1
|
Cui Y, Yun T, Zhao GG. Bioinformatics analysis of clinical significance of STMN1 gene in prognosis and immune infiltration in hepatocellular carcinoma. Shijie Huaren Xiaohua Zazhi 2022; 30:411-419. [DOI: 10.11569/wcjd.v30.i9.411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND STMN1 (stathmin1), a member of the microtubule destabilizing protein family, is transcriptionally repressed by the functional tumor suppressor protein p53. STMN1 was first identified as a cellular phosphorylated protein overexpressed in leukemia in 1983. STMN1 was found to be upregulated in a variety of cancers, such as non-small cell lung cancer, breast cancer, and gastric cancer, and it can induce cell differentiation, proliferation, and migration in solid tumors and is associated with a poor clinical prognosis. However, the role and mechanism of STMN1 in hepatocellular carcinoma remain unclear.
AIM To assess the expression of STMN1 gene in hepatocellular carcinoma and its relationship with the clinical characteristics and prognosis of patients, and to explore the molecular mechanism of STMN1 gene in this malignancy.
METHODS Clinical information and high-throughput RNA-sequencing data of hepatocellular carcinoma patients were downloaded from The Cancer Genome Atlas (https://portal.gdc.cancer.gov, TCGA) database. R (v3.6.2) software was used to analyze the expression of STMN1 gene in hepatocellular carcinoma tissues, and its relationship with clinical characteristics of patients was analyzed. Univariate and multivariate Cox regression models were used to analyze the prognostic value of STMN1 in hepatocellular carcinoma. Gene enrichment analysis was used to analyze STMN1 expression-related pathway mechanisms. ssGSEA method was used to analyze the infiltration of 24 immune cell types in tumors, and to explore the relationship between STMN1 and immune cells.
RESULTS STMN1 was highly expressed in tumor tissues (P < 0.001). The diagnostic value of STMN1 was assessed by ROC curve analysis, and the area under the ROC curve (AUC) was 0.971. The predictive ability of STMN1 had high accuracy. Chi-square test or Fisher exact test revealed that STMN1 expression was significantly associated with overall survival rate (P = 0.013), T stage (P = 0.002), TNM stage (P = 0.008), age (P = 0.006), and tumor grade (P < 0.001). The overall survival rate, progression-free survival rate, and disease-specific survival rate of the high STMN1 gene expression group were lower than those of the low expression group (P < 0.01). Cox regression analysis showed that the high expression of STMN1 was an independent risk factor for the prognosis of patients with hepatocellular carcinoma (hazard ratio = 1.808, 95% confidence interval: 1.288-2.234, P = 0.014). GSEA analysis showed that STMN1 gene in hepatocellular carcinoma is mainly involved in cell cycle, oocyte meiosis, T cell receptor signaling pathway, natural killer cell-mediated cytotoxicity, and spliceosome. Immune infiltration analysis showed that STMN1 expression was positively correlated with helper T cells 2, follicular helper T cells, and helper T cells, and was negatively correlated with neutrophils, helper T cells 17, and dendritic cells.
CONCLUSION The expression of STMN1 gene is up-regulated in hepato-cellular carcinoma, which is associated with a poor prognosis of patients. It is an independent prognostic factor for hepatocellular carcinoma patients and is expected to become a potential molecular marker of this malignancy.
Collapse
Affiliation(s)
- Yu Cui
- Department of General Surgery, The Fifth Central Hospital of Tianjin, Tianjin 300450, China
| | - Tao Yun
- Department of General Surgery, The Fifth Central Hospital of Tianjin, Tianjin 300450, China
| | - Guo-Gang Zhao
- Department of General Surgery, The Fifth Central Hospital of Tianjin, Tianjin 300450, China
| |
Collapse
|
2
|
Sellin ME, Stenmark S, Gullberg M. Cell type-specific expression of SEPT3-homology subgroup members controls the subunit number of heteromeric septin complexes. Mol Biol Cell 2014; 25:1594-607. [PMID: 24648497 PMCID: PMC4019491 DOI: 10.1091/mbc.e13-09-0553] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Heteromeric septin complexes serve as building blocks of filaments that organize the cortex of fungal and animal cells. This report addresses determinants that direct hetero-oligomerization of the 13 septin paralogues of mammals. It shows that three distinct septins direct assembly of tissue-specific heterotetramers. Septins are filament-forming proteins important for organizing the cortex of animal and fungal cells. In mammals, 13 septin paralogues were recently shown to assemble into core heterohexamer and heterooctamer complexes, which serve as building blocks for apolar filamentous structures that differ among cell types. To determine how tissue-specific septin paralogue expression may shape core heteromer repertoires and thereby modulate properties of septin filaments, we devised protocols to analyze native septin heteromers with distinct numbers of subunits. Our evidence based on genetically manipulated human cells supports and extends recent concepts of homology subgroup–restricted assembly into distinct categories of apolar heterohexamers and heterooctamers. We also identify a category of tetramers that have a subunit composition equivalent to an octameric building block. These atypical tetramers are prevalent in lymphocytes and neural tissues, in which octamers are abundant but hexamers are rare. Our results can be explained by tissue-specific expression of SEPT3 subgroup members: SEPT3, SEPT9, and SEPT12. These serve as cognate subunits in either heterooctamers or atypical tetramers but exhibit different preferences in various tissues. The identified tissue-specific repertoires of septin heteromers provide insights into how higher-order septin structures with differential properties and stabilities may form in diverse animal cell types.
Collapse
Affiliation(s)
- Mikael E Sellin
- Department of Molecular Biology, Umeå University, SE-901 87 Umeå, SwedenInstitute of Microbiology, D-BIOL, ETH Zürich, 8093 Zürich, Switzerland
| | - Sonja Stenmark
- Department of Molecular Biology, Umeå University, SE-901 87 Umeå, Sweden
| | - Martin Gullberg
- Department of Molecular Biology, Umeå University, SE-901 87 Umeå, Sweden
| |
Collapse
|
3
|
Silva VC, Cassimeris L. Stathmin and microtubules regulate mitotic entry in HeLa cells by controlling activation of both Aurora kinase A and Plk1. Mol Biol Cell 2013; 24:3819-31. [PMID: 24152729 PMCID: PMC3861079 DOI: 10.1091/mbc.e13-02-0108] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Revised: 09/18/2013] [Accepted: 10/16/2013] [Indexed: 12/11/2022] Open
Abstract
Depletion of stathmin, a microtubule (MT) destabilizer, delays mitotic entry by ∼4 h in HeLa cells. Stathmin depletion reduced the activity of CDC25 and its upstream activators, Aurora A and Plk1. Chemical inhibition of both Aurora A and Plk1 was sufficient to delay mitotic entry by 4 h, while inhibiting either kinase alone did not cause a delay. Aurora A and Plk1 are likely regulated downstream of stathmin, because the combination of stathmin knockdown and inhibition of Aurora A and Plk1 was not additive and again delayed mitotic entry by 4 h. Aurora A localization to the centrosome required MTs, while stathmin depletion spread its localization beyond that of γ-tubulin, indicating an MT-dependent regulation of Aurora A activation. Plk1 was inhibited by excess stathmin, detected in in vitro assays and cells overexpressing stathmin-cyan fluorescent protein. Recruitment of Plk1 to the centrosome was delayed in stathmin-depleted cells, independent of MTs. It has been shown that depolymerizing MTs with nocodazole abrogates the stathmin-depletion induced cell cycle delay; in this study, depolymerization with nocodazole restored Plk1 activity to near normal levels, demonstrating that MTs also contribute to Plk1 activation. These data demonstrate that stathmin regulates mitotic entry, partially via MTs, to control localization and activation of both Aurora A and Plk1.
Collapse
Affiliation(s)
- Victoria C. Silva
- Department of Biological Sciences, Lehigh University, Bethlehem, PA 18015
| | - Lynne Cassimeris
- Department of Biological Sciences, Lehigh University, Bethlehem, PA 18015
| |
Collapse
|
4
|
Abstract
The post-genomic era has produced a variety of new investigation technologies, techniques and approaches that may offer exciting insights into many long-standing questions of scientific research. The microtubule cytoskeleton is a highly conserved system that shows a high degree of internal complexity, is known to be integral to many cell systems and functions on a fundamental level. After decades of study, much is still unknown about microtubules in vivo from the control of dynamics in living cells to their responses to environmental changes and responses to other cellular processes. In the present article, we examine some outstanding questions in the microtubule field and propose a combination of emerging interdisciplinary approaches, i.e. high-throughput functional genomics techniques, quantitative and super-resolution microscopy, and in silico modelling, that could shed light on the systemic regulation of microtubules in cells by networks of regulatory factors. We propose that such an integrative approach is key to elucidate the function of the microtubule cytoskeleton as a complete responsive integral biological system.
Collapse
|
5
|
Chen YL, Uen YH, Li CF, Horng KC, Chen LR, Wu WR, Tseng HY, Huang HY, Wu LC, Shiue YL. The E2F transcription factor 1 transactives stathmin 1 in hepatocellular carcinoma. Ann Surg Oncol 2013; 20:4041-4054. [PMID: 22911364 DOI: 10.1245/s10434-012-2519-8] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2011] [Indexed: 12/31/2022]
Abstract
BACKGROUND Through data mining the Stanford Microarray Database, the stathmin 1 (STMN1) transcript was found to be frequently upregulated in the hepatocellular carcinoma (HCC) with low alpha-fetoprotein level. The molecular mechanism of STMN1 upregulation in HCCs remained unclear. METHODS Quantitative RT-PCR, immunoblotting, immunohistochemistry, and transfection of expression or small hairpin RNA interference plasmids, chromatin immunoprecipitation (ChIP), and quantitative ChIP assays were performed in HCC specimens or 2 distinct HCC-derived cell lines. Dual luciferase assay and site-directed mutagenesis were applied to analyze the activities of STMN1 proximal promoter region. RESULTS STMN1 mRNA and proteins were significantly associated with several clinicopathological features. High STMN1 or E2F1 immunoexpression was predictive of poor overall survival (OS) rate (P < .01). In HCC-derived cell lines, E2F1 was elevated before STMN1 mRNA during the cell cycle. Exogenous expression of E2F1 or both transcription factor DP-1 (TFDP1) and E2F1 genes induced E2F1 and STMN1 mRNA (P < .01). Knockdown of the E2F1 gene suppressed E2F1 and STMN1 mRNA and E2F1 and STMN1 protein levels (P < .05). The promoter activity of STMN1 gene increased with overexpression of both E2F1 and TFDP1 genes (P < .05); however, it decreased when mutations were introduced in the E2F1-binding sites (P < .05). CONCLUSIONS Upregulation of E2F1 and STMN1 proteins associate with worse outcomes in patients with HCC. E2F1 significantly correlates with STMN1 protein level in HCC lesions and in vitro transactivation assays, suggesting that STMN1 gene is transactivated by the E2F1 protein.
Collapse
MESH Headings
- Blotting, Western
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Cell Cycle
- Cell Proliferation
- Chromatin Immunoprecipitation
- E2F1 Transcription Factor/antagonists & inhibitors
- E2F1 Transcription Factor/genetics
- E2F1 Transcription Factor/metabolism
- Female
- Flow Cytometry
- Follow-Up Studies
- Gene Expression Regulation, Neoplastic
- Humans
- Immunoenzyme Techniques
- Liver/metabolism
- Liver/pathology
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Luciferases/metabolism
- Male
- Mutagenesis, Site-Directed
- Mutation/genetics
- Neoplasm Staging
- Prognosis
- Promoter Regions, Genetic/genetics
- RNA, Messenger/genetics
- RNA, Small Interfering/genetics
- Real-Time Polymerase Chain Reaction
- Retrospective Studies
- Reverse Transcriptase Polymerase Chain Reaction
- Stathmin/genetics
- Stathmin/metabolism
- Transcription Factor DP1/genetics
- Transcription Factor DP1/metabolism
- Transcriptional Activation
- Tumor Cells, Cultured
- alpha-Fetoproteins/genetics
- alpha-Fetoproteins/metabolism
Collapse
Affiliation(s)
- Yi-Ling Chen
- Institute of Biomedical Science, National Sun Yat-sen University, Kaohsiung, Taiwan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Duncan JE, Lytle NK, Zuniga A, Goldstein LSB. The Microtubule Regulatory Protein Stathmin Is Required to Maintain the Integrity of Axonal Microtubules in Drosophila. PLoS One 2013; 8:e68324. [PMID: 23840848 PMCID: PMC3694009 DOI: 10.1371/journal.pone.0068324] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Accepted: 05/22/2013] [Indexed: 12/25/2022] Open
Abstract
Axonal transport, a form of long-distance, bi-directional intracellular transport that occurs between the cell body and synaptic terminal, is critical in maintaining the function and viability of neurons. We have identified a requirement for the stathmin (stai) gene in the maintenance of axonal microtubules and regulation of axonal transport in Drosophila. The stai gene encodes a cytosolic phosphoprotein that regulates microtubule dynamics by partitioning tubulin dimers between pools of soluble tubulin and polymerized microtubules, and by directly binding to microtubules and promoting depolymerization. Analysis of stai function in Drosophila, which has a single stai gene, circumvents potential complications with studies performed in vertebrate systems in which mutant phenotypes may be compensated by genetic redundancy of other members of the stai gene family. This has allowed us to identify an essential function for stai in the maintenance of the integrity of axonal microtubules. In addition to the severe disruption in the abundance and architecture of microtubules in the axons of stai mutant Drosophila, we also observe additional neurological phenotypes associated with loss of stai function including a posterior paralysis and tail-flip phenotype in third instar larvae, aberrant accumulation of transported membranous organelles in stai deficient axons, a progressive bang-sensitive response to mechanical stimulation reminiscent of the class of Drosophila mutants used to model human epileptic seizures, and a reduced adult lifespan. Reductions in the levels of Kinesin-1, the primary anterograde motor in axonal transport, enhance these phenotypes. Collectively, our results indicate that stai has an important role in neuronal function, likely through the maintenance of microtubule integrity in the axons of nerves of the peripheral nervous system necessary to support and sustain long-distance axonal transport.
Collapse
Affiliation(s)
- Jason E. Duncan
- Department of Biology, Willamette University, Salem, Oregon, United States of America
- * E-mail:
| | - Nikki K. Lytle
- Department of Biology, Willamette University, Salem, Oregon, United States of America
| | - Alfredo Zuniga
- Department of Biology, Willamette University, Salem, Oregon, United States of America
| | - Lawrence S. B. Goldstein
- Howard Hughes Medical Institute, Department of Cellular and Molecular Medicine, School of Medicine, University of California San Diego, La Jolla, California, United States of America
| |
Collapse
|
7
|
Sellin ME, Stenmark S, Gullberg M. Mammalian SEPT9 isoforms direct microtubule-dependent arrangements of septin core heteromers. Mol Biol Cell 2012; 23:4242-55. [PMID: 22956766 PMCID: PMC3484102 DOI: 10.1091/mbc.e12-06-0486] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Cell type–specific alternative splicing results in six confirmed mammalian SEPT9 isoforms. SEPT9 expression levels dictate the hexamer-to-octamer ratio of septin core heteromers, and isoform compositions and expression levels together determine higher-order arrangements of septin filaments. Septin-family proteins assemble into rod-shaped heteromeric complexes that form higher-order arrangements at the cell cortex, where they serve apparently conserved functions as diffusion barriers and molecular scaffolds. There are 13 confirmed septin paralogues in mammals, which may be ubiquitous or tissue specific. Septin hetero-oligomerization appears homology subgroup directed, which in turn determines the subunit arrangement of six- to eight-subunit core heteromers. Here we address functional properties of human SEPT9, which, due to variable mRNA splicing, exists as multiple isoforms that differ between tissues. Myeloid K562 cells express three SEPT9 isoforms, all of which have an equal propensity to hetero-oligomerize with SEPT7-containing hexamers to generate octameric heteromers. However, due to limiting amounts of SEPT9, K562 cells contain both hexameric and octameric heteromers. To generate cell lines with controllable hexamer-to-octamer ratios and that express single SEPT9 isoforms, we developed a gene product replacement strategy. By this means we identified SEPT9 isoform–specific properties that either facilitate septin heteromer polymerization along microtubules or modulate the size range of submembranous septin disks—a prevalent septin structure in nonadhered cells. Our findings show that the SEPT9 expression level directs the hexamer-to-octamer ratio, and that the isoform composition and expression level together determine higher-order arrangements of septins.
Collapse
Affiliation(s)
- Mikael E Sellin
- Department of Molecular Biology, Umeå University, SE-901 87 Umeå, Sweden
| | | | | |
Collapse
|
8
|
Wang R, Wang Z, Yang J, Liu X, Wang L, Guo X, Zeng F, Wu M, Li G. LRRC4 inhibits the proliferation of human glioma cells by modulating the expression of STMN1 and microtubule polymerization. J Cell Biochem 2012; 112:3621-9. [PMID: 21809374 DOI: 10.1002/jcb.23293] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
LRRC4 is a tumor suppressor of glioma, and it is epigenetically inactivated commonly in glioma. Our previous study has shown that induction of LRRC4 expression inhibits the proliferation of glioma cells. However, little is known about the mechanisms underlying the action of LRRC4 in glioma cells. We employed two-dimensional fluorescence differential gel electrophoresis (2-D DIGE) and MALDI -TOF/TOF-MS/MS to identify 11 differentially expressed proteins, including the significantly down-regulated STMN1 expression in the LRRC4-expressing U251 glioma cells. The levels of STMN1 expression appeared to be positively associated with the pathogenic degrees of human glioma. Furthermore, induction of LRRC4 over-expression inhibited the STMN1 expression and U251 cell proliferation in vitro, and the glioma growth in vivo. In addition, induction of LRRC4 or knockdown of STMN1 expression induced cell cycle arrest in U251 cells, which was associated with modulating the p21, cyclin D1, and cyclin B expression, and the ERK phosphorylation, and inhibiting the CDK5 and cdc2 kinase activities, but increasing the microtubulin polymerization in U251 cells. LRRC4, at least partially by down-regulating the STMN1expression, acts as a major glioma suppressor, induces cell cycle arrest and modulates the dynamic process of microtubulin, leading to the inhibition of glioma cell proliferation and growth. Potentially, modulation of LRRC4 or STMN1 expression may be useful for design of new therapies for the intervention of glioma.
Collapse
Affiliation(s)
- Rong Wang
- Cancer Research Institute, Central South University, Changsha, Hunan, China
| | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Carney BK, Caruso Silva V, Cassimeris L. The microtubule cytoskeleton is required for a G2 cell cycle delay in cancer cells lacking stathmin and p53. Cytoskeleton (Hoboken) 2012; 69:278-89. [PMID: 22407961 DOI: 10.1002/cm.21024] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Revised: 02/24/2012] [Accepted: 02/29/2012] [Indexed: 12/25/2022]
Abstract
In several cancer cell lines, depleting the microtubule (MT)-destabilizing protein stathmin/oncoprotein18 leads to a G2 cell cycle delay and apoptosis. These phenotypes are observed only in synergy with low levels of p53, but the pathway(s) activated by stathmin depletion to delay the cell cycle are unknown. We found that stathmin depletion caused greater MT stability in synergy with loss of p53, measured by the levels of acetylated α-tubulin and the rate of centrosomal MT nucleation. Nocodazole or vinblastine-induced MT depolymerization abrogated the stathmin-depletion induced G2 delay, measured by the percentage of cells staining positive for several markers (TPX2, CDK1 with inhibitory phosphorylation), indicating that MTs are required to lengthen G2. Live cell imaging showed that stathmin depletion increased time in G2 without an impact on the duration of mitosis, indicating that the longer interphase duration is not simply a consequence of a previous slowed mitosis. In contrast, stabilization of MTs with paclitaxel (8 nM) slowed mitosis without lengthening the duration of interphase, demonstrating that increased MT stability alone is not sufficient to delay cells in G2.
Collapse
Affiliation(s)
- Bruce K Carney
- Department of Biological Sciences, Lehigh University, Bethlehem, Pennsylvania 18015, USA
| | | | | |
Collapse
|
10
|
Sellin ME, Holmfeldt P, Stenmark S, Gullberg M. Microtubules support a disk-like septin arrangement at the plasma membrane of mammalian cells. Mol Biol Cell 2011; 22:4588-601. [PMID: 21998205 PMCID: PMC3226477 DOI: 10.1091/mbc.e11-09-0754] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Septin assemblies during the interphase of animal cells remain poorly defined and are the topic of this report. The data point to a general model for assembly of higher-order septin arrangements at locations providing the greatest opportunity for binding cooperativity, which depends on both the cell type and external cues. Septin family proteins oligomerize through guanosine 5′-triphosphate–binding domains into core heteromers, which in turn polymerize at the cleavage furrow of dividing fungal and animal cells. Septin assemblies during the interphase of animal cells remain poorly defined and are the topic of this report. In this study, we developed protocols for visualization of authentic higher-order assemblies using tagged septins to effectively replace the endogenous gene product within septin core heteromers in human cells. Our analysis revealed that septins assemble into microtubule-supported, disk-like structures at the plasma membrane. In the absence of cell substrate adhesion, this is the predominant higher-order arrangement in interphase cells and each of the seven to eight septin family members expressed by the two analyzed cell types appears equally represented. However, studies of myeloid and lymphoid cell model systems revealed cell type–specific alterations of higher-order septin arrangements in response to substrate adhesion. Live-cell observations suggested that all higher-order septin assemblies are mutually exclusive with plasma membrane regions undergoing remodeling. The combined data point to a mechanism by which densely arranged cortical microtubules, which are typical for nonadhered spherical cells, support plasma membrane–bound, disk-like septin assemblies.
Collapse
Affiliation(s)
- Mikael E Sellin
- Department of Molecular Biology, Umeå University, SE-901 87 Umeå, Sweden.
| | | | | | | |
Collapse
|
11
|
Sellin ME, Sandblad L, Stenmark S, Gullberg M. Deciphering the rules governing assembly order of mammalian septin complexes. Mol Biol Cell 2011; 22:3152-64. [PMID: 21737677 PMCID: PMC3164462 DOI: 10.1091/mbc.e11-03-0253] [Citation(s) in RCA: 131] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Vertebrates express 9–17 septin family members known to oligomerize into diverse structures, but their native assembly states have remained elusive. The results presented suggest a generic model for how the temporal order of septin assembly directs the subunit arrangement within distinct pools of six- to eight-subunit core heteromers. Septins are conserved GTP-binding proteins that assemble into lateral diffusion barriers and molecular scaffolds. Vertebrate genomes contain 9–17 septin genes that encode both ubiquitous and tissue-specific septins. Expressed septins may assemble in various combinations through both heterotypic and homotypic G-domain interactions. However, little is known regarding assembly states of mammalian septins and mechanisms directing ordered assembly of individual septins into heteromeric units, which is the focus of this study. Our analysis of the septin system in cells lacking or overexpressing selected septins reveals interdependencies coinciding with previously described homology subgroups. Hydrodynamic and single-particle data show that individual septins exist solely in the context of stable six- to eight-subunit core heteromers, all of which contain SEPT2 and SEPT6 subgroup members and SEPT7, while heteromers comprising more than six subunits also contain SEPT9. The combined data suggest a generic model for how the temporal order of septin assembly is homology subgroup-directed, which in turn determines the subunit arrangement of native heteromers. Because mammalian cells normally express multiple members and/or isoforms of some septin subgroups, our data also suggest that only a minor fraction of native heteromers are arranged as perfect palindromes.
Collapse
Affiliation(s)
- Mikael E Sellin
- Department of Molecular Biology, Umeå University, SE-901 87 Umeå, Sweden.
| | | | | | | |
Collapse
|
12
|
Ng DCH, Zhao TT, Yeap YYC, Ngoei KR, Bogoyevitch MA. c-Jun N-terminal kinase phosphorylation of stathmin confers protection against cellular stress. J Biol Chem 2010; 285:29001-13. [PMID: 20630875 DOI: 10.1074/jbc.m110.128454] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The cell stress response encompasses the range of intracellular events required for adaptation to stimuli detrimental to cell survival. Although the c-Jun N-terminal kinase (JNK) is a stress-activated kinase that can promote either cell survival or death in response to detrimental stimuli, the JNK-regulated mechanisms involved in survival are not fully characterized. Here we show that in response to hyperosmotic stress, JNK phosphorylates a key cytoplasmic microtubule regulatory protein, stathmin (STMN), on conserved Ser-25 and Ser-38 residues. In in vitro biochemical studies, we identified STMN Ser-38 as the critical residue required for efficient phosphorylation by JNK and identified a novel kinase interaction domain in STMN required for recognition by JNK. We revealed that JNK was required for microtubule stabilization in response to hyperosmotic stress. Importantly, we also demonstrated a novel cytoprotective function for STMN, as the knockdown of STMN levels by siRNA was sufficient to augment viability in response to hyperosmotic stress. Our findings show that JNK targeting of STMN represents a novel stress-activated cytoprotective mechanism involving microtubule network changes.
Collapse
Affiliation(s)
- Dominic C H Ng
- Department of Biochemistry, Bio21 Institute, University of Melbourne, Parkville, 3010 Victoria, Australia
| | | | | | | | | |
Collapse
|
13
|
Hsieh SY, Huang SF, Yu MC, Yeh TS, Chen TC, Lin YJ, Chang CJ, Sung CM, Lee YL, Hsu CY. Stathmin1 overexpression associated with polyploidy, tumor-cell invasion, early recurrence, and poor prognosis in human hepatoma. Mol Carcinog 2010; 49:476-87. [PMID: 20232364 DOI: 10.1002/mc.20627] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Frequent intrahepatic metastasis causes early tumor recurrence and dismaying prognosis of human hepatocellular carcinoma (HCC). We recently identified overexpression of stathmin1 (STMN1) in human HCC. This study was designed to elucidate the clinical and biological significance of overexpression of STMN1 in HCC. Expression of STMN1 was conducted by quantitative reverse transcription-polymerase chain reaction and immunoblotting assays on 58 pairs of HCC and para-tumor liver tissues from patients with HCC along with normal liver tissues as the controls. Association of STMN1 overexpression with tumor recurrence and prognosis was investigated by Kaplan-Meier cumulative survival and Cox Regression analyses. Roles of STMN1 in cell cycle, cell motility, and invasion were determined by in vitro assays. STMN1 overexpression in hepatoma was strongly associated with local invasion (P = 0.031), early recurrence (P = 0.002), and poor prognosis (P = 0.005), and was an independent indicator for tumor recurrence (P = 0.0045). STMN1 overexpression further identified subgroups of HCC patients with higher tumor recurrence and worse prognosis among HCC patients with early tumor stage (T1) or intermediate histological grades (G2 and G3), both of whom represent the majority of HCC patients receiving primary curative hepatectomy. Silencing STMN1 expression via RNA interference suppressed invasion activity, while ectopic expression of STMN1 enhanced cell invasion and caused polyploidy of cells. In conclusion, STMN1 overexpression could predict early tumor recurrence and poor prognosis, particularly at early stage of hepatoma. Overexpression of STMN1 promoted polyploidy formation, tumor-cell invasion, and intrahepatic metastasis, suggesting that STMN1 can be a target for anti-cancer therapy of human hepatoma.
Collapse
Affiliation(s)
- Sen-Yung Hsieh
- Liver Research Unit, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Holmfeldt P, Sellin ME, Gullberg M. Upregulated Op18/stathmin activity causes chromosomal instability through a mechanism that evades the spindle assembly checkpoint. Exp Cell Res 2010; 316:2017-26. [PMID: 20399773 DOI: 10.1016/j.yexcr.2010.04.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2010] [Revised: 04/09/2010] [Accepted: 04/12/2010] [Indexed: 02/04/2023]
Abstract
Op18/stathmin (Op18) is a microtubule-destabilizing protein that is phosphorylation-inactivated during mitosis and its normal function is to govern tubulin subunit partitioning during interphase. Human tumors frequently overexpress Op18 and a tumor-associated Q18-->E mutation has been identified that confers hyperactivity, destabilizes spindle microtubules, and causes mitotic aberrancies, polyploidization, and chromosome loss in K562 leukemia cells. Here we determined whether wild-type and mutant Op18 have the potential to cause chromosomal instability by some means other than interference with spindle assembly, and thereby bypassing the spindle assembly checkpoint. Our approach was based on Op18 derivatives with distinct temporal order of activity during mitosis, conferred either by differential phosphorylation inactivation or by anaphase-specific degradation through fusion with the destruction box of cyclin B1. We present evidence that excessive Op18 activity generates chromosomal instability through interference occurring subsequent to the metaphase-to-anaphase transition, which reduces the fidelity of chromosome segregation to spindle poles during anaphase. Similar to uncorrected merotelic attachment, this mechanism evades detection by the spindle assembly checkpoint and thus provides an additional route to chromosomal instability.
Collapse
Affiliation(s)
- Per Holmfeldt
- Department of Molecular Biology, Umeå University, Umeå, Sweden
| | | | | |
Collapse
|
15
|
Functional and spatial regulation of mitotic centromere-associated kinesin by cyclin-dependent kinase 1. Mol Cell Biol 2010; 30:2594-607. [PMID: 20368358 DOI: 10.1128/mcb.00098-10] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Mitotic centromere-associated kinesin (MCAK) plays an essential role in spindle formation and in correction of improper microtubule-kinetochore attachments. The localization and activity of MCAK at the centromere/kinetochore are controlled by Aurora B kinase. However, MCAK is also abundant in the cytosol and at centrosomes during mitosis, and its regulatory mechanism at these sites is unknown. We show here that cyclin-dependent kinase 1 (Cdk1) phosphorylates T537 in the core domain of MCAK and attenuates its microtubule-destabilizing activity in vitro and in vivo. Phosphorylation of MCAK by Cdk1 promotes the release of MCAK from centrosomes and is required for proper spindle formation. Interfering with the regulation of MCAK by Cdk1 causes dramatic defects in spindle formation and in chromosome positioning. This is the first study demonstrating that Cdk1 regulates the localization and activity of MCAK in mitosis by directly phosphorylating the catalytic core domain of MCAK.
Collapse
|
16
|
Lundin VF, Leroux MR, Stirling PC. Quality control of cytoskeletal proteins and human disease. Trends Biochem Sci 2010; 35:288-97. [PMID: 20116259 DOI: 10.1016/j.tibs.2009.12.007] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2009] [Revised: 12/22/2009] [Accepted: 12/23/2009] [Indexed: 11/25/2022]
Abstract
Actins and tubulins are abundant cytoskeletal proteins that support diverse cellular processes. Owing to the unique properties of these filament-forming proteins, an intricate cellular machinery consisting minimally of the chaperonin CCT, prefoldin, phosducin-like proteins, and tubulin cofactors has evolved to facilitate their biogenesis. More recent evidence also suggests that regulated degradation pathways exist for actin (via TRIM32) and tubulin (via parkin or cofactor E-like). Collectively, these pathways maintain the quality control of cytoskeletal proteins ('proteostasis'), ensuring the appropriate function of microfilaments and microtubules. Here, we focus on the molecular mechanisms of the quality control of actin and tubulin, and discuss emerging links between cytoskeletal proteostasis and human diseases.
Collapse
Affiliation(s)
- Victor F Lundin
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | | | | |
Collapse
|
17
|
Abstract
Proper regulation of MT (microtubule) dynamics is essential for various vital processes, including the segregation of chromosomes, directional cell migration and differentiation. MT assembly and disassembly is modulated by a complex network of intracellular factors that co-operate or antagonize each other, are highly regulated in space and time and are thus attuned to the cell cycle and differentiation processes. While we only begin to appreciate how the concerted action of MT stabilizers and destabilizers shapes different MT patterns, a clear picture of how individual factors affect the MT structure is emerging. In this paper, we review the current knowledge about proteins that modulate MT dynamic instability.
Collapse
|
18
|
Holmfeldt P, Sellin ME, Gullberg M. Predominant regulators of tubulin monomer-polymer partitioning and their implication for cell polarization. Cell Mol Life Sci 2009; 66:3263-76. [PMID: 19585080 PMCID: PMC11115727 DOI: 10.1007/s00018-009-0084-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2009] [Revised: 06/19/2009] [Accepted: 06/22/2009] [Indexed: 12/15/2022]
Abstract
The microtubule-system organizes the cytoplasm during interphase and segregates condensed chromosomes during mitosis. Four unrelated conserved proteins, XMAP215/Dis1/TOGp, MCAK, MAP4 and Op18/stathmin, have all been implicated as predominant regulators of tubulin monomer-polymer partitioning in animal cells. However, while studies employing the Xenopus egg extract model system indicate that the partitioning is largely governed by the counteractive activities of XMAP215 and MCAK, studies of human cell lines indicate that MAP4 and Op18 are the predominant regulators of the interphase microtubule-array. Here, we review functional interplay of these proteins during interphase and mitosis in various cell model systems. We also review the evidence that MAP4 and Op18 have interphase-specific, counteractive and phosphorylation-inactivated activities that govern tubulin subunit partitioning in many mammalian cell types. Finally, we discuss evidence indicating that partitioning regulation by MAP4 and Op18 may be of significance to establish cell polarity.
Collapse
Affiliation(s)
- Per Holmfeldt
- Department of Molecular Biology, University of Umeå, Umeå, Sweden.
| | | | | |
Collapse
|
19
|
Harris G, Schaefer KL. The microtubule-targeting agent T0070907 induces proteasomal degradation of tubulin. Biochem Biophys Res Commun 2009; 388:345-9. [PMID: 19665001 DOI: 10.1016/j.bbrc.2009.08.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2009] [Accepted: 08/02/2009] [Indexed: 01/14/2023]
Abstract
Current microtubule-targeting agents interfere with the regulated assembly of microtubules from alpha/beta tubulin heterodimers but do not markedly alter tubulin levels. Previously, we showed that the compound T0070907 interferes with microtubule function by reversibly decreasing alpha and beta tubulin protein levels by more than 50% in multiple CRC cell lines. Since tubulin levels are generally relatively stable, and cells lack regulatory networks to respond to decreased tubulin levels by increasing synthesis, our result suggested the possibility of cancer therapies that act directly on tubulin homeostasis. The aim of this study was to determine whether T0070907 caused tubulin loss by increasing the degradation rate, and determine the proteases responsible for any increased degradation. T0070907 increased tubulin degradation rates in HT-29 cells. The proteasomal inhibitors MG132, epoxomicin, lactacystin, and ALLN suppressed T0070907-mediated tubulin loss, although epoxomicin and lactacystin were less effective than MG132, even at concentrations that completely inhibited TNFalpha-induced IkappaBalpha degradation. Inhibitors of lysosomal, aggresomal, and calpain-mediated degradation, as well as the caspase inhibitor zVAD-fmk had no effect on tubulin loss, and the cathepsin and calpain inhibitor E64d was unable to increase epoxomicin's ability to suppress tubulin loss. We conclude that T0070907-induced tubulin degradation proceeds through a proteasome-dependent pathway.
Collapse
Affiliation(s)
- Gianni Harris
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | | |
Collapse
|
20
|
Ringhoff DN, Cassimeris L. Stathmin regulates centrosomal nucleation of microtubules and tubulin dimer/polymer partitioning. Mol Biol Cell 2009; 20:3451-8. [PMID: 19515833 DOI: 10.1091/mbc.e09-02-0140] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Stathmin is a microtubule-destabilizing protein ubiquitously expressed in vertebrates and highly expressed in many cancers. In several cell types, stathmin regulates the partitioning of tubulin between unassembled and polymer forms, but the mechanism responsible for partitioning has not been determined. We examined stathmin function in two cell systems: mouse embryonic fibroblasts (MEFs) isolated from embryos +/+, +/-, and -/- for the stathmin gene and porcine kidney epithelial (LLCPK) cells expressing stathmin-cyan fluorescent protein (CFP) or injected with stathmin protein. In MEFs, the relative amount of stathmin corresponded to genotype, where cells heterozygous for stathmin expressed half as much stathmin mRNA and protein as wild-type cells. Reduction or loss of stathmin resulted in increased microtubule polymer but little change to microtubule dynamics at the cell periphery. Increased stathmin level in LLCPK cells, sufficient to reduce microtubule density, but allowing microtubules to remain at the cell periphery, also did not have a major impact on microtubule dynamics. In contrast, stathmin level had a significant effect on microtubule nucleation rate from centrosomes, where lower stathmin levels increased nucleation and higher stathmin levels reduced nucleation. The stathmin-dependent regulation of nucleation is only active in interphase; overexpression of stathmin-CFP did not impact metaphase microtubule nucleation rate in LLCPK cells and the number of astral microtubules was similar in stathmin +/+ and -/- MEFs. These data support a model in which stathmin functions in interphase to control the partitioning of tubulins between dimer and polymer pools by setting the number of microtubules per cell.
Collapse
Affiliation(s)
- Danielle N Ringhoff
- Department of Chemistry and Biological Sciences, Lehigh University, Bethlehem, PA 18015, USA
| | | |
Collapse
|
21
|
Verma NK, Dourlat J, Davies AM, Long A, Liu WQ, Garbay C, Kelleher D, Volkov Y. STAT3-stathmin interactions control microtubule dynamics in migrating T-cells. J Biol Chem 2009; 284:12349-62. [PMID: 19251695 DOI: 10.1074/jbc.m807761200] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
T-cell migration is a complex highly coordinated process that involves cell adhesion to the high endothelial venules or to the extracellular matrix by surface receptor/ligand interactions, cytoskeletal rearrangements, and phosphorylation-dependent signaling cascades. The mechanism(s) that regulates T-cell migration is of considerable relevance for understanding the pathogenesis of various diseases, such as chronic inflammatory diseases and cancer metastasis. This study was designed to identify potential involvement of STAT3, a latent transcription factor, in mediating integrin-induced T-cell migration. Using our previously characterized in vitro model for lymphocyte migration, we demonstrate that STAT3 is activated and translocated to the nucleus during the process of active motility of Hut78 T-lymphoma cells triggered via LFA-1. Blocking STAT3 signaling by multiple approaches inhibited LFA-1-induced T-cell locomotion via destabilization of microtubules and post-translational modification of tubulin. Here, we show that STAT3 physically interacts with stathmin to regulate microtubule dynamics in migrating T-cells. These observations strongly indicate that STAT3 is critically important for T-cell migration and associated signaling events.
Collapse
Affiliation(s)
- Navin K Verma
- Department of Clinical Medicine, Institute of Molecular Medicine, Trinity College Dublin, Dublin 8, Ireland.
| | | | | | | | | | | | | | | |
Collapse
|