1
|
Wu J, Li Y, Nabi G, Huang X, Zhang X, Wang Y, Huang L. Exosome and lipid metabolism-related genes in pancreatic adenocarcinoma: a prognosis analysis. Aging (Albany NY) 2023; 15:11331-11368. [PMID: 37857015 PMCID: PMC10637811 DOI: 10.18632/aging.205130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 09/27/2023] [Indexed: 10/21/2023]
Abstract
OBJECTIVE The purpose of the study was to investigate the role of exosome and lipid metabolism-related genes (EALMRGs) mRNA levels in the diagnosis and prognosis of Pancreatic Adenocarcinoma (PAAD). METHODS The mRNA expression pattern of PAAD and pan-cancers with prognostic data were obtained from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) database. EALMRGs were acquired from GeneCards and MSigDB database after merging and deduplication. Prognostic EALMRGs were screened through univariate COX regression analysis, and a prognostic model was constructed based on these genes by least absolute shrinkage and selection operator (LASSO) regression. The prognostic value of EALMRGs was then validated in pan-cancer data. The time characteristics ROC curve analysis was performed to evaluate the effectiveness of the prognostic genes. RESULTS We identified 5 hub genes (ABCB1, CAP1, EGFR, PPARG, SNCA) according to high and low-risk groups of prognoses. The risk formula was verified in three other cohort of pancreatic cancer patients and was explored in pan-cancer data. Additionally, T cell and dendritic cell infiltration was significantly increased in low-risk group. The expression of the 5 hub genes was also identified in single-cell sequencing data of pancreatic cancer with pivotal pathways. Additionally, functional enrichment analysis based on pancreatic cancer data in pancreatic cancer showed that protein serine/threonine kinase activity, focal adhesion, actin binding, cell-substrate junction, organic acid transport, and regulation of transporter activity were significant related to the expression of genes in EALMRGs. CONCLUSIONS Our risk formula shows potential prognostic value in multiple cancers and manifest pivotal alterations in immune infiltration and biological pathway in pancreatic cancer.
Collapse
Affiliation(s)
- Jia Wu
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Yajun Li
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Ghulam Nabi
- Institute of Nature Conservation, Polish Academy of Sciences, Krakow, Poland
| | - Xin Huang
- Department of Gastroenterology, Traditional Chinese Medicine Hospital of Yinchuan, Yinchuan, Ningxia, China
| | - Xu Zhang
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Yuanzhen Wang
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Liya Huang
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| |
Collapse
|
2
|
Ramsey A, Akana L, Miyajima E, Douglas S, Gray J, Rowland A, Sharma KD, Xu J, Xie JY, Zhou GL. CAP1 (cyclase-associated protein 1) mediates the cyclic AMP signals that activate Rap1 in stimulating matrix adhesion of colon cancer cells. Cell Signal 2023; 104:110589. [PMID: 36621727 PMCID: PMC9908859 DOI: 10.1016/j.cellsig.2023.110589] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 12/12/2022] [Accepted: 01/03/2023] [Indexed: 01/07/2023]
Abstract
We previously reported that CAP1 (Cyclase-Associated Protein 1) regulates matrix adhesion in mammalian cells through FAK (Focal Adhesion Kinase). More recently, we discovered a phosphor-regulation mechanism for CAP1 through the Ser307/Ser309 tandem site that is of critical importance for all CAP1 functions. However, molecular mechanisms underlying the CAP1 function in adhesion and its regulation remain largely unknown. Here we report that Rap1 also facilitates the CAP1 function in adhesion, and more importantly, we identify a novel signaling pathway where CAP1 mediates the cAMP signals, through the cAMP effectors Epac (Exchange proteins directly activated by cAMP) and PKA (Protein Kinase A), to activate Rap1 in stimulating matrix adhesion in colon cancer cells. Knockdown of CAP1 led to opposite adhesion phenotypes in SW480 and HCT116 colon cancer cells, with reduced matrix adhesion and reduced FAK and Rap1 activities in SW480 cells while it stimulated matrix adhesion as well as FAK and Rap1 activities in HCT116 cells. Importantly, depletion of CAP1 abolished the stimulatory effects of the cAMP activators forskolin and isoproterenol, as well as that of Epac and PKA, on matrix adhesion in both cell types. Our results consistently support a required role for CAP1 in the cAMP activation of Rap1. Identification of the key role for CAP1 in linking the major second messenger cAMP to activation of Rap1 in stimulating adhesion, which may potentially also regulate proliferation in other cell types, not only vertically extends our knowledge on CAP biology, but also carries important translational potential for targeting CAP1 in cancer therapeutics.
Collapse
Affiliation(s)
- Auburn Ramsey
- Department of Biological Sciences, Arkansas State University, State University, AR 72467, USA
| | - Lokesh Akana
- Department of Biological Sciences, Arkansas State University, State University, AR 72467, USA
| | - Erina Miyajima
- Department of Biological Sciences, Arkansas State University, State University, AR 72467, USA
| | - Spencer Douglas
- Department of Biological Sciences, Arkansas State University, State University, AR 72467, USA
| | - Joshua Gray
- Department of Biological Sciences, Arkansas State University, State University, AR 72467, USA
| | - Alyssa Rowland
- Department of Biological Sciences, Arkansas State University, State University, AR 72467, USA
| | - Krishna Deo Sharma
- Molecular Biosciences Graduate Program, Arkansas State University, State University, AR 72467, USA
| | - Jianfeng Xu
- Molecular Biosciences Graduate Program, Arkansas State University, State University, AR 72467, USA; Arkansas Biosciences Institute, Arkansas State University, Jonesboro, AR 72401, USA; College of Agriculture, Arkansas State University, State University, AR 72467, USA
| | - Jennifer Y Xie
- Molecular Biosciences Graduate Program, Arkansas State University, State University, AR 72467, USA; Department of Basic Sciences, New York Institute of Technology College of Osteopathic Medicine at Arkansas State University, Jonesboro, AR 72401, USA
| | - Guo-Lei Zhou
- Department of Biological Sciences, Arkansas State University, State University, AR 72467, USA; Molecular Biosciences Graduate Program, Arkansas State University, State University, AR 72467, USA.
| |
Collapse
|
3
|
Nakada-Tsukui K, Watanabe N, Shibata K, Wahyuni R, Miyamoto E, Nozaki T. Proteomic analysis of Atg8-dependent recruitment of phagosomal proteins in the enteric protozoan parasite Entamoeba histolytica. Front Cell Infect Microbiol 2022; 12:961645. [PMID: 36262186 PMCID: PMC9575557 DOI: 10.3389/fcimb.2022.961645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 08/11/2022] [Indexed: 11/13/2022] Open
Abstract
Autophagy is one of the bulk degradation systems and is conserved throughout eukaryotes. In the enteric protozoan parasite Entamoeba histolytica, the causative agent of human amebiasis, Atg8 is not exclusively involved in autophagy per se but also in other membrane traffic-related pathways such as phagosome biogenesis. We previously reported that repression of atg8 gene expression by antisense small RNA-mediated transcriptional gene silencing (gs) resulted in growth retardation, delayed endocytosis, and reduced acidification of endosomes and phagosomes. In this study, to better understand the role of Atg8 in phagocytosis and trogocytosis, we conducted a comparative proteomic analysis of phagosomes isolated from wild type and atg8-gs strains. We found that 127 and 107 proteins were detected >1.5-fold less or more abundantly, respectively, in phagosomes isolated from the atg8-gs strain, compared to the control strain. Among 127 proteins whose abundance was reduced in phagosomes from atg8-gs, a panel of proteins related to fatty acid metabolism, phagocytosis, and endoplasmic reticulum (ER) homeostasis was identified. Various lysosomal hydrolases and their receptors also tend to be excluded from phagosomes by atg8-gs, reinforcing the notion that Atg8 is involved in phagosomal acidification and digestion. On the contrary, among 107 proteins whose abundance increased in phagosomes from atg8-gs strain, ribosome-related proteins and metabolite interconversion enzymes are enriched. We further investigated the localization of several representative proteins, including adenylyl cyclase-associated protein and plasma membrane calcium pump, both of which were demonstrated to be recruited to phagosomes and trogosomes via an Atg8-dependent mechanism. Taken together, our study has provided the basis of the phagosome proteome to further elucidate molecular events in the Atg8-dependent regulatory network of phagosome/trogosome biogenesis in E. histolytica.
Collapse
Affiliation(s)
- Kumiko Nakada-Tsukui
- Department of Parasitology, National Institute of Infectious Diseases, Tokyo, Japan
- *Correspondence: Kumiko Nakada-Tsukui, ; Tomoyoshi Nozaki,
| | - Natsuki Watanabe
- Department of Biomedical Chemistry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kumiko Shibata
- Department of Biomedical Chemistry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Ratna Wahyuni
- Department of Biomedical Chemistry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Eri Miyamoto
- Department of Parasitology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Tomoyoshi Nozaki
- Department of Biomedical Chemistry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- *Correspondence: Kumiko Nakada-Tsukui, ; Tomoyoshi Nozaki,
| |
Collapse
|
4
|
Wang Q, Yuan W, Yang X, Wang Y, Li Y, Qiao H. Role of Cofilin in Alzheimer's Disease. Front Cell Dev Biol 2020; 8:584898. [PMID: 33324642 PMCID: PMC7726191 DOI: 10.3389/fcell.2020.584898] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 10/26/2020] [Indexed: 01/14/2023] Open
Abstract
Alzheimer's disease (AD) is a degenerative neurological disease and has an inconspicuous onset and progressive development. Clinically, it is characterized by severe dementia manifestations, including memory impairment, aphasia, apraxia, loss of recognition, impairment of visual-spatial skills, executive dysfunction, and changes in personality and behavior. Its etiology is unknown to date. However, several cellular biological signatures of AD have been identified such as synaptic dysfunction, β-amyloid plaques, hyperphosphorylated tau, cofilin-actin rods, and Hirano bodies which are related to the actin cytoskeleton. Cofilin is one of the most affluent and common actin-binding proteins and plays a role in cell motility, migration, shape, and metabolism. They also play an important role in severing actin filament, nucleating, depolymerizing, and bundling activities. In this review, we summarize the structure of cofilins and their functional and regulating roles, focusing on the synaptic dysfunction, β-amyloid plaques, hyperphosphorylated tau, cofilin-actin rods, and Hirano bodies of AD.
Collapse
Affiliation(s)
- Qiang Wang
- College of Acupuncture and Massage, Shaanxi University of Chinese Medicine, Xianyang, China
- Shaanxi Key Laboratory of Acupuncture and Medicine, Xianyang, China
| | - Wei Yuan
- College of Acupuncture and Massage, Shaanxi University of Chinese Medicine, Xianyang, China
- Shaanxi Key Laboratory of Acupuncture and Medicine, Xianyang, China
| | - Xiaohang Yang
- College of Acupuncture and Massage, Shaanxi University of Chinese Medicine, Xianyang, China
- College of Medical Technology, Shaanxi University of Chinese Medicine, Xi’an, China
| | - Yuan Wang
- College of Acupuncture and Massage, Shaanxi University of Chinese Medicine, Xianyang, China
- Shaanxi Key Laboratory of Acupuncture and Medicine, Xianyang, China
| | - Yongfeng Li
- College of Acupuncture and Massage, Shaanxi University of Chinese Medicine, Xianyang, China
- Shaanxi Key Laboratory of Acupuncture and Medicine, Xianyang, China
| | - Haifa Qiao
- College of Acupuncture and Massage, Shaanxi University of Chinese Medicine, Xianyang, China
- Shaanxi Key Laboratory of Acupuncture and Medicine, Xianyang, China
- Xianyang Key Laboratory of Neurobiology and Acupuncture, Xi’an, China
| |
Collapse
|
5
|
Rust MB, Khudayberdiev S, Pelucchi S, Marcello E. CAPt'n of Actin Dynamics: Recent Advances in the Molecular, Developmental and Physiological Functions of Cyclase-Associated Protein (CAP). Front Cell Dev Biol 2020; 8:586631. [PMID: 33072768 PMCID: PMC7543520 DOI: 10.3389/fcell.2020.586631] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 08/26/2020] [Indexed: 12/11/2022] Open
Abstract
Cyclase-associated protein (CAP) has been discovered three decades ago in budding yeast as a protein that associates with the cyclic adenosine monophosphate (cAMP)-producing adenylyl cyclase and that suppresses a hyperactive RAS2 variant. Since that time, CAP has been identified in all eukaryotic species examined and it became evident that the activity in RAS-cAMP signaling is restricted to a limited number of species. Instead, its actin binding activity is conserved among eukaryotes and actin cytoskeleton regulation emerged as its primary function. However, for many years, the molecular functions as well as the developmental and physiological relevance of CAP remained unknown. In the present article, we will compile important recent progress on its molecular functions that identified CAP as a novel key regulator of actin dynamics, i.e., the spatiotemporally controlled assembly and disassembly of actin filaments (F-actin). These studies unraveled a cooperation with ADF/Cofilin and Twinfilin in F-actin disassembly, a nucleotide exchange activity on globular actin monomers (G-actin) that is required for F-actin assembly and an inhibitory function towards the F-actin assembly factor INF2. Moreover, by focusing on selected model organisms, we will review current literature on its developmental and physiological functions, and we will present studies implicating CAP in human pathologies. Together, this review article summarizes and discusses recent achievements in understanding the molecular, developmental and physiological functions of CAP, which led this protein emerge as a novel CAPt'n of actin dynamics.
Collapse
Affiliation(s)
- Marco B Rust
- Molecular Neurobiology Group, Institute of Physiological Chemistry, University of Marburg, Marburg, Germany.,DFG Research Training Group, Membrane Plasticity in Tissue Development and Remodeling, GRK 2213, University of Marburg, Marburg, Germany.,Center for Mind, Brain and Behavior, University of Marburg and Justus-Liebig-University Giessen, Giessen, Germany
| | - Sharof Khudayberdiev
- Molecular Neurobiology Group, Institute of Physiological Chemistry, University of Marburg, Marburg, Germany
| | - Silvia Pelucchi
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Elena Marcello
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| |
Collapse
|
6
|
Adachi M, Masugi Y, Yamazaki K, Emoto K, Kobayashi Y, Tominaga E, Banno K, Aoki D, Sakamoto M. Upregulation of cyclase-associated actin cytoskeleton regulatory protein 2 in epithelial ovarian cancer correlates with aggressive histologic types and worse outcomes. Jpn J Clin Oncol 2020; 50:643-652. [PMID: 32211793 DOI: 10.1093/jjco/hyaa026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 01/22/2020] [Accepted: 02/08/2020] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE Cyclase-associated actin cytoskeleton regulatory protein 2 (CAP2) regulates actin dynamics to control cell cycles and cell migration. CAP2 overexpression contributes to cancer progression in several tumor types; however, the role of CAP2 expression in ovarian cancer remains unclear. This study aimed to clarify the significance of CAP2 expression in epithelial ovarian tumor. METHODS We evaluated CAP2 expression in ovarian cancer cell lines using quantitative real-time polymerase chain reaction, western blotting and immunocytochemistry and examined the effect of CAP2 silencing in migration and proliferation assays. CAP2 immunohistochemistry was conducted using tissue specimens from 432 ovarian carcinoma patients; a further 55 borderline and benign 65 lesions were analyzed. CAP2 expression levels were defined as low, intermediate or high, for correlation analysis with clinicopathological factors. RESULTS CAP2 expression was significantly higher in cell lines from Type II ovarian cancer than in those in Type I, and knockdown of CAP2 showed decreased migration and proliferation. Higher levels of CAP2 expression in human tissues were associated with Type II histology, residual lesion, lymph node metastasis, ascites cytology and higher clinical stage. High CAP2 expression levels were observed in 26 (23.4%) of 111 Type II ovarian cancers and in 16 (5.0%) of 321 Type I cancers but not in any borderline or benign lesions. Multivariate analyses showed that CAP2 expression in ovarian cancer is an independent prognostic factor for recurrence-free survival (P = 0.019). CONCLUSION CAP2 expression is upregulated in aggressive histologic types of epithelial ovarian cancer and serves as a novel prognostic biomarker for patient survival.
Collapse
Affiliation(s)
- Masataka Adachi
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan.,Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Yohei Masugi
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Ken Yamazaki
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Katsura Emoto
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Yusuke Kobayashi
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Eiichiro Tominaga
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Kouji Banno
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Daisuke Aoki
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Michiie Sakamoto
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
7
|
Dynamic Phosphorylation and Dephosphorylation of Cyclase-Associated Protein 1 by Antagonistic Signaling through Cyclin-Dependent Kinase 5 and cAMP Are Critical for the Protein Functions in Actin Filament Disassembly and Cell Adhesion. Mol Cell Biol 2020; 40:MCB.00282-19. [PMID: 31791978 DOI: 10.1128/mcb.00282-19] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 11/25/2019] [Indexed: 12/15/2022] Open
Abstract
Cyclase-associated protein 1 (CAP1) is a conserved actin-regulating protein that enhances actin filament dynamics and also regulates adhesion in mammalian cells. We previously found that phosphorylation at the Ser307/Ser309 tandem site controls its association with cofilin and actin and is important for CAP1 to regulate the actin cytoskeleton. Here, we report that transient Ser307/Ser309 phosphorylation is required for CAP1 function in both actin filament disassembly and cell adhesion. Both the phosphomimetic and the nonphosphorylatable CAP1 mutant, which resist transition between phosphorylated and dephosphorylated forms, had defects in rescuing the reduced rate of actin filament disassembly in the CAP1 knockdown HeLa cells. The phosphorylation mutants also had defects in alleviating the elevated focal adhesion kinase (FAK) activity and the enhanced focal adhesions in the knockdown cells. In dissecting further phosphoregulatory cell signals for CAP1, we found that cyclin-dependent kinase 5 (CDK5) phosphorylates both Ser307 and Ser309 residues, whereas cAMP signaling induces dephosphorylation at the tandem site, through its effectors protein kinase A (PKA) and exchange proteins directly activated by cAMP (Epac). No evidence supports an involvement of activated protein phosphatase in executing the dephosphorylation downstream from cAMP, whereas preventing CAP1 from accessing its kinase CDK5 appears to underlie CAP1 dephosphorylation induced by cAMP. Therefore, this study provides direct cellular evidence that transient phosphorylation is required for CAP1 functions in both actin filament turnover and adhesion, and the novel mechanistic insights substantially extend our knowledge of the cell signals that function in concert to regulate CAP1 by facilitating its transient phosphorylation.
Collapse
|
8
|
Avtanski D, Garcia A, Caraballo B, Thangeswaran P, Marin S, Bianco J, Lavi A, Poretsky L. Resistin induces breast cancer cells epithelial to mesenchymal transition (EMT) and stemness through both adenylyl cyclase-associated protein 1 (CAP1)-dependent and CAP1-independent mechanisms. Cytokine 2019; 120:155-164. [PMID: 31085453 DOI: 10.1016/j.cyto.2019.04.016] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 03/21/2019] [Accepted: 04/22/2019] [Indexed: 01/08/2023]
Abstract
Breast cancer incidence and metastasis in postmenopausal women are known to associate with obesity, but the molecular mechanisms behind this association are largely unknown. We investigated the effect of adipokine resistin on epithelial to mesenchymal transition (EMT) and stemness in breast cancer cells in vitro. Previous reports demonstrated that the inflammatory actions of resistin are mediated by the adenylyl cyclase-associated protein 1 (CAP1), which serves as its receptor. As a model for our study, we used MCF-7 and MDA-MB-231 breast cancer and MCF-10A breast epithelial cells. We showed that in MCF-7 cells resistin increases the migration of MCF-7 and MDA-MB-231 cells and induces the formation of cellular protrusions through reorganization of F-actin filaments. Resistin upregulated the expression of mesenchymal markers involved in EMT (SNAIL, SLUG, ZEB1, TWIST1, fibronectin, and vimentin), and downregulated those of epithelial markers (E-cadherin and claudin-1). Resistin also potentiated the nuclear translocation of SNAIL protein, indicating initiation of EMT reprogramming. We further induced EMT in non-carcinogenic breast epithelial MCF-10A cells demonstrating that the effects of resistin on EMT were not breast cancer cell specific. In order to assess whether resistin-induced EMT depends on CAP1, we used siRNA approach to silence CAP1 gene in MCF-7 cells. Results demonstrated that when CAP1 was silenced, the induction of SNAIL, ZEB1 and vimentin expression by resistin as well as SNAIL and ZEB1 nuclear translocation, were abolished. Additionally, CAP1 silencing resulted in a suppression of MCF-7 cells migration. We performed quantitative PCR array profiling the expression of 84 genes related to cancer stem cells (CSC), pluripotency and metastasis and selected a set of genes (ALDH1A1, ITGA4, LIN28B, SMO, KLF17, PTPRC, PROM1, SIRT1, and PECAM1) that were modulated by resistin. Further experiments demonstrated that the effect of resistin on the expression of some of these genes (PROM1, PTPRC, KLF17, SIRT1, and PECAM1) was also dependent on CAP1. Our results demonstrate that resistin promotes the metastatic potential of breast cancer cells by inducing EMT and stemness and some of these effects are mediated by CAP1.
Collapse
Affiliation(s)
- Dimiter Avtanski
- Friedman Diabetes Institute at Lenox Hill Hospital, Northwell Health, New York, NY, USA; The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, USA; Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA.
| | - Anabel Garcia
- Friedman Diabetes Institute at Lenox Hill Hospital, Northwell Health, New York, NY, USA
| | - Beatriz Caraballo
- Friedman Diabetes Institute at Lenox Hill Hospital, Northwell Health, New York, NY, USA
| | | | - Sela Marin
- Friedman Diabetes Institute at Lenox Hill Hospital, Northwell Health, New York, NY, USA
| | - Julianna Bianco
- Friedman Diabetes Institute at Lenox Hill Hospital, Northwell Health, New York, NY, USA
| | - Aaron Lavi
- Friedman Diabetes Institute at Lenox Hill Hospital, Northwell Health, New York, NY, USA
| | - Leonid Poretsky
- Friedman Diabetes Institute at Lenox Hill Hospital, Northwell Health, New York, NY, USA; The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, USA; Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| |
Collapse
|
9
|
Kolegova ES, Kakurina GV, Kondakova IV, Dobrodeev AY, Kostromitskii DN, Zhuikova LD. Adenylate Cyclase-Associated Protein 1 and Cofilin in Progression of Non-Small Cell Lung Cancer. Bull Exp Biol Med 2019; 167:393-395. [PMID: 31346877 DOI: 10.1007/s10517-019-04534-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Indexed: 10/26/2022]
Abstract
We studied the expression of mRNA and the level of CAP1 (adenylate cyclase-associated protein 1) and cofilin proteins in the tissues of patients with non-small cell lung cancer. The expression of mRNA and the level of CAP1 in tumor tissue increased during growth of the primary tumor and its metastasis. It was shown that with the growth of the primary tumor, the content of cofilin in the tumor tissue decreases against the background of increased expression of its mRNA; in regional metastasis, the content of cofilin and expression of the corresponding mRNA increased. It was found that increased content of the studied proteins in the tumor tissue increased the risk of metastasis.
Collapse
Affiliation(s)
- E S Kolegova
- Tomsk National Research Medical Center, Russian Academy of Science, Tomsk, Russia.
| | - G V Kakurina
- Tomsk National Research Medical Center, Russian Academy of Science, Tomsk, Russia
| | - I V Kondakova
- Tomsk National Research Medical Center, Russian Academy of Science, Tomsk, Russia
| | - A Yu Dobrodeev
- Tomsk National Research Medical Center, Russian Academy of Science, Tomsk, Russia
| | | | - L D Zhuikova
- Tomsk National Research Medical Center, Russian Academy of Science, Tomsk, Russia
| |
Collapse
|
10
|
ADF/cofilin regulation from a structural viewpoint. J Muscle Res Cell Motil 2019; 41:141-151. [DOI: 10.1007/s10974-019-09546-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 07/17/2019] [Indexed: 01/11/2023]
|
11
|
Hasan R, Zhou GL. The Cytoskeletal Protein Cyclase-Associated Protein 1 (CAP1) in Breast Cancer: Context-Dependent Roles in Both the Invasiveness and Proliferation of Cancer Cells and Underlying Cell Signals. Int J Mol Sci 2019; 20:E2653. [PMID: 31151140 PMCID: PMC6600220 DOI: 10.3390/ijms20112653] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 05/20/2019] [Accepted: 05/28/2019] [Indexed: 12/22/2022] Open
Abstract
As a conserved actin-regulating protein, CAP (adenylyl Cyclase-Associated Protein) functions to facilitate the rearrangement of the actin cytoskeleton. The ubiquitously expressed isoform CAP1 drives mammalian cell migration, and accordingly, most studies on the involvement of CAP1 in human cancers have largely been based on the rationale that up-regulated CAP1 will stimulate cancer cell migration and invasiveness. While findings from some studies reported so far support this case, lines of evidence largely from our recent studies point to a more complex and profound role for CAP1 in the invasiveness of cancer cells, where the potential activation of cell adhesion signaling is believed to play a key role. Moreover, CAP1 was also found to control proliferation in breast cancer cells, through the regulation of ERK (External signal-Regulated Kinase). Alterations in the activities of FAK (Focal Adhesion Kinase) and ERK from CAP1 depletion that are consistent to the opposite adhesion and proliferation phenotypes were detected in the metastatic and non-metastatic breast cancer cells. In this review, we begin with the overview of the literature on CAP, by highlighting the molecular functions of mammalian CAP1 in regulating the actin cytoskeleton and cell adhesion. We will next discuss the role of the FAK/ERK axis, and possibly Rap1, in mediating CAP1 signals to control breast cancer cell adhesion, invasiveness, and proliferation, largely based on our latest findings. Finally, we will discuss the relevance of these novel mechanistic insights to ultimately realizing the translational potential of CAP1 in targeted therapeutics for breast cancer.
Collapse
Affiliation(s)
- Rokib Hasan
- Molecular Biosciences Graduate Program, Arkansas State University, State University, AR 72467, USA.
| | - Guo-Lei Zhou
- Molecular Biosciences Graduate Program, Arkansas State University, State University, AR 72467, USA.
- Department of Biological Sciences, Arkansas State University, State University, AR 72467, USA.
| |
Collapse
|
12
|
Phosphorylation Regulates CAP1 (Cyclase-Associated Protein 1) Functions in the Motility and Invasion of Pancreatic Cancer Cells. Sci Rep 2019; 9:4925. [PMID: 30894654 PMCID: PMC6426867 DOI: 10.1038/s41598-019-41346-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 03/05/2019] [Indexed: 12/28/2022] Open
Abstract
Pancreatic cancer has the worst prognosis among major malignancies, largely due to its highly invasive property and difficulty in early detection. Mechanistic insights into cancerous transformation and especially metastatic progression are imperative for developing novel treatment strategies. The actin-regulating protein CAP1 is implicated in human cancers, while the role still remains elusive. In this study, we investigated roles for CAP1 and its phosphor-regulation in pancreatic cancer cells. No evidence supports remarkable up-regulation of CAP1 in the panel of cancer cell lines examined. However, knockdown of CAP1 in cancer cells led to enhanced stress fibers, reduced cell motility and invasion into Matrigel. Phosphorylation of CAP1 at the S308/S310 tandem regulatory site was elevated in cancer cells, consistent with hyper-activated GSK3 reported in pancreatic cancer. Inhibition of GSK3, a kinase for S310, reduced cell motility and invasion. Moreover, phosphor mutants had defects in alleviating actin stress fibers and rescuing the reduced invasiveness in the CAP1-knockdown PANC-1 cells. These results suggest a required role for transient phosphorylation for CAP1 function in controlling cancer cell invasiveness. Depletion of CAP1 also reduced FAK activity and cell adhesion, but did not cause significant alterations in ERK or cell proliferation. CAP1 likely regulates cancer cell invasiveness through effects on both actin filament turnover and cell adhesion. Finally, the growth factor PDGF induced CAP1 dephosphorylation, suggesting CAP1 may mediate extracellular signals to control cancer cell invasiveness. These findings may ultimately help develop strategies targeting CAP1 or its regulatory signals for controlling the invasive cycle of the disease.
Collapse
|
13
|
Cigarette smoke exposure decreases CFLAR expression in the bronchial epithelium, augmenting susceptibility for lung epithelial cell death and DAMP release. Sci Rep 2018; 8:12426. [PMID: 30127367 PMCID: PMC6102306 DOI: 10.1038/s41598-018-30602-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 07/30/2018] [Indexed: 01/29/2023] Open
Abstract
Cigarette smoking is a major risk factor for the inflammatory disease, chronic obstructive pulmonary disease (COPD). The mechanism by which cigarette smoke (CS) induces chronic lung inflammation is still largely unknown. We hypothesize that immunogenic airway epithelial cell death is involved in the initiation of the inflammatory response. We previously identified CFLAR, the gene encoding the cell death regulator protein c-FLIP, to be associated with CS-induced release of damage-associated molecular patterns (DAMPs). Here, we investigated the effect of CS on expression levels of CFLAR in bronchial biopsies from smokers and non-smokers and CFLAR transcript isoform-expression in a dataset of air-liquid interface-differentiated bronchial epithelial cells. Furthermore, CFLAR was down-regulated by siRNA in lung epithelial A549 cells, followed by investigation of the effects on apoptosis, necrosis and DAMP release. CS exposure significantly decreased CFLAR expression in bronchial epithelial cells. Moreover, we observed a shift in relative abundance of the isoforms c-FLIPS and c-FLIPL transcripts in bronchial biopsies of current smokers compared to non-smokers, consistent with a shift towards necroptosis. In vitro, down-regulation of CFLAR increased apoptosis at baseline as well as CS extract-induced necrosis and DAMP release. In conclusion, CS exposure decreases CFLAR expression, which might increase susceptibility to immunogenic cell death.
Collapse
|
14
|
Xie S, Liu Y, Li X, Tan M, Wang C, Field J, Zhou GL. Phosphorylation of the Cytoskeletal Protein CAP1 Regulates Non-Small Cell Lung Cancer Survival and Proliferation by GSK3β. J Cancer 2018; 9:2825-2833. [PMID: 30123351 PMCID: PMC6096378 DOI: 10.7150/jca.25993] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Accepted: 06/13/2018] [Indexed: 12/12/2022] Open
Abstract
Adenylate cyclase-associated protein 1 (CAP1) is an evolutionarily conserved protein that regulates actin dynamics. Our previous study indicates that CAP1 is overexpressed in NSCLC tissues and correlated with poor clinical outcomes. Further establishing the role and dissecting underlying mechanisms are imperative before targeting CAP1 can become a possibility for cancer treatment. Here we report our findings that knockdown of CAP1 inhibited cell proliferation and induced apoptosis in vitro and in vivo. Moreover, phosphor mutants of CAP1 at the S307/S309 regulatory site had compromised rescue effects for both the invasiveness and the proliferation in CAP1-knockdown cells and GSK3β kinase inhibitor LiCl inhibited cell phosphorylation site S307/S309 by up-regulating the expression of p53, BAK, BAD and cleaved PARP induced ROS production, decreased lung cancer cell viability, adhesion, proliferation, migration and invasion, and induction of apoptosis. These novel mechanistic insights may ultimately open up avenues for strategies targeting CAP1 in the treatment of lung cancer, tailored for specific types of the highly diverse disease.
Collapse
Affiliation(s)
- Shuanshuan Xie
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| | - Yang Liu
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| | - Xuan Li
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| | - Min Tan
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| | - Changhui Wang
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| | - Jeffrey Field
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Guo-Lei Zhou
- Department of Biological Sciences, Arkansas State University, State University, AR 72467, USA
| |
Collapse
|
15
|
Kakurina GV, Kolegova ES, Kondakova IV. Adenylyl Cyclase-Associated Protein 1: Structure, Regulation, and Participation in Cellular Processes. BIOCHEMISTRY (MOSCOW) 2018. [PMID: 29534668 DOI: 10.1134/s0006297918010066] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
This review summarizes information available to date about the structural organization, regulation of functional activity of adenylyl cyclase-associated protein 1 (CAP1), and its participation in cellular processes. Numerous data are generalized on the role of CAP1 in the regulation of actin cytoskeleton and its interactions with many actin-binding proteins. Attention is drawn to the similarity of the structure of CAP1 and its contribution to the remodeling of actin filaments in prokaryotes and eukaryotes, as well as to the difference in the interaction of CAP1 with adenylyl cyclase in these cells. In addition, we discuss the participation of CAP1 in various pathological processes.
Collapse
Affiliation(s)
- G V Kakurina
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, 634050, Russia.
| | | | | |
Collapse
|
16
|
Xie S, Shen C, Tan M, Li M, Song X, Wang C. Systematic analysis of gene expression alterations and clinical outcomes of adenylate cyclase-associated protein in cancer. Oncotarget 2018; 8:27216-27239. [PMID: 28423713 PMCID: PMC5432330 DOI: 10.18632/oncotarget.16111] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 02/20/2017] [Indexed: 12/21/2022] Open
Abstract
Adenylate Cyclase-associated protein (CAP) is an evolutionarily conserved protein that regulates actin dynamics. Our previous study indicates that CAP1 is overexpressed in NSCLC tissues and correlated with poor clinical outcomes, but CAP1 in HeLa cells actually inhibited migration and invasion, the role of CAP was discrepancy in different cancer types. The present study aims to determine whether CAP can serve as a prognostic marker in human cancers. The CAP expression was assessed using Oncomine database to determine the gene alteration during carcinogenesis, the copy number alteration, or mutations of CAP using cBioPortal, International Cancer Genome Consortium, and Tumorscape database investigated, and the association between CAP expression and the survival of cancer patient using Kaplan-Meier plotter and PrognoScan database evaluated. Therefore, the functional correlation between CAP expression and cancer phenotypes can be established; wherein CAP might serve as a diagnostic marker or therapeutic target for certain types of cancers.
Collapse
Affiliation(s)
- Shuanshuan Xie
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| | - Changxing Shen
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| | - Min Tan
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| | - Ming Li
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| | - Xiaolian Song
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| | - Changhui Wang
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| |
Collapse
|
17
|
Retrospective Proteomic Screening of 100 Breast Cancer Tissues. Proteomes 2017; 5:proteomes5030015. [PMID: 28686225 PMCID: PMC5620532 DOI: 10.3390/proteomes5030015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 06/20/2017] [Accepted: 07/04/2017] [Indexed: 12/18/2022] Open
Abstract
The present investigation has been conducted on one hundred tissue fragments of breast cancer, collected and immediately cryopreserved following the surgical resection. The specimens were selected from patients with invasive ductal carcinoma of the breast, the most frequent and potentially aggressive type of mammary cancer, with the objective to increase the knowledge of breast cancer molecular markers potentially useful for clinical applications. The proteomic screening; by 2D-IPG and mass spectrometry; allowed us to identify two main classes of protein clusters: proteins expressed ubiquitously at high levels in all patients; and proteins expressed sporadically among the same patients. Within the group of ubiquitous proteins, glycolytic enzymes and proteins with anti-apoptotic activity were predominant. Among the sporadic ones, proteins involved in cell motility, molecular chaperones and proteins involved in the detoxification appeared prevalent. The data of the present study indicates that the primary tumor growth is reasonably supported by concurrent events: the inhibition of apoptosis and stimulation of cellular proliferation, and the increased expression of glycolytic enzymes with multiple functions. The second phase of the evolution of the tumor can be prematurely scheduled by the occasional presence of proteins involved in cell motility and in the defenses of the oxidative stress. We suggest that this approach on large-scale 2D-IPG proteomics of breast cancer is currently a valid tool that offers the opportunity to evaluate on the same assay the presence and recurrence of individual proteins, their isoforms and short forms, to be proposed as prognostic indicators and susceptibility to metastasis in patients operated on for invasive ductal carcinoma of the breast.
Collapse
|
18
|
Cofilin-1 and Other ADF/Cofilin Superfamily Members in Human Malignant Cells. Int J Mol Sci 2016; 18:ijms18010010. [PMID: 28025492 PMCID: PMC5297645 DOI: 10.3390/ijms18010010] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 11/18/2016] [Accepted: 12/01/2016] [Indexed: 12/12/2022] Open
Abstract
Identification of actin-depolymerizing factor homology (ADF-H) domains in the structures of several related proteins led first to the formation of the ADF/cofilin family, which then expanded to the ADF/cofilin superfamily. This superfamily includes the well-studied cofilin-1 (Cfl-1) and about a dozen different human proteins that interact directly or indirectly with the actin cytoskeleton, provide its remodeling, and alter cell motility. According to some data, Cfl-1 is contained in various human malignant cells (HMCs) and is involved in the formation of malignant properties, including invasiveness, metastatic potential, and resistance to chemotherapeutic drugs. The presence of other ADF/cofilin superfamily proteins in HMCs and their involvement in the regulation of cell motility were discovered with the use of various OMICS technologies. In our review, we discuss the results of the study of Cfl-1 and other ADF/cofilin superfamily proteins, which may be of interest for solving different problems of molecular oncology, as well as for the prospects of further investigations of these proteins in HMCs.
Collapse
|
19
|
Xu L, Peng S, Huang Q, Liu Y, Jiang H, Li X, Wang J. Expression status of cyclase‑associated protein 2 as a prognostic marker for human breast cancer. Oncol Rep 2016; 36:1981-8. [PMID: 27573674 DOI: 10.3892/or.2016.5051] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 04/22/2016] [Indexed: 11/05/2022] Open
Abstract
Cyclase-associated protein 2 (CAP2) protein is reported to be upregulated in hepatocellular carcinoma (HCC). However, data regarding its expression pattern and clinical relevance in breast cancer are unknown. The aim of this study was to investigate CAP2 expression and its prognostic significance in breast cancer. CAP2 expression at the mRNA and protein levels was examined by real‑time quantitative-polymerase chain reaction and western blotting in 10 paired breast cancer tissues and adjacent normal tissues. The expression level of CAP2 protein in normal breast epithelial cells and breast cancer cell lines was quantified by western blotting. CAP2 protein expression was analyzed in paraffin‑embedded breast cancer samples, paired adjacent non‑tumor and normal breast tissues by immunohistochemical analysis. Statistical analyses were also performed to evaluate the clinicopathological significance of CAP2 expression. The results showed that the expression of CAP2 mRNA and protein was higher in breast cancer than that noted in the adjacent normal tissues in 10 paired samples. The expression level of CAP2 protein in breast cancer cell lines was higher than that in normal breast epithelial cells. In paraffin‑embedded tissue samples, the expression of CAP2 was higher in breast cancer than that found in the adjacent non‑cancerous tissues and normal breast tissues. Compared with the adjacent non‑cancerous tissues, overexpression of CAP2 was detected in 29.4% (37/126) of the patients. Overexpression of CAP2 was significantly associated with progesterone receptor (PR) expression (p<0.05), and decreased overall survival (OS) (p<0.05). In multivariate analysis, expression of CAP2 was an independent prognostic factor for OS [hazard ratio (HR), 4.821; 95% confidence interval (CI), 2.442‑9.518; p<0.001]. CAP2 is upregulated in breast cancer and is associated with the expression of PR and patient survival. CAP2 may serve as a prognostic indicator for patients with breast cancer.
Collapse
Affiliation(s)
- Lihua Xu
- Department of Hematology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510230, P.R. China
| | - Sida Peng
- Department of Hematology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510230, P.R. China
| | - Qunai Huang
- Breast Cancer Center, The Third Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Yu Liu
- Breast Cancer Center, The Third Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Hua Jiang
- Breast Cancer Center, The Third Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Xi Li
- Breast Cancer Center, The Third Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Jiani Wang
- Breast Cancer Center, The Third Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510630, P.R. China
| |
Collapse
|
20
|
Zhang H, Zhou GL. CAP1 (Cyclase-Associated Protein 1) Exerts Distinct Functions in the Proliferation and Metastatic Potential of Breast Cancer Cells Mediated by ERK. Sci Rep 2016; 6:25933. [PMID: 27173014 PMCID: PMC4865817 DOI: 10.1038/srep25933] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 04/22/2016] [Indexed: 12/14/2022] Open
Abstract
The actin-regulating protein CAP1 is implicated in the invasiveness of human cancers. However, the exact role remains elusive and controversial given lines of conflicting evidence. Moreover, a potential role in the proliferative transformation has largely been overlooked. Further establishing the role and dissecting underlying mechanisms are imperative before targeting CAP1 can become a possibility for cancer treatment. Here we report our findings that CAP1 exerts cell type-dependent functions in the invasiveness of breast cancer cells. Depletion of CAP1 in the metastatic MDA-MB-231 and BT-549 cancer cells stimulated the metastatic potential while it actually inhibited it in the non-metastatic MCF-7 cancer cells or in normal cells. Moreover, we demonstrate functions for CAP1 in cancer cell proliferation and anchorage-independent growth, again in a cell context-dependent manner. Importantly, we identify pivotal roles for the ERK-centered signaling in mediating both CAP1 functions. Phosphor mutants of CAP1 at the S307/S309 regulatory site had compromised rescue effects for both the invasiveness and proliferation in CAP1-knockdown cells, suggesting that CAP1 likely mediates upstream cell signals to control both functions. These novel mechanistic insights may ultimately open up avenues for strategies targeting CAP1 in the treatment of breast cancer, tailored for specific types of the highly diverse disease.
Collapse
Affiliation(s)
- Haitao Zhang
- Department of Biological Sciences, Arkansas State University, State University, AR 72467, USA.,Molecular Biosciences Program, Arkansas State University, State University, AR 72467, USA
| | - Guo-Lei Zhou
- Department of Biological Sciences, Arkansas State University, State University, AR 72467, USA.,Molecular Biosciences Program, Arkansas State University, State University, AR 72467, USA
| |
Collapse
|
21
|
Bao Z, Qiu X, Wang D, Ban N, Fan S, Chen W, Sun J, Xing W, Wang Y, Cui G. High expression of adenylate cyclase-associated protein 1 accelerates the proliferation, migration and invasion of neural glioma cells. Pathol Res Pract 2016; 212:264-73. [PMID: 26810579 DOI: 10.1016/j.prp.2015.12.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 10/26/2015] [Accepted: 12/31/2015] [Indexed: 11/26/2022]
Abstract
Adenylate cyclase-associated protein 1 (CAP1), a conserved member of cyclase-associated proteins was reported to be associated with the proliferation, migration or invasion of the tumors of pancreas, breast and liver, and was involved in astrocyte proliferation after acute Traumatic Brain Injury (TBI). In this study, we sought to investigate the character of CAP1 in the pathological process of human glioma by detecting human glioma specimens and cell lines. 43 of 100 specimens showed high expression of CAP1 via immunohistochemistry. With statistics analysis, we found out the expression level of CAP1 was correlated with the WHO grades of human glioma and was great positively related to Ki-67 (p<0.01). In vitro, silencing CAP1 in U251 and U87MG, the glioma cell lines with the relatively higher expression of CAP1, induced the proliferation of the cells significantly retarded, migration and invasion as well. Obviously, our results indicated that CAP1 participated in the molecular pathological process of glioma indeed, and in a certain sense, CAP1 might be a potential and promising molecular target for glioma diagnosis and therapies in the future.
Collapse
Affiliation(s)
- Zhen Bao
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, People's Republic of China
| | - Xiaojun Qiu
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, People's Republic of China
| | - Donglin Wang
- Department of Pathology, Medical College of Nantong University, Nantong, Jiangsu Province, People's Republic of China
| | - Na Ban
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong, Jiangsu Province, People's Republic of China
| | - Shaochen Fan
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong, Jiangsu Province, People's Republic of China
| | - Wenjuan Chen
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong, Jiangsu Province, People's Republic of China
| | - Jie Sun
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong, Jiangsu Province, People's Republic of China
| | - Weikang Xing
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, People's Republic of China
| | - Yunfeng Wang
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, People's Republic of China
| | - Gang Cui
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, People's Republic of China.
| |
Collapse
|
22
|
Zawadzka AM, Schilling B, Held JM, Sahu AK, Cusack MP, Drake PM, Fisher SJ, Gibson BW. Variation and quantification among a target set of phosphopeptides in human plasma by multiple reaction monitoring and SWATH-MS2 data-independent acquisition. Electrophoresis 2014; 35:3487-97. [PMID: 24853916 PMCID: PMC4565165 DOI: 10.1002/elps.201400167] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2014] [Revised: 04/26/2014] [Accepted: 05/13/2014] [Indexed: 11/07/2022]
Abstract
Human plasma contains proteins that reflect overall health and represents a rich source of proteins for identifying and understanding disease pathophysiology. However, few studies have investigated changes in plasma phosphoproteins. In addition, little is known about the normal variations in these phosphoproteins, especially with respect to specific sites of modification. To address these questions, we evaluated variability in plasma protein phosphorylation in healthy individuals using multiple reaction monitoring (MRM) and SWATH-MS2 data-independent acquisition. First, we developed a discovery workflow for phosphopeptide enrichment from plasma and identified targets for MRM assays. Next, we analyzed plasma from healthy donors using an analytical workflow consisting of MRM and SWATH-MS2 that targeted phosphopeptides from 58 and 68 phosphoproteins, respectively. These two methods produced similar results showing low variability in 13 phosphosites from 10 phosphoproteins (CVinter < 30%) and high interpersonal variation of 16 phosphosites from 14 phosphoproteins (CVinter > 30%). Moreover, these phosphopeptides originate from phosphoproteins involved in cellular processes governing homeostasis, immune response, cell-extracellular matrix interactions, lipid and sugar metabolism, and cell signaling. This limited assessment of technical and biological variability in phosphopeptides generated from plasma phosphoproteins among healthy volunteers constitutes a reference for future studies that target protein phosphorylation as biomarkers.
Collapse
Affiliation(s)
- Anna M. Zawadzka
- Buck Institute for Research on Aging, 8001 Redwood Blvd., Novato, CA 94945
| | - Birgit Schilling
- Buck Institute for Research on Aging, 8001 Redwood Blvd., Novato, CA 94945
| | - Jason M. Held
- Division of Oncology and Department of Anesthesiology, Washington University School of Medicine, Campus Box 8069, 660 S. Euclid Avenue, St. Louis, MO 63110
| | - Alexandria K. Sahu
- Buck Institute for Research on Aging, 8001 Redwood Blvd., Novato, CA 94945
| | - Michael P. Cusack
- Buck Institute for Research on Aging, 8001 Redwood Blvd., Novato, CA 94945
| | - Penelope M. Drake
- Department of Obstetrics, Gynecology and Reproductive Sciences, 513 Parnassus Ave., Box 0556, University of California San Francisco, San Francisco, CA 94143
| | - Susan J. Fisher
- Department of Obstetrics, Gynecology and Reproductive Sciences, 513 Parnassus Ave., Box 0556, University of California San Francisco, San Francisco, CA 94143
| | - Bradford W. Gibson
- Buck Institute for Research on Aging, 8001 Redwood Blvd., Novato, CA 94945
- Department of Pharmaceutical Chemistry, 513 Parnassus Ave., Box 0556, University of California San Francisco, San Francisco, CA 94143
| |
Collapse
|
23
|
Zhou GL, Zhang H, Wu H, Ghai P, Field J. Phosphorylation of the cytoskeletal protein CAP1 controls its association with cofilin and actin. J Cell Sci 2014; 127:5052-65. [PMID: 25315833 DOI: 10.1242/jcs.156059] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Cell signaling can control the dynamic balance between filamentous and monomeric actin by modulating actin regulatory proteins. One family of actin regulating proteins that controls actin dynamics comprises cyclase-associated proteins 1 and 2 (CAP1 and 2, respectively). However, cell signals that regulate CAPs remained unknown. We mapped phosphorylation sites on mouse CAP1 and found S307 and S309 to be regulatory sites. We further identified glycogen synthase kinase 3 as a kinase phosphorylating S309. The phosphomimetic mutant S307D/S309D lost binding to its partner cofilin and, when expressed in cells, caused accumulation of actin stress fibers similar to that in cells with reduced CAP expression. In contrast, the non-phosphorylatable S307A/S309A mutant showed drastically increased cofilin binding and reduced binding to actin. These results suggest that the phosphorylation serves to facilitate release of cofilin for a subsequent cycle of actin filament severing. Moreover, our results suggest that S307 and S309 function in tandem; neither the alterations in binding cofilin and/or actin, nor the defects in rescuing the phenotype of the enlarged cell size in CAP1 knockdown cells was observed in point mutants of either S307 or S309. In summary, we identify a novel regulatory mechanism of CAP1 through phosphorylation.
Collapse
Affiliation(s)
- Guo-Lei Zhou
- Department of Biological Sciences, Arkansas State University, State University, AR 72467, USA Molecular Biosciences Program, Arkansas State University, State University, AR 72467, USA
| | - Haitao Zhang
- Department of Biological Sciences, Arkansas State University, State University, AR 72467, USA Molecular Biosciences Program, Arkansas State University, State University, AR 72467, USA
| | - Huhehasi Wu
- Department of Biological Sciences, Arkansas State University, State University, AR 72467, USA
| | - Pooja Ghai
- Department of Biological Sciences, Arkansas State University, State University, AR 72467, USA Molecular Biosciences Program, Arkansas State University, State University, AR 72467, USA
| | - Jeffrey Field
- Department of Pharmacology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| |
Collapse
|