1
|
Lorenzen L, Frank D, Schwan C, Grosse R. Spatiotemporal Regulation of FMNL2 by N-Terminal Myristoylation and C-Terminal Phosphorylation Drives Rapid Filopodia Formation. Biomolecules 2023; 13:biom13030548. [PMID: 36979484 PMCID: PMC10046779 DOI: 10.3390/biom13030548] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/10/2023] [Accepted: 03/14/2023] [Indexed: 03/19/2023] Open
Abstract
The actin nucleating and polymerizing formin-like 2 (FMNL2) is upregulated in several cancers and has been shown to play important roles in cell migration, invasion, cell–cell adhesion and filopodia formation. Here, using structured illumination microscopy we show that FMNL2 promotes rapid and highly dynamic filopodia formation in epithelial cells while remaining on the tip of the growing filopodia. This filopodia tip localization depends fully on its N-terminal myristoylation. We further show that FMNL2-dependent filopodia formation requires its serine 1072 phosphorylation within the diaphanous-autoregulatory domain (DAD) by protein kinase C (PKC) α. Consistent with this, filopodia formation depends on PKC activity and PKCα localizes to the base of growing filopodia. Thus, a PKCα–FMNL2 signaling module spatiotemporally controls dynamic filopodia formation.
Collapse
Affiliation(s)
- Lina Lorenzen
- Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty, University of Freiburg, 79104 Freiburg, Germany
| | - Dennis Frank
- Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty, University of Freiburg, 79104 Freiburg, Germany
| | - Carsten Schwan
- Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty, University of Freiburg, 79104 Freiburg, Germany
- Correspondence: (C.S.); (R.G.)
| | - Robert Grosse
- Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty, University of Freiburg, 79104 Freiburg, Germany
- Centre for Integrative Biological Signalling Studies—CIBSS, 79104 Freiburg, Germany
- Correspondence: (C.S.); (R.G.)
| |
Collapse
|
2
|
Vakhrusheva A, Murashko A, Trifonova E, Efremov Y, Timashev P, Sokolova O. Role of Actin-binding Proteins in the Regulation of Cellular Mechanics. Eur J Cell Biol 2022; 101:151241. [DOI: 10.1016/j.ejcb.2022.151241] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 04/18/2022] [Accepted: 05/19/2022] [Indexed: 12/25/2022] Open
|
3
|
Jiao X, Wang B, Yang L, Zhao Q, Zhang M, Liu X, Zhou C, Wang R, Chen H, Wang J, Ren Y, Liu P. FMNL2 suppresses cell migration and invasion of breast cancer: a reduction of cytoplasmic p27 via RhoA/LIMK/Cofilin pathway. Cell Death Dis 2022; 8:155. [PMID: 35379791 PMCID: PMC8980084 DOI: 10.1038/s41420-022-00964-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 03/03/2022] [Accepted: 03/17/2022] [Indexed: 11/24/2022]
Abstract
Formin-like protein 2 (FMNL2) belongs to a highly conserved family of cytoskeletal remodeling proteins that have been reported to be implicated in various actin-dependent physiological and cancer-associated processes. In this study, we mainly investigated the effects of FMNL2 on breast cancer cell migration and invasion, and the underlying mechanisms involved. We found that FMNL2 reduced cell migration and invasion of breast cancer in vitro and in vivo. Further, FMNL2 disrupted actin cytoskeleton rearrangement and hampered the RhoA/LIMK/Cofilin pathway in breast cancer cells. Critically, both Rho inhibitor ZOL and LIMK inhibitor BMS3 significantly abrogated these migration-promoting effects in FMNL2-silencing MDA-MB-231 and BT549 cells. RhoA/LIMK/Cofilin pathway was involved in FMNL2 silencing-induced actin cytoskeleton rearrangement in MDA-MB-231 and BT549 cells. More importantly, cytoplasmic p27 promoted FMNL2-mediated cell migration and invasion through RhoA/LIMK/Cofilin pathway in MCF7 and MDA-MB-231 cells. In addition, the expression and prognosis of FMNL2 were associated with ER in breast cancer. Furthermore, ERα overexpression reduced the protein levels of FMNL2 in breast cancer cells, which were reversed by MG132. In conclusion, FMNL2 suppressed cell migration and invasion of breast cancer by inhibiting RhoA/LIMK/Cofilin pathway through a reduction of cytoplasmic p27. This finding implies that the interference of FMNL2-mediated RhoA/LIMK/Cofilin pathway involving the cytoplasmic p27 may be a promising strategy for ameliorating breast cancer metastasis and prognosis.
Collapse
Affiliation(s)
- Xinyan Jiao
- Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, P.R. China.,Key Laboratory for Tumor Precision Medicine of Shaanxi Province, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, P.R. China
| | - Bo Wang
- Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, P.R. China.,Key Laboratory for Tumor Precision Medicine of Shaanxi Province, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, P.R. China
| | - Lu Yang
- Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, P.R. China.,Key Laboratory for Tumor Precision Medicine of Shaanxi Province, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, P.R. China
| | - Qingbin Zhao
- Department of Geratology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, P.R. China
| | - Miao Zhang
- Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, P.R. China.,Key Laboratory for Tumor Precision Medicine of Shaanxi Province, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, P.R. China
| | - Xiaoxu Liu
- Department of Breast Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, P.R. China
| | - Can Zhou
- Department of Breast Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, P.R. China
| | - Ruiqi Wang
- Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, P.R. China.,Key Laboratory for Tumor Precision Medicine of Shaanxi Province, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, P.R. China
| | - He Chen
- Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, P.R. China.,Key Laboratory for Tumor Precision Medicine of Shaanxi Province, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, P.R. China
| | - Jichang Wang
- Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, P.R. China.,Department of Vascular Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, P.R. China
| | - Yu Ren
- Department of Breast Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, P.R. China.
| | - Peijun Liu
- Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, P.R. China. .,Key Laboratory for Tumor Precision Medicine of Shaanxi Province, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, P.R. China.
| |
Collapse
|
4
|
Zhao Y, Zhang H, Wang H, Ye M, Jin X. Role of formin INF2 in human diseases. Mol Biol Rep 2021; 49:735-746. [PMID: 34698992 DOI: 10.1007/s11033-021-06869-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 10/21/2021] [Indexed: 01/08/2023]
Abstract
Formin proteins catalyze actin nucleation and microfilament polymerization. Inverted formin 2 (INF2) is an atypical diaphanous-related formin characterized by polymerization and depolymerization of actin. Accumulating evidence showed that INF2 is associated with kidney disease focal segmental glomerulosclerosis and cancers, such as colorectal and thyroid cancer where it functions as a tumor suppressor, glioblastoma, breast, prostate, and gastric cancer, via its oncogenic function. However, studies on the underlying molecular mechanisms of the different roles of INF2 in diverse cancers are limited. This review comprehensively describes the structure, biochemical features, and primary pathogenic mutations of INF2.
Collapse
Affiliation(s)
- Yiting Zhao
- Department of Hepato-Biliary-Pancreatic Surgery, The Affiliated Ningbo Medical Center of LiHuiLi Hospital of Medical School of Ningbo University, Ningbo, 315048, China.,The Affiliated Hospital of Medical School, Ningbo University, Ningbo, 315020, China
| | - Hui Zhang
- Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathophysiology, Medical School of Ningbo University, Ningbo, 315211, China.,The Affiliated Hospital of Medical School, Ningbo University, Ningbo, 315020, China
| | - Haibiao Wang
- Department of Hepato-Biliary-Pancreatic Surgery, The Affiliated Ningbo Medical Center of LiHuiLi Hospital of Medical School of Ningbo University, Ningbo, 315048, China. .,Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathophysiology, Medical School of Ningbo University, Ningbo, 315211, China.
| | - Meng Ye
- Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathophysiology, Medical School of Ningbo University, Ningbo, 315211, China. .,The Affiliated Hospital of Medical School, Ningbo University, Ningbo, 315020, China.
| | - Xiaofeng Jin
- Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathophysiology, Medical School of Ningbo University, Ningbo, 315211, China. .,The Affiliated Hospital of Medical School, Ningbo University, Ningbo, 315020, China.
| |
Collapse
|
5
|
Abstract
Almost 25 years have passed since a mutation of a formin gene, DIAPH1, was identified as being responsible for a human inherited disorder: a form of sensorineural hearing loss. Since then, our knowledge of the links between formins and disease has deepened considerably. Mutations of DIAPH1 and six other formin genes (DAAM2, DIAPH2, DIAPH3, FMN2, INF2 and FHOD3) have been identified as the genetic cause of a variety of inherited human disorders, including intellectual disability, renal disease, peripheral neuropathy, thrombocytopenia, primary ovarian insufficiency, hearing loss and cardiomyopathy. In addition, alterations in formin genes have been associated with a variety of pathological conditions, including developmental defects affecting the heart, nervous system and kidney, aging-related diseases, and cancer. This review summarizes the most recent discoveries about the involvement of formin alterations in monogenic disorders and other human pathological conditions, especially cancer, with which they have been associated. In vitro results and experiments in modified animal models are discussed. Finally, we outline the directions for future research in this field.
Collapse
Affiliation(s)
| | - Miguel A. Alonso
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, 28049 Madrid, Spain;
| |
Collapse
|
6
|
Formin-like protein 2 promotes cell proliferation by a p27-related mechanism in human breast cancer cells. BMC Cancer 2021; 21:760. [PMID: 34193109 PMCID: PMC8247103 DOI: 10.1186/s12885-021-08533-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 06/23/2021] [Indexed: 12/25/2022] Open
Abstract
Background Breast cancer is the leading cause of cancer-related deaths in females worldwide. Formin-like protein 2 (FMNL2) is a member of formin family that governs cytokinesis, cell polarity, morphogenesis and cell division. To our knowledge, the function of FMNL2 in breast cancer proliferation still remains uncovered. Methods Tumor immune estimation resource (TIMER) analysis was used to detect the correlation between FMNL2 and Ki67 in breast cancer tissues. Quantitative real-time transcription polymerase chain reaction (qRT-PCR) and western blotting were performed to analyze the expression in human breast cancer cells. Moreover, RNA interference (RNAi) and plasmids were performed to silence and overexpress FMNL2 and p27. The CCK8, MTT, cell counting, colony formation, and 5-ethynyl-2-deoxyuridine (EdU) incorporation assays were used to detect cell proliferation, respectively. Flow cytometry analysis was used to detect cell cycle distribution. Further, the distribution of p27 was examined using immunofluorescence. Results We found that FMNL2 expression was positively associated with Ki67 among collected breast cancer tissues and in TCGA database. Compared to lower proliferative cells MCF7 and T47D, FMNL2 was overexpressed in highly proliferative breast cancer cells MDA-MB-231, BT549 and SUM159, accompanied by reduced levels of p27 and p21, and elevated CyclinD1 and Ki67 expression. FMNL2 silencing significantly inhibited the cell proliferation of MDA-MB-231 and BT549 cells. Meanwhile, FMNL2 overexpression distinctly promoted the cell proliferation of MCF7 cells. Furthermore, FMNL2 suppressed the nuclear levels of p27 and promoted p27 proteasomal degradation in human breast cancer cells. The ubiquitination of p27 was inhibited by FMNL2 silencing in BT549 cells. Besides, p27 silencing markedly elevated Ki67 expression and cell viability, which could be blocked by additionally FMNL2 silencing in MDA-MB-231 and BT549 cells. Furthermore, overexpression of p27WT significantly reversed the increased levels of FMNL2 and Ki67, cell viability and cell cycle progression induced by FMNL2 overexpression in MCF7 cells. More importantly, compared to p27WT group, those effects could be significantly reversed by p27△NLS overexpression. Conclusions These results demonstrated that FMNL2 promoted cell proliferation partially by reducing p27 nuclear localization and p27 protein stability in human breast cancer cells, suggesting the pivotal role of FMNL2 in breast cancer progression. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-08533-w.
Collapse
|
7
|
Yang M, Wu X, Li L, Li S, Li N, Mao M, Pan S, Du R, Wang X, Chen M, Xiao N, Zhu X, He G, Zhang L, Huang W, Pan H, Deng L, Chen L, Liang L, Guan J. COMMD10 inhibits tumor progression and induces apoptosis by blocking NF-κB signal and values up BCLC staging in predicting overall survival in hepatocellular carcinoma. Clin Transl Med 2021; 11:e403. [PMID: 34047468 PMCID: PMC8093973 DOI: 10.1002/ctm2.403] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 04/11/2021] [Accepted: 04/18/2021] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the third leading cause of cancer mortality worldwide. Currently, there is limited knowledge of dysregulation of cellular proliferation and apoptosis that contribute to the malignant phenotype in HCC. Copper metabolism gene MURR1 domain 10 (COMMD10) is initially identified as a suppressor gene in the pathogenesis of HCC in our observations. Here we aimed to explore its function and prognostic value in the progression of HCC. METHODS Functional experiments were performed to explore the role of COMMD10 in HCC. The molecular mechanisms of COMMD10 were determined by luciferase assay, immunofluorescence, and immunoprecipitation. The nomogram was based on a retrospective and multicenter study of 516 patients who were pathologically diagnosed with HCC from three Chinese hospitals. The predictive accuracy and discriminative ability of the nomogram were determined by a C-index and calibration curve and were compared with COMMD10 and the Barcelona Clinic Liver Cancer (BCLC) staging system. The primary endpoint was overall survival (OS). RESULTS COMMD10 expression was significantly lower in HCC than that in normal liver tissues. In vitro and in vivo experiments revealed that COMMD10 suppressed cell proliferation and induced apoptosis in HCC. Mechanistically, COMMD10 inhibits TNFα mediated ubiquitination of IκBα and p65 nuclear translocation through the combination of COMMD10-N terminal to the Rel homology domain of p65, which inhibited NF-κB activity and increased expression of cleaved caspase9/3 in HCC. Clinically, COMMD10 stratifies early-stage HCC patients into two risk groups with significantly different OS. Additionally, the nomogram based on COMMD10 and BCLC stage yielded more accuracy than BCLC stage alone for predicting OS of HCC patients in three cohorts. CONCLUSIONS COMMD10 suppresses proliferation and promotes apoptosis by inhibiting NF-κB signaling and values up BCLC staging in predicting OS, which provides evidence for the identification of potential therapeutic targets and the accurate prediction of prognosis for patients with HCC.
Collapse
Affiliation(s)
- Mi Yang
- Department of Radiation Oncology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Xixi Wu
- Department of Radiation Oncology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Lu Li
- Department of Radiation Oncology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Shaoqun Li
- Department of Radiation Oncology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Nan Li
- Department of Radiation Oncology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Mengyuan Mao
- Department of Radiation Oncology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Suming Pan
- Department of RadiotherapyYue Bei People's Hospital of Guangdong provinceShaoguanGuangdongChina
| | - Richang Du
- Department of PathologyYue Bei People's Hospital of Guangdong provinceShaoguanGuangdongChina
| | - Xiaoqing Wang
- Department of Radiation Oncology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Min Chen
- Department of Radiation Oncology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Nanjie Xiao
- Department of Radiation Oncology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Xiaohui Zhu
- Department of Patholog, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
- Department of Patholog, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdongChina
- Guangdong Province Key Laboratory of Molecular Tumor PathologyGuangzhouGuangdongChina
| | - Guoyang He
- Department of Patholog, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
- Department of Patholog, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdongChina
- Guangdong Province Key Laboratory of Molecular Tumor PathologyGuangzhouGuangdongChina
| | - Longshan Zhang
- Department of Radiation Oncology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Weiqiang Huang
- Department of Radiation Oncology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Hua Pan
- Department of Radiation Oncology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Lan Deng
- Department of Hematology, Zhujiang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Longhua Chen
- Department of Radiation Oncology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Li Liang
- Department of Patholog, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
- Department of Patholog, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdongChina
- Guangdong Province Key Laboratory of Molecular Tumor PathologyGuangzhouGuangdongChina
| | - Jian Guan
- Department of Radiation Oncology, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| |
Collapse
|
8
|
Valente LJ, Tarangelo A, Li AM, Naciri M, Raj N, Boutelle AM, Li Y, Mello SS, Bieging-Rolett K, DeBerardinis RJ, Ye J, Dixon SJ, Attardi LD. p53 deficiency triggers dysregulation of diverse cellular processes in physiological oxygen. J Cell Biol 2021; 219:152074. [PMID: 32886745 PMCID: PMC7594498 DOI: 10.1083/jcb.201908212] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 06/17/2020] [Accepted: 07/28/2020] [Indexed: 12/20/2022] Open
Abstract
The mechanisms by which TP53, the most frequently mutated gene in human cancer, suppresses tumorigenesis remain unclear. p53 modulates various cellular processes, such as apoptosis and proliferation, which has led to distinct cellular mechanisms being proposed for p53-mediated tumor suppression in different contexts. Here, we asked whether during tumor suppression p53 might instead regulate a wide range of cellular processes. Analysis of mouse and human oncogene-expressing wild-type and p53-deficient cells in physiological oxygen conditions revealed that p53 loss concurrently impacts numerous distinct cellular processes, including apoptosis, genome stabilization, DNA repair, metabolism, migration, and invasion. Notably, some phenotypes were uncovered only in physiological oxygen. Transcriptomic analysis in this setting highlighted underappreciated functions modulated by p53, including actin dynamics. Collectively, these results suggest that p53 simultaneously governs diverse cellular processes during transformation suppression, an aspect of p53 function that would provide a clear rationale for its frequent inactivation in human cancer.
Collapse
Affiliation(s)
- Liz J Valente
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA
| | - Amy Tarangelo
- Department of Biology, Stanford University, Stanford, CA
| | - Albert Mao Li
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA
| | - Marwan Naciri
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA.,École Normale Supérieure de Lyon, Université Claude Bernard Lyon I, Université de Lyon, Lyon, France
| | - Nitin Raj
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA
| | - Anthony M Boutelle
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA
| | - Yang Li
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA
| | - Stephano Spano Mello
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA.,Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY
| | - Kathryn Bieging-Rolett
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA
| | - Ralph J DeBerardinis
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX.,Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX
| | - Jiangbin Ye
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA
| | - Scott J Dixon
- Department of Biology, Stanford University, Stanford, CA
| | - Laura D Attardi
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA.,Department of Genetics, Stanford University School of Medicine, Stanford, CA.,Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
9
|
Biber G, Ben-Shmuel A, Sabag B, Barda-Saad M. Actin regulators in cancer progression and metastases: From structure and function to cytoskeletal dynamics. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 356:131-196. [PMID: 33066873 DOI: 10.1016/bs.ircmb.2020.05.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The cytoskeleton is a central factor contributing to various hallmarks of cancer. In recent years, there has been increasing evidence demonstrating the involvement of actin regulatory proteins in malignancy, and their dysregulation was shown to predict poor clinical prognosis. Although enhanced cytoskeletal activity is often associated with cancer progression, the expression of several inducers of actin polymerization is remarkably reduced in certain malignancies, and it is not completely clear how these changes promote tumorigenesis and metastases. The complexities involved in cytoskeletal induction of cancer progression therefore pose considerable difficulties for therapeutic intervention; it is not always clear which cytoskeletal regulator should be targeted in order to impede cancer progression, and whether this targeting may inadvertently enhance alternative invasive pathways which can aggravate tumor growth. The entire constellation of cytoskeletal machineries in eukaryotic cells are numerous and complex; the system is comprised of and regulated by hundreds of proteins, which could not be covered in a single review. Therefore, we will focus here on the actin cytoskeleton, which encompasses the biological machinery behind most of the key cellular functions altered in cancer, with specific emphasis on actin nucleating factors and nucleation-promoting factors. Finally, we discuss current therapeutic strategies for cancer which aim to target the cytoskeleton.
Collapse
Affiliation(s)
- G Biber
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - A Ben-Shmuel
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - B Sabag
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - M Barda-Saad
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel.
| |
Collapse
|
10
|
Nie H, Mei J, Zhang Q, An F, Zhan Q. Systematic Characterization of the Expression and Prognostic Values of Formin-Like Gene Family in Gastric Cancer. DNA Cell Biol 2020; 39:1664-1677. [PMID: 32551946 DOI: 10.1089/dna.2020.5508] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Formin-like genes (FMNLs) are members of formins family and have been implicated to the development and progression of multiple cancers. This research aims to analyze the expression profiles, prognostic values, and immune infiltrating associations of FMNLs in gastric cancer (GC) using multiple online bioinformatics website, including Oncomine, UALCAN, Kaplan-Meier Plotter, TIMER, GeneMANIA, DAVID, and LinkedOmics databases. The mRNA levels of FMNL1/2/3 were higher in GC tissues than normal. Meanwhile, FMNLs expressions tend to be upregulated in advanced and poorly differentiated GC. Prognostic value analysis suggested that high transcription levels of FMNL1/3 were associated with poor overall survival in GC patients. Correlation analysis between FMNLs expressions and immune infiltrating GC revealed that the expressions of FMNLs were significantly associated with immune infiltrating. Protein-protein interaction network and enrichment analysis of FMNLs in GC showed that FMNLs coexpressed genes mainly participated in organizing actin cytoskeleton through affecting small G proteins activity. Moreover, Gene Set Enrichment Analysis (GSEA) analysis uncovered FMNLs and their coexpressed genes was tightly associated with immune-related cellular functions. These findings demonstrate that FMNLs might play significant immunomodulatory roles in tumor immunity and could be novel therapeutic targets and potential prognostic biomarkers in GC.
Collapse
Affiliation(s)
- He Nie
- Department of Gastroenterology and Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Jie Mei
- Department of Oncology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Qinglin Zhang
- Department of Gastroenterology and Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Fangmei An
- Department of Gastroenterology and Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Qiang Zhan
- Department of Gastroenterology and Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| |
Collapse
|
11
|
Zhong B, Wang K, Xu H, Kong F. Silencing Formin-like 2 inhibits growth and metastasis of gastric cancer cells through suppressing internalization of integrins. Cancer Cell Int 2018; 18:79. [PMID: 29881327 PMCID: PMC5984784 DOI: 10.1186/s12935-018-0576-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 05/26/2018] [Indexed: 12/21/2022] Open
Abstract
Background Formin-like 2 (FMNL2) is a member of Formin family which governs cytokinesis, cellular polarity and morphogenesis. Dysregulation of FMNL2 has been discovered in cancers and is closely related to cancers. However, the role of FMNL2 in gastric cancer remains unclear. In this study, we aimed to investigate the role of FMNL2 in gastric cancer cells. Methods A FMNL2-specific shRNA was employed to decrease the endogenous expression of FMNL2. Then the degree of proliferation, apoptosis, migration and invasion of gastric cancer cells was assessed by MTT assay, flow cytometry, wound healing assay and transwell assay, respectively. The expression and distribution of FMNL2 and protein kinase C (PKC) α was detected by immunofluorescence. The internalization of integrins was detected by enzyme-linked immunosorbent assay. Results Our results showed that silencing FMNL2 suppressed proliferation, migration and invasion, and induced apoptosis of gastric cancer cells. The integrin internalization induced by PKC was declined by FMNL2 silencing. Conclusions Our study reveals that silencing FMNL2 suppresses growth and metastasis of gastric cancer cells. Modulation on integrin internalization may be implicated in the role of FMNL2 in growth and migration of gastric cancer cells. Our study indicates that FMNL2 may become a potential therapeutic target for gastric cancer.
Collapse
Affiliation(s)
- Banghua Zhong
- Department of Gastric, Intestine and Hernia Surgery, The First Affiliated Hospital of China Medical University, Shenyang, 110001 People's Republic of China
| | - Kewei Wang
- Department of Gastric, Intestine and Hernia Surgery, The First Affiliated Hospital of China Medical University, Shenyang, 110001 People's Republic of China
| | - Hao Xu
- Department of Gastric, Intestine and Hernia Surgery, The First Affiliated Hospital of China Medical University, Shenyang, 110001 People's Republic of China
| | - Fanmin Kong
- Department of Gastric, Intestine and Hernia Surgery, The First Affiliated Hospital of China Medical University, Shenyang, 110001 People's Republic of China
| |
Collapse
|
12
|
Yang SS, Li XM, Yang M, Ren XL, Hu JL, Zhu XH, Wang FF, Zeng ZC, Li JY, Cheng ZQ, Liao WT, Ding YQ, Guan J, Liang L. FMNL2 destabilises COMMD10 to activate NF-κB pathway in invasion and metastasis of colorectal cancer. Br J Cancer 2017; 117:1164-1175. [PMID: 28817833 PMCID: PMC5674093 DOI: 10.1038/bjc.2017.260] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 06/19/2017] [Accepted: 07/13/2017] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Diaphanous-related formins (DRFs), actin necleator, have been known to participate in the progression of cancer cells. We previously reported that FMNL2 (Formin-like2), a member of DRFs, was a positive regulator in colorectal cancer (CRC) metastasis, yet proteins and pathways required for the function of this pro-invasive DRFs remain to be identified. METHODS The relationship between FMNL2 and COMMD10 was examined using Co-IP, GST pull-down, immunofluorescence and in vitro ubiquitination assay. The in vitro and in vivo function of COMMD10 in CRC was evaluated using CCK-8 proliferation assay, plate colony formation, cell cycle, apoptosis and animal models. The inhibition of NF-κB signalling by COMMD10 was detected using dual-luciferase reporter assay and western blotting. Co-IP, GST pull-down and nuclear protein extraction assay were performed to evaluate the effect on p65 by COMMD10. Real-time PCR and western blotting were performed to detect expressions of FMNL2, COMMD10 and p65 in paired tissues. RESULTS FMNL2 targets COMMD10 for ubiquitin-mediated proteasome degradation in CRC cells. COMMD10 targets p65 NF-κB (nuclear factor-κB) subunit and reduces its nuclear translocation, thereby leading to the inactivation of NF-κB pathway and suppression of CRC invasion and metastasis. Inhibition of NF-κB signalling by COMMD10 is necessary for FMNL2-mediated CRC cell behaviours. Downregulation of COMMD10 predicts poor prognosis of CRC patients. The expressions of FMNL2, COMMD10 and p65 are highly linked in CRC tissues. CONCLUSIONS These data demonstrate that the FMNL2/COMMD10/p65 axis acts as a critical regulator in the maintenance of metastatic phenotypes and is strongly associated with negative clinical outcomes.
Collapse
Affiliation(s)
- S S Yang
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, Guangdong Province, People’s Republic of China
- Department of Pathology, The Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, Guangdong Province, People’s Republic of China
| | - X M Li
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, Guangdong Province, People’s Republic of China
- Department of Pathology, Shenzhen Baoan Maternal and Child Health Hospital, Shenzhen 518100, Guangdong Province, People’s Republic of China
| | - M Yang
- Department of Radiotherapy, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, People’s Republic of China
| | - X L Ren
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, Guangdong Province, People’s Republic of China
- Guangdong Provincial Key Laboratory of Molecular Oncologic Pathology, Guangzhou 510515, Guangdong Province, People’s Republic of China
| | - J L Hu
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, Guangdong Province, People’s Republic of China
- Guangdong Provincial Key Laboratory of Molecular Oncologic Pathology, Guangzhou 510515, Guangdong Province, People’s Republic of China
| | - X H Zhu
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, Guangdong Province, People’s Republic of China
- Guangdong Provincial Key Laboratory of Molecular Oncologic Pathology, Guangzhou 510515, Guangdong Province, People’s Republic of China
| | - F F Wang
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, Guangdong Province, People’s Republic of China
- Guangdong Provincial Key Laboratory of Molecular Oncologic Pathology, Guangzhou 510515, Guangdong Province, People’s Republic of China
| | - Z C Zeng
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, Guangdong Province, People’s Republic of China
- Guangdong Provincial Key Laboratory of Molecular Oncologic Pathology, Guangzhou 510515, Guangdong Province, People’s Republic of China
| | - J Y Li
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, Guangdong Province, People’s Republic of China
- Guangdong Provincial Key Laboratory of Molecular Oncologic Pathology, Guangzhou 510515, Guangdong Province, People’s Republic of China
| | - Z Q Cheng
- Department of Pathology, Shenzhen people’s Hospital, Second Clinical Medical College of Jinan University, Shenzhen 518020, Guangdong Province, People’s Republic of China
| | - W T Liao
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, Guangdong Province, People’s Republic of China
- Guangdong Provincial Key Laboratory of Molecular Oncologic Pathology, Guangzhou 510515, Guangdong Province, People’s Republic of China
| | - Y Q Ding
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, Guangdong Province, People’s Republic of China
- Guangdong Provincial Key Laboratory of Molecular Oncologic Pathology, Guangzhou 510515, Guangdong Province, People’s Republic of China
| | - J Guan
- Department of Radiotherapy, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, People’s Republic of China
- E-mail:
| | - L Liang
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, Guangdong Province, People’s Republic of China
- Guangdong Provincial Key Laboratory of Molecular Oncologic Pathology, Guangzhou 510515, Guangdong Province, People’s Republic of China
- E-mail:
| |
Collapse
|
13
|
Xu D, Xu R, He L, Xu T, Zhang Z, Han D, Du J. Comparison of Pathogenic Mechanisms Underlying Single and Recurrent Venous Thromboembolism Based on Gene Expression Profiling. Ann Vasc Surg 2016; 36:252-259. [PMID: 27423718 DOI: 10.1016/j.avsg.2016.05.088] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 05/16/2016] [Accepted: 05/23/2016] [Indexed: 11/19/2022]
Abstract
BACKGROUND Unprovoked venous thromboembolism (VTE), generally divided into single and recurrent categories, is a common leading cause of morbidity and mortality in a real-world population. This study was aimed to explore the similarities and differences in the mechanisms of single and recurrent VTE. METHODS Gene expression data (GSE19151) generated from 63 healthy controls, 32 single, and 38 recurrent VTE patients were analyzed. Differentially expressed genes (DEGs) were screened by Affy package and Kyoto Encyclopedia of Genes and Genomes and Gene Ontology enrichment analysis were performed using database for annotation, visualization, and integrated discovery. Based on the Search Tool for the Retrieval of Interacting Genes/Proteins database, protein-protein interaction network was visualized by Cytoscape, and modules were identified by CFinder. Finally, transcription factor regulatory networks were constructed. RESULTS Totally, 559 and 294 DEGs were obtained from recurrent and single VTE, respectively. There were 202 upregulated and 58 downregulated genes overlapped between them. Terms of regulation of actin cytoskeleton enriched by downregulated genes and oxidative phosphorylation enriched by upregulated genes were found in 2 types of VTE. Leukocyte transendothelial migration and Jak-STAT signaling pathway were found related with recurrent VTE. In addition, genes including signal transducer and activator of transcription 3 (STAT3) involving in the Jak-STAT signaling pathway were highly connected nodes. CONCLUSIONS Actin cytoskeleton and oxidative phosphorylation may be involved in the common mechanisms of recurrent VTE and single VTE. Leukocyte migration and Jak-STAT signaling pathway and their related genes may be important for the development and recurrence of VTE.
Collapse
Affiliation(s)
- Dejun Xu
- Department of Vascular and Lymphatic Surgery, Vascular and Lymphatic Surgery, China Japan Union Hospital of Jilin University, Changchun, Jilin, PR China
| | - Rutao Xu
- Department of Vascular Surgery, Henan Provincial People's Hospital, Zhengzhou, Henan, PR China
| | - Liu He
- Department of Anesthesiology, China Japan Union Hospital of Jilin University, Changchun, Jilin, PR China
| | - Tao Xu
- Department of Gastroenterology, The Fourth Clinical Hospital of Jilin University, Changchun, Jilin, PR China
| | - Zhenyu Zhang
- The Second Department of General Surgery, Armed Police Corps Hospital in Jilin Province, Changchun, Jilin, PR China
| | - Dongmei Han
- Department of Vascular and Lymphatic Surgery, Vascular and Lymphatic Surgery, China Japan Union Hospital of Jilin University, Changchun, Jilin, PR China
| | - Jianshi Du
- Department of Vascular and Lymphatic Surgery, Vascular and Lymphatic Surgery, China Japan Union Hospital of Jilin University, Changchun, Jilin, PR China.
| |
Collapse
|
14
|
Liu XR, Cai CX, Luo LM, Zheng WL, Shi R, Zeng J, Xu YQ, Wei M, Ma WL. Decreased expression of Sushi Domain Containing 2 correlates to progressive features in patients with hepatocellular carcinoma. Cancer Cell Int 2016; 16:15. [PMID: 26933386 PMCID: PMC4772460 DOI: 10.1186/s12935-016-0286-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 02/03/2016] [Indexed: 12/21/2022] Open
Abstract
Background Sushi Domain Containing 2 (SUSD2) has been identified as a regulator of colon and breast cancer. Increasing evidence suggests that SUSD2 plays a key role in tumorigenesis. However, the SUSD2 expression status and its functions in hepatocellular carcinoma (HCC) are still unrevealed. In the present study, we intended to investigate SUSD2 expression status and its correlation with the clinicopathological features in HCC patients. Furthermore,we examined the influence of SUSD2 on the proliferation, apoptosis, invasion and migration of the HCC cell lines HepG2 and SMMC7721. Methods We evaluated the SUSD2 expression in HCC tissues and paired normal liver tissues by quantitative real-time PCR and western blotting analysis. The clinicopathological significance of SUSD2 was investigated by immunohistochemistry (IHC) on a HCC tissue microarray. Receiver operating characteristic (ROC) analysis was applied to determine the optimal cut-off score for positive expression of SUSD2. The correlation between SUSD2 protein expression and clinicopathological features of HCC was analyzed by Chi square test. The cell proliferation, apoptosis, invasion and migration potential were observed to detect the functions of SUSD2 in HCC cells. Results Decreased expression of SUSD2 mRNA and protein were observed in the majority of HCC tissues, compared with paired normal liver tissues. When SUSD2 high expression percentage was determined to be above 52.5 % (area under ROC curve = 0.769, P = 0.000), low expression of SUSD2 was observed in 62.2 % (112/180) of HCC tissues and high expression of SUSD2 was observed in all normal liver tissues (16/16) by IHC. Decreased expression of SUSD2 in patients was correlated with high histological grade (χ2 = 5.198, P = 0.023), advanced clinical stage (χ2 = 30.244, P = 0.000), pT status (χ2 = 33.175, P = 0.000), pN status (χ2 = 4.785, P = 0.029), pM status (χ2 = 4.620, P = 0.032). Down-regulation of SUSD2 promoted cell proliferation,invasion and migration,reduced the cell apoptosis. Conclusions Our findings suggest that SUSD2 may play as a tumor suppressor in HCC cells and could be served as an additional potential marker for diagnosis.
Collapse
Affiliation(s)
- Xin-Rui Liu
- Institute of Genetic Engineering, School of Basic Medical Sciences, Southern Medical University, No.1838, Baiyun Road North, Guangzhou, China
| | - Cui-Xia Cai
- Institute of Genetic Engineering, School of Basic Medical Sciences, Southern Medical University, No.1838, Baiyun Road North, Guangzhou, China
| | - Li-Min Luo
- Centre for Liver Disease, 458th Hospital of PLA, Guangzhou, 510602 China
| | - Wen-Ling Zheng
- Institute of Genetic Engineering, School of Basic Medical Sciences, Southern Medical University, No.1838, Baiyun Road North, Guangzhou, China
| | - Rong Shi
- Institute of Genetic Engineering, School of Basic Medical Sciences, Southern Medical University, No.1838, Baiyun Road North, Guangzhou, China
| | - Jun Zeng
- Institute of Genetic Engineering, School of Basic Medical Sciences, Southern Medical University, No.1838, Baiyun Road North, Guangzhou, China
| | - You-Qin Xu
- Institute of Genetic Engineering, School of Basic Medical Sciences, Southern Medical University, No.1838, Baiyun Road North, Guangzhou, China
| | - Min Wei
- Institute of Genetic Engineering, School of Basic Medical Sciences, Southern Medical University, No.1838, Baiyun Road North, Guangzhou, China
| | - Wen-Li Ma
- Institute of Genetic Engineering, School of Basic Medical Sciences, Southern Medical University, No.1838, Baiyun Road North, Guangzhou, China
| |
Collapse
|
15
|
Zeng Y, Xie H, Qiao Y, Wang J, Zhu X, He G, Li Y, Ren X, Wang F, Liang L, Ding Y. Formin-like2 regulates Rho/ROCK pathway to promote actin assembly and cell invasion of colorectal cancer. Cancer Sci 2016; 106:1385-93. [PMID: 26258642 PMCID: PMC4638017 DOI: 10.1111/cas.12768] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 07/16/2015] [Accepted: 08/02/2015] [Indexed: 12/26/2022] Open
Abstract
Formin-like2 (FMNL2) is a member of the diaphanous-related formins family, which act as effectors and upstream modulators of Rho GTPases signaling and control the actin-dependent processes, such as cell motility or invasion. FMNL2 has been identified as promoting the motility and metastasis in colorectal carcinoma (CRC). However, whether FMNL2 regulates Rho signaling to promote cancer cell invasion remains unclear. In this study, we demonstrated an essential role for FMNL2 in the activations of Rho/ROCK pathway, SRF transcription or actin assembly, and subsequent CRC cell invasion. FMNL2 could activate Rho/ROCK pathway, and required ROCK to promote CRC cell invasion. Moreover, FMNL2 promoted the formation of filopodia and stress fiber, and activated the SRF transcription in a Rho-dependent manner. We also demonstrated that FMNL2 was necessary for LPA-induced invasion, RhoA/ROCK activation, actin assembly and SRF activation. FMNL2 was an essential component of LPA signal transduction toward RhoA by directly interacting with LARG. LARG silence inhibited RhoA/ROCK pathway and CRC cell invasion. Collectively, these data indicate that FMNL2, acting as upstream of RhoA by interacting with LARG, can promote actin assembly and CRC cell invasion through a Rho/ROCK-dependent mechanism.
Collapse
Affiliation(s)
- Yuanfeng Zeng
- Department of Pathology, Southern Medical University, Guangzhou, China.,Department of Pathology, the People's Hospital, Nanchang, China
| | - Huijun Xie
- Department of Pathology, Southern Medical University, Guangzhou, China.,Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Yudan Qiao
- Department of Pathology, Southern Medical University, Guangzhou, China.,Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Jianmei Wang
- Department of Pathology, Southern Medical University, Guangzhou, China.,Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Xiling Zhu
- Department of Pathology, Southern Medical University, Guangzhou, China.,Department of Oncology, General Hospital of Armed Police Forces, Beijing, China
| | - Guoyang He
- Department of Pathology, Southern Medical University, Guangzhou, China.,Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Yuling Li
- Department of Pathology, Southern Medical University, Guangzhou, China.,Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Xiaoli Ren
- Department of Pathology, Southern Medical University, Guangzhou, China.,Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Feifei Wang
- Department of Pathology, Southern Medical University, Guangzhou, China.,Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Li Liang
- Department of Pathology, Southern Medical University, Guangzhou, China.,Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Yanqing Ding
- Department of Pathology, Southern Medical University, Guangzhou, China.,Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| |
Collapse
|
16
|
Gardberg M, Heuser VD, Koskivuo I, Koivisto M, Carpén O. FMNL2/FMNL3 formins are linked with oncogenic pathways and predict melanoma outcome. JOURNAL OF PATHOLOGY CLINICAL RESEARCH 2016; 2:41-52. [PMID: 27499915 PMCID: PMC4858127 DOI: 10.1002/cjp2.34] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 12/02/2015] [Indexed: 01/19/2023]
Abstract
While most early (stage I‐II) melanomas are cured by surgery, recurrence is not uncommon. Prognostication by current clinicopathological parameters does not provide sufficient means for identifying patients who are at risk of developing metastases and in need of adjuvant therapy. Actin‐regulating formins may account for invasive properties of cancer cells, including melanoma. Here, we studied formin‐like protein 2 and 3 (FMNL2 and FMNL3) in melanoma by analysing their role in the invasive properties of melanoma cells and by evaluating whether FMNL2 expression is associated with melanoma outcome. Immunohistochemical characterization of FMNL2 in a cohort of 175 primary cutaneous stage I‐II melanomas indicated that high FMNL2 reactivity correlates with poor outcome as evaluated by recurrence free survival (p < 0.0001) or disease specific survival (p < 0.0001). In multivariate analysis, Breslow's thickness (p < 0.05) and FMNL2 expression (p < 0.001) remained as independent prognostic factors. Cellular studies revealed that FMNL2 is a component of filopodia in many melanoma cell lines. Inhibition of either FMNL2 or the closely related FMNL3 affected the maintenance of melanoma cell morphology and reduced migration. Finally, inhibition of the BRAF, PI3K and MAPK oncogenic pathways markedly reduced expression of both FMNL2 and FMNL3 in melanoma cells. The results suggest a major role for FMNL2/FMNL3 formins in melanoma biology and raise the possibility that the novel targeted melanoma drugs may interfere with the cellular properties regulated by these formins.
Collapse
Affiliation(s)
- Maria Gardberg
- Department of Pathology University of Turku and Turku University Hospital Turku Finland
| | - Vanina D Heuser
- Department of Pathology University of Turku and Turku University Hospital Turku Finland
| | - Ilkka Koskivuo
- Department of Plastic and General Surgery Turku University Hospital Turku Finland
| | - Mari Koivisto
- Department of Biostatistics University of Turku Turku Finland
| | - Olli Carpén
- Department of PathologyUniversity of Turku and Turku University HospitalTurkuFinland; Auria Biobank, Turku University HospitalTurkuFinland
| |
Collapse
|
17
|
Randall TS, Ehler E. A formin-g role during development and disease. Eur J Cell Biol 2014; 93:205-11. [PMID: 24342720 DOI: 10.1016/j.ejcb.2013.11.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 11/15/2013] [Accepted: 11/18/2013] [Indexed: 11/22/2022] Open
Abstract
Several different protein families were shown to be involved in the regulation of actin filament formation and have been studied extensively in processes such as cell migration. Among them are members of the formin family, which tend to promote the formation of linear actin filaments. Studies in recent years, often using loss of function animal models, have indicated that formin family members play roles beyond cell motility in vitro and are involved in processes ranging from tissue morphogenesis and cell differentiation to diseases such as cancer and cardiomyopathy. Therefore the aim of this review is to discuss these findings and to start putting them into a subcellular context.
Collapse
Affiliation(s)
- Thomas S Randall
- Randall Division of Cell and Molecular Biophysics, Cardiovascular Division, British Heart Foundation Centre of Research Excellence, King's College London, London SE1 1UL, United Kingdom
| | - Elisabeth Ehler
- Randall Division of Cell and Molecular Biophysics, Cardiovascular Division, British Heart Foundation Centre of Research Excellence, King's College London, London SE1 1UL, United Kingdom.
| |
Collapse
|
18
|
Lv Z, Zou H, Peng K, Wang J, Ding Y, Li Y, Ren X, Wang F, Chang R, Liang L, Ding Y. The suppressive role and aberrent promoter methylation of BTG3 in the progression of hepatocellular carcinoma. PLoS One 2013; 8:e77473. [PMID: 24147003 PMCID: PMC3798399 DOI: 10.1371/journal.pone.0077473] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Accepted: 09/02/2013] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND BTG3 (B-cell translocation gene 3) has been identified as a tumor suppressor and hypermethylation contributes to its down-regulation in some tumors, but its role in hepatocellular carcinoma (HCC) remain unknown. This study aimed to detect the expression and methylation status of BTG3 in HCC cell lines or tissues, and determine its function in HCC progression. METHODOLOGY The expression of BTG3 was detected in HCC cell lines and HCC tissue by real-time RT-PCR, Western blot or immunohistochemistry. The promoter methylation status of BTG3 was measured by using methylation-specific PCR in HCC cell lines. A series of assays were performed to evaluate the effect of BTG3 on proliferation, invasion and cell cycle transition in vitro. RESULTS BTG3 expression was lower in HCC cell lines than in hepatocyte cell line LO2 (P<0.05). BTG3 was also down-regulated in HCC tissues. Its expression was positively correlated with differentiation and distant metastasis (P<0.05). Patients with lower BTG3 expression had shorter overall survival time (P=0.029). DNA methylation directed repression of BTG3 mRNA expression in HCC cell lines. BTG3 suppressed proliferation, invasion and induces G1/S cycle arrest of HCC cells in vitro. CONCLUSION Down-regulation of BTG3 due to the promoter hypermethylation is closely associated with proliferation, invasion and cell cycle arrest of HCC cells. It may be a novel prognostic biomarker for HCC patients.
Collapse
Affiliation(s)
- Zhenbing Lv
- Department of Pathology, Southern Medical University, Guangzhou, Guangdong Province, People’s Republic of China
- Department of General Surgery, Nanchong Central Hospital, Nanchong City, Sichuan Province, People’s Republic of China
| | - Huichun Zou
- Graduate School, Southern Medical University, Guangzhou City, Guangdong Province, People’s Republic of China
| | - Kaiwen Peng
- Graduate School, Southern Medical University, Guangzhou City, Guangdong Province, People’s Republic of China
| | - Jianmei Wang
- Department of Pathology, Southern Medical University, Guangzhou, Guangdong Province, People’s Republic of China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, People’s Republic of China
| | - Yi Ding
- Department of Radiotherapy, Nanfang Hospital, Southern Medical University, Guangzhou City, Guangdong, People’s Republic of China
| | - Yuling Li
- Department of Pathology, Southern Medical University, Guangzhou, Guangdong Province, People’s Republic of China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, People’s Republic of China
| | - Xiaoli Ren
- Department of Pathology, Southern Medical University, Guangzhou, Guangdong Province, People’s Republic of China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, People’s Republic of China
| | - Feifei Wang
- Department of Pathology, Southern Medical University, Guangzhou, Guangdong Province, People’s Republic of China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, People’s Republic of China
| | - Rui Chang
- Second School of Clinical Medicine, Southern Medical University, Guangzhou City, Guangdong Province, People’s Republic of China
| | - Li Liang
- Department of Pathology, Southern Medical University, Guangzhou, Guangdong Province, People’s Republic of China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, People’s Republic of China
- * E-mail: (YD); (LL)
| | - Yanqing Ding
- Department of Pathology, Southern Medical University, Guangzhou, Guangdong Province, People’s Republic of China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, People’s Republic of China
- * E-mail: (YD); (LL)
| |
Collapse
|
19
|
Pharmacological Inhibition of Actin Assembly to Target Tumor Cell Motility. Rev Physiol Biochem Pharmacol 2013; 166:23-42. [DOI: 10.1007/112_2013_16] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
20
|
Liang WS, Craig DW, Carpten J, Borad MJ, Demeure MJ, Weiss GJ, Izatt T, Sinari S, Christoforides A, Aldrich J, Kurdoglu A, Barrett M, Phillips L, Benson H, Tembe W, Braggio E, Kiefer JA, Legendre C, Posner R, Hostetter GH, Baker A, Egan JB, Han H, Lake D, Stites EC, Ramanathan RK, Fonseca R, Stewart AK, Von Hoff D. Genome-wide characterization of pancreatic adenocarcinoma patients using next generation sequencing. PLoS One 2012; 7:e43192. [PMID: 23071490 PMCID: PMC3468610 DOI: 10.1371/journal.pone.0043192] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Accepted: 07/19/2012] [Indexed: 12/24/2022] Open
Abstract
Pancreatic adenocarcinoma (PAC) is among the most lethal malignancies. While research has implicated multiple genes in disease pathogenesis, identification of therapeutic leads has been difficult and the majority of currently available therapies provide only marginal benefit. To address this issue, our goal was to genomically characterize individual PAC patients to understand the range of aberrations that are occurring in each tumor. Because our understanding of PAC tumorigenesis is limited, evaluation of separate cases may reveal aberrations, that are less common but may provide relevant information on the disease, or that may represent viable therapeutic targets for the patient. We used next generation sequencing to assess global somatic events across 3 PAC patients to characterize each patient and to identify potential targets. This study is the first to report whole genome sequencing (WGS) findings in paired tumor/normal samples collected from 3 separate PAC patients. We generated on average 132 billion mappable bases across all patients using WGS, and identified 142 somatic coding events including point mutations, insertion/deletions, and chromosomal copy number variants. We did not identify any significant somatic translocation events. We also performed RNA sequencing on 2 of these patients' tumors for which tumor RNA was available to evaluate expression changes that may be associated with somatic events, and generated over 100 million mapped reads for each patient. We further performed pathway analysis of all sequencing data to identify processes that may be the most heavily impacted from somatic and expression alterations. As expected, the KRAS signaling pathway was the most heavily impacted pathway (P<0.05), along with tumor-stroma interactions and tumor suppressive pathways. While sequencing of more patients is needed, the high resolution genomic and transcriptomic information we have acquired here provides valuable information on the molecular composition of PAC and helps to establish a foundation for improved therapeutic selection.
Collapse
Affiliation(s)
- Winnie S. Liang
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - David W. Craig
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - John Carpten
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | | | - Michael J. Demeure
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
- Virginia G. Piper Cancer Center Clinical Trials, Scottsdale Healthcare, Scottsdale, Arizona, United States of America
| | - Glen J. Weiss
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
- Virginia G. Piper Cancer Center Clinical Trials, Scottsdale Healthcare, Scottsdale, Arizona, United States of America
| | - Tyler Izatt
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Shripad Sinari
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Alexis Christoforides
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Jessica Aldrich
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Ahmet Kurdoglu
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Michael Barrett
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Lori Phillips
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Hollie Benson
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Waibhav Tembe
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | | | - Jeffrey A. Kiefer
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Christophe Legendre
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Richard Posner
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Galen H. Hostetter
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Angela Baker
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Jan B. Egan
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
- Mayo Clinic, Scottsdale, Arizona, United States of America
| | - Haiyong Han
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Douglas Lake
- Arizona State University, Tempe, Arizona, United States of America
| | - Edward C. Stites
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Ramesh K. Ramanathan
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
- Virginia G. Piper Cancer Center Clinical Trials, Scottsdale Healthcare, Scottsdale, Arizona, United States of America
| | - Rafael Fonseca
- Mayo Clinic, Scottsdale, Arizona, United States of America
| | | | - Daniel Von Hoff
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
- Mayo Clinic, Scottsdale, Arizona, United States of America
- Virginia G. Piper Cancer Center Clinical Trials, Scottsdale Healthcare, Scottsdale, Arizona, United States of America
- * E-mail:
| |
Collapse
|
21
|
Liang WS, Craig DW, Carpten J, Borad MJ, Demeure MJ, Weiss GJ, Izatt T, Sinari S, Christoforides A, Aldrich J, Kurdoglu A, Barrett M, Phillips L, Benson H, Tembe W, Braggio E, Kiefer JA, Legendre C, Posner R, Hostetter GH, Baker A, Egan JB, Han H, Lake D, Stites EC, Ramanathan RK, Fonseca R, Stewart AK, Von Hoff D. Genome-wide characterization of pancreatic adenocarcinoma patients using next generation sequencing. PLoS One 2012. [PMID: 23071490 DOI: 10.137/journal.pone.0043192] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Pancreatic adenocarcinoma (PAC) is among the most lethal malignancies. While research has implicated multiple genes in disease pathogenesis, identification of therapeutic leads has been difficult and the majority of currently available therapies provide only marginal benefit. To address this issue, our goal was to genomically characterize individual PAC patients to understand the range of aberrations that are occurring in each tumor. Because our understanding of PAC tumorigenesis is limited, evaluation of separate cases may reveal aberrations, that are less common but may provide relevant information on the disease, or that may represent viable therapeutic targets for the patient. We used next generation sequencing to assess global somatic events across 3 PAC patients to characterize each patient and to identify potential targets. This study is the first to report whole genome sequencing (WGS) findings in paired tumor/normal samples collected from 3 separate PAC patients. We generated on average 132 billion mappable bases across all patients using WGS, and identified 142 somatic coding events including point mutations, insertion/deletions, and chromosomal copy number variants. We did not identify any significant somatic translocation events. We also performed RNA sequencing on 2 of these patients' tumors for which tumor RNA was available to evaluate expression changes that may be associated with somatic events, and generated over 100 million mapped reads for each patient. We further performed pathway analysis of all sequencing data to identify processes that may be the most heavily impacted from somatic and expression alterations. As expected, the KRAS signaling pathway was the most heavily impacted pathway (P<0.05), along with tumor-stroma interactions and tumor suppressive pathways. While sequencing of more patients is needed, the high resolution genomic and transcriptomic information we have acquired here provides valuable information on the molecular composition of PAC and helps to establish a foundation for improved therapeutic selection.
Collapse
Affiliation(s)
- Winnie S Liang
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Zhang SH, Qian YM, Liu AW, Cai J, Zhao XL, Wei JJ, Zhu MH. Clinicopathologic significance and function of S-phase kinase-associated protein 2 overexpression in hepatocellular carcinoma. Hum Pathol 2012; 43:1084-93. [DOI: 10.1016/j.humpath.2011.08.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2011] [Revised: 08/27/2011] [Accepted: 08/31/2011] [Indexed: 12/14/2022]
|
23
|
High expression of trimethylated histone H3 lysine 4 is associated with poor prognosis in hepatocellular carcinoma. Hum Pathol 2012; 43:1425-35. [PMID: 22406368 DOI: 10.1016/j.humpath.2011.11.003] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Revised: 10/30/2011] [Accepted: 11/02/2011] [Indexed: 12/21/2022]
Abstract
Tumor-associated epigenetic alterations including DNA methylation and histone modifications are important determinants in the initiation and progression of hepatocellular cancer (HCC) and represent promising biomarkers and therapeutic targets. Locus-specific trimethylation of histone H3 lysine 4 (H3K4me) is a well-known modification linked to the enhanced transcriptional expression of many genes activated in HCC. Our aim was to assess the cellular expression pattern of H3K4me3 in HCC and its association with clinicopathologic variables and outcome. Expression of H3K4me3 and the histone methyltransferase (HMT) SET and MYND domain-containing protein 3 (SMYD3) was studied by Western blotting and immunohistochemistry in cell lines and tumor tissue microarray from a well-characterized series of HCC patients (n = 168). The optimal cut-point of H3K4me3 expression for prognosis was determined by the X-tile program. The prognostic significance was evaluated using Kaplan-Meier survival estimates and log-rank tests. Tumor tissue microarray from another independent HCC patients cohort (n = 147) was used for validation studies. Expression of H3K4me3 and SMYD3 were enhanced in HCC cell lines. In tumor specimens, enhanced expression of H3K4me3 was correlated with reduced overall survival (P < .0001), especially in early-stage HCC patients (TNM I/II). Furthermore, both univariate and multivariate analyses revealed that H3K4me3 level was a significant and independent predictor of poor survival (hazard ratio, 3.592; 95% confidence interval, 2.302-5.605). In addition, H3K4m3 expression was positively correlated with SMYD3 expression in both testing and validation cohorts (P < .0001). In conclusion, H3K4me3 level defines unrecognized subsets of HCC patients with distinct epigenetic phenotype and clinical outcome and can thus be a novel predictor for poor prognosis of HCC patients, especially at TNM I/II stage.
Collapse
|
24
|
Stevenson RP, Veltman D, Machesky LM. Actin-bundling proteins in cancer progression at a glance. J Cell Sci 2012; 125:1073-9. [PMID: 22492983 DOI: 10.1242/jcs.093799] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- Richard P Stevenson
- The Beatson Institute for Cancer Research, Garscube Estate, Switchback Rd, Bearsden, Glasgow G61 1BD, UK
| | | | | |
Collapse
|