Bhaskar S, Gowda J, Prasanna J, Kumar A. Does altering proteasomal activity and trafficking reduce the arborization mediated specific vulnerability of SNpc dopaminergic neurons of Parkinson's disease? Med Hypotheses 2020; 143:110062. [PMID: 32652429 DOI: 10.1016/j.mehy.2020.110062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 06/27/2020] [Indexed: 01/15/2023]
Abstract
Parkinson's disease (PD) is a late-onset degenerative neuronal disorder and stands second among the neurological disorders with 1% of the total world population being affected. The disease originates majorly due to compromised function of the dopaminergic (DA) neurons in the Substantia Nigra pars compacta (SNpc), but not the ventral tegmental area (VTA) region of the midbrain. The differential susceptibility for degeneration is majorly attributed to morphological, molecular, and electrophysiological heterogeneity existing in DA neurons of SNpc and VTA. Long-range axonal arborization and a higher number of synapses in SNpc DA neurons make it more vulnerable compared to VTA DA neurons. Studies have shown that a decrease in such axonal arborization places DA neurons at decreased risk in PD. The two well established underlying mechanisms are a) As arborization is an energy-demanding process, increased redistribution of mitochondria to the axonal terminals occurs to satisfy the bioenergetic requirement b) The stabilization of axon-promoting factors at the axonal tip is an essential component for enhancing the arborization process. Interfering with any of these two processes would probably alleviate the degeneration of SNpc DA neurons. To accomplish the decreased stability of arborizing factors and thereby increase the resilience of SNpc DA neurons, we hypothesize the activation of anterograde transport-dependent recruitment of proteasomes to axon terminals as one of the most favorable approaches. Understanding this putative avenue of enhancing proteasomal activity and migration to the axonal tip could provide insight into the progression of neurodegeneration in PD and possibly offer a novel therapeutic strategy.
Collapse
Affiliation(s)
- Smitha Bhaskar
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education, Allalsandra, Yelahanka, Bengaluru 560065, Karnataka, India
| | - Jeevan Gowda
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education, Allalsandra, Yelahanka, Bengaluru 560065, Karnataka, India
| | - Jyothi Prasanna
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education, Allalsandra, Yelahanka, Bengaluru 560065, Karnataka, India
| | - Anujith Kumar
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education, Allalsandra, Yelahanka, Bengaluru 560065, Karnataka, India.
| |
Collapse
|
35
|
Zheng C, Atlas E, Lee HMT, Jao SLJ, Nguyen KCQ, Hall DH, Chalfie M. Opposing effects of an F-box protein and the HSP90 chaperone network on microtubule stability and neurite growth in Caenorhabditis elegans. Development 2020; 147:dev189886. [PMID: 32467239 PMCID: PMC7328132 DOI: 10.1242/dev.189886] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 05/04/2020] [Indexed: 01/08/2023]
Abstract
Molecular chaperones often work collaboratively with the ubiquitylation-proteasome system (UPS) to facilitate the degradation of misfolded proteins, which typically safeguards cellular differentiation and protects cells from stress. In this study, however, we report that the Hsp70/Hsp90 chaperone machinery and an F-box protein, MEC-15, have opposing effects on neuronal differentiation, and that the chaperones negatively regulate neuronal morphogenesis and functions. Using the touch receptor neurons (TRNs) of Caenorhabditis elegans, we find that mec-15(-) mutants display defects in microtubule formation, neurite growth, synaptic development and neuronal functions, and that these defects can be rescued by the loss of Hsp70/Hsp90 chaperones and co-chaperones. MEC-15 probably functions in a Skp-, Cullin- and F-box- containing complex to degrade DLK-1, which is an Hsp90 client protein stabilized by the chaperones. The abundance of DLK-1, and likely other Hsp90 substrates, is fine-tuned by the antagonism between MEC-15 and the chaperones; this antagonism regulates TRN development, as well as synaptic functions of GABAergic motor neurons. Therefore, a balance between the UPS and the chaperones tightly controls neuronal differentiation.
Collapse
Affiliation(s)
- Chaogu Zheng
- School of Biological Sciences, The University of Hong Kong, Hong Kong SAR, China
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Emily Atlas
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Ho Ming Terence Lee
- School of Biological Sciences, The University of Hong Kong, Hong Kong SAR, China
| | | | - Ken C Q Nguyen
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - David H Hall
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Martin Chalfie
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| |
Collapse
|
36
|
Advances in defining signaling networks for the establishment of neuronal polarity. Curr Opin Cell Biol 2020; 63:76-87. [DOI: 10.1016/j.ceb.2019.12.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 12/22/2019] [Accepted: 12/24/2019] [Indexed: 12/18/2022]
|
37
|
Kullmann JA, Trivedi N, Howell D, Laumonnerie C, Nguyen V, Banerjee SS, Stabley DR, Shirinifard A, Rowitch DH, Solecki DJ. Oxygen Tension and the VHL-Hif1α Pathway Determine Onset of Neuronal Polarization and Cerebellar Germinal Zone Exit. Neuron 2020; 106:607-623.e5. [PMID: 32183943 DOI: 10.1016/j.neuron.2020.02.025] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 02/04/2020] [Accepted: 02/19/2020] [Indexed: 02/06/2023]
Abstract
Postnatal brain circuit assembly is driven by temporally regulated intrinsic and cell-extrinsic cues that organize neurogenesis, migration, and axo-dendritic specification in post-mitotic neurons. While cell polarity is an intrinsic organizer of morphogenic events, environmental cues in the germinal zone (GZ) instructing neuron polarization and their coupling during postnatal development are unclear. We report that oxygen tension, which rises at birth, and the von Hippel-Lindau (VHL)-hypoxia-inducible factor 1α (Hif1α) pathway regulate polarization and maturation of post-mitotic cerebellar granule neurons (CGNs). At early postnatal stages with low GZ vascularization, Hif1α restrains CGN-progenitor cell-cycle exit. Unexpectedly, cell-intrinsic VHL-Hif1α pathway activation also delays the timing of CGN differentiation, germinal zone exit, and migration initiation through transcriptional repression of the partitioning-defective (Pard) complex. As vascularization proceeds, these inhibitory mechanisms are downregulated, implicating increasing oxygen tension as a critical switch for neuronal polarization and cerebellar GZ exit.
Collapse
Affiliation(s)
- Jan A Kullmann
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps University of Marburg, 35032 Marburg, Germany
| | - Niraj Trivedi
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Danielle Howell
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Christophe Laumonnerie
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Vien Nguyen
- Department of Pediatrics and Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Shalini S Banerjee
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Daniel R Stabley
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Abbas Shirinifard
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - David H Rowitch
- Department of Pediatrics and Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Pediatrics and Wellcome Trust-MRC Stem Cell Institute, University of Cambridge, Hills Road, Cambridge CB2 0AN, UK
| | - David J Solecki
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
38
|
Scott JL, Frick CT, Johnson KA, Liu H, Yong SS, Varney AG, Wiest O, Stahelin RV. Molecular Analysis of Membrane Targeting by the C2 Domain of the E3 Ubiquitin Ligase Smurf1. Biomolecules 2020; 10:biom10020229. [PMID: 32033048 PMCID: PMC7072158 DOI: 10.3390/biom10020229] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 01/28/2020] [Accepted: 01/29/2020] [Indexed: 12/18/2022] Open
Abstract
SMAD ubiquitination regulatory factor 1 (Smurf1) is a Nedd4 family E3 ubiquitin ligase that regulates cell motility, polarity and TGFβ signaling. Smurf1 contains an N-terminal protein kinase C conserved 2 (C2) domain that targets cell membranes and is required for interactions with membrane-localized substrates such as RhoA. Here, we investigated the lipid-binding mechanism of Smurf1 C2, revealing a general affinity for anionic membranes in addition to a selective affinity for phosphoinositides (PIPs). We found that Smurf1 C2 localizes not only to the plasma membrane but also to negatively charged intracellular sites, acting as an anionic charge sensor and selective PIP-binding domain. Site-directed mutagenesis combined with docking/molecular dynamics simulations revealed that the Smurf1 C2 domain loop region primarily interacts with PIPs and cell membranes, as opposed to the β-surface cationic patch employed by other C2 domains. By depleting PIPs from the inner leaflet of the plasma membrane, we found that PIP binding is necessary for plasma membrane localization. Finally, we used a Smurf1 cellular ubiquitination assay to show that the amount of ubiquitin at the plasma membrane interface depends on the lipid-binding properties of Smurf1. This study shows the mechanism by which Smurf1 C2 targets membrane-based substrates and reveals a novel interaction for non-calcium-dependent C2 domains and membrane lipids.
Collapse
Affiliation(s)
- Jordan L. Scott
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA; (J.L.S.); (C.T.F.); (K.A.J.); (H.L.); (S.S.Y.); (A.G.V.); (O.W.)
| | - Cary T. Frick
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA; (J.L.S.); (C.T.F.); (K.A.J.); (H.L.); (S.S.Y.); (A.G.V.); (O.W.)
| | - Kristen A. Johnson
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA; (J.L.S.); (C.T.F.); (K.A.J.); (H.L.); (S.S.Y.); (A.G.V.); (O.W.)
| | - Haining Liu
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA; (J.L.S.); (C.T.F.); (K.A.J.); (H.L.); (S.S.Y.); (A.G.V.); (O.W.)
| | - Sylvia S. Yong
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA; (J.L.S.); (C.T.F.); (K.A.J.); (H.L.); (S.S.Y.); (A.G.V.); (O.W.)
| | - Allyson G. Varney
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA; (J.L.S.); (C.T.F.); (K.A.J.); (H.L.); (S.S.Y.); (A.G.V.); (O.W.)
| | - Olaf Wiest
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA; (J.L.S.); (C.T.F.); (K.A.J.); (H.L.); (S.S.Y.); (A.G.V.); (O.W.)
| | - Robert V. Stahelin
- Department of Medicinal Chemistry and Molecular Pharmacology and the Purdue Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
- Correspondence: ; Tel.: +1-765-494-4152
| |
Collapse
|
39
|
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
|
40
|
Abstract
Neurons develop polarity by the formation of specialized dendritic and axonal structural compartments. A new report now provides evidence that reveals how neurons regulate the initiation and further maintenance of axonal growth, challenging our currently held view of RhoA function in axogenesis.
Collapse
Affiliation(s)
- Anton Omelchenko
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ 08854, USA
| | - Bonnie L Firestein
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ 08854, USA.
| |
Collapse
|
41
|
Petrasek J, Erhartova D, Levine B. Protective Effect of SMAD-Specific E3 Ubiquitin Protein Ligase 1 in Alcoholic Steatohepatitis in Mice. Hepatol Commun 2019; 3:1450-1458. [PMID: 31701069 PMCID: PMC6824063 DOI: 10.1002/hep4.1427] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 08/26/2019] [Indexed: 12/11/2022] Open
Abstract
Excessive accumulation of lipids in the liver is crucial in the pathogenesis of alcoholic steatohepatitis and may be partly mediated by impaired degradation of lipid droplets by autophagy. The E3 ubiquitin ligase SMAD-specific E3 ubiquitin protein ligase 1 (SMURF1) regulates selective autophagy by ubiquitinating proteins on cargo destined for autophagic delivery to the lysosome for degradation. Here, we evaluated the role of SMURF1 in the regulation of hepatic lipid degradation in alcoholic steatohepatitis. In patients with severe alcoholic hepatitis, SMURF1 colocalized with lipid droplet membranes in liver explants. In a mouse model of alcoholic steatohepatitis, Smurf1 -/- mice fed an alcohol diet displayed increased hepatocyte accumulation of lipid droplets and triglycerides as well as more severe liver injury compared to wild-type mice. The increased severity of liver steatosis in alcohol-fed Smurf1 -/- mice was rescued by adeno-associated virus (AAV) serotype 8-mediated hepatic expression of wild-type Smurf1 protein but not by mutant Smurf1 proteins either lacking the catalytically active cysteine 699 required for ubiquitin transfer or the N-terminal C2 phospholipid membrane-binding domain. Conclusion: Smurf1 plays a protective role in the pathogenesis of alcoholic steatohepatitis through a mechanism that requires both its ubiquitin-ligase activity and C2 phospholipid-binding domains. These findings have implications for understanding the roles of ubiquitin ligases in fatty liver disease.
Collapse
Affiliation(s)
- Jan Petrasek
- Digestive and Liver Diseases Division and Center for Autophagy Research, Department of Internal MedicineUniversity of Texas Southwestern Medical CenterDallasTX
| | - Denisa Erhartova
- Institute for Clinical and Experimental MedicinePragueCzech Republic
| | - Beth Levine
- Howard Hughes Medical Institute and Center for Autophagy Research, Department of Internal MedicineUniversity of Texas Southwestern Medical CenterDallasTX
| |
Collapse
|
42
|
Dupraz S, Hilton BJ, Husch A, Santos TE, Coles CH, Stern S, Brakebusch C, Bradke F. RhoA Controls Axon Extension Independent of Specification in the Developing Brain. Curr Biol 2019; 29:3874-3886.e9. [PMID: 31679934 DOI: 10.1016/j.cub.2019.09.040] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 08/22/2019] [Accepted: 09/16/2019] [Indexed: 12/22/2022]
Abstract
The specification of an axon and its subsequent outgrowth are key steps during neuronal polarization, a prerequisite to wire the brain. The Rho-guanosine triphosphatase (GTPase) RhoA is believed to be a central player in these processes. However, its physiological role has remained undefined. Here, genetic loss- and gain-of-function experiments combined with time-lapse microscopy, cell culture, and in vivo analysis show that RhoA is not involved in axon specification but confines the initiation of neuronal polarization and axon outgrowth during development. Biochemical analysis and super-resolution microscopy together with molecular and pharmacological manipulations reveal that RhoA restrains axon growth by activating myosin-II-mediated actin arc formation in the growth cone to prevent microtubules from protruding toward the leading edge. Through this mechanism, RhoA regulates the duration of axon growth and pause phases, thus controlling the tightly timed extension of developing axons. Thereby, this work unravels physiologically relevant players coordinating actin-microtubule interactions during axon growth.
Collapse
Affiliation(s)
- Sebastian Dupraz
- Axonal Growth and Regeneration Group, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Building 99, 53127 Bonn, Germany
| | - Brett J Hilton
- Axonal Growth and Regeneration Group, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Building 99, 53127 Bonn, Germany
| | - Andreas Husch
- Axonal Growth and Regeneration Group, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Building 99, 53127 Bonn, Germany
| | - Telma E Santos
- Axonal Growth and Regeneration Group, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Building 99, 53127 Bonn, Germany
| | - Charlotte H Coles
- Axonal Growth and Regeneration Group, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Building 99, 53127 Bonn, Germany
| | - Sina Stern
- Axonal Growth and Regeneration Group, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Building 99, 53127 Bonn, Germany
| | - Cord Brakebusch
- Biotech Research & Innovation Centre, University of Copenhagen, Ole Maaløes Vej 5, 2200 Copenhagen, Denmark
| | - Frank Bradke
- Axonal Growth and Regeneration Group, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Building 99, 53127 Bonn, Germany.
| |
Collapse
|
43
|
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: 6] [Impact Index Per Article: 1.0] [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
|
44
|
Ubiquitination and Long Non-coding RNAs Regulate Actin Cytoskeleton Regulators in Cancer Progression. Int J Mol Sci 2019; 20:ijms20122997. [PMID: 31248165 PMCID: PMC6627692 DOI: 10.3390/ijms20122997] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 06/16/2019] [Accepted: 06/17/2019] [Indexed: 12/15/2022] Open
Abstract
Actin filaments are a major component of the cytoskeleton in eukaryotic cells and play an important role in cancer metastasis. Dynamics and reorganization of actin filaments are regulated by numerous regulators, including Rho GTPases, PAKs (p21-activated kinases), ROCKs (Rho-associated coiled-coil containing kinases), LIMKs (LIM domain kinases), and SSH1 (slingshot family protein phosphate 1). Ubiquitination, as a ubiquitous post-transcriptional modification, deceases protein levels of actin cytoskeleton regulatory factors and thereby modulates the actin cytoskeleton. There is increasing evidence showing cytoskeleton regulation by long noncoding RNAs (lncRNAs) in cancer metastasis. However, which E3 ligases are activated for the ubiquitination of actin-cytoskeleton regulators involved in tumor metastasis remains to be fully elucidated. Moreover, it is not clear how lncRNAs influence the expression of actin cytoskeleton regulators. Here, we summarize physiological and pathological mechanisms of lncRNAs and ubiquitination control mediators of actin cytoskeleton regulators which that are involved in tumorigenesis and tumor progression. Finally, we briefly discuss crosstalk between ubiquitination and lncRNA control mediators of actin-cytoskeleton regulators in cancer.
Collapse
|
45
|
Hakanen J, Ruiz-Reig N, Tissir F. Linking Cell Polarity to Cortical Development and Malformations. Front Cell Neurosci 2019; 13:244. [PMID: 31213986 PMCID: PMC6558068 DOI: 10.3389/fncel.2019.00244] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 05/16/2019] [Indexed: 01/23/2023] Open
Abstract
Cell polarity refers to the asymmetric distribution of signaling molecules, cellular organelles, and cytoskeleton in a cell. Neural progenitors and neurons are highly polarized cells in which the cell membrane and cytoplasmic components are compartmentalized into distinct functional domains in response to internal and external cues that coordinate polarity and behavior during development and disease. In neural progenitor cells, polarity has a prominent impact on cell shape and coordinate several processes such as adhesion, division, and fate determination. Polarity also accompanies a neuron from the beginning until the end of its life. It is essential for development and later functionality of neuronal circuitries. During development, polarity governs transitions between multipolar and bipolar during migration of postmitotic neurons, and directs the specification and directional growth of axons. Once reaching final positions in cortical layers, neurons form dendrites which become compartmentalized to ensure proper establishment of neuronal connections and signaling. Changes in neuronal polarity induce signaling cascades that regulate cytoskeletal changes, as well as mRNA, protein, and vesicle trafficking, required for synapses to form and function. Hence, defects in establishing and maintaining cell polarity are associated with several neural disorders such as microcephaly, lissencephaly, schizophrenia, autism, and epilepsy. In this review we summarize the role of polarity genes in cortical development and emphasize the relationship between polarity dysfunctions and cortical malformations.
Collapse
Affiliation(s)
- Janne Hakanen
- Université catholique de Louvain, Institute of Neuroscience, Developmental Neurobiology, Brussels, Belgium
| | - Nuria Ruiz-Reig
- Université catholique de Louvain, Institute of Neuroscience, Developmental Neurobiology, Brussels, Belgium
| | - Fadel Tissir
- Université catholique de Louvain, Institute of Neuroscience, Developmental Neurobiology, Brussels, Belgium
| |
Collapse
|
46
|
Cui CP, Zhang Y, Wang C, Yuan F, Li H, Yao Y, Chen Y, Li C, Wei W, Liu CH, He F, Liu Y, Zhang L. Dynamic ubiquitylation of Sox2 regulates proteostasis and governs neural progenitor cell differentiation. Nat Commun 2018; 9:4648. [PMID: 30405104 PMCID: PMC6220269 DOI: 10.1038/s41467-018-07025-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 10/05/2018] [Indexed: 12/31/2022] Open
Abstract
Sox2 is a key transcriptional factor for maintaining pluripotency of stem cells. Sox2 deficiency causes neurodegeneration and impairs neurogenesis. Although the transcriptional regulation of Sox2 has been extensively studied, the mechanisms that control Sox2 protein turnover are yet to be clarified. Here we show that the RING-finger ubiquitin ligase complex CUL4ADET1-COP1 and the deubiquitylase OTUD7B govern Sox2 protein stability during neural progenitor cells (NPCs) differentiation. Sox2 expression declines concordantly with OTUD7B and reciprocally with CUL4A and COP1 levels upon NPCs differentiation. COP1, as the substrate receptor, interacts directly with and ubiquitylates Sox2, while OTUD7B removes polyUb conjugates from Sox2 and increases its stability. COP1 knockdown stabilizes Sox2 and prevents differentiation, while OTUD7B knockdown destabilizes Sox2 and induces differentiation. Thus, CUL4ADET1-COP1 and OTUD7B exert opposite roles in regulating Sox2 protein stability at the post-translational level, which represents a critical regulatory mechanism involved in the maintenance and differentiation of NPCs. Sox2 regulates pluripotency in neural progenitor cells (NPC) but how protein stability affects this is unclear. Here, the authors identify changes in ubiquitylation of Sox2 (by CUL4A-DET1-COP1 ligase and OTUD7B deubiquitylase) as controlling protein stability and so the differentiation state of NPCs.
Collapse
Affiliation(s)
- Chun-Ping Cui
- State Key Laboratory of Proteomics, National Center of Protein Sciences (Beijing), Beijing Institute of Lifeomics, 100850, Beijing, China
| | - Yuan Zhang
- State Key Laboratory of Proteomics, National Center of Protein Sciences (Beijing), Beijing Institute of Lifeomics, 100850, Beijing, China
| | - Chanjuan Wang
- State Key Laboratory of Proteomics, National Center of Protein Sciences (Beijing), Beijing Institute of Lifeomics, 100850, Beijing, China
| | - Fang Yuan
- State Key Laboratory of Reproductive Medicine, Institute for Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Science, 100101, Beijing, China
| | - Hongchang Li
- State Key Laboratory of Proteomics, National Center of Protein Sciences (Beijing), Beijing Institute of Lifeomics, 100850, Beijing, China
| | - Yuying Yao
- State Key Laboratory of Proteomics, National Center of Protein Sciences (Beijing), Beijing Institute of Lifeomics, 100850, Beijing, China
| | - Yuhan Chen
- State Key Laboratory of Proteomics, National Center of Protein Sciences (Beijing), Beijing Institute of Lifeomics, 100850, Beijing, China
| | - Chunnan Li
- State Key Laboratory of Proteomics, National Center of Protein Sciences (Beijing), Beijing Institute of Lifeomics, 100850, Beijing, China
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| | - Cui Hua Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Fuchu He
- State Key Laboratory of Proteomics, National Center of Protein Sciences (Beijing), Beijing Institute of Lifeomics, 100850, Beijing, China
| | - Yan Liu
- State Key Laboratory of Reproductive Medicine, Institute for Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing, China. .,Institute for Stem Cell and Regeneration, Chinese Academy of Science, 100101, Beijing, China.
| | - Lingqiang Zhang
- State Key Laboratory of Proteomics, National Center of Protein Sciences (Beijing), Beijing Institute of Lifeomics, 100850, Beijing, China.
| |
Collapse
|
47
|
Essmann CL, Ryan KR, Elmi M, Bryon-Dodd K, Porter A, Vaughan A, McMullan R, Nurrish S. Activation of RHO-1 in cholinergic motor neurons competes with dopamine signalling to control locomotion. PLoS One 2018; 13:e0204057. [PMID: 30240421 PMCID: PMC6150489 DOI: 10.1371/journal.pone.0204057] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 08/31/2018] [Indexed: 12/14/2022] Open
Abstract
The small GTPase RhoA plays a crucial role in the regulation of neuronal signalling to generate behaviour. In the developing nervous system RhoA is known to regulate the actin cytoskeleton, however the effectors of RhoA-signalling in adult neurons remain largely unidentified. We have previously shown that activation of the RhoA ortholog (RHO-1) in C. elegans cholinergic motor neurons triggers hyperactivity of these neurons and loopy locomotion with exaggerated body bends. This is achieved in part through increased diacylglycerol (DAG) levels and the recruitment of the synaptic vesicle protein UNC-13 to synaptic release sites, however other pathways remain to be identified. Dopamine, which is negatively regulated by the dopamine re-uptake transporter (DAT), has a central role in modulating locomotion in both humans and C. elegans. In this study we identify a new pathway in which RHO-1 regulates locomotory behaviour by repressing dopamine signalling, via DAT-1, linking these two pathways together. We observed an upregulation of dat-1 expression when RHO-1 is activated and show that loss of DAT-1 inhibits the loopy locomotion phenotype caused by RHO-1 activation. Reducing dopamine signalling in dat-1 mutants through mutations in genes involved in dopamine synthesis or in the dopamine receptor DOP-1 restores the ability of RHO-1 to trigger loopy locomotion in dat-1 mutants. Taken together, we show that negative regulation of dopamine signalling via DAT-1 is necessary for the neuronal RHO-1 pathway to regulate locomotion.
Collapse
Affiliation(s)
- Clara L. Essmann
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Katie R. Ryan
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Muna Elmi
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Kimberley Bryon-Dodd
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Andrew Porter
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Andrew Vaughan
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Rachel McMullan
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Stephen Nurrish
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| |
Collapse
|
48
|
Hapak SM, Rothlin CV, Ghosh S. PAR3-PAR6-atypical PKC polarity complex proteins in neuronal polarization. Cell Mol Life Sci 2018; 75:2735-2761. [PMID: 29696344 PMCID: PMC11105418 DOI: 10.1007/s00018-018-2828-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 04/03/2018] [Accepted: 04/23/2018] [Indexed: 01/01/2023]
Abstract
Polarity is a fundamental feature of cells. Protein complexes, including the PAR3-PAR6-aPKC complex, have conserved roles in establishing polarity across a number of eukaryotic cell types. In neurons, polarity is evident as distinct axonal versus dendritic domains. The PAR3, PAR6, and aPKC proteins also play important roles in neuronal polarization. During this process, either aPKC kinase activity, the assembly of the PAR3-PAR6-aPKC complex or the localization of these proteins is regulated downstream of a number of signaling pathways. In turn, the PAR3, PAR6, and aPKC proteins control various effector molecules to establish neuronal polarity. Herein, we discuss the many signaling mechanisms and effector functions that have been linked to PAR3, PAR6, and aPKC during the establishment of neuronal polarity.
Collapse
Affiliation(s)
- Sophie M Hapak
- Department of Medicine, School of Medicine, University of Minnesota, 401 East River Parkway, Minneapolis, MN, 55455, USA.
| | - Carla V Rothlin
- Department of Immunobiology, School of Medicine, Yale University, 300 Cedar Street, New Haven, CT, 06520, USA
- Department of Pharmacology, School of Medicine, Yale University, 333 Cedar Street, New Haven, CT, 06520, USA
| | - Sourav Ghosh
- Department of Neurology, School of Medicine, Yale University, 300 George Street, New Haven, CT, 06511, USA
- Department of Pharmacology, School of Medicine, Yale University, 333 Cedar Street, New Haven, CT, 06520, USA
| |
Collapse
|
49
|
Chen Y, Huang Q, Liu W, Zhu Q, Cui CP, Xu L, Guo X, Wang P, Liu J, Dong G, Wei W, Liu CH, Feng Z, He F, Zhang L. Mutually exclusive acetylation and ubiquitylation of the splicing factor SRSF5 control tumor growth. Nat Commun 2018; 9:2464. [PMID: 29942010 PMCID: PMC6018636 DOI: 10.1038/s41467-018-04815-3] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 05/18/2018] [Indexed: 12/30/2022] Open
Abstract
Most tumor cells take up more glucose than normal cells. Splicing dysregulation is one of the molecular hallmarks of cancer. However, the role of splicing factor in glucose metabolism and tumor development remains poorly defined. Here, we show that upon glucose intake, the splicing factor SRSF5 is specifically induced through Tip60-mediated acetylation on K125, which antagonizes Smurf1-mediated ubiquitylation. SRSF5 promotes the alternative splicing of CCAR1 to produce CCAR1S proteins, which promote tumor growth by enhancing glucose consumption and acetyl-CoA production. Conversely, upon glucose starvation, SRSF5 is deacetylated by HDAC1, and ubiquitylated by Smurf1 on the same lysine, resulting in proteasomal degradation of SRSF5. The CCAR1L proteins accumulate to promote apoptosis. Importantly, SRSF5 is hyperacetylated and upregulated in human lung cancers, which correlates with increased CCAR1S expression and tumor progression. Thus, SRSF5 responds to high glucose to promote cancer development, and SRSF5-CCAR1 axis may be valuable targets for cancer therapeutics.
Collapse
Affiliation(s)
- Yuhan Chen
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center of Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 100850, China.,Department of Genomics and Proteomics, Beijing Institute of Radiation Medicine, Beijing, 100850, China.,Affiliated BaYi Children's Hospital, PLA Army General Hospital, National Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing Key Laboratory of Pediatric Organ Failure, Beijing, 100700, China
| | - Qingyang Huang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center of Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 100850, China.,Department of Genomics and Proteomics, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Wen Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center of Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 100850, China.,Department of Genomics and Proteomics, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Qiong Zhu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center of Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 100850, China.,Department of Genomics and Proteomics, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Chun-Ping Cui
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center of Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 100850, China.,Department of Genomics and Proteomics, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Liang Xu
- Department of Genomics and Proteomics, Beijing Institute of Radiation Medicine, Beijing, 100850, China.,Department of Biochemistry and Molecular Biology, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Xing Guo
- Department of Genomics and Proteomics, Beijing Institute of Radiation Medicine, Beijing, 100850, China.,Department of Neurobiology, Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Ping Wang
- Department of Central Laboratory, Shanghai Tenth People's Hospital, School of Life Science and Technology, Tongji University, Shanghai, 200072, China
| | - Jingwen Liu
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, 100853, China
| | - Guanglong Dong
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, 100853, China
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
| | - Cui Hua Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Zhichun Feng
- Affiliated BaYi Children's Hospital, PLA Army General Hospital, National Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing Key Laboratory of Pediatric Organ Failure, Beijing, 100700, China
| | - Fuchu He
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center of Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 100850, China. .,Department of Genomics and Proteomics, Beijing Institute of Radiation Medicine, Beijing, 100850, China.
| | - Lingqiang Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center of Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 100850, China. .,Department of Genomics and Proteomics, Beijing Institute of Radiation Medicine, Beijing, 100850, China. .,School of Life Science, Jiangsu Normal University, Xuzhou, 221116, Jiangsu, China.
| |
Collapse
|
50
|
Lee A, Rayner SL, De Luca A, Gwee SSL, Morsch M, Sundaramoorthy V, Shahheydari H, Ragagnin A, Shi B, Yang S, Williams KL, Don EK, Walker AK, Zhang KY, Yerbury JJ, Cole NJ, Atkin JD, Blair IP, Molloy MP, Chung RS. Casein kinase II phosphorylation of cyclin F at serine 621 regulates the Lys48-ubiquitylation E3 ligase activity of the SCF (cyclin F) complex. Open Biol 2018; 7:rsob.170058. [PMID: 29021214 PMCID: PMC5666078 DOI: 10.1098/rsob.170058] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 09/15/2017] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder that is characterized by progressive weakness, paralysis and muscle loss often resulting in patient death within 3–5 years of diagnosis. Recently, we identified disease-linked mutations in the CCNF gene, which encodes the cyclin F protein, in cohorts of patients with familial and sporadic ALS and frontotemporal dementia (FTD) (Williams KL et al. 2016 Nat. Commun.7, 11253. (doi:10.1038/ncomms11253)). Cyclin F is a part of a Skp1-Cul-F-box (SCF) E3 ubiquitin-protein ligase complex and is responsible for ubiquitylating proteins for degradation by the proteasome. In this study, we investigated the phosphorylation status of cyclin F and the effect of the serine to glycine substitution at site 621 (S621G) on E3 ligase activity. This specific mutation (S621G) was found in a multi-generational Australian family with ALS/FTD. We identified seven phosphorylation sites on cyclin F, of which five are newly reported including Ser621. These phosphorylation sites were mostly identified within the PEST (proline, glutamic acid, serine and threonine) sequence located at the C-terminus of cyclin F. Additionally, we determined that casein kinase II (CK2) can phosphorylate Ser621 and thereby regulate the E3 ligase activity of the SCF(cyclin F) complex. Furthermore, the S621G mutation in cyclin F prevents phosphorylation by CK2 and confers elevated Lys48-ubiquitylation activity, a hallmark of ALS/FTD pathology. These findings highlight the importance of phosphorylation in regulating the activity of the SCF(cyclin F) E3 ligase complex that can affect downstream processes and may lead to defective motor neuron development, neuron degeneration and ultimately ALS and FTD.
Collapse
Affiliation(s)
- Albert Lee
- Department of Biomedical Sciences, Centre for Motor Neuron Disease Research, Faculty of Medicine and Health Sciences, Macquarie University, 2 Technology Place, North Ryde, NSW 2109, Australia .,Australian Proteome Analysis Facility, Research Park Drive, Macquarie University, North Ryde, NSW 2109, Australia
| | - Stephanie L Rayner
- Department of Biomedical Sciences, Centre for Motor Neuron Disease Research, Faculty of Medicine and Health Sciences, Macquarie University, 2 Technology Place, North Ryde, NSW 2109, Australia.,Faculty of Science and Engineering, Department of Chemistry and Biomolecular Sciences, Research Park Drive, Macquarie University, North Ryde, NSW 2109, Australia
| | - Alana De Luca
- Department of Biomedical Sciences, Centre for Motor Neuron Disease Research, Faculty of Medicine and Health Sciences, Macquarie University, 2 Technology Place, North Ryde, NSW 2109, Australia
| | - Serene S L Gwee
- Department of Biomedical Sciences, Centre for Motor Neuron Disease Research, Faculty of Medicine and Health Sciences, Macquarie University, 2 Technology Place, North Ryde, NSW 2109, Australia
| | - Marco Morsch
- Department of Biomedical Sciences, Centre for Motor Neuron Disease Research, Faculty of Medicine and Health Sciences, Macquarie University, 2 Technology Place, North Ryde, NSW 2109, Australia
| | - Vinod Sundaramoorthy
- Department of Biomedical Sciences, Centre for Motor Neuron Disease Research, Faculty of Medicine and Health Sciences, Macquarie University, 2 Technology Place, North Ryde, NSW 2109, Australia
| | - Hamideh Shahheydari
- Department of Biomedical Sciences, Centre for Motor Neuron Disease Research, Faculty of Medicine and Health Sciences, Macquarie University, 2 Technology Place, North Ryde, NSW 2109, Australia
| | - Audrey Ragagnin
- Department of Biomedical Sciences, Centre for Motor Neuron Disease Research, Faculty of Medicine and Health Sciences, Macquarie University, 2 Technology Place, North Ryde, NSW 2109, Australia
| | - Bingyang Shi
- Department of Biomedical Sciences, Centre for Motor Neuron Disease Research, Faculty of Medicine and Health Sciences, Macquarie University, 2 Technology Place, North Ryde, NSW 2109, Australia
| | - Shu Yang
- Department of Biomedical Sciences, Centre for Motor Neuron Disease Research, Faculty of Medicine and Health Sciences, Macquarie University, 2 Technology Place, North Ryde, NSW 2109, Australia
| | - Kelly L Williams
- Department of Biomedical Sciences, Centre for Motor Neuron Disease Research, Faculty of Medicine and Health Sciences, Macquarie University, 2 Technology Place, North Ryde, NSW 2109, Australia
| | - Emily K Don
- Department of Biomedical Sciences, Centre for Motor Neuron Disease Research, Faculty of Medicine and Health Sciences, Macquarie University, 2 Technology Place, North Ryde, NSW 2109, Australia
| | - Adam K Walker
- Department of Biomedical Sciences, Centre for Motor Neuron Disease Research, Faculty of Medicine and Health Sciences, Macquarie University, 2 Technology Place, North Ryde, NSW 2109, Australia
| | - Katharine Y Zhang
- Department of Biomedical Sciences, Centre for Motor Neuron Disease Research, Faculty of Medicine and Health Sciences, Macquarie University, 2 Technology Place, North Ryde, NSW 2109, Australia
| | - Justin J Yerbury
- Illawarra Health and Medical Research Institute, School of Biological Sciences, University of Wollongong, Northfields Avenue, Wollongong, NSW 2522, Australia
| | - Nicholas J Cole
- Department of Biomedical Sciences, Centre for Motor Neuron Disease Research, Faculty of Medicine and Health Sciences, Macquarie University, 2 Technology Place, North Ryde, NSW 2109, Australia
| | - Julie D Atkin
- Department of Biomedical Sciences, Centre for Motor Neuron Disease Research, Faculty of Medicine and Health Sciences, Macquarie University, 2 Technology Place, North Ryde, NSW 2109, Australia.,Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, Victoria, Australia
| | - Ian P Blair
- Department of Biomedical Sciences, Centre for Motor Neuron Disease Research, Faculty of Medicine and Health Sciences, Macquarie University, 2 Technology Place, North Ryde, NSW 2109, Australia
| | - Mark P Molloy
- Australian Proteome Analysis Facility, Research Park Drive, Macquarie University, North Ryde, NSW 2109, Australia.,Faculty of Science and Engineering, Department of Chemistry and Biomolecular Sciences, Research Park Drive, Macquarie University, North Ryde, NSW 2109, Australia
| | - Roger S Chung
- Department of Biomedical Sciences, Centre for Motor Neuron Disease Research, Faculty of Medicine and Health Sciences, Macquarie University, 2 Technology Place, North Ryde, NSW 2109, Australia
| |
Collapse
|