1
|
Aerbajinai W, Zhu J, Chin K, Rodgers GP. Glia maturation factor-γ regulates amyloid-β42 phagocytosis through scavenger receptor class A type I in murine macrophages. J Leukoc Biol 2024; 117:qiae197. [PMID: 39243388 PMCID: PMC11685041 DOI: 10.1093/jleuko/qiae197] [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: 07/09/2024] [Accepted: 09/05/2024] [Indexed: 09/09/2024] Open
Abstract
Dysfunctional phagocytic clearance of β-amyloid (Aβ) in microglia and peripheral macrophages/monocytes has been implicated in Alzheimer's disease, but the mechanisms underlying this dysfunction are not yet well understood. In this study, we examined the role of glia maturation factor-γ (GMFG), an actin-disassembly protein, i.e. highly expressed in immune cells, in macrophage Aβ phagocytosis and in regulating type I class A scavenger receptor, a cell-surface receptor that has previously been implicated in Aβ clearance. GMFG knockdown-increased phagocytosis of Aβ42 in bone marrow-derived macrophages and RAW264.7 murine macrophages, while GMFG overexpression reduced Aβ42 uptake in these cells. Blocking with anti-type I class A scavenger receptor antibodies inhibited Aβ42 uptake in GMFG-knockdown cells, establishing a role for type I class A scavenger receptor in Aβ42 phagocytosis. GMFG knockdown-increased type I class A scavenger receptor protein expression under both basal conditions and in response to Aβ42 treatment via both the transcriptional and posttranscriptional levels in RAW264.7 macrophages. GMFG knockdown modulated Aβ42-induced K48-linked and K63-polyubiquitination of type I class A scavenger receptor, the phosphorylation of type I class A scavenger receptor and c-Jun N-Terminal kinase (JNK), suggesting that GMFG plays a role for intracellular signaling in the type I class A scavenger receptor--mediated uptake of Aβ. Further, GMFG-knockdown cells displayed increased levels of the transcriptional factor MafB, and silencing of MafB in these cells reduced their type I class A scavenger receptor expression. Finally, GMFG was found to interact with the nuclear pore complex component RanBP2, and silencing of RanBP2 in GMFG-knockdown cells reduced their type I class A scavenger receptor expression. Collectively, these data support the role of GMFG as a novel regulator of type I class A scavenger receptor in macrophage Aβ phagocytosis and may provide insight into therapeutic approaches to potentially slow or prevent the progression of Alzheimer's disease.
Collapse
Affiliation(s)
- Wulin Aerbajinai
- Molecular and Clinical Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, BG 10, RM 9N113, 10 Center Dr., Bethesda, MD 20892, United States
| | - Jianqiong Zhu
- Molecular and Clinical Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, BG 10, RM 9N113, 10 Center Dr., Bethesda, MD 20892, United States
| | - Kyung Chin
- Molecular and Clinical Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, BG 10, RM 9N113, 10 Center Dr., Bethesda, MD 20892, United States
| | - Griffin P Rodgers
- Molecular and Clinical Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, BG 10, RM 9N113, 10 Center Dr., Bethesda, MD 20892, United States
| |
Collapse
|
2
|
Zhang T, Zheng B, Xia C, Wu P, Zheng B, Jiang L, Li J, Lv G, Zhou H, Huang W, Zou M. Hypoxic Upregulation of IER2 Increases Paracrine GMFG Signaling of Endoplasmic Reticulum Stress-CAF to Promote Chordoma Progression via Targeting ITGB1. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405421. [PMID: 39207055 PMCID: PMC11515918 DOI: 10.1002/advs.202405421] [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: 05/17/2024] [Revised: 07/27/2024] [Indexed: 09/04/2024]
Abstract
Currently, the oncogenic mechanism of endoplasmic reticulum stress-CAF (ERS-CAF) subpopulation in chordoma remains unknown. Here, single-cell RNA sequencing, spatial transcriptomics, GeoMx Digital Spatial Profiler, data-independent acquisition proteomics, bulk RNA-seq, and multiplexed quantitative immunofluorescence are used to unveil the precise molecular mechanism of how ERS-CAF affected chordoma progression. Results show that hypoxic microenvironment reprograms CAFs into ERS-CAF subtype. Mechanistically, this occurrs via hypoxia-mediated transcriptional upregulation of IER2. Overexpression of IER2 in CAFs promotes chordoma progression, which can be impeded by IER2 knockdown or use of ERS inhibitors. IER2 also induces expression of ERS-CAF marker genes and results in production of a pro-tumorigenic paracrine GMFG signaling, which exert its biological function via directly binding to ITGB1 on tumor cells. ITGB1 inhibition attenuates tumor malignant progression, which can be partially reversed by exogenous GMFG intervention. Further analyses reveal a positive correlation between ITGB1high tumor cell counts and SPP1+ macrophage density, as well as the spatial proximity of these two cell types. Clinically, a significant correlation of high IER2/ITGB1 expression with tumor aggressive phenotype and poor patient survival is observed. Collectively, the findings suggest that ERS-CAF regulates SPP1+ macrophage to aggravate chordoma progression via the IER2/GMFG/ITGB1 axis, which may be targeted therapeutically in future.
Collapse
Affiliation(s)
- Tao‐Lan Zhang
- Department of PharmacyThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyang421001China
| | - Bo‐Wen Zheng
- Department of PharmacyThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyang421001China
- Musculoskeletal Tumor CenterPeking University People's HospitalPeking UniversityBeijing100044China
| | - Chao Xia
- Department of Spine SurgeryThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyang421001China
| | - Peng‐Fei Wu
- Department of Genetics and EndocrinologyNational Children's Medical Center for South Central RegionGuangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhouGuangdong510623China
| | - Bo‐Yv Zheng
- Department of Orthopedics SurgeryGeneral Hospital of the Central Theater CommandWuhan430061China
| | - Ling‐Xiang Jiang
- Department of Radiation OncologyMelvin and Bren Simon Comprehensive Cancer CenterIndiana University School of MedicineIndianapolisIN46202USA
| | - Jing Li
- Department of Spine SurgeryThe Second Xiangya HospitalCentral South UniversityChangsha410011China
| | - Guo‐Hua Lv
- Department of Spine SurgeryThe Second Xiangya HospitalCentral South UniversityChangsha410011China
| | - Hong Zhou
- Department of RadiologyThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyang421001China
| | - Wei Huang
- The First Affiliated HospitalHealth Management CenterHengyang Medical SchoolUniversity of South ChinaHengyang421001China
| | - Ming‐Xiang Zou
- Department of Spine SurgeryThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyang421001China
| |
Collapse
|
3
|
Yang Y, Duan Y, Jiang H, Li J, Bai W, Zhang Q, Li J, Shao J. Bioinformatics-driven identification and validation of diagnostic biomarkers for cerebral ischemia reperfusion injury. Heliyon 2024; 10:e28565. [PMID: 38601664 PMCID: PMC11004763 DOI: 10.1016/j.heliyon.2024.e28565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 03/20/2024] [Indexed: 04/12/2024] Open
Abstract
Objective This article aims to identify genetic features associated with immune cell infiltration in cerebral ischemia-reperfusion injury (CIRI) development through bioinformatics, with the goal of discovering diagnostic biomarkers and potential therapeutic targets. Methods We obtained two datasets from the Gene Expression Omnibus (GEO) database to identify immune-related differentially expressed genes (IRDEGs). These genes' functions were analyzed via Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG). Tools such as CIBERSORT and ssGSEA assessed immune cell infiltration. The Starbase and miRDB databases predicted miRNAs interacting with hub genes, and Cytoscape software mapped mRNA-miRNA interaction networks. The ENCORI database was employed to predict RNA binding proteins interacting with hub genes. Key genes were identified using a random forest algorithm and constructing a Support Vector Machine (SVM) model. LASSO regression analysis constructed a diagnostic model for hub genes to determine their diagnostic value, and PCR analysis validated their expression in cerebral ischemia-reperfusion. Results We identified 10 IRDEGs (C1qa, Ccl4, Cd74, Cd8a, Cxcl10, Gmfg, Grp, Lgals3bp, Timp1, Vim). The random forest algorithm, and SVM model intersection revealed three key genes (Ccl4, Gmfg, C1qa) as diagnostic biomarkers for CIRI. LASSO regression analysis, further refined this to two key genes (Ccl4 and C1qa), With ROC curve, analysis confirming their diagnostic efficacy (C1qa AUC = 0.75, Ccl4 AUC = 0.939). PCR analysis corroborated these findings. Conclusions Our study elucidates immune and metabolic response mechanisms in CIRI, identifying two immune-related genes as key biomarkers and potential therapeutic targets in response to cerebral ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Yuan Yang
- Department of Anesthesiology, The First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Yushan Duan
- Department of Critical Care Medicine, The Second Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Huan Jiang
- Department of Anesthesiology, The First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Junjie Li
- Department of Anesthesiology, The First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Wenya Bai
- Department of Anesthesiology, The First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Qi Zhang
- Department of Anesthesiology, The First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Junming Li
- Department of Anesthesiology, The First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Jianlin Shao
- Department of Anesthesiology, The First Affiliated Hospital, Kunming Medical University, Kunming, China
| |
Collapse
|
4
|
McGuirk ER, Koundinya N, Nagarajan P, Padrick SB, Goode BL. Direct observation of cortactin protecting Arp2/3-actin filament branch junctions from GMF-mediated destabilization. Eur J Cell Biol 2024; 103:151378. [PMID: 38071835 PMCID: PMC10843626 DOI: 10.1016/j.ejcb.2023.151378] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 12/02/2023] [Accepted: 12/02/2023] [Indexed: 01/28/2024] Open
Abstract
How cells tightly control the formation and turnover of branched actin filament arrays to drive cell motility, endocytosis, and other cellular processes is still not well understood. Here, we investigated the mechanistic relationship between two binding partners of the Arp2/3 complex, glia maturation factor (GMF) and cortactin. Individually, GMF and cortactin have opposite effects on the stability of actin filament branches, but it is unknown how they work in concert with each other to govern branch turnover. Using TIRF microscopy, we observe that GMF's branch destabilizing activities are potently blocked by cortactin (IC50 = 1.3 nM) and that this inhibition requires direct interactions of cortactin with Arp2/3 complex. The simplest model that would explain these results is competition for binding Arp2/3 complex. However, we find that cortactin and GMF do not compete for free Arp2/3 complex in solution. Further, we use single molecule analysis to show that cortactin's on-rate (3 ×107 s-1 M-1) and off-rate (0.03 s-1) at branch junctions are minimally affected by excess GMF. Together, these results show that cortactin binds with high affinity to branch junctions, where it blocks the destabilizing effects of GMF, possibly by a mechanism that is allosteric in nature. In addition, the affinities we measure for cortactin at actin filament branch junctions (Kd = 0.9 nM) and filament sides (Kd = 206 nM) are approximately 20-fold stronger than previously reported. These observations contribute to an emerging view of molecular complexity in how Arp2/3 complex is regulated through the integration of multiple inputs.
Collapse
Affiliation(s)
- Emma R McGuirk
- Department of Biology, Rosenstiel Basic Medical Science Research Center, Brandeis University, 415 South Street, Waltham, MA 02454, USA
| | - Neha Koundinya
- Department of Biology, Rosenstiel Basic Medical Science Research Center, Brandeis University, 415 South Street, Waltham, MA 02454, USA
| | - Priyashree Nagarajan
- Department of Biochemistry and Molecular Biology, Drexel University, Philadelphia, PA 19104, USA
| | - Shae B Padrick
- Department of Biochemistry and Molecular Biology, Drexel University, Philadelphia, PA 19104, USA
| | - Bruce L Goode
- Department of Biology, Rosenstiel Basic Medical Science Research Center, Brandeis University, 415 South Street, Waltham, MA 02454, USA.
| |
Collapse
|
5
|
Amlerova Z, Chmelova M, Anderova M, Vargova L. Reactive gliosis in traumatic brain injury: a comprehensive review. Front Cell Neurosci 2024; 18:1335849. [PMID: 38481632 PMCID: PMC10933082 DOI: 10.3389/fncel.2024.1335849] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 02/01/2024] [Indexed: 01/03/2025] Open
Abstract
Traumatic brain injury (TBI) is one of the most common pathological conditions impacting the central nervous system (CNS). A neurological deficit associated with TBI results from a complex of pathogenetic mechanisms including glutamate excitotoxicity, inflammation, demyelination, programmed cell death, or the development of edema. The critical components contributing to CNS response, damage control, and regeneration after TBI are glial cells-in reaction to tissue damage, their activation, hypertrophy, and proliferation occur, followed by the formation of a glial scar. The glial scar creates a barrier in damaged tissue and helps protect the CNS in the acute phase post-injury. However, this process prevents complete tissue recovery in the late/chronic phase by producing permanent scarring, which significantly impacts brain function. Various glial cell types participate in the scar formation, but this process is mostly attributed to reactive astrocytes and microglia, which play important roles in several brain pathologies. Novel technologies including whole-genome transcriptomic and epigenomic analyses, and unbiased proteomics, show that both astrocytes and microglia represent groups of heterogenic cell subpopulations with different genomic and functional characteristics, that are responsible for their role in neurodegeneration, neuroprotection and regeneration. Depending on the representation of distinct glia subpopulations, the tissue damage as well as the regenerative processes or delayed neurodegeneration after TBI may thus differ in nearby or remote areas or in different brain structures. This review summarizes TBI as a complex process, where the resultant effect is severity-, region- and time-dependent and determined by the model of the CNS injury and the distance of the explored area from the lesion site. Here, we also discuss findings concerning intercellular signaling, long-term impacts of TBI and the possibilities of novel therapeutical approaches. We believe that a comprehensive study with an emphasis on glial cells, involved in tissue post-injury processes, may be helpful for further research of TBI and be the decisive factor when choosing a TBI model.
Collapse
Affiliation(s)
- Zuzana Amlerova
- Department of Neuroscience, Second Faculty of Medicine, Charles University, Prague, Czechia
| | - Martina Chmelova
- Department of Neuroscience, Second Faculty of Medicine, Charles University, Prague, Czechia
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
| | - Miroslava Anderova
- Department of Neuroscience, Second Faculty of Medicine, Charles University, Prague, Czechia
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
| | - Lydia Vargova
- Department of Neuroscience, Second Faculty of Medicine, Charles University, Prague, Czechia
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
6
|
Goode BL, Eskin J, Shekhar S. Mechanisms of actin disassembly and turnover. J Cell Biol 2023; 222:e202309021. [PMID: 37948068 PMCID: PMC10638096 DOI: 10.1083/jcb.202309021] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/21/2023] [Accepted: 10/23/2023] [Indexed: 11/12/2023] Open
Abstract
Cellular actin networks exhibit a wide range of sizes, shapes, and architectures tailored to their biological roles. Once assembled, these filamentous networks are either maintained in a state of polarized turnover or induced to undergo net disassembly. Further, the rates at which the networks are turned over and/or dismantled can vary greatly, from seconds to minutes to hours or even days. Here, we review the molecular machinery and mechanisms employed in cells to drive the disassembly and turnover of actin networks. In particular, we highlight recent discoveries showing that specific combinations of conserved actin disassembly-promoting proteins (cofilin, GMF, twinfilin, Srv2/CAP, coronin, AIP1, capping protein, and profilin) work in concert to debranch, sever, cap, and depolymerize actin filaments, and to recharge actin monomers for new rounds of assembly.
Collapse
Affiliation(s)
- Bruce L. Goode
- Department of Biology, Rosenstiel Basic Medical Science Research Center, Brandeis University, Waltham, MA, USA
| | - Julian Eskin
- Department of Biology, Rosenstiel Basic Medical Science Research Center, Brandeis University, Waltham, MA, USA
| | - Shashank Shekhar
- Departments of Physics, Cell Biology and Biochemistry, Emory University, Atlanta, GA, USA
| |
Collapse
|
7
|
Zhao Y, Wei X, Li J, Diao Y, Shan C, Li W, Zhang S, Wu F. High Level of GMFG Correlated to Poor Clinical Outcome and Promoted Cell Migration and Invasion through EMT Pathway in Triple-Negative Breast Cancer. Genes (Basel) 2023; 14:1157. [PMID: 37372337 DOI: 10.3390/genes14061157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 05/13/2023] [Accepted: 05/23/2023] [Indexed: 06/29/2023] Open
Abstract
Triple-negative breast cancer (TNBC) has a very poor prognosis due to the disease's lack of established targeted treatment options. Glia maturation factor γ (GMFG), a novel ADF/cofilin superfamily protein, has been reported to be differentially expressed in tumors, but its expression level in TNBC remains unknown. The question of whether GMFG correlates with the TNBC prognosis is also unclear. In this study, data from the Cancer Genome Atlas (TCGA), Clinical Proteomic Tumor Analysis Consortium (CPTAC), Human Protein Atlas (HPA), and Genotype-Tissue Expression (GTEx) databases were used to analyze the expression of GMFG in pan-cancer and the correlation between clinical factors. Gene Set Cancer Analysis (GSCA) and Gene Set Enrichment Analysis (GSEA) were also used to analyze the functional differences between the different expression levels and predict the downstream pathways. GMFG expression in breast cancer tissues, and its related biological functions, were further analyzed by immunohistochemistry (IHC), immunoblotting, RNAi, and function assay; we found that TNBC has a high expression of GMFG, and this higher expression was correlated with a poorer prognosis in TCGA and collected specimens of the TNBC. GMFG was also related to TNBC patients' clinicopathological data, especially those with histological grade and axillary lymph node metastasis. In vitro, GMFG siRNA inhibited cell migration and invasion through the EMT pathway. The above data indicate that high expression of GMFG in TNBC is related to malignancy and that GMFG could be a biomarker for the detection of TNBC metastasis.
Collapse
Affiliation(s)
- Yonglin Zhao
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Xing Wei
- Department of Gynaecology and Obstetrics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Jia Li
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Yan Diao
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Changyou Shan
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Weimiao Li
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Shuqun Zhang
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Fei Wu
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China, Xi'an 710061, China
| |
Collapse
|
8
|
Li H, Luo Q, Cai S, Tie R, Meng Y, Shan W, Xu Y, Zeng X, Qian P, Huang H. Glia maturation factor-γ is required for initiation and maintenance of hematopoietic stem and progenitor cells. Stem Cell Res Ther 2023; 14:117. [PMID: 37122014 PMCID: PMC10150485 DOI: 10.1186/s13287-023-03328-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 04/05/2023] [Indexed: 05/02/2023] Open
Abstract
BACKGROUND In vertebrates, hematopoietic stem and progenitor cells (HSPCs) emerge from hemogenic endothelium in the floor of the dorsal aorta and subsequently migrate to secondary niches where they expand and differentiate into committed lineages. Glia maturation factor γ (gmfg) is a key regulator of actin dynamics that was shown to be highly expressed in hematopoietic tissue. Our goal is to investigate the role and mechanism of gmfg in embryonic HSPC development. METHODS In-depth bioinformatics analysis of our published RNA-seq data identified gmfg as a cogent candidate gene implicated in HSPC development. Loss and gain-of-function strategies were applied to study the biological function of gmfg. Whole-mount in situ hybridization, confocal microscopy, flow cytometry, and western blotting were used to evaluate changes in the number of various hematopoietic cells and expression levels of cell proliferation, cell apoptosis and hematopoietic-related markers. RNA-seq was performed to screen signaling pathways responsible for gmfg deficiency-induced defects in HSPC initiation. The effect of gmfg on YAP sublocalization was assessed in vitro by utilizing HUVEC cell line. RESULTS We took advantage of zebrafish embryos to illustrate that loss of gmfg impaired HSPC initiation and maintenance. In gmfg-deficient embryos, the number of hemogenic endothelium and HSPCs was significantly reduced, with the accompanying decreased number of erythrocytes, myelocytes and lymphocytes. We found that blood flow modulates gmfg expression and gmfg overexpression could partially rescue the reduction of HSPCs in the absence of blood flow. Assays in zebrafish and HUVEC showed that gmfg deficiency suppressed the activity of YAP, a well-established blood flow mediator, by preventing its shuttling from cytoplasm to nucleus. During HSPC initiation, loss of gmfg resulted in Notch inactivation and the induction of Notch intracellular domain could partially restore the HSPC loss in gmfg-deficient embryos. CONCLUSIONS We conclude that gmfg mediates blood flow-induced HSPC maintenance via regulation of YAP, and contributes to HSPC initiation through the modulation of Notch signaling. Our findings reveal a brand-new aspect of gmfg function and highlight a novel mechanism for embryonic HSPC development.
Collapse
Affiliation(s)
- Honghu Li
- Bone Marrow Transplantation Center, School of Medicine, The First Affiliated Hospital, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310012, Zhejiang, People's Republic of China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, People's Republic of China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, People's Republic of China
| | - Qian Luo
- Bone Marrow Transplantation Center, School of Medicine, The First Affiliated Hospital, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310012, Zhejiang, People's Republic of China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, People's Republic of China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, People's Republic of China
| | - Shuyang Cai
- Bone Marrow Transplantation Center, School of Medicine, The First Affiliated Hospital, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310012, Zhejiang, People's Republic of China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, People's Republic of China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, People's Republic of China
| | - Ruxiu Tie
- Bone Marrow Transplantation Center, School of Medicine, The First Affiliated Hospital, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310012, Zhejiang, People's Republic of China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, People's Republic of China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, People's Republic of China
| | - Ye Meng
- Bone Marrow Transplantation Center, School of Medicine, The First Affiliated Hospital, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310012, Zhejiang, People's Republic of China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, People's Republic of China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, People's Republic of China
| | - Wei Shan
- Bone Marrow Transplantation Center, School of Medicine, The First Affiliated Hospital, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310012, Zhejiang, People's Republic of China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, People's Republic of China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, People's Republic of China
| | - Yulin Xu
- Bone Marrow Transplantation Center, School of Medicine, The First Affiliated Hospital, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310012, Zhejiang, People's Republic of China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, People's Republic of China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, People's Republic of China
| | - Xiangjun Zeng
- Bone Marrow Transplantation Center, School of Medicine, The First Affiliated Hospital, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310012, Zhejiang, People's Republic of China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, People's Republic of China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, People's Republic of China
| | - Pengxu Qian
- Bone Marrow Transplantation Center, School of Medicine, The First Affiliated Hospital, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310012, Zhejiang, People's Republic of China.
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, People's Republic of China.
- Institute of Hematology, Zhejiang University, Hangzhou, People's Republic of China.
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, People's Republic of China.
- Center of Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, 310012, People's Republic of China.
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, 310012, Zhejiang, People's Republic of China.
- School of Medicine, Zhejiang University, No. 866 Yuhangtang Road, Hangzhou, People's Republic of China.
| | - He Huang
- Bone Marrow Transplantation Center, School of Medicine, The First Affiliated Hospital, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310012, Zhejiang, People's Republic of China.
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, People's Republic of China.
- Institute of Hematology, Zhejiang University, Hangzhou, People's Republic of China.
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, People's Republic of China.
- Center of Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, 310012, People's Republic of China.
| |
Collapse
|
9
|
Liu J, Zhu X, Gao L, Geng R, Tao X, Xu H, Chen Z. Expression and Prognostic Role of Glia Maturation Factor-γ in Gliomas. Front Mol Neurosci 2022; 15:906762. [PMID: 35845613 PMCID: PMC9277395 DOI: 10.3389/fnmol.2022.906762] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 05/13/2022] [Indexed: 11/16/2022] Open
Abstract
Background Glia maturation factor-γ (GMFG) regulates actin cytoskeletal organization and promotes the invasion of cancer cells. However, its expression pattern and molecular function in gliomas have not been clearly defined. Methods In this study, public datasets comprising 2,518 gliomas samples were used to explore GMFG expression and its correlation with malignancy in gliomas. Immunohistochemistry (IHC) staining was performed to determine the expression of GMFG in gliomas using an in-house cohort that contained 120 gliomas samples. Gene ontology enrichment analysis was conducted using the DAVID tool. The correlation between GMFG expression and immune cell infiltration was evaluated using TIMER, Tumor Immune Single-Cell Hub (TISCH) database, and IHC staining assays. The Kaplan-Meier analysis was performed to determine the prognostic role of GMFG and its association with temozolomide (TMZ) response in gliomas. Results The GMFG expression was higher in gliomas compared with non-tumor brain tissues both in public datasets and in-house cohort. High expression of GMFG was significantly associated with WHO grade IV, IDH 1/2 wild-type, and mesenchymal (ME) subtypes. Bioinformatic prediction and IHC analysis revealed that GMFG expression obviously correlated with the macrophage marker CD163 in gliomas. Moreover, both lower grade glioma (LGG) and glioblastoma multiforme (GBM) patients with high GMFG expression had shorter overall survival than those with low GMFG expression. These results indicate that GMFG may be a therapeutic target for the treatment of such patients. Patients with low GMFG expression who received chemotherapy had a longer survival time than those with high GMFG expression. For patients who received ion radiotherapy (IR) only, the GMFG expression level had no effect on the overall survival neither in CGGA and TCGA datasets. Conclusion The GMFG is a novel prognostic biomarker for patients with both LGG and GBM. Increased GMFG expression is associated with tumor-associated macrophages (TAMs) infiltration and with a bad response to TMZ treatment.
Collapse
Affiliation(s)
- Junhui Liu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaonan Zhu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lun Gao
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Rongxin Geng
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiang Tao
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Haitao Xu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhibiao Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
10
|
Tang H, Liu J, Huang J. GMFG (glia maturation factor gamma) inhibits lung cancer growth by activating p53 signaling pathway. Bioengineered 2022; 13:9284-9293. [PMID: 35383531 PMCID: PMC9161896 DOI: 10.1080/21655979.2022.2049958] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 02/28/2022] [Accepted: 03/01/2022] [Indexed: 12/26/2022] Open
Abstract
The tumor-promoting or tumor-suppressing functions of Glia maturation factor gamma (GMFG) were described in several cancers. However, how GMFG regulates lung cancer progression is elusive. Bioinformatics analysis was employed to analyze GMFG expression in lung adenocarcinoma (LUAD) and lung squamous cancer (LUSC) as well as its significance in prognosis prediction and diagnosis in lung cancer patients. CCK8 and colony formation assays were adopted to evaluate the impact of GMFG overexpressing and depleting on lung cancer cell proliferation. And in vivo experiments were implemented. Luciferase reporter assays were used to disclose the signaling pathway mediated by GMFG in lung cancer. GMFG expression was lower in LUSC and LUAD tissues compared with normal lung tissues based on TCGA and GTEx databases. Low GMFG expression was associated with lower overall survival and shorter disease specific survival compared high GMFG expression. In vitro loss and gain functions assays demonstrated that ectopically GMFG expression dampened the lung cancer cell proliferation while GMFG knockout escalated the cell proliferation. The promoting effect of GMFG knockout on lung cancer tumorgenesis was also observed in vivo. More interesting, GMFG overexpression reinforced the p53 signaling pathway in lung cancer cells, conversely GMFG deficiency disrupted p53 signaling pathway. In conclusion, we revealed that GMFG is fundamental to p53 signaling pathway to inhibit lung cancer progression, highlighting the importance of GMFG as a p53 inducer for lung cancer patient's diagnosis and therapy.
Collapse
Affiliation(s)
- Hua Tang
- Department of Thoracic Surgery, Shanghai Changzheng Hospital, Navy Military Medical University, Shanghai, Shanghai, China
| | - Jie Liu
- Department of Thoracic Surgery, Army medical university, Southwest hospital, Chongqing, Sichuan , China
| | - Jun Huang
- Department of Thoracic Oncology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
11
|
Zhang YL, Cao JL, Zhang Y, Liao L, Deng L, Yang SY, Hu SY, Ning Y, Zhang FL, Li DQ. RNF144A exerts tumor suppressor function in breast cancer through targeting YY1 for proteasomal degradation to downregulate GMFG expression. Med Oncol 2022; 39:48. [PMID: 35103856 PMCID: PMC8807444 DOI: 10.1007/s12032-021-01631-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 12/13/2021] [Indexed: 11/13/2022]
Abstract
Ring finger protein 144A (RNF144A), a poorly characterized member of the RING-in-between-RING family of E3 ubiquitin ligases, is an emerging tumor suppressor, but its underlying mechanism remains largely elusive. To address this issue, we used Affymetrix GeneChip Human Transcriptome Array 2.0 to profile gene expression in MDA-MB-231 cells stably expressing empty vector pCDH and Flag-RNF144A, and found that 128 genes were differentially expressed between pCDH- and RNF144A-expressing cells with fold change over 1.5. We further demonstrated that RNF144A negatively regulated the protein and mRNA levels of glial maturation factor γ (GMFG). Mechanistical investigations revealed that transcription factor YY1 transcriptionally activated GMFG expression, and RNF144A interacted with YY1 and promoted its ubiquitination-dependent degradation, thus blocking YY1-induced GMFG expression. Functional rescue assays showed that ectopic expression of RNF144A suppressed the proliferative, migratory, and invasive potential of breast cancer cells, and the noted effects were partially restored by re-expression of GMFG in RNF144A-overexpressing breast cancer cells. Collectively, these findings reveal that RNF144A negatively regulates GMFG expression by targeting YY1 for proteasomal degradation, thus inhibiting the proliferation, migration, and invasion of breast cancer cells.
Collapse
Affiliation(s)
- Yin-Ling Zhang
- Fudan University Shanghai Cancer Center, Shanghai Key Laboratory of Medical Epigenetics, International Co-Laboratory of Medical Epigenetics and Metabolism of Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.,Cancer Institute, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Jin-Ling Cao
- Fudan University Shanghai Cancer Center, Shanghai Key Laboratory of Medical Epigenetics, International Co-Laboratory of Medical Epigenetics and Metabolism of Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Ye Zhang
- Fudan University Shanghai Cancer Center, Shanghai Key Laboratory of Medical Epigenetics, International Co-Laboratory of Medical Epigenetics and Metabolism of Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Li Liao
- Fudan University Shanghai Cancer Center, Shanghai Key Laboratory of Medical Epigenetics, International Co-Laboratory of Medical Epigenetics and Metabolism of Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.,Cancer Institute, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Ling Deng
- Fudan University Shanghai Cancer Center, Shanghai Key Laboratory of Medical Epigenetics, International Co-Laboratory of Medical Epigenetics and Metabolism of Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.,Cancer Institute, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Shao-Ying Yang
- Fudan University Shanghai Cancer Center, Shanghai Key Laboratory of Medical Epigenetics, International Co-Laboratory of Medical Epigenetics and Metabolism of Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.,Cancer Institute, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Shu-Yuan Hu
- Fudan University Shanghai Cancer Center, Shanghai Key Laboratory of Medical Epigenetics, International Co-Laboratory of Medical Epigenetics and Metabolism of Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.,Cancer Institute, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yan Ning
- Department of Pathology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China.
| | - Fang-Lin Zhang
- Fudan University Shanghai Cancer Center, Shanghai Key Laboratory of Medical Epigenetics, International Co-Laboratory of Medical Epigenetics and Metabolism of Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China. .,Cancer Institute, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Da-Qiang Li
- Fudan University Shanghai Cancer Center, Shanghai Key Laboratory of Medical Epigenetics, International Co-Laboratory of Medical Epigenetics and Metabolism of Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China. .,Cancer Institute, Shanghai Medical College, Fudan University, Shanghai, 200032, China. .,Department of Breast Surgery, Shanghai Medical College, Fudan University, Shanghai, 200032, China. .,Shanghai Key Laboratory of Breast Cancer, Shanghai Medical College, Fudan University, Shanghai, 200032, China. .,Shanghai Key Laboratory of Radiation Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
12
|
Fujimoto K, Nakano K, Kuwayama H, Yumura S. Deletion of gmfA induces keratocyte-like migration in Dictyostelium. FEBS Open Bio 2021; 12:306-319. [PMID: 34855306 PMCID: PMC8727941 DOI: 10.1002/2211-5463.13339] [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: 10/05/2021] [Revised: 11/15/2021] [Accepted: 11/30/2021] [Indexed: 11/06/2022] Open
Abstract
Glia maturation factor (GMF) has been established as an inactivating factor of the actin‐related protein 2/3 (Arp2/3) complex, which regulates actin assembly. Regulation of actin assembly and reorganization is crucial for various cellular events, such as cell migration, cell division, and development. Here, to examine the roles of ADF‐H domain‐containing protein (also known as glia maturation factor; GmfA), the product of a single GMF homologous gene in Dictyostelium, gmfA‐null cells were generated. They had moderate defects in cell growth and cytokinesis. Interestingly, they showed a keratocyte‐like fan shape with a broader pseudopod, where Arp3 accumulated at higher levels than in wild‐type cells. They migrated with higher persistence, but their velocities were comparable to those of wild‐type cells. The polar pseudopods during cell division were also broader than those in wild‐type cells. However, GmfA did not localize at the pseudopods in migrating cells or the polar pseudopods in dividing cells. Adhesions of mutant cells to the substratum were much stronger than that of wild‐type cells. Although the mutant cells showed chemotaxis comparable to that of wild‐type cells, they formed disconnected streams during the aggregation stage; however, they finally formed normal fruiting bodies. These results suggest that GmfA plays a crucial role in cell migration.
Collapse
Affiliation(s)
- Koushiro Fujimoto
- Graduate School of Sciences and Technology for Innovation, Yamaguchi University, Japan
| | - Kentaro Nakano
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Japan
| | - Hidekazu Kuwayama
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Japan
| | - Shigehiko Yumura
- Graduate School of Sciences and Technology for Innovation, Yamaguchi University, Japan
| |
Collapse
|
13
|
Li Y, Tang Y, Liu J, Meng X, Wang Y, Min Q, Hong R, Tsubata T, Hase K, Wang JY. Glia maturation factor-γ is involved in S1P-induced marginal zone B cell chemotaxis and optimal T-independent type II antigen-induced IgM production. Int Immunol 2021; 34:35-43. [PMID: 34673932 DOI: 10.1093/intimm/dxab097] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 10/20/2021] [Indexed: 11/14/2022] Open
Abstract
Marginal zone B cells (MZB) represent a unique B cell subpopulation that rapidly differentiate into IgM-secreting plasma cells in response to T-independent (T-I) antigen. Sphingosine 1-phosphate (S1P) promotes MZB localization to the marginal zone. However, intracellular molecules involved in MZB localization and migration remain largely unknown. Here we show that MZB lacking the Glia maturation factor-γ (GMFG) are impaired in chemotaxis toward S1P under both in vitro and in vivo conditions, suggesting that GMFG is an effector downstream of S1P receptors. GMFG undergoes serine phosphorylation upon S1P stimulation and is required for S1P-induced desensitization of S1P receptor 1 (S1PR1). Compared with wild type mice, Gmfg -/- mice produce elevated levels of 4-hydroxy-3-nitrophenyl-acetyl (NP)-specific IgM against a T-I type II antigen, NP-Ficoll, accompanied by dysregulated MZB localization. These results identify GMFG as a regulator of S1P-induced MZB chemotaxis and reveal a role for MZB localization in the marginal zone for optimal IgM production against a T-I antigen.
Collapse
Affiliation(s)
- Yingqian Li
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Yue Tang
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Jun Liu
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Xin Meng
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Ying Wang
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Qing Min
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Rongjian Hong
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai200030, China
| | - Takeshi Tsubata
- Department of Immunology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Koji Hase
- Division of Biochemistry, Faculty of Pharmacy, Keio University, Tokyo, Japan
| | - Ji-Yang Wang
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China.,Department of Immunology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo 113-8510, Japan.,Department of Clinical Immunology, Children's Hospital of Fudan University, Shanghai, 200032, China.,Department of Microbiology and Immunology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
14
|
Yang Y, He X, Tang QQ, Shao YC, Song WJ, Gong PJ, Zeng YF, Huang SR, Zhou JY, Wan HF, Wei L, Zhang JW. GMFG Has Potential to Be a Novel Prognostic Marker and Related to Immune Infiltrates in Breast Cancer. Front Oncol 2021; 11:629633. [PMID: 34367945 PMCID: PMC8343142 DOI: 10.3389/fonc.2021.629633] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 06/30/2021] [Indexed: 12/24/2022] Open
Abstract
A growing amount of evidence has indicated immune genes perform a crucial position in the development and progression of breast cancer microenvironment. The purpose of our study was to identify immunogenic prognostic marker and explore potential regulatory mechanisms for breast cancer. We identified the genes related to ImmuneScore using ESTIMATE algorithm and WGCNA analysis, and we identified the differentially expressed gene (DEGs). Then, Glia maturation factor γ (GMFG) was determined as a predictive factor by intersecting immune-related genes with DEGs and survival analysis. We found the expression of GMFG was lower in breast cancer tissues compared with normal breast tissues, which was further verified by immunohistochemical (IHC). Moreover, the decreased expression of GMFG was significantly related to the poor prognosis. Besides, the expression of GMFG was related to the age, ER status, PR status, HER2 status and tumor size, which further suggested that the expression of GMFG was correlated with the subtype and the growth of tumor. The univariate and multivariate Cox regression analyses revealed that age, stage, the expression level of GMFG and radiotherapy were independent factors for predicting the prognosis of breast cancer patients. Subsequently, a prognostic model to predict the 3-year, 5-year and 10-year overall survival rate was developed based on the above four variables, and visualized as a nomogram. The values of area under the curve of the nomogram at 3-year, 5-year and 10-year were 0.897, 0.873 and 0.922, respectively, which was higher than stage in prognostic accuracy. In addition, we also found that GMFG expression level was correlated with sensitivity of some breast cancer chemotherapy drugs. Furthermore, the results of GSEA indicated immune-related pathways were mainly enriched in GMFG-high-expression group. CIBERSORT analysis for the proportion of tumor-infiltrating immune cells (TIICs) suggested that expression of GMFG was positively association with multiple kinds T-cell in BC. Among them, CD8+ T cells had the strongest correlation with GMFG expression, which revealed that GMFG might has an antitumor effect by increasing the infiltration of CD8+ T cells in breast cancer. Accordingly, GMFG has the potential to become a novel immune biomarker for the diagnosis and treatment of breast cancer.
Collapse
Affiliation(s)
- Yan Yang
- Department of Breast and Thyroid Surgery, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Xin He
- Department of Breast and Thyroid Surgery, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Qian-Qian Tang
- Department of Breast and Thyroid Surgery, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - You-Cheng Shao
- Department of Pathology and Pathophysiology, School of Basic Medicine, Wuhan University, Wuhan, China
| | - Wen-Jing Song
- Department of Breast and Thyroid Surgery, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Peng-Ju Gong
- Department of Breast and Thyroid Surgery, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Yi-Fan Zeng
- Department of Breast and Thyroid Surgery, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Si-Rui Huang
- Department of Breast and Thyroid Surgery, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Jiang-Yao Zhou
- Department of Breast and Thyroid Surgery, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Hui-Fang Wan
- Department of Breast and Thyroid Surgery, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Lei Wei
- Department of Pathology and Pathophysiology, School of Basic Medicine, Wuhan University, Wuhan, China
| | - Jing-Wei Zhang
- Department of Breast and Thyroid Surgery, Zhongnan Hospital, Wuhan University, Wuhan, China
| |
Collapse
|
15
|
Deretic N, Bolger-Munro M, Choi K, Abraham L, Gold MR. The Actin-Disassembly Protein Glia Maturation Factor γ Enhances Actin Remodeling and B Cell Antigen Receptor Signaling at the Immune Synapse. Front Cell Dev Biol 2021; 9:647063. [PMID: 34336818 PMCID: PMC8318000 DOI: 10.3389/fcell.2021.647063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 06/07/2021] [Indexed: 11/16/2022] Open
Abstract
Signaling by the B cell antigen receptor (BCR) initiates actin remodeling. The assembly of branched actin networks that are nucleated by the Arp2/3 complex exert outward force on the plasma membrane, allowing B cells to form membrane protrusions that can scan the surface of antigen-presenting cells (APCs). The resulting Arp2/3 complex-dependent actin retrograde flow promotes the centripetal movement and progressive coalescence of BCR microclusters, which amplifies BCR signaling. Glia maturation factor γ (GMFγ) is an actin disassembly-protein that releases Arp2/3 complex-nucleated actin filaments from actin networks. By doing so, GMFγ could either oppose the actions of the Arp2/3 complex or support Arp2/3 complex-nucleated actin polymerization by contributing to the recycling of actin monomers and Arp2/3 complexes. We now show that reducing the levels of GMFγ in human B cell lines via transfection with a specific siRNA impairs the ability of B cells to spread on antigen-coated surfaces, decreases the velocity of actin retrograde flow, diminishes the coalescence of BCR microclusters into a central cluster at the B cell-APC contact site, and decreases APC-induced BCR signaling. These effects of depleting GMFγ are similar to what occurs when the Arp2/3 complex is inhibited. This suggests that GMFγ cooperates with the Arp2/3 complex to support BCR-induced actin remodeling and amplify BCR signaling at the immune synapse.
Collapse
Affiliation(s)
- Nikola Deretic
- Department of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Madison Bolger-Munro
- Department of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Kate Choi
- Department of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Libin Abraham
- Department of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Michael R Gold
- Department of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
16
|
Lan A, Ren C, Wang X, Tong G, Yang G. Bioinformatics and survival analysis of glia maturation factor-γ in pan-cancers. BMC Cancer 2021; 21:423. [PMID: 33863293 PMCID: PMC8052856 DOI: 10.1186/s12885-021-08163-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 04/06/2021] [Indexed: 11/16/2022] Open
Abstract
Background Glia maturation factor-γ (GMFG) is reported to inhibit the actin nucleation through binding to the actin-related protein-2/3 complex (Arp2/3). Considering the main function of GMFG in actin remodeling, which is vital for immune response, angiogenesis, cell division and motility, GMFG is supposed to have important roles in tumor development, while up to now, only two studies described the role of GMFG in cancers. By investigating the clinical values of GMFG using The Cancer Genome Atlas (TCGA) data and the functional mechanisms of GMFG through analyses of Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichments, this study was aimed to better understand the impact of GMFG in pan-cancers and to draw more attentions for the future research of GMFG. Methods RNA-seq and clinical data of cancer patients were collected from TCGA and analyzed by the Kaplan-Meier methods. GO and KEGG analyses were conducted using the online tools from the Database for Annotation, Visualization and Integrated Discovery (DAVID). Results Compared to the corresponding normal samples, GMFG was significantly upregulated in glioblastoma (GBM), kidney clear cell carcinoma (KIRC), lower grade glioma (LGG), acute myeloid leukemia (LAML), and pancreatic cancer (PAAD), testicular cancer (TGCT), but was downregulated in kidney chromophobe (KICH), lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC) (P < 0.05 for all). High expression of GMFG predicted worse OS in GBM (HR = 1.5, P = 0.017), LGG (HR = 2.2, P < 0.001), LUSC (HR = 1.4, P = 0.022) and ocular melanomas (UVM) (HR = 7, P < 0.001), as well as worse DFS in LGG (HR = 1.8, P < 0.001) and prostate cancer (PRAD) (HR = 1.9, P = 0.004). In contrast, high expression of GMFG was associated with better OS in skin cutaneous melanoma (SKCM) (HR = 0.59, P < 0.001) and thymoma (THYM) (HR = 0.098, P = 0.031), as well as better DFS in bile duct cancer (CHOL) (HR = 0.2, P = 0.003). GMFG was mainly involved in the immune response, protein binding and cytokine-cytokine receptor interaction pathways, and was positively associated with multiple immunomodulators in most cancers. Conclusion Our study preliminarily identified that GMFG may cause different survivals for different cancers through modulating tumor progression, immune response status and tissue-specific tumor microenvironment (TME). Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-08163-2.
Collapse
Affiliation(s)
- Aihua Lan
- Central Laboratory, the Fifth People's Hospital of Shanghai, Fudan University, Shanghai, 200240, China
| | - Chunxia Ren
- Center for Reproductive Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200120, China
| | - Xiaoling Wang
- Center for Reproductive Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200120, China
| | - Guoqing Tong
- Center for Reproductive Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200120, China.
| | - Gong Yang
- Central Laboratory, the Fifth People's Hospital of Shanghai, Fudan University, Shanghai, 200240, China. .,Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Fudan University Shanghai Medical College, Shanghai, 200032, China.
| |
Collapse
|
17
|
Fabian C, Han M, Bjerkvig R, Niclou SP. Novel facets of glioma invasion. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 360:33-64. [PMID: 33962750 DOI: 10.1016/bs.ircmb.2020.08.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Malignant gliomas including Glioblastoma (GBM) are characterized by extensive diffuse tumor cell infiltration throughout the brain, which represents a major challenge in clinical disease management. While surgical resection is beneficial for patient outcome, it is well recognized that tumor cells at the invasive front or beyond stay behind and constitute a major source of tumor recurrence. Invasive glioma cells also represent a difficult therapeutic target since they are localized within normal functional brain areas with an intact blood brain barrier (BBB), thereby excluding most systemic drug treatments. Cell movement is mediated via the actin cytoskeleton where corresponding membrane protrusions play essential roles. This review provides an overview of the various paths of glioma cell invasion and underlines the specific aspects of the brain microenvironment. We highlight recent insight into tumor microtubes, neuro-glioma synapses and tumor metabolism which can regulate collective invasion processes. We also focus on the deregulation of actin cytoskeleton-related components in the context of glioma invasion, a deregulation that may be controlled by genomic alterations in tumor cells as well as by various external factors, including extracellular matrix (ECM) components and non-malignant stromal cells. Finally we critically assess the challenges and opportunities for therapeutically targeting glioma cell invasion.
Collapse
Affiliation(s)
- Carina Fabian
- NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg; Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Mingzhi Han
- Department of Biomedicine, University of Bergen, Bergen, Norway; Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University; Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Rolf Bjerkvig
- NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg; Department of Biomedicine, University of Bergen, Bergen, Norway.
| | - Simone P Niclou
- NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg; Department of Biomedicine, University of Bergen, Bergen, Norway.
| |
Collapse
|
18
|
Glia maturation factor-γ regulates murine macrophage iron metabolism and M2 polarization through mitochondrial ROS. Blood Adv 2020; 3:1211-1225. [PMID: 30971398 DOI: 10.1182/bloodadvances.2018026070] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 02/16/2019] [Indexed: 12/19/2022] Open
Abstract
In macrophages, cellular iron metabolism status is tightly integrated with macrophage phenotype and associated with mitochondrial function. However, how molecular events regulate mitochondrial activity to integrate regulation of iron metabolism and macrophage phenotype remains unclear. Here, we explored the important role of the actin-regulatory protein glia maturation factor-γ (GMFG) in the regulation of cellular iron metabolism and macrophage phenotype. We found that GMFG was downregulated in murine macrophages by exposure to iron and hydrogen peroxide. GMFG knockdown altered the expression of iron metabolism proteins and increased iron levels in murine macrophages and concomitantly promoted their polarization toward an anti-inflammatory M2 phenotype. GMFG-knockdown macrophages exhibited moderately increased levels of mitochondrial reactive oxygen species (mtROS), which were accompanied by decreased expression of some mitochondrial respiration chain components, including the iron-sulfur cluster assembly scaffold protein ISCU as well as the antioxidant enzymes SOD1 and SOD2. Importantly, treatment of GMFG-knockdown macrophages with the antioxidant N-acetylcysteine reversed the altered expression of iron metabolism proteins and significantly inhibited the enhanced gene expression of M2 macrophage markers, suggesting that mtROS is mechanistically linked to cellular iron metabolism and macrophage phenotype. Finally, GMFG interacted with the mitochondrial membrane ATPase ATAD3A, suggesting that GMFG knockdown-induced mtROS production might be attributed to alteration of mitochondrial function in macrophages. Our findings suggest that GMFG is an important regulator in cellular iron metabolism and macrophage phenotype and could be a novel therapeutic target for modulating macrophage function in immune and metabolic disorders.
Collapse
|
19
|
Chánez-Paredes S, Montoya-García A, Schnoor M. Cellular and pathophysiological consequences of Arp2/3 complex inhibition: role of inhibitory proteins and pharmacological compounds. Cell Mol Life Sci 2019; 76:3349-3361. [PMID: 31073744 PMCID: PMC11105272 DOI: 10.1007/s00018-019-03128-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 04/30/2019] [Accepted: 05/02/2019] [Indexed: 02/06/2023]
Abstract
The actin-related protein complex 2/3 (Arp2/3) generates branched actin networks important for many cellular processes such as motility, vesicular trafficking, cytokinesis, and intercellular junction formation and stabilization. Activation of Arp2/3 requires interaction with actin nucleation-promoting factors (NPFs). Regulation of Arp2/3 activity is achieved by endogenous inhibitory proteins through direct binding to Arp2/3 and competition with NPFs or by binding to Arp2/3-induced actin filaments and disassembly of branched actin networks. Arp2/3 inhibition has recently garnered more attention as it has been associated with attenuation of cancer progression, neurotoxic effects during drug abuse, and pathogen invasion of host cells. In this review, we summarize current knowledge on expression, inhibitory mechanisms and function of endogenous proteins able to inhibit Arp2/3 such as coronins, GMFs, PICK1, gadkin, and arpin. Moreover, we discuss cellular consequences of pharmacological Arp2/3 inhibition.
Collapse
Affiliation(s)
- Sandra Chánez-Paredes
- Department for Molecular Biomedicine, CINVESTAV-IPN, Av. IPN 2508, San Pedro Zacatenco, GAM, 07360, Mexico City, Mexico
| | - Armando Montoya-García
- Department for Molecular Biomedicine, CINVESTAV-IPN, Av. IPN 2508, San Pedro Zacatenco, GAM, 07360, Mexico City, Mexico
| | - Michael Schnoor
- Department for Molecular Biomedicine, CINVESTAV-IPN, Av. IPN 2508, San Pedro Zacatenco, GAM, 07360, Mexico City, Mexico.
| |
Collapse
|
20
|
Giannopoulou AF, Konstantakou EG, Velentzas AD, Avgeris SN, Avgeris M, Papandreou NC, Zoi I, Filippa V, Katarachia S, Lampidonis AD, Prombona A, Syntichaki P, Piperi C, Basdra EK, Iconomidou V, Papadavid E, Anastasiadou E, Papassideri IS, Papavassiliou AG, Voutsinas GE, Scorilas A, Stravopodis DJ. Gene-Specific Intron Retention Serves as Molecular Signature that Distinguishes Melanoma from Non-Melanoma Cancer Cells in Greek Patients. Int J Mol Sci 2019; 20:937. [PMID: 30795533 PMCID: PMC6412294 DOI: 10.3390/ijms20040937] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 02/15/2019] [Accepted: 02/20/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Skin cancer represents the most common human malignancy, and it includes BCC, SCC, and melanoma. Since melanoma is one of the most aggressive types of cancer, we have herein attempted to develop a gene-specific intron retention signature that can distinguish BCC and SCC from melanoma biopsy tumors. METHODS Intron retention events were examined through RT-sqPCR protocols, using total RNA preparations derived from BCC, SCC, and melanoma Greek biopsy specimens. Intron-hosted miRNA species and their target transcripts were predicted via the miRbase and miRDB bioinformatics platforms, respectively. Ιntronic ORFs were recognized through the ORF Finder application. Generation and visualization of protein interactomes were achieved by the IntAct and Cytoscape softwares, while tertiary protein structures were produced by using the I-TASSER online server. RESULTS c-MYC and Sestrin-1 genes proved to undergo intron retention specifically in melanoma. Interaction maps of proteins encoded by genes being potentially targeted by retained intron-accommodated miRNAs were generated and SRPX2 was additionally delivered to our melanoma-specific signature. Novel ORFs were identified in MCT4 and Sestrin-1 introns, with potentially critical roles in melanoma development. CONCLUSIONS The property of c-MYC, Sestrin-1, and SRPX2 genes to retain specific introns could be clinically used to molecularly differentiate non-melanoma from melanoma tumors.
Collapse
Affiliation(s)
- Aikaterini F Giannopoulou
- Section of Cell Biology and Biophysics, Department of Biology, School of Science, National and Kapodistrian University of Athens, 15701 Athens, Greece.
| | - Eumorphia G Konstantakou
- Section of Cell Biology and Biophysics, Department of Biology, School of Science, National and Kapodistrian University of Athens, 15701 Athens, Greece.
| | - Athanassios D Velentzas
- Section of Cell Biology and Biophysics, Department of Biology, School of Science, National and Kapodistrian University of Athens, 15701 Athens, Greece.
| | - Socratis N Avgeris
- Laboratory of Molecular Carcinogenesis and Rare Disease Genetics, Institute of Biosciences and Applications, National Center for Scientific Research "Demokritos", 15310 Athens, Greece.
| | - Margaritis Avgeris
- Section of Biochemistry and Molecular Biology, Department of Biology, School of Science, National and Kapodistrian University of Athens, 15701 Athens, Greece.
| | - Nikos C Papandreou
- Section of Cell Biology and Biophysics, Department of Biology, School of Science, National and Kapodistrian University of Athens, 15701 Athens, Greece.
| | - Ilianna Zoi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece.
| | - Vicky Filippa
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece.
| | - Stamatia Katarachia
- Section of Cell Biology and Biophysics, Department of Biology, School of Science, National and Kapodistrian University of Athens, 15701 Athens, Greece.
| | - Antonis D Lampidonis
- Section of Cell Biology and Biophysics, Department of Biology, School of Science, National and Kapodistrian University of Athens, 15701 Athens, Greece.
| | - Anastasia Prombona
- Laboratory of Chronobiology, Institute of Biosciences and Applications, National Center for Scientific Research "Demokritos", 15310 Athens, Greece.
| | - Popi Syntichaki
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece.
| | - Christina Piperi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece.
| | - Efthimia K Basdra
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece.
| | - Vassiliki Iconomidou
- Section of Cell Biology and Biophysics, Department of Biology, School of Science, National and Kapodistrian University of Athens, 15701 Athens, Greece.
| | - Evangelia Papadavid
- 2nd Department of Dermatology and Venereology, Medical School, National and Kapodistrian University of Athens, "Attikon" University Hospital, 12462 Athens, Greece.
| | - Ema Anastasiadou
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece.
| | - Issidora S Papassideri
- Section of Cell Biology and Biophysics, Department of Biology, School of Science, National and Kapodistrian University of Athens, 15701 Athens, Greece.
| | - Athanasios G Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece.
| | - Gerassimos E Voutsinas
- Laboratory of Molecular Carcinogenesis and Rare Disease Genetics, Institute of Biosciences and Applications, National Center for Scientific Research "Demokritos", 15310 Athens, Greece.
| | - Andreas Scorilas
- Section of Biochemistry and Molecular Biology, Department of Biology, School of Science, National and Kapodistrian University of Athens, 15701 Athens, Greece.
| | - Dimitrios J Stravopodis
- Section of Cell Biology and Biophysics, Department of Biology, School of Science, National and Kapodistrian University of Athens, 15701 Athens, Greece.
| |
Collapse
|
21
|
Shroff N, Ander BP, Zhan X, Stamova B, Liu D, Hull H, Hamade FR, Dykstra-Aiello C, Ng K, Sharp FR, Jickling GC. HDAC9 Polymorphism Alters Blood Gene Expression in Patients with Large Vessel Atherosclerotic Stroke. Transl Stroke Res 2019; 10:19-25. [PMID: 29651704 PMCID: PMC6186202 DOI: 10.1007/s12975-018-0619-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 01/06/2018] [Accepted: 03/06/2018] [Indexed: 12/20/2022]
Abstract
The histone deacetylase 9 (HDAC9) polymorphism rs2107595 is associated with an increased risk for large vessel atherosclerotic stroke (LVAS). In humans, there remains a need to better understand this HDAC9 polymorphism's contribution to large vessel stroke. In this pilot study, we evaluated whether the HDAC9 polymorphism rs2107595 is associated with differences in leukocyte gene expression in patients with LVAS. HDAC9 SNP rs2107595 was genotyped in 155 patients (43 LVAS and 112 vascular risk factor controls). RNA isolated from blood was processed on whole genome microarrays. Gene expression was compared between HDAC9 risk allele-positive and risk allele-negative LVAS patients and controls. Functional analysis identified canonical pathways and molecular functions associated with rs2107595 in LVAS. In HDAC9 SNP rs2107595 risk allele-positive LVAS patients, there were 155 genes differentially expressed compared to risk allele-negative patients (fold change > |1.2|, p < 0.05). The 155 genes separated the risk allele-positive and risk allele-negative LVAS patients on a principal component analysis. Pathways associated with HDAC9 risk allele-positive status involved IL-6 signaling, cholesterol efflux, and platelet aggregation. These preliminary data suggest an association with the HDAC9 rs2107595 risk allele and peripheral immune, lipid, and clotting systems in LVAS. Further study is required to evaluate whether these differences are related to large vessel atherosclerosis and stroke risk.
Collapse
Affiliation(s)
- Natasha Shroff
- Department of Neurology, University of California at Davis School of Medicine, Sacramento, CA, 95817, USA.
- MIND Institute Wet Labs, Room 2415, 2805 50th Street, Sacramento, CA, 95817, USA.
| | - Bradley P Ander
- Department of Neurology, University of California at Davis School of Medicine, Sacramento, CA, 95817, USA
| | - Xinhua Zhan
- Department of Neurology, University of California at Davis School of Medicine, Sacramento, CA, 95817, USA
| | - Boryana Stamova
- Department of Neurology, University of California at Davis School of Medicine, Sacramento, CA, 95817, USA
| | - DaZhi Liu
- Department of Neurology, University of California at Davis School of Medicine, Sacramento, CA, 95817, USA
| | - Heather Hull
- Department of Neurology, University of California at Davis School of Medicine, Sacramento, CA, 95817, USA
| | - Farah R Hamade
- Department of Neurology, University of California at Davis School of Medicine, Sacramento, CA, 95817, USA
| | - Cheryl Dykstra-Aiello
- Department of Neurology, University of California at Davis School of Medicine, Sacramento, CA, 95817, USA
| | - Kwan Ng
- Department of Neurology, University of California at Davis School of Medicine, Sacramento, CA, 95817, USA
| | - Frank R Sharp
- Department of Neurology, University of California at Davis School of Medicine, Sacramento, CA, 95817, USA
| | - Glen C Jickling
- Department of Neurology, University of California at Davis School of Medicine, Sacramento, CA, 95817, USA
| |
Collapse
|
22
|
Maheshwari D, Shukla VK, Jain A, Tripathi S, Kumar D, Arora A. Solution structure and dynamics of glia maturation factor from Caenorhabditis elegans. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2018; 1866:1008-1020. [PMID: 29981887 DOI: 10.1016/j.bbapap.2018.06.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 06/11/2018] [Accepted: 06/27/2018] [Indexed: 10/28/2022]
Abstract
BACKGROUND The GMF class of the ADF-H domain family proteins regulate actin dynamics by binding to the Arp2/3 complex and F-actin through their Site-1 and Site-2, respectively. CeGMF of C. elegans is analogous to GMFγ of human and mouse and is 138 amino acids in length. METHODS We have characterized the solution structure and dynamics of CeGMF by solution NMR spectroscopy and its thermal stability by DSC. RESULTS The solution structure of CeGMF shows canonical ADF-H fold with two additional β-strands in the β4-β5 loop region. The Site-1 of CeGMF is well formed and residues of all three regions of Site-1 show dynamic flexibility. However, the β4-β5 loop of Site-2 is less inclined towards the C-terminal, as the latter is truncated by four residues in comparison to GMF isoforms of human and mouse. Regions of Site-2 show motions on ns-ps timescale, but dynamic flexibility of β4-β5 loop is low in comparison to corresponding F-loop region of ADF/cofilin UNC-60B. A general difference in packing of α3 and α1 between GMF and ADF/cofilins was noticed. Additionally, thermal stability of CeGMF was significantly higher than its ADF/cofilin homologs. CONCLUSION We have presented the first solution structure of GMF from C. elegans, which highlights the structural differences between the Site-2 of CeGMF and mammalian GMF isoforms. Further, we have seen the differences in structure, dynamics, and thermal stability of GMF and ADF/cofilin. GENERAL SIGNIFICANCE This study provides a useful insight to structural and dynamics factors that define the specificity of GMF towards Arp2/3 complex.
Collapse
Affiliation(s)
- Diva Maheshwari
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific and Innovative Research, New Delhi 110025, India
| | - Vaibhav Kumar Shukla
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Anupam Jain
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Sarita Tripathi
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Dinesh Kumar
- Centre of Biomedical Research (CBMR), Sanjay Gandhi Post-Graduate Institute of Medical Sciences, Raibareli Road, Lucknow, Uttar Pradesh 226014, India
| | - Ashish Arora
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific and Innovative Research, New Delhi 110025, India.
| |
Collapse
|
23
|
Goode BL, Sweeney MO, Eskin JA. GMF as an Actin Network Remodeling Factor. Trends Cell Biol 2018; 28:749-760. [PMID: 29779865 DOI: 10.1016/j.tcb.2018.04.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 04/22/2018] [Accepted: 04/23/2018] [Indexed: 10/24/2022]
Abstract
Glia maturation factor (GMF) has recently been established as a regulator of the actin cytoskeleton with a unique role in remodeling actin network architecture. Conserved from yeast to mammals, GMF is one of five members of the ADF-H family of actin regulatory proteins, which includes ADF/cofilin, Abp1/Drebrin, Twinfilin, and Coactosin. GMF does not bind actin, but instead binds the Arp2/3 complex with high affinity. Through this association, GMF catalyzes the debranching of actin filament networks and inhibits actin nucleation by Arp2/3 complex. Here, we discuss GMF's emerging role in controlling actin filament spatial organization and dynamics underlying cell motility, endocytosis, and other biological processes. Further, we attempt to reconcile these functions with its earlier characterization as a cell differentiation factor.
Collapse
Affiliation(s)
- Bruce L Goode
- Rosenstiel Basic Medical Sciences Research Center, Brandeis University, 415 South Street, Waltham, MA 02454 USA.
| | - Meredith O Sweeney
- Rosenstiel Basic Medical Sciences Research Center, Brandeis University, 415 South Street, Waltham, MA 02454 USA
| | - Julian A Eskin
- Rosenstiel Basic Medical Sciences Research Center, Brandeis University, 415 South Street, Waltham, MA 02454 USA
| |
Collapse
|
24
|
Molinie N, Gautreau A. The Arp2/3 Regulatory System and Its Deregulation in Cancer. Physiol Rev 2017; 98:215-238. [PMID: 29212790 DOI: 10.1152/physrev.00006.2017] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 05/10/2017] [Accepted: 05/11/2017] [Indexed: 02/07/2023] Open
Abstract
The Arp2/3 complex is an evolutionary conserved molecular machine that generates branched actin networks. When activated, the Arp2/3 complex contributes the actin branched junction and thus cross-links the polymerizing actin filaments in a network that exerts a pushing force. The different activators initiate branched actin networks at the cytosolic surface of different cellular membranes to promote their protrusion, movement, or scission in cell migration and membrane traffic. Here we review the structure, function, and regulation of all the direct regulators of the Arp2/3 complex that induce or inhibit the initiation of a branched actin network and that controls the stability of its branched junctions. Our goal is to present recent findings concerning novel inhibitory proteins or the regulation of the actin branched junction and place these in the context of what was previously known to provide a global overview of how the Arp2/3 complex is regulated in human cells. We focus on the human set of Arp2/3 regulators to compare normal Arp2/3 regulation in untransformed cells to the deregulation of the Arp2/3 system observed in patients affected by various cancers. In many cases, these deregulations promote cancer progression and have a direct impact on patient survival.
Collapse
Affiliation(s)
- Nicolas Molinie
- Ecole Polytechnique, Université Paris-Saclay, CNRS UMR 7654, Palaiseau, France; and Moscow Institute of Physics and Technology, Life Sciences Center, Dolgoprudny, Russia
| | - Alexis Gautreau
- Ecole Polytechnique, Université Paris-Saclay, CNRS UMR 7654, Palaiseau, France; and Moscow Institute of Physics and Technology, Life Sciences Center, Dolgoprudny, Russia
| |
Collapse
|
25
|
Kempuraj D, Thangavel R, Selvakumar GP, Zaheer S, Ahmed ME, Raikwar SP, Zahoor H, Saeed D, Natteru PA, Iyer S, Zaheer A. Brain and Peripheral Atypical Inflammatory Mediators Potentiate Neuroinflammation and Neurodegeneration. Front Cell Neurosci 2017; 11:216. [PMID: 28790893 PMCID: PMC5522882 DOI: 10.3389/fncel.2017.00216] [Citation(s) in RCA: 263] [Impact Index Per Article: 32.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Accepted: 07/05/2017] [Indexed: 12/18/2022] Open
Abstract
Neuroinflammatory response is primarily a protective mechanism in the brain. However, excessive and chronic inflammatory responses can lead to deleterious effects involving immune cells, brain cells and signaling molecules. Neuroinflammation induces and accelerates pathogenesis of Parkinson’s disease (PD), Alzheimer’s disease (AD) and Multiple sclerosis (MS). Neuroinflammatory pathways are indicated as novel therapeutic targets for these diseases. Mast cells are immune cells of hematopoietic origin that regulate inflammation and upon activation release many proinflammatory mediators in systemic and central nervous system (CNS) inflammatory conditions. In addition, inflammatory mediators released from activated glial cells induce neurodegeneration in the brain. Systemic inflammation-derived proinflammatory cytokines/chemokines and other factors cause a breach in the blood brain-barrier (BBB) thereby allowing for the entry of immune/inflammatory cells including mast cell progenitors, mast cells and proinflammatory cytokines and chemokines into the brain. These peripheral-derived factors and intrinsically generated cytokines/chemokines, α-synuclein, corticotropin-releasing hormone (CRH), substance P (SP), beta amyloid 1–42 (Aβ1–42) peptide and amyloid precursor proteins can activate glial cells, T-cells and mast cells in the brain can induce additional release of inflammatory and neurotoxic molecules contributing to chronic neuroinflammation and neuronal death. The glia maturation factor (GMF), a proinflammatory protein discovered in our laboratory released from glia, activates mast cells to release inflammatory cytokines and chemokines. Chronic increase in the proinflammatory mediators induces neurotoxic Aβ and plaque formation in AD brains and neurodegeneration in PD brains. Glial cells, mast cells and T-cells can reactivate each other in neuroinflammatory conditions in the brain and augment neuroinflammation. Further, inflammatory mediators from the brain can also enter into the peripheral system through defective BBB, recruit immune cells into the brain, and exacerbate neuroinflammation. We suggest that mast cell-associated inflammatory mediators from systemic inflammation and brain could augment neuroinflammation and neurodegeneration in the brain. This review article addresses the role of some atypical inflammatory mediators that are associated with mast cell inflammation and their activation of glial cells to induce neurodegeneration.
Collapse
Affiliation(s)
- Duraisamy Kempuraj
- Harry S. Truman Memorial Veteran's Hospital, U.S. Department of Veterans AffairsColumbia, MO, United States.,Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of MissouriColumbia, MO, United States
| | - Ramasamy Thangavel
- Harry S. Truman Memorial Veteran's Hospital, U.S. Department of Veterans AffairsColumbia, MO, United States.,Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of MissouriColumbia, MO, United States
| | - Govindhasamy P Selvakumar
- Harry S. Truman Memorial Veteran's Hospital, U.S. Department of Veterans AffairsColumbia, MO, United States.,Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of MissouriColumbia, MO, United States
| | - Smita Zaheer
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of MissouriColumbia, MO, United States
| | - Mohammad E Ahmed
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of MissouriColumbia, MO, United States
| | - Sudhanshu P Raikwar
- Harry S. Truman Memorial Veteran's Hospital, U.S. Department of Veterans AffairsColumbia, MO, United States.,Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of MissouriColumbia, MO, United States
| | - Haris Zahoor
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of MissouriColumbia, MO, United States
| | - Daniyal Saeed
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of MissouriColumbia, MO, United States
| | - Prashant A Natteru
- Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of MissouriColumbia, MO, United States
| | - Shankar Iyer
- Harry S. Truman Memorial Veteran's Hospital, U.S. Department of Veterans AffairsColumbia, MO, United States.,Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of MissouriColumbia, MO, United States
| | - Asgar Zaheer
- Harry S. Truman Memorial Veteran's Hospital, U.S. Department of Veterans AffairsColumbia, MO, United States.,Department of Neurology and the Center for Translational Neuroscience, School of Medicine, University of MissouriColumbia, MO, United States
| |
Collapse
|
26
|
Wang H, Chen Z, Chang H, Mu X, Deng W, Yuan Z, Yao F, Liu Y, Mai R, Wu B. Expression of glia maturation factor γ is associated with colorectal cancer metastasis and its downregulation suppresses colorectal cancer cell migration and invasion in vitro. Oncol Rep 2017; 37:929-936. [PMID: 28075454 DOI: 10.3892/or.2017.5361] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 11/11/2016] [Indexed: 11/05/2022] Open
Abstract
Glia maturation factor γ (GMFG) functions to reorganize the actin cytoskeleton and appears to play a causative role in cell migration and adherence. The present study assessed GMFG expression in colorectal cancer cells and tissue specimens and then explored the role of GMFG in colorectal cancer progression in vitro. GMFG protein was highly expressed in colorectal cancer tissues and a metastatic colon cancer cell line. Knockdown of GMFG expression using GMFG siRNA or anti-GMFG antibody decreased the capacity of colon cancer LoVo cell migration and invasion in vitro, while recombinant GMFG treatment induced LoVo cell migration. Furthermore, GMFG knockdown also decreased expression of MMP2 protein and reversed epithelial-mesenchymal transition (EMT) phenotypes in LoVo cells. Co-culture of LoVo cells with human umbilical vein endothelial cells (HUVECs) and exogenous GMFG treatment promoted LoVo cell migration and invasion. The data from the present study indicate that GMFG should be further evaluated as a biomarker for detection of colorectal cancer metastasis and that the targeting of GMFG expression or function could be a novel strategy in the future control of colorectal cancer.
Collapse
Affiliation(s)
- Huili Wang
- Research Center of Clinical Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510000, P.R. China
| | - Zhijiang Chen
- Pediatric Center of Zhujiang Hospital, Southern Medical University, Guangzhou 510000, P.R. China
| | - Hongen Chang
- Department of Neurology, Liuzhou Hospital of Traditional Chinese Medicine, Guangxi 545001, P.R. China
| | - Xiaoping Mu
- Guangdong Women and Children Hospital, Guangzhou 510000, P.R. China
| | - Wenyu Deng
- Guangdong Women and Children Hospital, Guangzhou 510000, P.R. China
| | - Zhaohu Yuan
- Department of Blood Transfusion Center, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou 510000, P.R. China
| | - Fang Yao
- Research Center of Clinical Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510000, P.R. China
| | - Yan Liu
- Guangdong Women and Children Hospital, Guangzhou 510000, P.R. China
| | - Rongjia Mai
- Guangdong Women and Children Hospital, Guangzhou 510000, P.R. China
| | - Bingyi Wu
- Research Center of Clinical Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510000, P.R. China
| |
Collapse
|
27
|
Cofilin-1 and Other ADF/Cofilin Superfamily Members in Human Malignant Cells. Int J Mol Sci 2016; 18:ijms18010010. [PMID: 28025492 PMCID: PMC5297645 DOI: 10.3390/ijms18010010] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 11/18/2016] [Accepted: 12/01/2016] [Indexed: 12/12/2022] Open
Abstract
Identification of actin-depolymerizing factor homology (ADF-H) domains in the structures of several related proteins led first to the formation of the ADF/cofilin family, which then expanded to the ADF/cofilin superfamily. This superfamily includes the well-studied cofilin-1 (Cfl-1) and about a dozen different human proteins that interact directly or indirectly with the actin cytoskeleton, provide its remodeling, and alter cell motility. According to some data, Cfl-1 is contained in various human malignant cells (HMCs) and is involved in the formation of malignant properties, including invasiveness, metastatic potential, and resistance to chemotherapeutic drugs. The presence of other ADF/cofilin superfamily proteins in HMCs and their involvement in the regulation of cell motility were discovered with the use of various OMICS technologies. In our review, we discuss the results of the study of Cfl-1 and other ADF/cofilin superfamily proteins, which may be of interest for solving different problems of molecular oncology, as well as for the prospects of further investigations of these proteins in HMCs.
Collapse
|
28
|
ox-LDL induces endothelial dysfunction by promoting Arp2/3 complex expression. Biochem Biophys Res Commun 2016; 475:182-8. [DOI: 10.1016/j.bbrc.2016.05.068] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 05/12/2016] [Indexed: 01/27/2023]
|
29
|
Aerbajinai W, Liu L, Zhu J, Kumkhaek C, Chin K, Rodgers GP. Glia Maturation Factor-γ Regulates Monocyte Migration through Modulation of β1-Integrin. J Biol Chem 2016; 291:8549-64. [PMID: 26895964 DOI: 10.1074/jbc.m115.674200] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Indexed: 12/30/2022] Open
Abstract
Monocyte migration requires the dynamic redistribution of integrins through a regulated endo-exocytosis cycle, but the complex molecular mechanisms underlying this process have not been fully elucidated. Glia maturation factor-γ (GMFG), a novel regulator of the Arp2/3 complex, has been shown to regulate directional migration of neutrophils and T-lymphocytes. In this study, we explored the important role of GMFG in monocyte chemotaxis, adhesion, and β1-integrin turnover. We found that knockdown of GMFG in monocytes resulted in impaired chemotactic migration toward formyl-Met-Leu-Phe (fMLP) and stromal cell-derived factor 1α (SDF-1α) as well as decreased α5β1-integrin-mediated chemoattractant-stimulated adhesion. These GMFG knockdown impaired effects could be reversed by cotransfection of GFP-tagged full-length GMFG. GMFG knockdown cells reduced the cell surface and total protein levels of α5β1-integrin and increased its degradation. Importantly, we demonstrate that GMFG mediates the ubiquitination of β1-integrin through knockdown or overexpression of GMFG. Moreover, GMFG knockdown retarded the efficient recycling of β1-integrin back to the plasma membrane following normal endocytosis of α5β1-integrin, suggesting that the involvement of GMFG in maintaining α5β1-integrin stability may occur in part by preventing ubiquitin-mediated degradation and promoting β1-integrin recycling. Furthermore, we observed that GMFG interacted with syntaxin 4 (STX4) and syntaxin-binding protein 4 (STXBP4); however, only knockdown of STXBP4, but not STX4, reduced monocyte migration and decreased β1-integrin cell surface expression. Knockdown of STXBP4 also substantially inhibited β1-integrin recycling in human monocytes. These results indicate that the effects of GMFG on monocyte migration and adhesion probably occur through preventing ubiquitin-mediated proteasome degradation of α5β1-integrin and facilitating effective β1-integrin recycling back to the plasma membrane.
Collapse
Affiliation(s)
- Wulin Aerbajinai
- From the Molecular and Clinical Hematology Branch, NHLBI, National Institutes of Health, Bethesda, Maryland 20892 and
| | - Lunhua Liu
- the Laboratory of Cellular and Molecular Biology, NCI, National Institutes of Health, Bethesda, Maryland 20892
| | - Jianqiong Zhu
- From the Molecular and Clinical Hematology Branch, NHLBI, National Institutes of Health, Bethesda, Maryland 20892 and
| | - Chutima Kumkhaek
- From the Molecular and Clinical Hematology Branch, NHLBI, National Institutes of Health, Bethesda, Maryland 20892 and
| | - Kyung Chin
- From the Molecular and Clinical Hematology Branch, NHLBI, National Institutes of Health, Bethesda, Maryland 20892 and
| | - Griffin P Rodgers
- From the Molecular and Clinical Hematology Branch, NHLBI, National Institutes of Health, Bethesda, Maryland 20892 and
| |
Collapse
|
30
|
Shimoda Y, Matsuo K, Kitamura Y, Ono K, Ueyama T, Matoba S, Yamada H, Wu T, Chen J, Emoto N, Ikeda K. Diabetes-Related Ankyrin Repeat Protein (DARP/Ankrd23) Modifies Glucose Homeostasis by Modulating AMPK Activity in Skeletal Muscle. PLoS One 2015; 10:e0138624. [PMID: 26398569 PMCID: PMC4580461 DOI: 10.1371/journal.pone.0138624] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 08/31/2015] [Indexed: 12/20/2022] Open
Abstract
Skeletal muscle is the major site for glucose disposal, the impairment of which closely associates with the glucose intolerance in diabetic patients. Diabetes-related ankyrin repeat protein (DARP/Ankrd23) is a member of muscle ankyrin repeat proteins, whose expression is enhanced in the skeletal muscle under diabetic conditions; however, its role in energy metabolism remains poorly understood. Here we report a novel role of DARP in the regulation of glucose homeostasis through modulating AMP-activated protein kinase (AMPK) activity. DARP is highly preferentially expressed in skeletal muscle, and its expression was substantially upregulated during myotube differentiation of C2C12 myoblasts. Interestingly, DARP-/- mice demonstrated better glucose tolerance despite similar body weight, while their insulin sensitivity did not differ from that in wildtype mice. We found that phosphorylation of AMPK, which mediates insulin-independent glucose uptake, in skeletal muscle was significantly enhanced in DARP-/- mice compared to that in wildtype mice. Gene silencing of DARP in C2C12 myotubes enhanced AMPK phosphorylation, whereas overexpression of DARP in C2C12 myoblasts reduced it. Moreover, DARP-silencing increased glucose uptake and oxidation in myotubes, which was abrogated by the treatment with AICAR, an AMPK activator. Of note, improved glucose tolerance in DARP-/- mice was abolished when mice were treated with AICAR. Mechanistically, gene silencing of DARP enhanced protein expression of LKB1 that is a major upstream kinase for AMPK in myotubes in vitro and the skeletal muscle in vivo. Together with the altered expression under diabetic conditions, our data strongly suggest that DARP plays an important role in the regulation of glucose homeostasis under physiological and pathological conditions, and thus DARP is a new therapeutic target for the treatment of diabetes mellitus.
Collapse
Affiliation(s)
- Yoshiaki Shimoda
- Department of Cardiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii, Kawaramachi-Hirokoji, Kamigyo, Kyoto 602–8566, Japan
| | - Kiyonari Matsuo
- Department of Cardiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii, Kawaramachi-Hirokoji, Kamigyo, Kyoto 602–8566, Japan
| | - Youhei Kitamura
- Department of Cardiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii, Kawaramachi-Hirokoji, Kamigyo, Kyoto 602–8566, Japan
| | - Kazunori Ono
- Department of Cardiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii, Kawaramachi-Hirokoji, Kamigyo, Kyoto 602–8566, Japan
| | - Tomomi Ueyama
- Department of Cardiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii, Kawaramachi-Hirokoji, Kamigyo, Kyoto 602–8566, Japan
| | - Satoaki Matoba
- Department of Cardiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii, Kawaramachi-Hirokoji, Kamigyo, Kyoto 602–8566, Japan
| | - Hiroyuki Yamada
- Department of Cardiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii, Kawaramachi-Hirokoji, Kamigyo, Kyoto 602–8566, Japan
| | - Tongbin Wu
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Ju Chen
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Noriaki Emoto
- Department of Clinical Pharmacy, Kobe Pharmaceutical University, 4-19-1 Motoyama-Kitamachi, Higashinada, Kobe6588558, Japan
| | - Koji Ikeda
- Department of Clinical Pharmacy, Kobe Pharmaceutical University, 4-19-1 Motoyama-Kitamachi, Higashinada, Kobe6588558, Japan
- * E-mail:
| |
Collapse
|
31
|
Haynes EM, Asokan SB, King SJ, Johnson HE, Haugh JM, Bear JE. GMFβ controls branched actin content and lamellipodial retraction in fibroblasts. J Cell Biol 2015; 209:803-12. [PMID: 26101216 PMCID: PMC4477851 DOI: 10.1083/jcb.201501094] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The primary activity of GMFβ in vivo is actin branch disassembly (and not inhibition of Arp2/3 activation), and this activity plays an important role in lamellipodial dynamics and directional migration toward ECM cues. The lamellipodium is an important structure for cell migration containing branched actin nucleated via the Arp2/3 complex. The formation of branched actin is relatively well studied, but less is known about its disassembly and how this influences migration. GMF is implicated in both Arp2/3 debranching and inhibition of Arp2/3 activation. Modulation of GMFβ, a ubiquitous GMF isoform, by depletion or overexpression resulted in changes in lamellipodial dynamics, branched actin content, and migration. Acute pharmacological inhibition of Arp2/3 by CK-666, coupled to quantitative live-cell imaging of the complex, showed that depletion of GMFβ decreased the rate of branched actin disassembly. These data, along with mutagenesis studies, suggest that debranching (not inhibition of Arp2/3 activation) is a primary activity of GMFβ in vivo. Furthermore, depletion or overexpression of GMFβ disrupted the ability of cells to directionally migrate to a gradient of fibronectin (haptotaxis). These data suggest that debranching by GMFβ plays an important role in branched actin regulation, lamellipodial dynamics, and directional migration.
Collapse
Affiliation(s)
- Elizabeth M Haynes
- UNC Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27514 Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Sreeja B Asokan
- UNC Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27514 Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Samantha J King
- UNC Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27514 Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Heath E Johnson
- Department of Chemical & Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695
| | - Jason M Haugh
- Department of Chemical & Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695
| | - James E Bear
- UNC Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27514 Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 Howard Hughes Medical Institute, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| |
Collapse
|
32
|
Abstract
Endocytosis, the process whereby the plasma membrane invaginates to form vesicles, is essential for bringing many substances into the cell and for membrane turnover. The mechanism driving clathrin-mediated endocytosis (CME) involves > 50 different protein components assembling at a single location on the plasma membrane in a temporally ordered and hierarchal pathway. These proteins perform precisely choreographed steps that promote receptor recognition and clustering, membrane remodeling, and force-generating actin-filament assembly and turnover to drive membrane invagination and vesicle scission. Many critical aspects of the CME mechanism are conserved from yeast to mammals and were first elucidated in yeast, demonstrating that it is a powerful system for studying endocytosis. In this review, we describe our current mechanistic understanding of each step in the process of yeast CME, and the essential roles played by actin polymerization at these sites, while providing a historical perspective of how the landscape has changed since the preceding version of the YeastBook was published 17 years ago (1997). Finally, we discuss the key unresolved issues and where future studies might be headed.
Collapse
Affiliation(s)
- Bruce L Goode
- Brandeis University, Department of Biology, Rosenstiel Center, Waltham, Massachusetts 02454
| | - Julian A Eskin
- Brandeis University, Department of Biology, Rosenstiel Center, Waltham, Massachusetts 02454
| | - Beverly Wendland
- The Johns Hopkins University, Department of Biology, Baltimore, Maryland 21218
| |
Collapse
|
33
|
Poukkula M, Hakala M, Pentinmikko N, Sweeney MO, Jansen S, Mattila J, Hietakangas V, Goode BL, Lappalainen P. GMF promotes leading-edge dynamics and collective cell migration in vivo. Curr Biol 2014; 24:2533-40. [PMID: 25308079 DOI: 10.1016/j.cub.2014.08.066] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 08/01/2014] [Accepted: 08/29/2014] [Indexed: 01/22/2023]
Abstract
Lamellipodia are dynamic actin-rich cellular extensions that drive advancement of the leading edge during cell migration. Lamellipodia undergo periodic extension and retraction cycles, but the molecular mechanisms underlying these dynamics and their role in cell migration have remained obscure. We show that glia-maturation factor (GMF), which is an Arp2/3 complex inhibitor and actin filament debranching factor, regulates lamellipodial protrusion dynamics in living cells. In cultured S2R(+) cells, GMF silencing resulted in an increase in the width of lamellipodial actin filament arrays. Importantly, live-cell imaging of mutant Drosophila egg chambers revealed that the dynamics of actin-rich protrusions in migrating border cells is diminished in the absence of GMF. Consequently, velocity of border cell clusters undergoing guided migration was reduced in GMF mutant flies. Furthermore, genetic studies demonstrated that GMF cooperates with the Drosophila homolog of Aip1 (flare) in promoting disassembly of Arp2/3-nucleated actin filament networks and driving border cell migration. These data suggest that GMF functions in vivo to promote the disassembly of Arp2/3-nucleated actin filament arrays, making an important contribution to cell migration within a 3D tissue environment.
Collapse
Affiliation(s)
- Minna Poukkula
- Institute of Biotechnology, University of Helsinki, P.O. Box 56, 00014 Helsinki, Finland
| | - Markku Hakala
- Institute of Biotechnology, University of Helsinki, P.O. Box 56, 00014 Helsinki, Finland
| | - Nalle Pentinmikko
- Institute of Biotechnology, University of Helsinki, P.O. Box 56, 00014 Helsinki, Finland
| | - Meredith O Sweeney
- Rosenstiel Center for Basic Biomedical Research, Brandeis University, Waltham, MA 02453, USA
| | - Silvia Jansen
- Rosenstiel Center for Basic Biomedical Research, Brandeis University, Waltham, MA 02453, USA
| | - Jaakko Mattila
- Institute of Biotechnology, University of Helsinki, P.O. Box 56, 00014 Helsinki, Finland; Department of Biosciences, University of Helsinki, P.O. Box 56, 00014 Helsinki, Finland
| | - Ville Hietakangas
- Institute of Biotechnology, University of Helsinki, P.O. Box 56, 00014 Helsinki, Finland; Department of Biosciences, University of Helsinki, P.O. Box 56, 00014 Helsinki, Finland
| | - Bruce L Goode
- Rosenstiel Center for Basic Biomedical Research, Brandeis University, Waltham, MA 02453, USA
| | - Pekka Lappalainen
- Institute of Biotechnology, University of Helsinki, P.O. Box 56, 00014 Helsinki, Finland.
| |
Collapse
|
34
|
Sender V, Stamme C. Lung cell-specific modulation of LPS-induced TLR4 receptor and adaptor localization. Commun Integr Biol 2014; 7:e29053. [PMID: 25136402 PMCID: PMC4134348 DOI: 10.4161/cib.29053] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2014] [Accepted: 04/29/2014] [Indexed: 01/07/2023] Open
Abstract
Lung infection by Gram-negative bacteria is a major cause of morbidity and mortality in humans. Lipopolysaccharide (LPS), located in the outer membrane of the Gram-negative bacterial cell wall, is a highly potent stimulus of immune and structural cells via the TLR4/MD2 complex whose function is sequentially regulated by defined subsets of adaptor proteins. Regulatory mechanisms of lung-specific defense pathways point at the crucial role of resident alveolar macrophages, alveolar epithelial cells, the TLR4 receptor pathway, and lung surfactant in shaping the innate immune response to Gram-negative bacteria and LPS. During the past decade intracellular spatiotemporal localization of TLR4 emerged as a key feature of TLR4 function. Here, we briefly review lung cell type- and compartment-specific mechanisms of LPS-induced TLR4 regulation with a focus on primary resident hematopoietic and structural cells as well as modifying microenvironmental factors involved.
Collapse
Affiliation(s)
- Vicky Sender
- Department of Microbiology, Tumor and Cell Biology; Karolinska Institutet; Stockholm, Sweden
| | - Cordula Stamme
- Division of Cellular Pneumology, Research Center Borstel, Leibniz-Center for Medicine and Biosciences; Borstel, Germany ; Department of Anesthesiology, University Hospital of Lübeck, Lübeck, Germany
| |
Collapse
|
35
|
Zuo P, Ma Y, Huang Y, Ye F, Wang P, Wang X, Zhou C, Lu W, Kong B, Xie X. High GMFG expression correlates with poor prognosis and promotes cell migration and invasion in epithelial ovarian cancer. Gynecol Oncol 2014; 132:745-51. [PMID: 24486602 DOI: 10.1016/j.ygyno.2014.01.044] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Revised: 01/18/2014] [Accepted: 01/23/2014] [Indexed: 10/25/2022]
Abstract
OBJECTIVE The aim of this study was to characterize the clinical significance of GMFG, a novel ADF/cofilin superfamily protein, and investigate its role in cell migration and invasion in epithelial ovarian cancer (EOC). METHODS The expression of GMFG in EOC tissues and ovarian cancer cell lines was evaluated by immunohistochemistry and immunoblotting respectively. The data were statistically analyzed for the associations of GMFG expression with clinicopathologic parameters and survival. In vitro cell migration and invasion assays were performed to determine the role of GMFG in cell migratory behaviors. The effect of GMFG on reorganization of actin cytoskeleton was investigated by immunostaining. RESULTS GMFG was overexpressed in EOC. Up-regulated GMFG expression was closely correlated with advanced FIGO stage and chemoresistance of the disease. EOC patients with higher GMFG expression showed poorer progression-free survival (PFS) and overall survival (OS). In vitro cellular assays revealed that GMFG promoted cell migration and invasion. GMFG expression altered actin cytoskeleton organization probably by interacting with the Arp2/3 complex. CONCLUSION GMFG expression independently predicts poorer prognosis in patients with EOC. Ectopic overexpression of GMFG contributes to the malignant biological behavior of ovarian cancer cells.
Collapse
Affiliation(s)
- Peng Zuo
- Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yuejiang Ma
- Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yongjie Huang
- Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Feng Ye
- Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Pei Wang
- Department of Gynecologic Oncology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xinyu Wang
- Department of Gynecologic Oncology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Caiyun Zhou
- Department of Pathology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Weiguo Lu
- Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China; Department of Gynecologic Oncology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Beihua Kong
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Ji'nan, Shandong, China
| | - Xing Xie
- Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China; Department of Gynecologic Oncology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
36
|
β-Glucuronidase is a suitable internal control gene for mRNA quantitation in pathophysiological and non-pathological livers. Exp Mol Pathol 2013; 95:131-5. [DOI: 10.1016/j.yexmp.2013.06.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Accepted: 06/05/2013] [Indexed: 11/18/2022]
|
37
|
Ecscr regulates insulin sensitivity and predisposition to obesity by modulating endothelial cell functions. Nat Commun 2013; 4:2389. [DOI: 10.1038/ncomms3389] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 08/02/2013] [Indexed: 01/08/2023] Open
|
38
|
Boczkowska M, Rebowski G, Dominguez R. Glia maturation factor (GMF) interacts with Arp2/3 complex in a nucleotide state-dependent manner. J Biol Chem 2013; 288:25683-25688. [PMID: 23897816 DOI: 10.1074/jbc.c113.493338] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Glia maturation factor (GMF) is a member of the actin-depolymerizing factor (ADF)/cofilin family. ADF/cofilin promotes disassembly of aged actin filaments, whereas GMF interacts specifically with Arp2/3 complex at branch junctions and promotes debranching. A distinguishing feature of ADF/cofilin is that it binds tighter to ADP-bound than to ATP-bound monomeric or filamentous actin. The interaction is also regulated by phosphorylation at Ser-3 of mammalian cofilin, which inhibits binding to actin. However, it is unknown whether these two factors play a role in the interaction of GMF with Arp2/3 complex. Here we show using isothermal titration calorimetry that mammalian GMF has very low affinity for ATP-bound Arp2/3 complex but binds ADP-bound Arp2/3 complex with 0.7 μM affinity. The phosphomimetic mutation S2E in GMF inhibits this interaction. GMF does not bind monomeric ATP- or ADP-actin, confirming its specificity for Arp2/3 complex. We further show that mammalian Arp2/3 complex nucleation activated by the WCA region of the nucleation-promoting factor N-WASP is not affected by GMF, whereas nucleation activated by the WCA region of WAVE2 is slightly inhibited at high GMF concentrations. Together, the results suggest that GMF functions by a mechanism similar to that of other ADF/cofilin family members, displaying a preference for ADP-Arp2/3 complex and undergoing inhibition by phosphorylation of a serine residue near the N terminus. Arp2/3 complex nucleation occurs in the ATP state, and nucleotide hydrolysis promotes debranching, suggesting that the higher affinity of GMF for ADP-Arp2/3 complex plays a physiological role by promoting debranching of aged branch junctions without interfering with Arp2/3 complex nucleation.
Collapse
Affiliation(s)
- Malgorzata Boczkowska
- From the Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Grzegorz Rebowski
- From the Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Roberto Dominguez
- From the Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104.
| |
Collapse
|
39
|
Structural basis for regulation of Arp2/3 complex by GMF. Nat Struct Mol Biol 2013; 20:1062-8. [PMID: 23893131 PMCID: PMC3766443 DOI: 10.1038/nsmb.2628] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2013] [Accepted: 06/05/2013] [Indexed: 01/15/2023]
Abstract
Arp2/3 complex mediates formation of complex cellular structures such as lamellapodia by nucleating branched actin filaments. Arp2/3 complex activity is precisely controlled by more than a dozen regulators, yet the structural mechanism by which regulators interact with the complex is unknown. GMF is a recently discovered regulator of Arp2/3 complex that can inhibit nucleation and dissemble branches. We solved the structure of the 240 kDa complex of Mus musculus GMF and Bos taurus Arp2/3 and found GMF binds to the barbed end of Arp2, overlapping with the proposed binding site of WASP family proteins. The structure suggests GMF can bind branch junctions like cofilin binds filament sides, consistent with a modified cofilin-like mechanism for debranching by GMF. The GMF-Arp2 interface reveals how the ADF-H actin-binding domain in GMF is exploited to specifically recognize Arp2/3 complex and not actin.
Collapse
|
40
|
Ydenberg CA, Padrick SB, Sweeney MO, Gandhi M, Sokolova O, Goode BL. GMF severs actin-Arp2/3 complex branch junctions by a cofilin-like mechanism. Curr Biol 2013; 23:1037-45. [PMID: 23727094 DOI: 10.1016/j.cub.2013.04.058] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Revised: 03/27/2013] [Accepted: 04/19/2013] [Indexed: 12/31/2022]
Abstract
BACKGROUND Branched actin filament networks driving cell motility, endocytosis, and intracellular transport are assembled in seconds by the Arp2/3 complex and must be equally rapidly debranched and turned over. One of the only factors known to promote debranching of actin networks is the yeast homolog of glia maturation factor (GMF), which is structurally related to the actin filament-severing protein cofilin. However, the identity of the molecular mechanism underlying debranching and whether this activity extends to mammalian GMF have remained open questions. RESULTS Using scanning mutagenesis and total internal reflection fluorescence microscopy, we show that GMF depends on two separate surfaces for debranching. One is analogous to the G-actin and F-actin binding site on cofilin, but we show using fluorescence anisotropy and chemical crosslinking that it instead interacts with actin-related proteins in the Arp2/3 complex. The other is analogous to a second F-actin binding site on cofilin, which in GMF appears to contact the first actin subunit in the daughter filament. We further show that GMF binds to the Arp2/3 complex with low nanomolar affinity and promotes the open conformation. Finally, we show that this debranching activity and mechanism are conserved for mammalian GMF. CONCLUSIONS GMF debranches filaments by a mechanism related to cofilin-mediated severing, but in which GMF has evolved to target molecular junctions between actin-related proteins in the Arp2/3 complex and actin subunits in the daughter filament of the branch. This activity and mechanism are conserved in GMF homologs from evolutionarily distant species.
Collapse
Affiliation(s)
- Casey A Ydenberg
- Rosenstiel Center for Basic Biomedical Research, Brandeis University, Waltham, MA 02453, USA
| | | | | | | | | | | |
Collapse
|
41
|
Das A, Das ND, Jung KH, Park JH, Lee HT, Han D, Choi MR, Kang SC, Chai YG. Proteomic changes induced by histone demethylase JMJD3 in TNF alpha-treated human monocytic (THP-1) cells. Mol Immunol 2013; 56:113-22. [PMID: 23711388 DOI: 10.1016/j.molimm.2013.04.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Revised: 03/16/2013] [Accepted: 04/23/2013] [Indexed: 01/03/2023]
Abstract
JMJD3, a Jumonji C family histone demethylase, plays an important role in the regulation of inflammation induced by the transcription factor nuclear factor-kappa B (NF-κB) in response to various stimuli. JMJD3 is a histone-3 lysine-27 trimethylation (H3K27me3) demethylase, a histone mark associated with transcriptional repression and activation of a diverse set of genes. The present study assessed stable JMJD3 knockdown (KD)-dependent proteomic profiling in human leukemia monocyte (THP-1) cells to analyze the JMJD3-mediated differential changes of marker expression in inflammatory cells. To analyze the protein expression profile of tumor necrosis factor-alpha (TNF-α)-stimulated JMJD3-kd THP-1 cells, we employed matrix-assisted-laser-desorption/ionization-time-of-flight mass spectrometry (MALDI-TOF MS). Additionally, Ingenuity Pathways Analysis (IPA) was applied to establish the molecular networks. A comparative proteomic profile was determined in TNF-α-treated both of JMJD3-kd THP-1 cells and THP-1 scrambled (sc) cells. The expression of tripartite motif protein (TRIM5), glutathione peroxidase (GPx), glia maturation factor-γ (GMFG), caspase recruitment domain family, member 14 (CARMA2), and dUTP pyrophosphatase were significantly down-regulated, whereas heat shock protein beta-1 (HspB1) and prohibition were significantly up-regulated in JMJD3-kd THP-1 cells. The molecular and signaling networks of the differentially expressed proteins in JMJD3-kd THP-1 cells were determined by IPA. The molecular network signatures and functional proteomics obtained in this study may facilitate the suppression of different key inflammatory regulators through JMJD3-attenuation, which would be crucial to evaluate potential therapeutic targets and to elucidate the molecular mechanism of JMJD3-kd dependent effects in THP-1 cells.
Collapse
Affiliation(s)
- Amitabh Das
- Department of Molecular & Life Science, Hanyang University, Ansan, Republic of Korea.
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Aerbajinai W, Lee K, Chin K, Rodgers GP. Glia maturation factor-γ negatively modulates TLR4 signaling by facilitating TLR4 endocytic trafficking in macrophages. THE JOURNAL OF IMMUNOLOGY 2013; 190:6093-103. [PMID: 23677465 DOI: 10.4049/jimmunol.1203048] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
TLR4 signaling must be tightly regulated to provide both effective immune protection and avoid inflammation-induced pathology. Thus, the mechanisms that negatively regulate the TLR4-triggered inflammatory response are of particular importance. Glia maturation factor-γ (GMFG), a novel actin depolymerization factor/cofilin superfamily protein that is expressed in inflammatory cells, has been implicated in mediating neutrophil and T cell migration, but its function in macrophage immune response remains unclear. In the current study, the role of GMFG in the LPS-induced TLR4-signaling pathway was investigated in THP-1 macrophages and human primary macrophages. LPS stimulation of macrophages decreased GMFG mRNA and protein expression. We show that GMFG negatively regulates LPS-induced activation of NF-κB-, MAPK-, and IRF3-signaling pathways and subsequent production of proinflammatory cytokines and type I IFN in human macrophages. We found that endogenous GMFG localized within early and late endosomes. GMFG knockdown delayed LPS-induced TLR4 internalization and caused prolonged TLR4 retention at the early endosome, suggesting that TLR4 transport from early to late endosomes is interrupted, which may contribute to enhanced LPS-induced TLR4 signaling. Taken together, our findings suggest that GMFG functions as a negative regulator of TLR4 signaling by facilitating TLR4 endocytic trafficking in macrophages.
Collapse
Affiliation(s)
- Wulin Aerbajinai
- Molecular and Clinical Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | |
Collapse
|
43
|
Zuo P, Fu Z, Tao T, Ye F, Chen L, Wang X, Lu W, Xie X. The expression of glia maturation factors and the effect of glia maturation factor-γ on angiogenic sprouting in zebrafish. Exp Cell Res 2013; 319:707-17. [PMID: 23333559 DOI: 10.1016/j.yexcr.2013.01.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Revised: 12/16/2012] [Accepted: 01/07/2013] [Indexed: 11/15/2022]
Abstract
Angiogenesis is a vital process for proper embryonic development, wound healing, malignant tumor growth and metastasis. Two glia maturation factor genes, glia maturation factor-β (GMFB) and glia maturation factor-γ (GMFG), presenting different expression patterns and distinct biological functions are found in vertebrates. But, the role of GMFB and GMFG in vascular development remains largely unknown. Here, we showed that both GMFB and GMFG are highly conserved in vertebrates. Whole-mount in situ hybridization and quantitative RT-PCR results revealed that GMFB and GMFG were differently expressed during zebrafish embryogenesis. GMFB was highly enriched in brain and GMFG was predominantly expressed in endothelial cells. By gene specific MO, knockdown of GMFG, but not GMFB, severely disrupted angiogenic sprouting of intersegmental vessels (ISVs), but this angiogenic defects were prevented by overexpression of a MO-resistant form of zebrafish GMFG mRNA. In addition, the expressions of angiogenic factors VEGF-A, STAT3, MMP2, MMP9, and MMP13 were significantly decreased in endothelial cells of GMFG morphants. Our findings provide the first in vivo evidence that GMFG is an important regulator for angiogenic sprouting during angiogenesis in zebrafish and suggest that GMFG may act as a novel potential target for anti-angiogenesis therapy in clinical settings.
Collapse
Affiliation(s)
- Peng Zuo
- Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Lippert DND, Wilkins JA. Glia maturation factor gamma regulates the migration and adherence of human T lymphocytes. BMC Immunol 2012; 13:21. [PMID: 22510515 PMCID: PMC3447661 DOI: 10.1186/1471-2172-13-21] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Accepted: 04/04/2012] [Indexed: 11/28/2022] Open
Abstract
Background Lymphocyte migration and chemotaxis are essential for effective immune surveillance. A critical aspect of migration is cell polarization and the extension of pseudopodia in the direction of movement. However, our knowledge of the underlying molecular mechanisms responsible for these events is incomplete. Proteomic analysis of the isolated leading edges of CXCL12 stimulated human T cell lines was used to identify glia maturation factor gamma (GMFG) as a component of the pseudopodia. This protein is predominantly expressed in hematopoietic cells and it has been shown to regulate cytoskeletal branching. The present studies were undertaken to examine the role of GMFG in lymphocyte migration. Results Microscopic analysis of migrating T-cells demonstrated that GMFG was distributed along the axis of movement with enrichment in the leading edge and behind the nucleus of these cells. Inhibition of GMFG expression in T cell lines and IL-2 dependent human peripheral blood T cells with shRNAmir reduced cellular basal and chemokine induced migration responses. The failure of the cells with reduced GMFG to migrate was associated with an apparent inability to detach from the substrates that they were moving on. It was also noted that these cells had an increased adherence to extracellular matrix proteins such as fibronectin. These changes in adherence were associated with altered patterns of β1 integrin expression and increased levels of activated integrins as detected with the activation specific antibody HUTS4. GMFG loss was also shown to increase the expression of the β2 integrin LFA-1 and to increase the adhesion of these cells to ICAM-1. Conclusions The present studies demonstrate that GMFG is a component of human T cell pseudopodia required for migration. The reduction in migration and increased adherence properties associated with inhibition of GMFG expression suggest that GMFG activity influences the regulation of integrin mediated adhesion.
Collapse
Affiliation(s)
- Dustin N D Lippert
- Manitoba Centre for Proteomics and Systems Biology, University of Manitoba, Winnipeg, Canada
| | | |
Collapse
|
45
|
Ydenberg CA, Smith BA, Breitsprecher D, Gelles J, Goode BL. Cease-fire at the leading edge: new perspectives on actin filament branching, debranching, and cross-linking. Cytoskeleton (Hoboken) 2011; 68:596-602. [PMID: 22002930 DOI: 10.1002/cm.20543] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2011] [Revised: 10/08/2011] [Accepted: 10/10/2011] [Indexed: 11/05/2022]
Abstract
Membrane protrusion at the leading edge of migrating cells is driven by the polymerization of actin. Recent studies using advanced imaging techniques raised a lively controversy about the morphology of these filaments; however, common ground between the two sides now appears to have been found. Here we discuss how the controversy has led to a deeper consideration of the architecture of actin networks underlying cell migration, and has helped define new challenges that lie ahead.
Collapse
Affiliation(s)
- Casey A Ydenberg
- Department of Biology and Rosenstiel Basic Medical Sciences Research Center, Brandeis University, Waltham, Massachusetts 02454, USA
| | | | | | | | | |
Collapse
|
46
|
Poukkula M, Kremneva E, Serlachius M, Lappalainen P. Actin-depolymerizing factor homology domain: a conserved fold performing diverse roles in cytoskeletal dynamics. Cytoskeleton (Hoboken) 2011; 68:471-90. [PMID: 21850706 DOI: 10.1002/cm.20530] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2011] [Revised: 06/29/2011] [Accepted: 08/05/2011] [Indexed: 11/09/2022]
Abstract
Actin filaments form contractile and protrusive structures that play central roles in many processes such as cell migration, morphogenesis, endocytosis, and cytokinesis. During these processes, the dynamics of the actin filaments are precisely regulated by a large array of actin-binding proteins. The actin-depolymerizing factor homology (ADF-H) domain is a structurally conserved protein motif, which promotes cytoskeletal dynamics by interacting with monomeric and/or filamentous actin, and with the Arp2/3 complex. Despite their structural homology, the five classes of ADF-H domain proteins display distinct biochemical activities and cellular roles, only parts of which are currently understood. ADF/cofilin promotes disassembly of aged actin filaments, whereas twinfilin inhibits actin filament assembly via sequestering actin monomers and interacting with filament barbed ends. GMF does not interact with actin, but instead binds Arp2/3 complex and promotes dissociation of Arp2/3-mediated filament branches. Abp1 and drebrin are multidomain proteins that interact with actin filaments and regulate the activities of other proteins during various actin-dependent processes. The exact function of coactosin is currently incompletely understood. In this review article, we discuss the biochemical functions, cellular roles, and regulation of the five groups of ADF-H domain proteins.
Collapse
Affiliation(s)
- Minna Poukkula
- Program in Cell and Molecular Biology, Institute of Biotechnology, University of Helsinki, Finland
| | | | | | | |
Collapse
|
47
|
Aerbajinai W, Liu L, Chin K, Zhu J, Parent CA, Rodgers GP. Glia maturation factor-γ mediates neutrophil chemotaxis. J Leukoc Biol 2011; 90:529-38. [PMID: 21653232 DOI: 10.1189/jlb.0710424] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Chemotaxis is fundamental to the directional migration of neutrophils toward endogenous and exogenous chemoattractants. Recent studies have demonstrated that ADF/cofilin superfamily members play important roles in reorganizing the actin cytoskeleton by disassembling actin filaments. GMFG, a novel ADF/cofilin superfamily protein that is expressed in inflammatory cells, has been implicated in regulating actin reorganization in microendothelial cells, but its function in neutrophils remains unclear. Here, we show that GMFG is an important regulator for cell migration and polarity in neutrophils. Knockdown of endogenous GMFG impaired fMLF- and IL-8 (CXCL8)-induced chemotaxis in dHL-60 cells. GMFG knockdown attenuated the formation of lamellipodia at the leading edge of cells exposed to fMLF or CXCL8, as well as the phosphorylation of p38 and PAK1/2 in response to fMLF or CXCL8. Live cell imaging revealed that GMFG was recruited to the leading edge of cells in response to fMLF, as well as CXCL8. Overexpression of GMFG enhanced phosphorylation of p38 but not of PAK1/2 in dHL-60 cells. In addition, we found that GMFG is associated with WAVE2. Taken together, our findings suggest that GMFG is a novel factor in regulating neutrophil chemotaxis by modulating actin cytoskeleton reorganization.
Collapse
Affiliation(s)
- Wulin Aerbajinai
- Molecular and Clinical Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892-2560, USA
| | | | | | | | | | | |
Collapse
|
48
|
D’Andrea FP, Safwat A, Kassem M, Gautier L, Overgaard J, Horsman MR. Cancer stem cell overexpression of nicotinamide N-methyltransferase enhances cellular radiation resistance. Radiother Oncol 2011; 99:373-8. [DOI: 10.1016/j.radonc.2011.05.086] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Revised: 05/26/2011] [Accepted: 05/27/2011] [Indexed: 12/29/2022]
|
49
|
Kutschera S, Weber H, Weick A, De Smet F, Genove G, Takemoto M, Prahst C, Riedel M, Mikelis C, Baulande S, Champseix C, Kummerer P, Conseiller E, Multon MC, Heroult M, Bicknell R, Carmeliet P, Betsholtz C, Augustin HG. Differential Endothelial Transcriptomics Identifies Semaphorin 3G as a Vascular Class 3 Semaphorin. Arterioscler Thromb Vasc Biol 2011; 31:151-9. [DOI: 10.1161/atvbaha.110.215871] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Objective—
To characterize the role of a vascular-expressed class 3 semaphorin (semaphorin 3G [Sema3G]).
Methods and Results—
Semaphorins have been identified as axon guidance molecules. Yet, they have more recently also been characterized as attractive and repulsive regulators of angiogenesis. Through a transcriptomic screen, we identified Sema3G as a molecule of angiogenic endothelial cells. Sema3G-deficient mice are viable and exhibit no overt vascular phenotype. Yet, LacZ expression in the Sema3G locus revealed intense arterial vascular staining in the angiogenic vasculature, starting at E9.5, which was detectable throughout adolescence and downregulated in adult vasculature. Sema3G is expressed as a full-length 100-kDa secreted molecule that is processed by furin proteases to yield 95- and a 65-kDa Sema domain–containing subunits. Full-length Sema3G binds to NP2, whereas processed Sema3G binds to NP1 and NP2. Expression profiling and cellular experiments identified autocrine effects of Sema3G on endothelial cells and paracrine effects on smooth muscle cells.
Conclusion—
Although the mouse knockout phenotype suggests compensatory mechanisms, the experiments identify Sema3G as a primarily endothelial cell–expressed class 3 semaphorin that controls endothelial and smooth muscle cell functions in autocrine and paracrine manners, respectively.
Collapse
Affiliation(s)
- Simone Kutschera
- From Vascular Oncology and Metastasis (S.K., A.W., C.P., M.R., C.M., M.H., and H.G.A.), German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), Heidelberg, Germany; Vascular Biology and Tumor Angiogenesis (S.K., A.W., C.P., M.H., and H.G.A.), Medical Faculty Mannheim (CBTM), Heidelberg University, Heidelberg, Germany; the Department of Vascular Biology and Angiogenesis Research (H.W., P.K., and H.G.A.), Tumor Biology Center, Freiburg, Germany; the Department for Transgene Technology and Gene
| | - Holger Weber
- From Vascular Oncology and Metastasis (S.K., A.W., C.P., M.R., C.M., M.H., and H.G.A.), German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), Heidelberg, Germany; Vascular Biology and Tumor Angiogenesis (S.K., A.W., C.P., M.H., and H.G.A.), Medical Faculty Mannheim (CBTM), Heidelberg University, Heidelberg, Germany; the Department of Vascular Biology and Angiogenesis Research (H.W., P.K., and H.G.A.), Tumor Biology Center, Freiburg, Germany; the Department for Transgene Technology and Gene
| | - Anja Weick
- From Vascular Oncology and Metastasis (S.K., A.W., C.P., M.R., C.M., M.H., and H.G.A.), German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), Heidelberg, Germany; Vascular Biology and Tumor Angiogenesis (S.K., A.W., C.P., M.H., and H.G.A.), Medical Faculty Mannheim (CBTM), Heidelberg University, Heidelberg, Germany; the Department of Vascular Biology and Angiogenesis Research (H.W., P.K., and H.G.A.), Tumor Biology Center, Freiburg, Germany; the Department for Transgene Technology and Gene
| | - Frederik De Smet
- From Vascular Oncology and Metastasis (S.K., A.W., C.P., M.R., C.M., M.H., and H.G.A.), German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), Heidelberg, Germany; Vascular Biology and Tumor Angiogenesis (S.K., A.W., C.P., M.H., and H.G.A.), Medical Faculty Mannheim (CBTM), Heidelberg University, Heidelberg, Germany; the Department of Vascular Biology and Angiogenesis Research (H.W., P.K., and H.G.A.), Tumor Biology Center, Freiburg, Germany; the Department for Transgene Technology and Gene
| | - Guillem Genove
- From Vascular Oncology and Metastasis (S.K., A.W., C.P., M.R., C.M., M.H., and H.G.A.), German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), Heidelberg, Germany; Vascular Biology and Tumor Angiogenesis (S.K., A.W., C.P., M.H., and H.G.A.), Medical Faculty Mannheim (CBTM), Heidelberg University, Heidelberg, Germany; the Department of Vascular Biology and Angiogenesis Research (H.W., P.K., and H.G.A.), Tumor Biology Center, Freiburg, Germany; the Department for Transgene Technology and Gene
| | - Minoru Takemoto
- From Vascular Oncology and Metastasis (S.K., A.W., C.P., M.R., C.M., M.H., and H.G.A.), German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), Heidelberg, Germany; Vascular Biology and Tumor Angiogenesis (S.K., A.W., C.P., M.H., and H.G.A.), Medical Faculty Mannheim (CBTM), Heidelberg University, Heidelberg, Germany; the Department of Vascular Biology and Angiogenesis Research (H.W., P.K., and H.G.A.), Tumor Biology Center, Freiburg, Germany; the Department for Transgene Technology and Gene
| | - Claudia Prahst
- From Vascular Oncology and Metastasis (S.K., A.W., C.P., M.R., C.M., M.H., and H.G.A.), German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), Heidelberg, Germany; Vascular Biology and Tumor Angiogenesis (S.K., A.W., C.P., M.H., and H.G.A.), Medical Faculty Mannheim (CBTM), Heidelberg University, Heidelberg, Germany; the Department of Vascular Biology and Angiogenesis Research (H.W., P.K., and H.G.A.), Tumor Biology Center, Freiburg, Germany; the Department for Transgene Technology and Gene
| | - Maria Riedel
- From Vascular Oncology and Metastasis (S.K., A.W., C.P., M.R., C.M., M.H., and H.G.A.), German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), Heidelberg, Germany; Vascular Biology and Tumor Angiogenesis (S.K., A.W., C.P., M.H., and H.G.A.), Medical Faculty Mannheim (CBTM), Heidelberg University, Heidelberg, Germany; the Department of Vascular Biology and Angiogenesis Research (H.W., P.K., and H.G.A.), Tumor Biology Center, Freiburg, Germany; the Department for Transgene Technology and Gene
| | - Constantinos Mikelis
- From Vascular Oncology and Metastasis (S.K., A.W., C.P., M.R., C.M., M.H., and H.G.A.), German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), Heidelberg, Germany; Vascular Biology and Tumor Angiogenesis (S.K., A.W., C.P., M.H., and H.G.A.), Medical Faculty Mannheim (CBTM), Heidelberg University, Heidelberg, Germany; the Department of Vascular Biology and Angiogenesis Research (H.W., P.K., and H.G.A.), Tumor Biology Center, Freiburg, Germany; the Department for Transgene Technology and Gene
| | - Sylvain Baulande
- From Vascular Oncology and Metastasis (S.K., A.W., C.P., M.R., C.M., M.H., and H.G.A.), German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), Heidelberg, Germany; Vascular Biology and Tumor Angiogenesis (S.K., A.W., C.P., M.H., and H.G.A.), Medical Faculty Mannheim (CBTM), Heidelberg University, Heidelberg, Germany; the Department of Vascular Biology and Angiogenesis Research (H.W., P.K., and H.G.A.), Tumor Biology Center, Freiburg, Germany; the Department for Transgene Technology and Gene
| | - Catherine Champseix
- From Vascular Oncology and Metastasis (S.K., A.W., C.P., M.R., C.M., M.H., and H.G.A.), German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), Heidelberg, Germany; Vascular Biology and Tumor Angiogenesis (S.K., A.W., C.P., M.H., and H.G.A.), Medical Faculty Mannheim (CBTM), Heidelberg University, Heidelberg, Germany; the Department of Vascular Biology and Angiogenesis Research (H.W., P.K., and H.G.A.), Tumor Biology Center, Freiburg, Germany; the Department for Transgene Technology and Gene
| | - Petra Kummerer
- From Vascular Oncology and Metastasis (S.K., A.W., C.P., M.R., C.M., M.H., and H.G.A.), German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), Heidelberg, Germany; Vascular Biology and Tumor Angiogenesis (S.K., A.W., C.P., M.H., and H.G.A.), Medical Faculty Mannheim (CBTM), Heidelberg University, Heidelberg, Germany; the Department of Vascular Biology and Angiogenesis Research (H.W., P.K., and H.G.A.), Tumor Biology Center, Freiburg, Germany; the Department for Transgene Technology and Gene
| | - Emmanuel Conseiller
- From Vascular Oncology and Metastasis (S.K., A.W., C.P., M.R., C.M., M.H., and H.G.A.), German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), Heidelberg, Germany; Vascular Biology and Tumor Angiogenesis (S.K., A.W., C.P., M.H., and H.G.A.), Medical Faculty Mannheim (CBTM), Heidelberg University, Heidelberg, Germany; the Department of Vascular Biology and Angiogenesis Research (H.W., P.K., and H.G.A.), Tumor Biology Center, Freiburg, Germany; the Department for Transgene Technology and Gene
| | - Marie-Christine Multon
- From Vascular Oncology and Metastasis (S.K., A.W., C.P., M.R., C.M., M.H., and H.G.A.), German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), Heidelberg, Germany; Vascular Biology and Tumor Angiogenesis (S.K., A.W., C.P., M.H., and H.G.A.), Medical Faculty Mannheim (CBTM), Heidelberg University, Heidelberg, Germany; the Department of Vascular Biology and Angiogenesis Research (H.W., P.K., and H.G.A.), Tumor Biology Center, Freiburg, Germany; the Department for Transgene Technology and Gene
| | - Melanie Heroult
- From Vascular Oncology and Metastasis (S.K., A.W., C.P., M.R., C.M., M.H., and H.G.A.), German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), Heidelberg, Germany; Vascular Biology and Tumor Angiogenesis (S.K., A.W., C.P., M.H., and H.G.A.), Medical Faculty Mannheim (CBTM), Heidelberg University, Heidelberg, Germany; the Department of Vascular Biology and Angiogenesis Research (H.W., P.K., and H.G.A.), Tumor Biology Center, Freiburg, Germany; the Department for Transgene Technology and Gene
| | - Roy Bicknell
- From Vascular Oncology and Metastasis (S.K., A.W., C.P., M.R., C.M., M.H., and H.G.A.), German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), Heidelberg, Germany; Vascular Biology and Tumor Angiogenesis (S.K., A.W., C.P., M.H., and H.G.A.), Medical Faculty Mannheim (CBTM), Heidelberg University, Heidelberg, Germany; the Department of Vascular Biology and Angiogenesis Research (H.W., P.K., and H.G.A.), Tumor Biology Center, Freiburg, Germany; the Department for Transgene Technology and Gene
| | - Peter Carmeliet
- From Vascular Oncology and Metastasis (S.K., A.W., C.P., M.R., C.M., M.H., and H.G.A.), German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), Heidelberg, Germany; Vascular Biology and Tumor Angiogenesis (S.K., A.W., C.P., M.H., and H.G.A.), Medical Faculty Mannheim (CBTM), Heidelberg University, Heidelberg, Germany; the Department of Vascular Biology and Angiogenesis Research (H.W., P.K., and H.G.A.), Tumor Biology Center, Freiburg, Germany; the Department for Transgene Technology and Gene
| | - Christer Betsholtz
- From Vascular Oncology and Metastasis (S.K., A.W., C.P., M.R., C.M., M.H., and H.G.A.), German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), Heidelberg, Germany; Vascular Biology and Tumor Angiogenesis (S.K., A.W., C.P., M.H., and H.G.A.), Medical Faculty Mannheim (CBTM), Heidelberg University, Heidelberg, Germany; the Department of Vascular Biology and Angiogenesis Research (H.W., P.K., and H.G.A.), Tumor Biology Center, Freiburg, Germany; the Department for Transgene Technology and Gene
| | - Hellmut G. Augustin
- From Vascular Oncology and Metastasis (S.K., A.W., C.P., M.R., C.M., M.H., and H.G.A.), German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), Heidelberg, Germany; Vascular Biology and Tumor Angiogenesis (S.K., A.W., C.P., M.H., and H.G.A.), Medical Faculty Mannheim (CBTM), Heidelberg University, Heidelberg, Germany; the Department of Vascular Biology and Angiogenesis Research (H.W., P.K., and H.G.A.), Tumor Biology Center, Freiburg, Germany; the Department for Transgene Technology and Gene
| |
Collapse
|
50
|
Hild G, Bugyi B, Nyitrai M. Conformational dynamics of actin: effectors and implications for biological function. Cytoskeleton (Hoboken) 2010; 67:609-29. [PMID: 20672362 PMCID: PMC3038201 DOI: 10.1002/cm.20473] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2010] [Accepted: 07/15/2010] [Indexed: 12/30/2022]
Abstract
Actin is a protein abundant in many cell types. Decades of investigations have provided evidence that it has many functions in living cells. The diverse morphology and dynamics of actin structures adapted to versatile cellular functions is established by a large repertoire of actin-binding proteins. The proper interactions with these proteins assume effective molecular adaptations from actin, in which its conformational transitions play essential role. This review attempts to summarise our current knowledge regarding the coupling between the conformational states of actin and its biological function.
Collapse
Affiliation(s)
- Gábor Hild
- Department of Biophysics, University of Pécs, Faculty of Medicine, Pécs, Szigeti str. 12, H-7624, Hungary
| | | | | |
Collapse
|