1
|
Garone ME, Chase SE, Zhang C, Krendel M. Myosin 1e deficiency affects migration of 4T1 breast cancer cells. Cytoskeleton (Hoboken) 2024; 81:723-736. [PMID: 38140937 PMCID: PMC11193843 DOI: 10.1002/cm.21819] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 11/03/2023] [Accepted: 12/01/2023] [Indexed: 12/24/2023]
Abstract
Metastasis of breast cancer cells to distant tissue sites is responsible for the majority of deaths associated with breast cancer. Previously we have examined the role of class I myosin motor protein, myosin 1e (myo1e), in cancer metastasis using the Mouse Mammary Tumor Virus-Polyoma Middle T Antigen (MMTV-PyMT) mouse model. Mice deficient in myo1e formed tumors with a more differentiated phenotype relative to the wild-type mice and formed no detectable lung metastases. In the current study, we investigated how the absence of myo1e affects cell migration and invasion in vitro, using the highly invasive and migratory breast cancer cell line, 4T1. 4T1 cells deficient in myo1e exhibited an altered morphology and slower rates of migration in the wound-healing and transwell migration assays compared to the WT 4T1 cells. While integrin trafficking and Golgi reorientation did not appear to be altered upon myo1e loss, we observed lower rates of focal adhesion disassembly in myo1e-deficient cells, which could help explain the cell migration defect.
Collapse
Affiliation(s)
- Michael E. Garone
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210
| | - Sharon E. Chase
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210
| | - Chunling Zhang
- Department of Neuroscience and Physiology, State University of New York Upstate Medical University, Syracuse, NY 13210
| | - Mira Krendel
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210
| |
Collapse
|
2
|
Lv HB, Wu QY, Zhang YJ, Quan SW, Ma N, Dai YQ, Sun Y. Study on the expression and prognostic relationship of MYL6B in liver cancer based on bioinformatics. World J Clin Oncol 2024; 15:1188-1197. [PMID: 39351463 PMCID: PMC11438851 DOI: 10.5306/wjco.v15.i9.1188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/21/2024] [Accepted: 08/02/2024] [Indexed: 08/29/2024] Open
Abstract
BACKGROUND Primary liver cancer is a prevalent and deadly cancer type. Despite treatment advances, prognosis remains poor, with high recurrence rates. Early detection is crucial but challenging due to the disease's insidious nature. Myosin proteins play significant roles in cancer development, influencing cell migration, invasion, and tumor suppression. MYL6B, a myosin light chain, is involved in various cellular processes and has been associated with poor prognosis in colorectal adenocarcinoma and potential as a biomarker in breast cancer. AIM To investigate the expression of MYL6B in liver hepatocellular carcinoma (LIHC) and its impact on prognosis and potential mechanisms of action using bioinformatics methods. METHODS The expression of MYL6B in pan-cancer and normal tissues was analyzed using the gene expression profiling interactive analysis 2 and tumor immune estimation resource databases. The expression level of MYL6B in LIHC tissues and its relationship with prognosis were analyzed, immunohistochemical analysis of MYL6B and its effect on immune cell infiltration, and the protein network were further studied. RESULTS MYL6B was highly expressed in diffuse large b-cell lymphoma, LIHC, pancreatic adenocarcinoma, skin cutaneous melanoma, thymoma, uterine corpus endometrial carcinoma, uterine carcinosarcoma, and lowly expressed in kidney chromophobe, acute myeloid leukemia, testicular germ cell tumors. The expression level of MYL6B was significantly different between cancer and normal tissues. It had a significant impact on both overall survival and disease-free survival. MYL6B is highly expressed in hepatocellular carcinoma and its expression level increases with cancer progression. High MYL6B expression is associated with poor prognosis in terms of overall survival and recurrence-free survival. The immunohistochemical level of MYL6B is high in hepatocellular carcinoma tissues, and MYL6B has a high level of immune infiltration inflammation. In protein network analysis, MYL6B is correlated with MYL2, MYL6, MYL9, MYLK4, MYLK2, MYL12A, MYL12B, MYH11, MYH9 and MYH10. CONCLUSION The expression level of MYL6B in LIHC was significantly higher than in normal liver tissues, and it was correlated with the degree of differentiation survival rate, and immune infiltration. MYL6B is a potential target for LIHC treatment.
Collapse
Affiliation(s)
- Hai-Bing Lv
- Department of General Surgery, Beidahuang Group General Hospital, Harbin 150000, Heilongjiang Province, China
| | - Qing-Yun Wu
- Department of General Surgery, Xianning Central Hospital, Xianning 437000, Hubei Province, China
| | - Yu-Jiao Zhang
- Department of Medical oncology, Beidahuang Group General Hospital, Harbin 150000, Heilongjiang Province, China
| | - Sheng-Wei Quan
- Department of General Surgery, Beidahuang Group General Hospital, Harbin 150000, Heilongjiang Province, China
| | - Ning Ma
- Department of General Surgery, Daqing Oilfield General Hospital, Daqing 163000, Heilongjiang Province, China
| | - Yu-Qing Dai
- College of Clinical Medicine, Bengbu Medical University, Bengbu 233000, Anhui Province, China
| | - Yan Sun
- Department of General Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin 150000, Heilongjiang Province, China
| |
Collapse
|
3
|
Xu S, Chen X, Ying H, Chen J, Ye M, Lin Z, Zhang X, Shen T, Li Z, Zheng Y, Zhang D, Ke Y, Chen Z, Lu Z. Multi‑omics identification of a signature based on malignant cell-associated ligand-receptor genes for lung adenocarcinoma. BMC Cancer 2024; 24:1138. [PMID: 39267056 PMCID: PMC11395699 DOI: 10.1186/s12885-024-12911-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 09/06/2024] [Indexed: 09/14/2024] Open
Abstract
PURPOSE Lung adenocarcinoma (LUAD) significantly contributes to cancer-related mortality worldwide. The heterogeneity of the tumor immune microenvironment in LUAD results in varied prognoses and responses to immunotherapy among patients. Consequently, a clinical stratification algorithm is necessary and inevitable to effectively differentiate molecular features and tumor microenvironments, facilitating personalized treatment approaches. METHODS We constructed a comprehensive single-cell transcriptional atlas using single-cell RNA sequencing data to reveal the cellular diversity of malignant epithelial cells of LUAD and identified a novel signature through a computational framework coupled with 10 machine learning algorithms. Our study further investigates the immunological characteristics and therapeutic responses associated with this prognostic signature and validates the predictive efficacy of the model across multiple independent cohorts. RESULTS We developed a six-gene prognostic model (MYO1E, FEN1, NMI, ZNF506, ALDOA, and MLLT6) using the TCGA-LUAD dataset, categorizing patients into high- and low-risk groups. This model demonstrates robust performance in predicting survival across various LUAD cohorts. We observed distinct molecular patterns and biological processes in different risk groups. Additionally, analysis of two immunotherapy cohorts (N = 317) showed that patients with a high-risk signature responded more favorably to immunotherapy compared to those in the low-risk group. Experimental validation further confirmed that MYO1E enhances the proliferation and migration of LUAD cells. CONCLUSION We have identified malignant cell-associated ligand-receptor subtypes in LUAD cells and developed a robust prognostic signature by thoroughly analyzing genomic, transcriptomic, and immunologic data. This study presents a novel method to assess the prognosis of patients with LUAD and provides insights into developing more effective immunotherapies.
Collapse
Affiliation(s)
- Shengshan Xu
- Department of Thoracic Surgery, Jiangmen Central Hospital, Jiangmen, Guangdong, China.
| | - Xiguang Chen
- Department of Medical Oncology, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Haoxuan Ying
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jiarong Chen
- Department of Oncology, Jiangmen Central Hospital, Jiangmen, Guangdong, China
| | - Min Ye
- Department of Thoracic Surgery, Jiangmen Central Hospital, Jiangmen, Guangdong, China
| | - Zhichao Lin
- Department of Thoracic Surgery, Jiangmen Central Hospital, Jiangmen, Guangdong, China
| | - Xin Zhang
- Clinical Experimental Center, Jiangmen Key Laboratory of Clinical Biobanks and Translational Research, Jiangmen Central Hospital, Jiangmen, Guangdong, China
| | - Tao Shen
- Department of Thoracic Surgery, Jiangmen Central Hospital, Jiangmen, Guangdong, China
| | - Zumei Li
- Department of Thoracic Surgery, Jiangmen Central Hospital, Jiangmen, Guangdong, China
| | - Youbin Zheng
- Department of Radiology, Jiangmen Wuyi Hospital of Traditional Chinese Medicine, Jiangmen, Guangdong, China
| | - Dongxi Zhang
- Department of Thoracic Surgery, Jiangmen Central Hospital, Jiangmen, Guangdong, China
| | - Yongwen Ke
- Department of Thoracic Surgery, Jiangmen Central Hospital, Jiangmen, Guangdong, China
| | - Zhuowen Chen
- Department of Thoracic Surgery, Jiangmen Central Hospital, Jiangmen, Guangdong, China
| | - Zhuming Lu
- Department of Thoracic Surgery, Jiangmen Central Hospital, Jiangmen, Guangdong, China.
| |
Collapse
|
4
|
Khair L, Hayes K, Tutto A, Samant A, Ferreira L, Nguyen TT, Brehm M, Messina LM. Physical activity regulates the immune response to breast cancer by a hematopoietic stem cell-autonomous mechanism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.30.560299. [PMID: 37873380 PMCID: PMC10592839 DOI: 10.1101/2023.09.30.560299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Physical activity is a modifiable lifestyle factor that is associated with a decreased risk for the development of breast cancer. While the exact mechanisms for the reduction in cancer risk due to physical activity are largely unknown, it is postulated that the biological reduction in cancer risk is driven by improvements in inflammation and immune function with exercise. Hematopoietic stem cells (HSCs) are the progenitor for all of the cells of the immune system and are involved in cancer immunosurveillance through differentiation into cytotoxic cell population. In this study, we investigate the role of physical activity (PA) in a spontaneously occurring model of breast cancer over time, with a focus on tumor incidence, circulating and tumor-infiltrating immune cells as well gene expression profiles of tumors and hematopoietic stem cells. Furthermore, we show that, in addition to a direct effect of PA on the immune cells of tumor-bearing mice, PA reduces the oxidative stress in HSCs of wildtype and tumor-bearing mice, and by doing so, alters the differentiation of the HSCs towards T cells in order to enhance cancer immunosurveillance.
Collapse
Affiliation(s)
- Lyne Khair
- Department of Surgery, Division of Vascular Surgery, UMass Memorial Medical Center
- Diabetes Center of Excellence, UMass Chan Medical School
| | - Katherine Hayes
- Department of Surgery, Division of Vascular Surgery, UMass Memorial Medical Center
| | - Amanda Tutto
- Department of Surgery, Division of Vascular Surgery, UMass Memorial Medical Center
| | - Amruta Samant
- Department of Surgery, Division of Vascular Surgery, UMass Memorial Medical Center
| | | | - Tammy T. Nguyen
- Department of Surgery, Division of Vascular Surgery, UMass Memorial Medical Center
- Diabetes Center of Excellence, UMass Chan Medical School
| | - Michael Brehm
- Diabetes Center of Excellence, UMass Chan Medical School
- Program in Molecular Medicine, UMass Chan Medical School
| | - Louis M. Messina
- Department of Surgery, Division of Vascular Surgery, UMass Memorial Medical Center
- Diabetes Center of Excellence, UMass Chan Medical School
| |
Collapse
|
5
|
Lei J, Pan Y, Gao R, He B, Wang Z, Lei X, Zhang Z, Yang N, Yan M. Rutaecarpine induces the differentiation of triple-negative breast cancer cells through inhibiting fumarate hydratase. J Transl Med 2023; 21:553. [PMID: 37592347 PMCID: PMC10436383 DOI: 10.1186/s12967-023-04396-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 07/29/2023] [Indexed: 08/19/2023] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is one of the most aggressive human cancers and has poor prognosis. Approximately 80% of TNBC cases belong to the molecular basal-like subtype, which can be exploited therapeutically by inducing differentiation. However, the strategies for inducing the differentiation of TNBC remain underexplored. METHODS A three-dimensional (3D) morphological screening model based on a natural compound library was used to identify possible candidate compounds that can induce TNBC cell differentiation. The efficacy of rutaecarpine was verified using assays: RT-qPCR, RNA-seq, flow cytometry, immunofluorescence, SCENITH and label-free LC-MS/MS. The direct targets of rutaecarpine were identified through drug affinity responsive target stability (DARTS) assay. A xenograft mice model was also constructed to confirm the effect of rutaecarpine in vivo. RESULTS We identified that rutaecarpine, an indolopyridoquinazolinone, induces luminal differentiation of basal TNBC cells in both 3D spheroids and in vivo mice models. Mechanistically, rutaecarpine treatment leads to global metabolic stress and elevated ROS in 3D cultured TNBC cells. Moreover, NAC, a scavenger of ROS, impedes rutaecarpine-induced differentiation of TNBC cells in 3D culture. Finally, we identified fumarate hydratase (FH) as the direct interacting target of rutaecarpine. The inhibition of FH and the knockdown of FH consistently induced the differentiation of TNBC cells in 3D culture. CONCLUSIONS Our results provide a platform for differentiation therapy drug discovery using 3D culture models and identify rutaecarpine as a potential compound for TNBC treatment.
Collapse
Affiliation(s)
- Jie Lei
- State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Yujia Pan
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, 116023, China
| | - Rui Gao
- Department of Medical Oncology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 510275, China
| | - Bin He
- State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Zifeng Wang
- State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Xinxing Lei
- State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Zijian Zhang
- State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Na Yang
- Department of Laboratory Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, 510180, China.
| | - Min Yan
- State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University, Guangzhou, 510060, China.
| |
Collapse
|
6
|
Liu S, Liu P, Fei X, Zhu C, Hou J, Wang X, Pan Y. Analysis and validation of the potential of the MYO1E gene in pancreatic adenocarcinoma based on a bioinformatics approach. Oncol Lett 2023; 26:285. [PMID: 37274465 PMCID: PMC10236097 DOI: 10.3892/ol.2023.13871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 03/22/2023] [Indexed: 06/06/2023] Open
Abstract
Pancreatic adenocarcinoma (PAAD) is a common digestive cancer, and its prognosis is poor. Myosin 1E (MYO1E) is a class I myosin family member whose expression and function have not been reported in PAAD. In the present study, bioinformatics analysis was used to explore the expression levels of MYO1E in PAAD and its prognostic value, and the immunological role of MYO1E in PAAD was analyzed. The study revealed that a variety of malignancies have substantially increased MYO1E expression. Further investigation demonstrated that PAAD tissues exhibited greater levels of MYO1E mRNA and protein expression than normal tissues. High MYO1E expression is associated with poor prognosis in patients with PAAD. MYO1E expression was also associated with pathological stage in patients with PAAD. Functional enrichment analysis demonstrated that MYO1E was linked to multiple tumor-related mechanisms in PAAD. The pancreatic adenocarcinoma tumor microenvironment (TME) was analyzed and it was revealed that MYO1E expression was positively associated with tumor immune cell infiltration. In addition, MYO1E was closely associated with some tumor chemokines/receptors and immune checkpoints. In vitro experiments revealed that the suppression of MYO1E expression could inhibit pancreatic adenocarcinoma cell proliferation, invasion and migration. Through preliminary analysis, the present study evaluated the potential function of MYO1E in PAAD and its function in TME, and MYO1E may become a potential biomarker for PAAD.
Collapse
Affiliation(s)
- Songbai Liu
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou 550000, P.R. China
| | - Peng Liu
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou 550000, P.R. China
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550000, P.R. China
| | - Xiaobin Fei
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou 550000, P.R. China
| | - Changhao Zhu
- Department of Hepatobiliary Surgery, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, Guizhou 550000, P.R. China
| | - Junyi Hou
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou 550000, P.R. China
| | - Xing Wang
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou 550000, P.R. China
- Department of Hepatobiliary Surgery, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, Guizhou 550000, P.R. China
| | - Yaozhen Pan
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou 550000, P.R. China
- Department of Hepatobiliary Surgery, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, Guizhou 550000, P.R. China
| |
Collapse
|
7
|
Jusue-Torres I, Tiv R, Ricarte-Filho JC, Mallisetty A, Contreras-Vargas L, Godoy-Calderon MJ, Khaddour K, Kennedy K, Valyi-Nagy K, David O, Menchaca M, Kottorou A, Koutras A, Dimitrakopoulos F, Abdelhady KM, Massad M, Rubinstein I, Feldman L, Stewart J, Shimamura T, Danilova L, Hulbert A. Myo1e overexpression in lung adenocarcinoma is associated with increased risk of mortality. Sci Rep 2023; 13:4107. [PMID: 36914720 PMCID: PMC10011530 DOI: 10.1038/s41598-023-30765-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 02/28/2023] [Indexed: 03/16/2023] Open
Abstract
This study aims to perform a comprehensive genomic analysis to assess the influence of overexpression of MYO1E in non-small cell lung carcinoma (NSCLC) and whether there are differences in survival and mortality risk in NSCLC patients depending on both DNA methylation and RNA expression of MYO1E. The DNA methylation probe cg13887966 was inversely correlated with MYO1E RNA expression in both LUAD and LUSC subpopulations showing that lower MYO1E RNA expression was associated with higher MYO1E DNA methylation. Late stages of lung cancer showed significantly lower MYO1E DNA methylation and significantly higher MYO1E RNA expression for LUAD but not for LUSC. Low DNA methylation as well as high RNA expression of MYO1E are associated with a shorter median survival time and an increased risk of mortality for LUAD, but not for LUSC. This study suggests that changes in MYO1E methylation and expression in LUAD patients may have an essential role in lung cancer's pathogenesis. It shows the utility of MYO1E DNA methylation and RNA expression in predicting survival for LUAD patients. Also, given the low normal expression of MYO1E in blood cells MYO1E DNA methylation has the potential to be used as circulating tumor marker in liquid biopsies.
Collapse
Affiliation(s)
| | - Richies Tiv
- Department of Surgery, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | | | - Apurva Mallisetty
- Department of Surgery, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | - Leglys Contreras-Vargas
- Department of Surgery, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | | | - Karam Khaddour
- Division of Hematology Oncology, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | - Kathleen Kennedy
- Division of Hematology Oncology, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | - Klara Valyi-Nagy
- Department of Pathology, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | - Odile David
- Department of Pathology, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | - Martha Menchaca
- Department of Radiology, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | - Anastasia Kottorou
- Molecular Oncology Laboratory, Division of Oncology, Medical School, University of Patras, Patras, Greece
| | - Angelos Koutras
- Molecular Oncology Laboratory, Division of Oncology, Medical School, University of Patras, Patras, Greece
| | - Foteinos Dimitrakopoulos
- Molecular Oncology Laboratory, Division of Oncology, Medical School, University of Patras, Patras, Greece
| | | | - Malek Massad
- Division of Cardiothoracic Surgery, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | - Israel Rubinstein
- Medical and Research Services, Jesse Brown VA Medical Center, Chicago, IL, USA
- Division of Pulmonary, Critical Care, Sleep, and Allergy Medicine, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | - Lawrence Feldman
- Division of Hematology Oncology, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
- Medical and Research Services, Jesse Brown VA Medical Center, Chicago, IL, USA
| | - John Stewart
- Department of Surgery, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
- Section of Surgical Oncology, Department of Surgery, Louisiana State University, New Orleans, LA, USA
| | - Takeshi Shimamura
- Cancer Center, University of Illinois at Chicago, Chicago, IL, USA
- Division of Cardiothoracic Surgery, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | - Ludmila Danilova
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Laboratory of Systems Biology and Computational Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow, Russia
| | - Alicia Hulbert
- Cancer Center, University of Illinois at Chicago, Chicago, IL, USA.
- Medical and Research Services, Jesse Brown VA Medical Center, Chicago, IL, USA.
- Department of Surgery, University of Illinois College of Medicine, 909 South Wolcott Ave, COMRB Suite 5140, Chicago, IL, 60612, USA.
| |
Collapse
|
8
|
Johnson CA, Behbehani R, Buss F. Unconventional Myosins from Caenorhabditis elegans as a Probe to Study Human Orthologues. Biomolecules 2022; 12:biom12121889. [PMID: 36551317 PMCID: PMC9775386 DOI: 10.3390/biom12121889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/09/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Unconventional myosins are a superfamily of actin-based motor proteins that perform a number of roles in fundamental cellular processes, including (but not limited to) intracellular trafficking, cell motility, endocytosis, exocytosis and cytokinesis. 40 myosins genes have been identified in humans, which belong to different 12 classes based on their domain structure and organisation. These genes are widely expressed in different tissues, and mutations leading to loss of function are associated with a wide variety of pathologies while over-expression often results in cancer. Caenorhabditis elegans (C. elegans) is a small, free-living, non-parasitic nematode. ~38% of the genome of C. elegans has predicted orthologues in the human genome, making it a valuable tool to study the function of human counterparts and human diseases. To date, 8 unconventional myosin genes have been identified in the nematode, from 6 different classes with high homology to human paralogues. The hum-1 and hum-5 (heavy chain of an unconventional myosin) genes encode myosin of class I, hum-2 of class V, hum-3 and hum-8 of class VI, hum-6 of class VII and hum-7 of class IX. The hum-4 gene encodes a high molecular mass myosin (307 kDa) that is one of the most highly divergent myosins and is a member of class XII. Mutations in many of the human orthologues are lethal, indicating their essential properties. However, a functional characterisation for many of these genes in C. elegans has not yet been performed. This article reviews the current knowledge of unconventional myosin genes in C. elegans and explores the potential use of the nematode to study the function and regulation of myosin motors to provide valuable insights into their role in diseases.
Collapse
|
9
|
Diaz-Valencia JD, Estrada-Abreo LA, Rodríguez-Cruz L, Salgado-Aguayo AR, Patiño-López G. Class I Myosins, molecular motors involved in cell migration and cancer. Cell Adh Migr 2022; 16:1-12. [PMID: 34974807 PMCID: PMC8741282 DOI: 10.1080/19336918.2021.2020705] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 12/02/2021] [Accepted: 12/16/2021] [Indexed: 01/13/2023] Open
Abstract
Class I Myosins are a subfamily of motor proteins with ATPase activity and a characteristic structure conserved in all myosins: A N-Terminal Motor Domain, a central Neck and a C terminal Tail domain. Humans have eight genes for these myosins. Class I Myosins have different functions: regulate membrane tension, participate in endocytosis, exocytosis, intracellular trafficking and cell migration. Cell migration is influenced by many cellular components including motor proteins, like myosins. Recently has been reported that changes in myosin expression have an impact on the migration of cancer cells, the formation of infiltrates and metastasis. We propose that class I myosins might be potential markers for future diagnostic, prognostic or even as therapeutic targets in leukemia and other cancers.Abbreviations: Myo1g: Myosin 1g; ALL: Acute Lymphoblastic Leukemia, TH1: Tail Homology 1; TH2: Tail Homology 2; TH3: Tail Homology 3.
Collapse
Affiliation(s)
- Juan D. Diaz-Valencia
- Immunology and Proteomics Laboratory, Children’s Hospital of Mexico, Mexico City, Mexico
| | - Laura A. Estrada-Abreo
- Immunology and Proteomics Laboratory, Children’s Hospital of Mexico, Mexico City, Mexico
- Cell Biology and Flow Cytometry Laboratory, Metropolitan Autonomous University, México City, Mexico
| | - Leonor Rodríguez-Cruz
- Cell Biology and Flow Cytometry Laboratory, Metropolitan Autonomous University, México City, Mexico
| | - Alfonso R. Salgado-Aguayo
- Rheumatic Diseases Laboratory, National Institute of Respiratory Diseases “Ismael Cosío Villegas”, Mexico City, Mexico
| | - Genaro Patiño-López
- Immunology and Proteomics Laboratory, Children’s Hospital of Mexico, Mexico City, Mexico
| |
Collapse
|
10
|
Jayathirtha M, Neagu AN, Whitham D, Alwine S, Darie CC. Investigation of the effects of downregulation of jumping translocation breakpoint (JTB) protein expression in MCF7 cells for potential use as a biomarker in breast cancer. Am J Cancer Res 2022; 12:4373-4398. [PMID: 36225631 PMCID: PMC9548009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 06/18/2022] [Indexed: 06/16/2023] Open
Abstract
MCF7 is a commonly used luminal type A non-invasive/poor-invasive human breast cancer cell line that does not usually migrate or invade compared with MDA-MB-231 highly metastatic cells, which emphasize an invasive and migratory behavior. Under special conditions, MCF7 cells might acquire invasive features. The aberration in expression and biological functions of the jumping translocation breackpoint (JTB) protein is associated with malignant transformation of cells, based on mitochondrial dysfunction, inhibition of tumor suppressive function of TGF-β, and involvement in cancer cell cycle. To investigate new putative functions of JTB by cellular proteomics, we analyzed the biological processes and pathways that are associated with the JTB protein downregulation. The results demonstrated that MCF7 cell line developed a more "aggressive" phenotype and behavior. Most of the proteins that were overexpressed in this experiment promoted the actin cytoskeleton reorganization that is involved in growth and metastatic dissemination of cancer cells. Some of these proteins are involved in the epithelial-mesenchymal transition (EMT) process (ACTBL2, TUBA4A, MYH14, CSPG5, PKM, UGDH, HSP90AA2, and MIF), in correlation with the energy metabolism reprogramming (PKM, UGDH), stress-response (HSP10, HSP70A1A, HSP90AA2), and immune and inflammatory response (MIF and ERp57-TAPBP). Almost all upregulated proteins in JTB downregulated condition promote viability, motility, proliferation, invasion, survival into a hostile microenvironment, metabolic reprogramming, and escaping of tumor cells from host immune control, leading to a more invasive phenotype for MCF7 cell line. Due to their downregulated condition, four proteins, such as CREBZF, KMT2B, SELENOS and CACNA1I are also involved in maintenance of the invasive phenotype of cancer cells, promoting cell proliferation, migration, invasion and tumorigenesis. Other downregulated proteins, such as MAZ, PLEKHG2, ENO1, TPI2, TOR2A, and CNNM1, may promote suppression of cancer cell growth, invasion, EMT, tumorigenic abilities, interacting with glucose and lipid metabolism, disrupting nuclear envelope stability, or suppressing apoptosis and developing anti-angiogenetic activities. Therefore, the main biological processes and pathways that may increase the tumorigenic potential of the MCF7 cells in JTB downregulated condition are related to the actin cytoskeleton organization, EMT, mitotic cell cycle, glycolysis and fatty acid metabolism, inflammatory response and macrophage activation, chemotaxis and migration, cellular response to stress condition (oxidative stress and hypoxia), transcription control, histone modification and ion transport.
Collapse
Affiliation(s)
- Madhuri Jayathirtha
- Biochemistry & Proteomics Group, Department of Chemistry and Biomolecular Science, Clarkson UniversityPotsdam, NY 13699-5810, USA
| | - Anca-Narcisa Neagu
- Laboratory of Animal Histology, Faculty of Biology, “Alexandru Ioan Cuza” University of IasiCarol I bvd. No. 22, Iasi 700505, Romania
| | - Danielle Whitham
- Biochemistry & Proteomics Group, Department of Chemistry and Biomolecular Science, Clarkson UniversityPotsdam, NY 13699-5810, USA
| | - Shelby Alwine
- Biochemistry & Proteomics Group, Department of Chemistry and Biomolecular Science, Clarkson UniversityPotsdam, NY 13699-5810, USA
| | - Costel C Darie
- Biochemistry & Proteomics Group, Department of Chemistry and Biomolecular Science, Clarkson UniversityPotsdam, NY 13699-5810, USA
| |
Collapse
|
11
|
Regua A, Papp C, Grageda A, Porter BA, Caza T, Bichindaritz I, Krendel M, Sivapiragasam A, Bratslavsky G, Kuznetsov VA, Kotula L. ABI1-based expression signature predicts breast cancer metastasis and survival. Mol Oncol 2022; 16:2632-2657. [PMID: 34967509 PMCID: PMC9297774 DOI: 10.1002/1878-0261.13175] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 12/29/2021] [Indexed: 11/05/2022] Open
Abstract
Despite the current standard of care, breast cancer remains one of the leading causes of mortality in women worldwide, thus emphasizing the need for better predictive and therapeutic targets. ABI1 is associated with poor survival and an aggressive breast cancer phenotype, although its role in tumorigenesis, metastasis, and the disease outcome remains to be elucidated. Here, we define the ABI1-based seven-gene prognostic signature that predicts survival of metastatic breast cancer patients; ABI1 is an essential component of the signature. Genetic disruption of Abi1 in primary breast cancer tumors of PyMT mice led to significant reduction of the number and size of lung metastases in a gene dose-dependent manner. The disruption of Abi1 resulted in deregulation of the WAVE complex at the mRNA and protein levels in mouse tumors. In conclusion, ABI1 is a prognostic metastatic biomarker in breast cancer. We demonstrate, for the first time, that lung metastasis is associated with an Abi1 gene dose and specific gene expression aberrations in primary breast cancer tumors. These results indicate that targeting ABI1 may provide a therapeutic advantage in breast cancer patients.
Collapse
Affiliation(s)
- Angelina Regua
- Department of UrologySUNY Upstate Medical UniversitySyracuseNYUSA
- Department of Biochemistry and Molecular BiologySUNY Upstate Medical UniversitySyracuseNYUSA
- Present address:
Department of Cancer BiologyWake Forest University School of MedicineWinston‐SalemNC27101USA
| | - Csaba Papp
- Department of UrologySUNY Upstate Medical UniversitySyracuseNYUSA
- Department of Biochemistry and Molecular BiologySUNY Upstate Medical UniversitySyracuseNYUSA
| | - Andre Grageda
- Department of UrologySUNY Upstate Medical UniversitySyracuseNYUSA
- Department of Biochemistry and Molecular BiologySUNY Upstate Medical UniversitySyracuseNYUSA
| | - Baylee A. Porter
- Department of UrologySUNY Upstate Medical UniversitySyracuseNYUSA
- Department of Biochemistry and Molecular BiologySUNY Upstate Medical UniversitySyracuseNYUSA
| | - Tiffany Caza
- Department of PathologySUNY Upstate Medical UniversitySyracuseNYUSA
| | | | - Mira Krendel
- Department of Cell and Developmental BiologySUNY Upstate Medical UniversitySyracuseNYUSA
| | | | - Gennady Bratslavsky
- Department of UrologySUNY Upstate Medical UniversitySyracuseNYUSA
- Department of Biochemistry and Molecular BiologySUNY Upstate Medical UniversitySyracuseNYUSA
| | - Vladimir A. Kuznetsov
- Department of UrologySUNY Upstate Medical UniversitySyracuseNYUSA
- Department of Biochemistry and Molecular BiologySUNY Upstate Medical UniversitySyracuseNYUSA
| | - Leszek Kotula
- Department of UrologySUNY Upstate Medical UniversitySyracuseNYUSA
- Department of Biochemistry and Molecular BiologySUNY Upstate Medical UniversitySyracuseNYUSA
| |
Collapse
|
12
|
Detection of Myosin 1g Overexpression in Pediatric Leukemia by Novel Monoclonal Antibodies. Int J Mol Sci 2022; 23:ijms23073912. [PMID: 35409272 PMCID: PMC8999415 DOI: 10.3390/ijms23073912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/23/2022] [Accepted: 03/01/2022] [Indexed: 01/27/2023] Open
Abstract
Myosin 1g (Myo1g) is a mechanoenzyme associated with actin filaments, expressed exclusively in hematopoietic cells, and involved in various cellular functions, including cell migration, adhesion, and membrane trafficking. Despite the importance of Myo1g in distinct functions, there is currently no monoclonal antibody (mAb) against Myo1g. mAbs are helpful tools for the detection of specific antigens in tumor cells and other tissues. The development of mAbs against targeted dysregulated molecules in cancer cells remains a crucial tool for aiding in the diagnosis and the treatment of patients. Using hybridoma technology, we generated a panel of hybridomas specific for Myo1g. ELISA, immunofluorescence, and Western blot assay results revealed the recognition of Myo1g by these novel monoclonal antibodies in normal and transformed T and B cells. Here, we report the development and application of new monoclonal antibodies against Myo1g for their potential use to detect its overexpression in acute lymphoblastic leukemia (ALL) patients.
Collapse
|
13
|
Estrada-Abreo LA, Rodríguez-Cruz L, Garfias-Gómez Y, Araujo-Cardenas JE, Antonio-Andrés G, Salgado-Aguayo AR, Orozco-Ruiz D, Torres-Nava JR, Díaz-Valencia JD, Huerta-Yépez S, Patiño-López G. High expression of Myosin 1g in pediatric acute lymphoblastic leukemia. Oncotarget 2021; 12:1937-1945. [PMID: 34548909 PMCID: PMC8448507 DOI: 10.18632/oncotarget.28055] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 08/13/2021] [Indexed: 12/29/2022] Open
Abstract
Acute Lymphoblastic Leukemia (ALL) is the most frequent cancer in pediatric population. Although the treatment has improved and almost 85% of the children are cured about 20% suffer relapse, therefore finding molecules that participate in the pathogenesis of the disease for the identification of relapse and patients at risk is an urgent unmet need. Class I myosins are molecular motors involved in membrane tension, endocytosis, phagocytosis and cell migration and recently they have been shown important for development and aggressiveness of diverse cancer types, however Myo1g an hematopoietic specific myosin has not been studied in cancer so far. We evaluated the expression of Myo1g by qRT-PCR, Immunocytochemistry and Immunofluorescence in a cohort of 133 ALL patients and correlated the expression at diagnosis and after treatment with clinical features and treatment outcomes. We found high expression levels of Myo1g in Peripheral Blood Mononuclear Cells (PBMCs) from patients with ALL at diagnosis and those levels decreased after complete remission; furthermore, we found an increase in Myo1g expression on patients with 9:22 translocation and those who relapse. This study show that Myo1g is over expressed in ALL and that may participate in the pathogenesis of the disease specially in high-risk patients.
Collapse
Affiliation(s)
- Laura A Estrada-Abreo
- Immunology and Proteomics Laboratory, Hospital Infantil de México Federico Gómez, México City, México.,Cell Biology and Flow Cytometry Laboratory, Department of Health Sciences, Universidad Autónoma Metropolitana, Iztapalapa, México
| | - Leonor Rodríguez-Cruz
- Cell Biology and Flow Cytometry Laboratory, Department of Health Sciences, Universidad Autónoma Metropolitana, Iztapalapa, México
| | - Yanelly Garfias-Gómez
- Immunology and Proteomics Laboratory, Hospital Infantil de México Federico Gómez, México City, México
| | - Janeth E Araujo-Cardenas
- Immunology and Proteomics Laboratory, Hospital Infantil de México Federico Gómez, México City, México
| | - Gabriela Antonio-Andrés
- Oncologic Diseases Research Unit, Hospital Infantil de México Federico Gómez, México City, México
| | - Alfonso R Salgado-Aguayo
- Laboratory of Research on Rheumatic Diseases, National Institute of Respiratory Diseases, Ismael Cosío Villegas, México City, México
| | | | | | - Juan D Díaz-Valencia
- Immunology and Proteomics Laboratory, Hospital Infantil de México Federico Gómez, México City, México
| | - Sara Huerta-Yépez
- Oncologic Diseases Research Unit, Hospital Infantil de México Federico Gómez, México City, México
| | - Genaro Patiño-López
- Immunology and Proteomics Laboratory, Hospital Infantil de México Federico Gómez, México City, México
| |
Collapse
|
14
|
Overexpression of Human Syndecan-1 Protects against the Diethylnitrosamine-Induced Hepatocarcinogenesis in Mice. Cancers (Basel) 2021; 13:cancers13071548. [PMID: 33801718 PMCID: PMC8037268 DOI: 10.3390/cancers13071548] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/18/2021] [Accepted: 03/24/2021] [Indexed: 12/20/2022] Open
Abstract
Simple Summary Syndecan-1 is a Janus-faced proteoglycan: depending on the type of cancer, it can promote or inhibit the development of tumors. Our previous in vitro experiments revealed that transfection of human syndecan-1 (hSDC1) into hepatoma cells, initiating hepatocyte-like differentiation. To further confirm the antitumor action of hSDC1 in the context of liver carcinogenesis, mice transgenic for albumin promoter-driven hSDC1 were created with exclusive expression of hSDC1 in the liver. Indeed, hSDC1 interfered with the development of liver cancer in diethylnitrosamine (DEN)-induced hepatocarcinogenesis experiments. The mechanism was found to be related to lipid metabolism that plays an important role in the induction of nonalcoholic liver cirrhosis. Nonalcoholic fatty liver disease is known to promote the development of cancer; therefore, the oncoprotective effect of hSDC1 may be mediated by a beneficial modulation of lipid metabolism. Abstract Although syndecan-1 (SDC1) is known to be dysregulated in various cancer types, its implication in tumorigenesis is poorly understood. Its effect may be detrimental or protective depending on the type of cancer. Our previous data suggest that SDC1 is protective against hepatocarcinogenesis. To further verify this notion, human SDC1 transgenic (hSDC1+/+) mice were generated that expressed hSDC1 specifically in the liver under the control of the albumin promoter. Hepatocarcinogenesis was induced by a single dose of diethylnitrosamine (DEN) at an age of 15 days after birth, which resulted in tumors without cirrhosis in wild-type and hSDC1+/+ mice. At the experimental endpoint, livers were examined macroscopically and histologically, as well as by immunohistochemistry, Western blot, receptor tyrosine kinase array, phosphoprotein array, and proteomic analysis. Liver-specific overexpression of hSDC1 resulted in an approximately six month delay in tumor formation via the promotion of SDC1 shedding, downregulation of lipid metabolism, inhibition of the mTOR and the β-catenin pathways, and activation of the Foxo1 and p53 transcription factors that lead to the upregulation of the cell cycle inhibitors p21 and p27. Furthermore, both of them are implicated in the regulation of intermediary metabolism. Proteomic analysis showed enhanced lipid metabolism, activation of motor proteins, and loss of mitochondrial electron transport proteins as promoters of cancer in wild-type tumors, inhibited in the hSDC1+/+ livers. These complex mechanisms mimic the characteristics of nonalcoholic steatohepatitis (NASH) induced human liver cancer successfully delayed by syndecan-1.
Collapse
|
15
|
Xu F, Zhang L, Xu Y, Song D, He W, Ji X, Shao J. Hypermethylation of SCAND3 and Myo1g Gene Are Potential Diagnostic Biomarkers for Hepatocellular Carcinoma. Cancers (Basel) 2020; 12:E2332. [PMID: 32824823 PMCID: PMC7465898 DOI: 10.3390/cancers12082332] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/13/2020] [Accepted: 08/14/2020] [Indexed: 12/15/2022] Open
Abstract
Presently, there is a lack of effective blood-based biomarkers facilitating the diagnosis of hepatocellular carcinoma (HCC). Thus, we aimed to investigate novel methylation markers for HCC diagnosis, and explore relationships between biomarker methylation and clinicopathology of HCC. The methylation status of the SCAN domain containing three (SCAND3) and myosin 1g (Myo1g) genes in HCC cell lines and tissues were detected by digital droplet PCR. The serum SCAND3 and Myo1g methylation levels were analyzed in HCC-afflicted patients and unafflicted controls. The results indicated SCAND3 and Myo1g methylation were abnormally high in the HCC cell lines and tissues. The values of serum SCAND3, Myo1g, and SCAND3 + Myo1g methylation with respect to facilitating the detection, and early detection of HCC were better than for alpha-fetoprotein (AFP) alone. Furthermore, when we combined SCAND3 + Myo1g with AFP, a high sensitivity and specificity resulted. Notably, in the AFP-negative HCC group, the methylation of SCAND3 and Myo1g also showed an excellent diagnostic performance. Besides this, a high serum SCAND3 methylation level was an independent risk factor for predicting portal vein tumor thrombus (PVTT) in HCC patients (OR = 4.746, p = 0.013). Finally, SCAND3 and Myo1g enhanced the HCC diagnostics as noninvasive serum methylation biomarkers, and the SCAND3 methylation status effectively indicated HCC accompanied by PVTT.
Collapse
Affiliation(s)
- Fei Xu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China; (F.X.); (L.Z.); (Y.X.); (D.S.); (W.H.); (X.J.)
- Department of Molecular Diagnostics, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Lulu Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China; (F.X.); (L.Z.); (Y.X.); (D.S.); (W.H.); (X.J.)
- Department of Molecular Diagnostics, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Yuxia Xu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China; (F.X.); (L.Z.); (Y.X.); (D.S.); (W.H.); (X.J.)
- Department of Molecular Diagnostics, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Di Song
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China; (F.X.); (L.Z.); (Y.X.); (D.S.); (W.H.); (X.J.)
- Department of Molecular Diagnostics, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Wenting He
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China; (F.X.); (L.Z.); (Y.X.); (D.S.); (W.H.); (X.J.)
- Department of Molecular Diagnostics, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Xiaomeng Ji
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China; (F.X.); (L.Z.); (Y.X.); (D.S.); (W.H.); (X.J.)
- Department of Molecular Diagnostics, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Jianyong Shao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China; (F.X.); (L.Z.); (Y.X.); (D.S.); (W.H.); (X.J.)
- Department of Molecular Diagnostics, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| |
Collapse
|
16
|
Abstract
Myosins constitute a superfamily of actin-based molecular motor proteins that mediates a variety of cellular activities including muscle contraction, cell migration, intracellular transport, the formation of membrane projections, cell adhesion, and cell signaling. The 12 myosin classes that are expressed in humans share sequence similarities especially in the N-terminal motor domain; however, their enzymatic activities, regulation, ability to dimerize, binding partners, and cellular functions differ. It is becoming increasingly apparent that defects in myosins are associated with diseases including cardiomyopathies, colitis, glomerulosclerosis, neurological defects, cancer, blindness, and deafness. Here, we review the current state of knowledge regarding myosins and disease.
Collapse
|
17
|
Expanding cancer predisposition genes with ultra-rare cancer-exclusive human variations. Sci Rep 2020; 10:13462. [PMID: 32778766 PMCID: PMC7418036 DOI: 10.1038/s41598-020-70494-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 07/28/2020] [Indexed: 12/18/2022] Open
Abstract
It is estimated that up to 10% of cancer incidents are attributed to inherited genetic alterations. Despite extensive research, there are still gaps in our understanding of genetic predisposition to cancer. It was theorized that ultra-rare variants partially account for the missing heritable component. We harness the UK BioBank dataset of ~ 500,000 individuals, 14% of which were diagnosed with cancer, to detect ultra-rare, possibly high-penetrance cancer predisposition variants. We report on 115 cancer-exclusive ultra-rare variations and nominate 26 variants with additional independent evidence as cancer predisposition variants. We conclude that population cohorts are valuable source for expanding the collection of novel cancer predisposition genes.
Collapse
|
18
|
Navinés-Ferrer A, Martín M. Long-Tailed Unconventional Class I Myosins in Health and Disease. Int J Mol Sci 2020; 21:ijms21072555. [PMID: 32272642 PMCID: PMC7177449 DOI: 10.3390/ijms21072555] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/03/2020] [Accepted: 04/04/2020] [Indexed: 01/21/2023] Open
Abstract
Long-tailed unconventional class I myosin, Myosin 1E (MYO1E) and Myosin 1F (MYO1F) are motor proteins that use chemical energy from the hydrolysis of adenosine triphosphate (ATP) to produce mechanical work along the actin cytoskeleton. On the basis of their motor properties and structural features, myosins perform a variety of essential roles in physiological processes such as endocytosis, exocytosis, cell adhesion, and migration. The long tailed unconventional class I myosins are characterized by having a conserved motor head domain, which binds actin and hydrolyzes ATP, followed by a short neck with an isoleucine-glutamine (IQ) motif, which binds calmodulin and is sensitive to calcium, and a tail that contains a pleckstrin homology domain (PH), a tail homology 1 domain (TH1), wherein these domains allow membrane binding, a tail homology 2 domain (TH2), an ATP-insensitive actin-binding site domain, and a single Src homology 3 domain (SH3) susceptible to binding proline rich regions in other proteins. Therefore, these motor proteins are able to bind actin, plasma membrane, and other molecules (adaptor, kinases, membrane proteins) that contribute to their function, ranging from increasing membrane tension to molecular trafficking and cellular adhesion. MYO1E and MYO1F function in host self-defense, with a better defined role in innate immunity in cell migration and phagocytosis. Impairments of their function have been identified in patients suffering pathologies ranging from tumoral processes to kidney diseases. In this review, we summarize our current knowledge of specific features and functions of MYO1E and MYO1F in various tissues, as well as their involvement in disease.
Collapse
Affiliation(s)
- A. Navinés-Ferrer
- Biochemistry and Molecular Biology Unit, Biomedicine Department, Faculty of Medicine, University of Barcelona, 08036 Barcelona, Spain
- Laboratory of Clinic and Experimental Respiratory Immunoallergy, IDIBAPS, 08036 Barcelona, Spain
| | - M. Martín
- Biochemistry and Molecular Biology Unit, Biomedicine Department, Faculty of Medicine, University of Barcelona, 08036 Barcelona, Spain
- Laboratory of Clinic and Experimental Respiratory Immunoallergy, IDIBAPS, 08036 Barcelona, Spain
- ARADyAL research network, Carlos III Health Institute, 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-93-4024541; Fax: +34-93-4035882
| |
Collapse
|
19
|
Brzeska H, Gonzalez J, Korn ED, Titus MA. Basic-hydrophobic sites are localized in conserved positions inside and outside of PH domains and affect localization of Dictyostelium myosin 1s. Mol Biol Cell 2020; 31:101-117. [PMID: 31774725 PMCID: PMC6960411 DOI: 10.1091/mbc.e19-08-0475] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 11/14/2019] [Accepted: 11/22/2019] [Indexed: 11/21/2022] Open
Abstract
Myosin 1s have critical roles in linking membranes to the actin cytoskeleton via direct binding to acidic lipids. Lipid binding may occur through PIP3/PIP2-specific PH domains or nonspecific ionic interactions involving basic-hydrophobic (BH) sites but the mechanism of myosin 1s distinctive lipid targeting is poorly understood. Now we show that PH domains occur in all Dictyostelium myosin 1s and that the BH sites of Myo1A, B, C, D, and F are in conserved positions near the β3/β4 loops of their PH domains. In spite of these shared lipid-binding sites, we observe significant differences in myosin 1s highly dynamic localizations. All myosin 1s except Myo1A are present in macropinocytic structures but only Myo1B and Myo1C are enriched at the edges of macropinocytic cups and associate with the actin in actin waves. In contrast, Myo1D, E, and F are enclosed by the actin wave. Mutations of BH sites affect localization of all Dictyostelium myosin 1s. Notably, mutation of the BH site located within the PH domains of PIP3-specific Myo1D and Myo1F completely eradicates membrane binding. Thus, BH sites are important determinants of motor targeting and may have a similar role in the localization of other myosin 1s.
Collapse
Affiliation(s)
- Hanna Brzeska
- Laboratory of Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Jesus Gonzalez
- Laboratory of Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Edward D. Korn
- Laboratory of Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Margaret A. Titus
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455
| |
Collapse
|
20
|
Intermittent rolling is a defect of the extravasation cascade caused by Myosin1e-deficiency in neutrophils. Proc Natl Acad Sci U S A 2019; 116:26752-26758. [PMID: 31811025 DOI: 10.1073/pnas.1902502116] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Neutrophil extravasation is a migratory event in response to inflammation that depends on cytoskeletal dynamics regulated by myosins. Myosin-1e (Myo1e) is a long-tailed class-I myosin that has not yet been studied in the context of neutrophil-endothelial interactions and neutrophil extravasation. Intravital microscopy of TNFα-inflamed cremaster muscles in Myo1e-deficient mice revealed that Myo1e is required for efficient neutrophil extravasation. Specifically, Myo1e deficiency caused increased rolling velocity, decreased firm adhesion, aberrant crawling, and strongly reduced transmigration. Interestingly, we observed a striking discontinuous rolling behavior termed "intermittent rolling," during which Myo1e-deficient neutrophils showed alternating rolling and jumping movements. Surprisingly, chimeric mice revealed that these effects were due to Myo1e deficiency in leukocytes. Vascular permeability was not significantly altered in Myo1e KO mice. Myo1e-deficient neutrophils showed diminished arrest, spreading, uropod formation, and chemotaxis due to defective actin polymerization and integrin activation. In conclusion, Myo1e critically regulates adhesive interactions of neutrophils with the vascular endothelium and neutrophil extravasation. Myo1e may therefore be an interesting target in chronic inflammatory diseases characterized by excessive neutrophil recruitment.
Collapse
|
21
|
Zhang D, Baldwin P, Leal AS, Carapellucci S, Sridhar S, Liby KT. A nano-liposome formulation of the PARP inhibitor Talazoparib enhances treatment efficacy and modulates immune cell populations in mammary tumors of BRCA-deficient mice. Am J Cancer Res 2019; 9:6224-6238. [PMID: 31534547 PMCID: PMC6735511 DOI: 10.7150/thno.36281] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 07/22/2019] [Indexed: 12/28/2022] Open
Abstract
Two recently approved PARP inhibitors provide an important new therapeutic option for patients with BRCA-mutated metastatic breast cancer. PARP inhibitors significantly prolong progression-free survival in patients, but conventional oral delivery of PARP inhibitors is hindered by limited bioavailability and off-target toxicities, thus compromising the therapeutic benefits and quality of life for patients. Here, we developed a new delivery system, in which the PARP inhibitor Talazoparib is encapsulated in the bilayer of a nano-liposome, to overcome these limitations. Methods: Nano-Talazoparib (NanoTLZ) was characterized both in vitro and in vivo. The therapeutic efficacy and toxicity of Nano-Talazoparib (NanoTLZ) were evaluated in BRCA-deficient mice. The regulation of NanoTLZ on gene transcription and immunomodulation were further investigated in spontaneous BRCA-deficient tumors. Results: NanoTLZ significantly (p<0.05) prolonged the overall survival of BRCA-deficient mice compared to all of the other experimental groups, including saline control, empty nanoparticles, and free Talazoparib groups (oral and i.v.). Moreover, NanoTLZ was better tolerated than treatment with free Talazoparib, with no significant weight lost or alopecia as was observed with the free drug. After 5 doses, NanoTLZ altered the expression of over 140 genes and induced DNA damage, cell cycle arrest and inhibition of cell proliferation in the tumor. In addition, NanoTLZ favorably modulated immune cell populations in vivo and significantly (p<0.05) decreased the percentage of myeloid derived suppressor cells in both the tumor and spleen compared to control groups. Conclusions: Our results demonstrate that delivering nanoformulated Talazoparib not only enhances treatment efficacy but also reduces off-target toxicities in BRCA-deficient mice; the same potential is predicted for patients with BRCA-deficient breast cancer.
Collapse
|
22
|
Roles of Myosin-Mediated Membrane Trafficking in TGF-β Signaling. Int J Mol Sci 2019; 20:ijms20163913. [PMID: 31408934 PMCID: PMC6719161 DOI: 10.3390/ijms20163913] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 08/08/2019] [Accepted: 08/09/2019] [Indexed: 12/17/2022] Open
Abstract
Recent findings have revealed the role of membrane traffic in the signaling of transforming growth factor-β (TGF-β). These findings originate from the pivotal function of TGF-β in development, cell proliferation, tumor metastasis, and many other processes essential in malignancy. Actin and unconventional myosin have crucial roles in subcellular trafficking of receptors; research has also revealed a growing number of unconventional myosins that have crucial roles in TGF-β signaling. Unconventional myosins modulate the spatial organization of endocytic trafficking and tether membranes or transport them along the actin cytoskeletons. Current models do not fully explain how membrane traffic forms a bridge between TGF-β and the downstream effectors that produce its functional responsiveness, such as cell migration. In this review, we present a brief overview of the current knowledge of the TGF-β signaling pathway and the molecular components that comprise the core pathway as follows: ligands, receptors, and Smad mediators. Second, we highlight key role(s) of myosin motor-mediated protein trafficking and membrane domain segregation in the modulation of the TGF-β signaling pathway. Finally, we review future challenges and provide future prospects in this field.
Collapse
|
23
|
Zhang Y, Cao F, Zhou Y, Feng Z, Sit B, Krendel M, Yu CH. Tail domains of myosin-1e regulate phosphatidylinositol signaling and F-actin polymerization at the ventral layer of podosomes. Mol Biol Cell 2019; 30:622-635. [PMID: 30601698 PMCID: PMC6589698 DOI: 10.1091/mbc.e18-06-0398] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
During podosome formation, distinct phosphatidylinositol 3,4,5-trisphosphate lipid (PI(3,4,5)P3) production and F-actin polymerization take place at integrin-mediated adhesions. Membrane-associated actin regulation factors, such as myosin-1, serve as key molecules to link phosphatidylinositol signals to podosome assembly. Here, we report that long-tailed myosin-1e (Myo1e) is enriched at the ventral layer of the podosome core in a PI(3,4,5)P3-dependent manner. The combination of TH1 and TH2 (TH12) of Myo1e tail domains contains the essential motif for PI(3,4,5)P3-dependent membrane association and ventral localization at the podosome. TH12 KR2A (K772A and R782A) becomes dissociated from the plasma membrane. While F-actin polymerizations are initialized from the ventral layer of the podosome, TH12 precedes the recruitment of N-WASP and Arp2/3 in the initial phase of podosome formation. Overexpression of TH12, not TH12 KR2A, impedes PI(3,4,5)P3 signaling, restrains F-actin polymerization, and inhibits podosome formation. TH12 also suppresses gelatin degradation and migration speed of invadopodia-forming A375 melanoma cells. Thus, TH12 domain of Myo1e serves as a regulatory component to connect phosphatidylinositol signaling to F-actin polymerization at the podosome.
Collapse
Affiliation(s)
- Yage Zhang
- School of Biomedical Sciences, Faculty of Medicine, University of Hong Kong, Hong Kong
| | - Fakun Cao
- School of Biomedical Sciences, Faculty of Medicine, University of Hong Kong, Hong Kong
| | - Yuhuan Zhou
- School of Biomedical Sciences, Faculty of Medicine, University of Hong Kong, Hong Kong
| | - Zhen Feng
- School of Biomedical Sciences, Faculty of Medicine, University of Hong Kong, Hong Kong
| | - Brian Sit
- School of Biomedical Sciences, Faculty of Medicine, University of Hong Kong, Hong Kong.,Randall Division of Cell and Molecular Biophysics, Faculty of Life Sciences and Medicine, King's College London, London WC2R 2LS, United Kingdom
| | - Mira Krendel
- SUNY Upstate Medical University, Syracuse, NY 13210
| | - Cheng-Han Yu
- School of Biomedical Sciences, Faculty of Medicine, University of Hong Kong, Hong Kong
| |
Collapse
|
24
|
Lyraki R, Lokaj M, Soares DC, Little A, Vermeren M, Marsh JA, Wittinghofer A, Hurd T. Characterization of a novel RP2-OSTF1 interaction and its implication for actin remodelling. J Cell Sci 2018; 131:jcs.211748. [PMID: 29361551 DOI: 10.1242/jcs.211748] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 12/21/2017] [Indexed: 11/20/2022] Open
Abstract
Retinitis pigmentosa 2 (RP2) is the causative gene for a form of X-linked retinal degeneration. RP2 was previously shown to have GTPase-activating protein (GAP) activity towards the small GTPase ARL3 via its N-terminus, but the function of the C-terminus remains elusive. Here, we report a novel interaction between RP2 and osteoclast-stimulating factor 1 (OSTF1), an intracellular protein that indirectly enhances osteoclast formation and activity and is a negative regulator of cell motility. Moreover, this interaction is abolished by a human pathogenic mutation in RP2. We utilized a structure-based approach to pinpoint the binding interface to a strictly conserved cluster of residues on the surface of RP2 that spans both the C- and N-terminal domains of the protein, and which is structurally distinct from the ARL3-binding site. In addition, we show that RP2 is a positive regulator of cell motility in vitro, recruiting OSTF1 to the cell membrane and preventing its interaction with the migration regulator Myo1E.
Collapse
Affiliation(s)
- Rodanthi Lyraki
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK
| | - Mandy Lokaj
- Structural Biology Group, Max-Planck Institut für Molekulare Physiologie, Abteilung Strukturelle Biologie, Otto-Hahn-Str. 11, 44227 Dortmund, Germany
| | - Dinesh C Soares
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK
| | - Abigail Little
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK
| | - Matthieu Vermeren
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK.,MRC Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Joseph A Marsh
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK
| | - Alfred Wittinghofer
- Structural Biology Group, Max-Planck Institut für Molekulare Physiologie, Abteilung Strukturelle Biologie, Otto-Hahn-Str. 11, 44227 Dortmund, Germany
| | - Toby Hurd
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK
| |
Collapse
|
25
|
Encinar Del Dedo J, Idrissi FZ, Fernandez-Golbano IM, Garcia P, Rebollo E, Krzyzanowski MK, Grötsch H, Geli MI. ORP-Mediated ER Contact with Endocytic Sites Facilitates Actin Polymerization. Dev Cell 2017; 43:588-602.e6. [PMID: 29173820 DOI: 10.1016/j.devcel.2017.10.031] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 09/11/2017] [Accepted: 10/27/2017] [Indexed: 11/18/2022]
Abstract
Oxysterol binding protein-related proteins (ORPs) are conserved lipid binding polypeptides, enriched at ER contacts sites. ORPs promote non-vesicular lipid transport and work as lipid sensors in the context of many cellular tasks, but the determinants of their distinct localization and function are not understood. Here, we demonstrate that the yeast endocytic invaginations associate with the ER and that this association specifically requires the ORPs Osh2 and Osh3, which bridge the endocytic myosin-I Myo5 to the ER integral-membrane VAMP-associated protein (VAP) Scs2. Disruption of the ER contact with endocytic sites using ORP, VAP, myosin-I, or reticulon mutants delays and weakens actin polymerization and interferes with vesicle scission. Finally, we provide evidence suggesting that ORP-dependent sterol transfer facilitates actin polymerization at endocytic sites.
Collapse
Affiliation(s)
- Javier Encinar Del Dedo
- Institute for Molecular Biology of Barcelona (CSIC), Baldiri Reixac 15, 08028 Barcelona, Spain
| | - Fatima-Zahra Idrissi
- Institute for Molecular Biology of Barcelona (CSIC), Baldiri Reixac 15, 08028 Barcelona, Spain
| | | | - Patricia Garcia
- Institute for Molecular Biology of Barcelona (CSIC), Baldiri Reixac 15, 08028 Barcelona, Spain
| | - Elena Rebollo
- Institute for Molecular Biology of Barcelona (CSIC), Baldiri Reixac 15, 08028 Barcelona, Spain
| | - Marek K Krzyzanowski
- Institute for Molecular Biology of Barcelona (CSIC), Baldiri Reixac 15, 08028 Barcelona, Spain
| | - Helga Grötsch
- Institute for Molecular Biology of Barcelona (CSIC), Baldiri Reixac 15, 08028 Barcelona, Spain
| | - Maria Isabel Geli
- Institute for Molecular Biology of Barcelona (CSIC), Baldiri Reixac 15, 08028 Barcelona, Spain.
| |
Collapse
|
26
|
Mohammed A, Biegert G, Adamec J, Helikar T. Identification of potential tissue-specific cancer biomarkers and development of cancer versus normal genomic classifiers. Oncotarget 2017; 8:85692-85715. [PMID: 29156751 PMCID: PMC5689641 DOI: 10.18632/oncotarget.21127] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 09/05/2017] [Indexed: 01/15/2023] Open
Abstract
Machine learning techniques for cancer prediction and biomarker discovery can hasten cancer detection and significantly improve prognosis. Recent “OMICS” studies which include a variety of cancer and normal tissue samples along with machine learning approaches have the potential to further accelerate such discovery. To demonstrate this potential, 2,175 gene expression samples from nine tissue types were obtained to identify gene sets whose expression is characteristic of each cancer class. Using random forests classification and ten-fold cross-validation, we developed nine single-tissue classifiers, two multi-tissue cancer-versus-normal classifiers, and one multi-tissue normal classifier. Given a sample of a specified tissue type, the single-tissue models classified samples as cancer or normal with a testing accuracy between 85.29% and 100%. Given a sample of non-specific tissue type, the multi-tissue bi-class model classified the sample as cancer versus normal with a testing accuracy of 97.89%. Given a sample of non-specific tissue type, the multi-tissue multi-class model classified the sample as cancer versus normal and as a specific tissue type with a testing accuracy of 97.43%. Given a normal sample of any of the nine tissue types, the multi-tissue normal model classified the sample as a particular tissue type with a testing accuracy of 97.35%. The machine learning classifiers developed in this study identify potential cancer biomarkers with sensitivity and specificity that exceed those of existing biomarkers and pointed to pathways that are critical to tissue-specific tumor development. This study demonstrates the feasibility of predicting the tissue origin of carcinoma in the context of multiple cancer classes.
Collapse
Affiliation(s)
- Akram Mohammed
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Greyson Biegert
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Jiri Adamec
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Tomáš Helikar
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| |
Collapse
|
27
|
Myosin-1E interacts with FAK proline-rich region 1 to induce fibronectin-type matrix. Proc Natl Acad Sci U S A 2017; 114:3933-3938. [PMID: 28348210 DOI: 10.1073/pnas.1614894114] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Focal adhesion kinase (FAK) is a nonreceptor tyrosine kinase involved in development and human disease, including cancer. It is currently thought that the four-point one, ezrin, radixin, moesin (FERM)-kinase domain linker, which contains autophosphorylation site tyrosine (Y) 397, is not required for in vivo FAK function until late midgestation. Here, we directly tested this hypothesis by generating mice with FAK Y397-to-phenylalanine (F) mutations in the germline. We found that Y397F embryos exhibited reduced mesodermal fibronectin (FN) and osteopontin expression and died during mesoderm development akin to FAK kinase-dead mice. We identified myosin-1E (MYO1E), an actin-dependent molecular motor, to interact directly with the FAK FERM-kinase linker and induce FAK kinase activity and Y397 phosphorylation. Active FAK in turn accumulated in the nucleus where it led to the expression of osteopontin and other FN-type matrix in both mouse embryonic fibroblasts and human melanoma. Our data support a model in which FAK Y397 autophosphorylation is required for FAK function in vivo and is positively regulated by MYO1E.
Collapse
|
28
|
Hic-5 remodeling of the stromal matrix promotes breast tumor progression. Oncogene 2016; 36:2693-2703. [PMID: 27893716 PMCID: PMC5541773 DOI: 10.1038/onc.2016.422] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 10/03/2016] [Accepted: 10/03/2016] [Indexed: 12/19/2022]
Abstract
The remodeling of the stromal extracellular matrix (ECM) plays a crucial, but incompletely understood role during tumor progression and metastasis. Hic-5, a focal adhesion scaffold protein, has previously been implicated in tumor cell invasion, proliferation and metastasis. To investigate the role of Hic-5 in breast tumor progression in vivo, Hic-5−/− mice were generated and crossed with the Mouse Mammary Tumor Virus-Polyoma Middle T Antigen (MMTV-PyMT) mouse. Tumors from the Hic-5−/−;PyMT mice exhibited increased latency and reduced growth, with fewer lung metastases, as compared to Hic-5+/−;PyMT mice. Immunohistochemical analysis showed that Hic-5 is primarily expressed in the cancer associated fibroblasts (CAFs). Further analysis revealed that the Hic-5−/−;PyMT tumor stroma contains fewer CAFs and exhibits reduced ECM deposition. The remodeling of the stromal matrix by CAFs has been shown to increase tumor rigidity to indirectly regulate FAK Y397 phosphorylation in tumor cells to promote their growth and invasion. Accordingly, the Hic-5−/−;PyMT tumor cells exhibited a reduction in FAK Y397 phosphorylation. Isolated Hic-5−/−;PyMT CAFs were defective in stress fiber organization and exhibited reduced contractility. These cells also failed to efficiently deposit and organize the ECM in two and three dimensions. This, in turn, impacted three dimensional MDA-MB-231 tumor cell migration behavior. Thus, using a new knockout mouse model, we have identified Hic-5 expression in CAFs as a key requirement for deposition and remodeling of the stromal ECM to promote non-cell autonomous breast tumor progression.
Collapse
|