1
|
Lingappa AF, Akintunde O, Samueli E, Ewald C, Michon M, Ziari N, Lu M, Yu SF, Froehlich M, Le PU, Fernandez Y, Mallesh S, Lin J, Kitaygorodskyy A, Solas D, Reed JC, Lingappa JR, Müller-Schiffmann A, Korth C, Prasad D, Nalca A, Aston E, Fabbri B, Anand SK, Campi TW, Petrouski E, Dey D, Andrews DW, Rubenstein JL, Lingappa VR. Small molecule protein assembly modulators with pan-cancer therapeutic efficacy. Open Biol 2024; 14:240210. [PMID: 39689856 DOI: 10.1098/rsob.240210] [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: 10/03/2023] [Revised: 10/05/2024] [Accepted: 10/08/2024] [Indexed: 12/19/2024] Open
Abstract
Two structurally unrelated small molecule chemotypes, represented by compounds PAV-617 and PAV-951, with antiviral activity in cell culture against Mpox virus (formerly known as monkeypox virus) and human immunodeficiency virus (HIV) respectively, were studied for anti-cancer efficacy. Each exhibited apparent pan-cancer cytotoxicity with reasonable pharmacokinetics. Non-toxicity is demonstrated in a non-cancer cell line and in mice at doses achieving drug exposure at active concentrations. Anti-tumour properties of both chemotypes were validated in mouse xenografts against A549 human lung cancer and, for one of the chemotypes, against HT-29 colorectal cancer. The targets of these compounds are unconventional: each binds to a different transient, energy-dependent multi-protein complex. Treatment with these compounds alters the target multi-protein complexes in a manner that appears to remove a block, crucial for cancer survival and progression, on a homeostatic linkage between uncontrolled proliferation and apoptosis. These compounds provide starting points for development of novel, next-generation, non-toxic, pan-cancer therapeutics.
Collapse
Affiliation(s)
| | | | | | | | - Maya Michon
- Prosetta Biosciences, San Francisco, CA, USA
| | | | - Ming Lu
- University of California San Francisco, San Francisco, CA, USA
| | | | | | | | | | | | - Jim Lin
- Prosetta Biosciences, San Francisco, CA, USA
| | | | | | - Jonathan C Reed
- Department of Global Health, University of Washington, Seattle, WA, USA
| | - Jaisri R Lingappa
- Department of Global Health, University of Washington, Seattle, WA, USA
| | | | - Carsten Korth
- Institute of Neuropathology, Heinrich Heine University, Dusseldorf, Germany
| | | | - Aysegul Nalca
- United States Army Medical Research Institute for Infectious Diseases, Frederick, MD, USA
| | | | | | | | | | | | | | | | | | - Vishwanath R Lingappa
- Prosetta Biosciences, San Francisco, CA, USA
- University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
2
|
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
|
3
|
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: 3.0] [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
|
4
|
Obi N, Jung AY, Maurer T, Huebner M, Johnson T, Behrens S, Jaskulski S, Becher H, Chang-Claude J. Association of circulating leptin, adiponectin, and resistin concentrations with long-term breast cancer prognosis in a German patient cohort. Sci Rep 2021; 11:23526. [PMID: 34876619 PMCID: PMC8651788 DOI: 10.1038/s41598-021-02958-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 11/19/2021] [Indexed: 12/23/2022] Open
Abstract
Adipokines including leptin, adiponectin and resistin have been linked to risk of obesity-related cancers potentially through low-grade chronic inflammation pathways. We aimed to assess the role of post-diagnosis circulating adipokines on long-term prognosis in a prospective breast cancer cohort. Adipokines were measured in blood collected at baseline shortly after diagnosis (2002-2005) and at follow-up (2009) from 3112 breast cancer patients enrolled in the population-based MARIE study. Half of the patients had measurements at both time-points. All-cause mortality, breast cancer specific mortality and recurrences were ascertained up to June 2015 (11 years median follow-up). Associations with time-varying adipokine concentrations overall and stratified by estrogen and progesterone receptor (ERPR) were evaluated using adjusted proportional hazard regression. At baseline (n = 2700) and follow-up (n = 2027), median concentrations for leptin, adiponectin and resistin were 4.6 and 2.7 ng/ml, 24.4 and 30.0 mg/l, 15.4 and 26.2 ng/ml, respectively. After adjustment, there was no evidence for associations between adipokines and any outcome overall. In ERPR negative tumors, highest vs. lowest quintile of adiponectin was significantly associated with increased breast cancer specific mortality (HR 2.51, 95%CI 1.07-5.92). Overall, post-diagnosis adipokines were not associated with long-term outcomes after breast cancer. In patients with ERPR negative tumors, higher concentrations of adiponectin may be associated with increased breast cancer specific mortality and warrant further investigation.
Collapse
Affiliation(s)
- Nadia Obi
- Institute for Medical Biometry and Epidemiology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany.
| | - Audrey Y Jung
- Division of Cancer Epidemiology/Unit of Genetic Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 581, 69120, Heidelberg, Germany
| | - Tabea Maurer
- Cancer Epidemiology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marianne Huebner
- Institute for Medical Biometry and Epidemiology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
- Department of Statistics and Probability, Michigan State University, East Lansing, MI, 48824, USA
| | - Theron Johnson
- Division of Cancer Epidemiology/Unit of Genetic Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 581, 69120, Heidelberg, Germany
| | - Sabine Behrens
- Division of Cancer Epidemiology/Unit of Genetic Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 581, 69120, Heidelberg, Germany
| | - Stefanie Jaskulski
- Division of Cancer Epidemiology/Unit of Genetic Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 581, 69120, Heidelberg, Germany
- Institute for Prevention and Cancer Epidemiology, Faculty of Medicine and Medical Center, University Medical Center, University of Freiburg, Freiburg, Germany
| | - Heiko Becher
- Institute for Medical Biometry and Epidemiology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Jenny Chang-Claude
- Division of Cancer Epidemiology/Unit of Genetic Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 581, 69120, Heidelberg, Germany
- Cancer Epidemiology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
5
|
Zeng J, Li X, Liang L, Duan H, Xie S, Wang C. Phosphorylation of CAP1 regulates lung cancer proliferation, migration, and invasion. J Cancer Res Clin Oncol 2021; 148:137-153. [PMID: 34636991 PMCID: PMC8752530 DOI: 10.1007/s00432-021-03819-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 09/27/2021] [Indexed: 01/13/2023]
Abstract
Purpose Cyclase-associated protein 1 (CAP1) is a ubiquitous protein which regulates actin dynamics. Previous studies have shown that S308 and S310 are the two major phosphorylated sites in human CAP1. In the present study, we aimed to investigate the role of CAP1 phosphorylation in lung cancer. Methods Massive bioinformatics analysis was applied to determine CAP1’s role in different cancers and especially in lung cancer. Lung cancer patients’ serum and tissue were collected and analyzed in consideration of clinical background. CAP1 shRNA-lentivirus and siRNA were applied to CAP1 gene knockdown, and plasmids were constructed for CAP1 phosphorylation and de-phosphorylation. Microarray analysis was used for CAP1-associated difference analysis. Both in vitro and in vivo experiments were performed to investigate the roles of CAP1 phosphorylation and de-phosphorylation in lung cancer A549 cells. Results CAP1 is a kind of cancer-related protein. Its mRNA was overexpressed in most types of cancer tissues when compared with normal tissues. CAP1 high expression correlated with poor prognosis. Our results showed that serum CAP1 protein concentrations were significantly upregulated in non-small cell lung cancer (NSCLC) patients when compared with the healthy control group, higher serum CAP1 protein concentration correlated with shorter overall survival (OS) in NSCLC patients, and higher pCAP1 and CAP1 protein level were observed in lung cancer patients’ tumor tissue compared with adjacent normal tissue. Knockdown CAP1 in A549 cells can inhibit proliferation and migration, and the effect is validated in H1975 cells. It can also lead to an increase ratio of F-actin/G-actin. In addition, phosphorylated S308 and S310 in CAP1 promoted lung cancer cell proliferation, migration, and metastasis both in vitro and in vivo. When de-phosphorylated, these two sites in CAP1 showed the opposite effect. Phosphorylation of CAP1 can promote epithelial–mesenchymal transition (EMT). Conclusion These findings indicated that CAP1 phosphorylation can promote lung cancer proliferation, migration, and invasion. Phosphorylation sites of CAP1 might be a novel target for lung cancer treatment. Supplementary Information The online version contains supplementary material available at 10.1007/s00432-021-03819-9.
Collapse
Affiliation(s)
- Jie Zeng
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No.301, Mid Yanchang Rd, Shanghai, 200072, People's Republic of China
| | - Xuan Li
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No.301, Mid Yanchang Rd, Shanghai, 200072, People's Republic of China
| | - Long Liang
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No.301, Mid Yanchang Rd, Shanghai, 200072, People's Republic of China
| | - Hongxia Duan
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No.301, Mid Yanchang Rd, Shanghai, 200072, People's Republic of China
| | - Shuanshuan Xie
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No.301, Mid Yanchang Rd, Shanghai, 200072, People's Republic of China.
| | - Changhui Wang
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No.301, Mid Yanchang Rd, Shanghai, 200072, People's Republic of China.
| |
Collapse
|
6
|
Bergqvist M, Elebro K, Borgquist S, Rosendahl AH. Adipocytes Under Obese-Like Conditions Change Cell Cycle Distribution and Phosphorylation Profiles of Breast Cancer Cells: The Adipokine Receptor CAP1 Matters. Front Oncol 2021; 11:628653. [PMID: 33738261 PMCID: PMC7962603 DOI: 10.3389/fonc.2021.628653] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 01/22/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Obesity and associated metabolic conditions impact adipocyte functionality with potential consequences for breast cancer risk and prognosis, but contributing mechanisms remain to be understood. The adipokine receptor adenylyl cyclase-associated protein-1 (CAP1) has been implicated in the progression of breast cancer, but results are conflicting and the underlying molecular mechanisms are still unknown. In this study, molecular and cellular effects in breast cancer cells by stimulation of adipocytes under normal or obese-like conditions, and potential involvement of CAP1, were assessed. MATERIAL AND METHODS Estrogen receptor (ER)-positive T47D and ER-negative MDA-MB-231 breast cancer cells were exposed to adipocyte-secretome from adipocytes placed under pressures mimicking normal and obese-like metabolic conditions. Changes in phosphorylated kinase proteins and related biological pathways were assessed by phospho-antibody array and PANTHER analysis, cell proliferation were investigated through sulforhodamine B, cell cycle distribution by flow cytometry. Functional effects of CAP1 were subsequently examined following small interfering (si)RNA-mediated knockdown. RESULTS Protein phosphorylations involved in important biological processes were enriched in T47D breast cancer cells in response to adipocyte secretome from obese-like compared with normal conditions. The obesity-associated adipocyte secretome further stimulated cell proliferation and a shift from cell cycle G1-phase to S- and G2/M-phase was observed. Silencing of CAP1 decreased cell proliferation in both T47D and MDA-MB-231 cells, and reduced the obesity-associated secretome-induction of phosphoproteins involved in cell proliferation pathways. CONCLUSIONS These results indicate that the adipocyte secretome and CAP1 are mechanistically important for the proliferation of both ER-positive and ER-negative breast cancer cells, and potential signaling mediators were identified. These studies provide biological insight into how obesity-associated factors could affect breast cancer.
Collapse
Affiliation(s)
- Malin Bergqvist
- Department of Clinical Sciences Lund, Oncology, Lund University, Skåne University Hospital, Lund, Sweden
| | - Karin Elebro
- Department of Clinical Sciences Lund, Oncology, Lund University, Skåne University Hospital, Lund, Sweden
- Department of Clinical Sciences Malmö, Surgery, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Signe Borgquist
- Department of Clinical Sciences Lund, Oncology, Lund University, Skåne University Hospital, Lund, Sweden
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Ann H. Rosendahl
- Department of Clinical Sciences Lund, Oncology, Lund University, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
7
|
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: 4.5] [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
|
8
|
Bergqvist M, Elebro K, Sandsveden M, Borgquist S, Rosendahl AH. Effects of tumor-specific CAP1 expression and body constitution on clinical outcomes in patients with early breast cancer. Breast Cancer Res 2020; 22:67. [PMID: 32560703 PMCID: PMC7304201 DOI: 10.1186/s13058-020-01307-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Accepted: 06/04/2020] [Indexed: 12/25/2022] Open
Abstract
Background Obesity induces molecular changes that may favor tumor progression and metastatic spread, leading to impaired survival outcomes in breast cancer. Adenylate cyclase-associated protein 1 (CAP1), an actin regulatory protein and functional receptor for the obesity-associated adipokine resistin, has been implicated with inferior cancer prognosis. Here, the objective was to investigate the interplay between body composition and CAP1 tumor expression regarding breast cancer outcome through long-term survival analyses. Methods Among 718 women with primary invasive breast cancer within the large population-based prospective Malmö Diet and Cancer Study, tumor-specific CAP1 levels were assessed following thorough antibody validation and immunohistochemical staining of tumor tissue microarrays. Antibody specificity and functional application validity were determined by CAP1 gene silencing, qRT-PCR, Western immunoblotting, and cell microarray immunostaining. Kaplan-Meier and multivariable Cox proportional hazard models were used to assess survival differences in terms of breast cancer-specific survival (BCSS) and overall survival (OS) according to body composition and CAP1 expression. Results Study participants were followed for up to 25 years (median 10.9 years), during which 239 deaths were observed. Patients with low CAP1 tumor expression were older at diagnosis, displayed anthropometric measurements indicating a higher adiposity status (wider waist and hip, higher body mass index and body fat percentage), and were more prone to have unfavorable tumor characteristics (higher histological grade, higher Ki67, and estrogen receptor (ER) negativity). Overall, patients with CAP1-low tumors had impaired BCSS (adjusted hazard ratio: HRadj = 0.52, 95% CI 0.31–0.88) and OS (HRadj = 0.64, 95% CI 0.44–0.92) compared with patients having high CAP1 tumor expression. Further, analyses stratified according to different anthropometric measures or ER status showed that the CAP1-associated survival outcomes were most pronounced among patients with low adiposity status or ER-positive disease. Conclusions Low CAP1 tumor expression was associated with higher body fatness and worse survival outcomes in breast cancer patients with effect modification by adiposity and ER status. CAP1 could be a novel marker for poorer survival outcome in leaner or ER-positive breast cancer patients, highlighting the need for considering body constitution in clinical decision making.
Collapse
Affiliation(s)
- Malin Bergqvist
- Department of Clinical Sciences Lund, Oncology, Skåne University Hospital, Lund University, Lund, Sweden.
| | - Karin Elebro
- Department of Clinical Sciences Lund, Oncology, Skåne University Hospital, Lund University, Lund, Sweden.,Department of Clinical Sciences Malmö, Surgery, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Malte Sandsveden
- Department of Clinical Sciences Malmö, Surgery, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Signe Borgquist
- Department of Clinical Sciences Lund, Oncology, Skåne University Hospital, Lund University, Lund, Sweden.,Department of Oncology, Clinical Medicine, Aarhus University Hospital, Aarhus University, Aarhus, Denmark
| | - Ann H Rosendahl
- Department of Clinical Sciences Lund, Oncology, Skåne University Hospital, Lund University, Lund, Sweden.
| |
Collapse
|
9
|
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: 4] [Impact Index Per Article: 1.0] [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
|
10
|
Saker Z, Bahmad HF, Fares Y, Al Najjar Z, Saad M, Harati H, Nabha S. Prognostic impact of adenylyl cyclase-associated protein 2 (CAP2) in glioma: A clinicopathological study. Heliyon 2020; 6:e03236. [PMID: 32042970 PMCID: PMC7002826 DOI: 10.1016/j.heliyon.2020.e03236] [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: 08/13/2019] [Revised: 11/05/2019] [Accepted: 01/13/2020] [Indexed: 12/21/2022] Open
Abstract
Background Gliomas are a group of diseases arising from intracranial neoplastic tissues that produce a wide spectrum of clinicopathological features and morphological changes. Key questions that intrigue neuro-oncology researchers include defining novel oncophenotypic signatures relevant to diagnosing such tumors and predicting prognoses among patients. One of the key regulators of the cellular actin dynamics is adenylyl cyclase-associated protein 2 (CAP2), a protein that has been studied before in the milieu of cancer and shown to be associated with tumor progression; yet, its expression levels in the context of gliomas have not been assessed. Hence, we were interested in investigating CAP2 expression in gliomas and evaluating its clinicopathological and prognostic significance. Materials and methods CAP2 expression at the protein level was analyzed in 47 human paraffin-embedded gliomas and normal brain tissues by automated immunohistochemical analysis. Statistical analysis was also performed to assess CAP2 expression level in normal and tumor tissues, and to evaluate its clinicopathological and prognostic significance. Results Our results revealed high expression of CAP2 protein in tumors of gliomas compared to normal tissues and normal areas adjacent to tumors. High expression of CAP2 was also associated with advanced tumor grades among gliomas. Kaplan-Meier analysis revealed that high CAP2 expression was associated with poor prognosis of patients with glioma (P < 0.05). In Cox regression analysis, CAP2 expression was indicated as an independent prognostic factor for overall survival (hazard ratio (HR) = 1.843, 95% confidence interval (CI), 1.252-2.714; P < 0.005). Conclusion CAP2 is overexpressed in glioma and it is proposed as a potential prognostic biomarker for patients with gliomas. CAP2 expression level may serve as a promising target for diagnosis and treatment of glioma.
Collapse
Affiliation(s)
- Zahraa Saker
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Hisham F Bahmad
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon.,Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.,Faculty of Medicine, Beirut Arab University, Beirut, Lebanon
| | - Youssef Fares
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon.,Department of Neurosurgery, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Zahraa Al Najjar
- Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Mohamad Saad
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon.,Qatar Computing Research Institute, Hamad Bin Khalifa University, Doha, Qatar
| | - Hayat Harati
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Sanaa Nabha
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| |
Collapse
|
11
|
Mechanism of synergistic actin filament pointed end depolymerization by cyclase-associated protein and cofilin. Nat Commun 2019; 10:5320. [PMID: 31757941 PMCID: PMC6876575 DOI: 10.1038/s41467-019-13213-2] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 10/25/2019] [Indexed: 12/02/2022] Open
Abstract
The ability of cells to generate forces through actin filament turnover was an early adaptation in evolution. While much is known about how actin filaments grow, mechanisms of their disassembly are incompletely understood. The best-characterized actin disassembly factors are the cofilin family proteins, which increase cytoskeletal dynamics by severing actin filaments. However, the mechanism by which severed actin filaments are recycled back to monomeric form has remained enigmatic. We report that cyclase-associated-protein (CAP) works in synergy with cofilin to accelerate actin filament depolymerization by nearly 100-fold. Structural work uncovers the molecular mechanism by which CAP interacts with actin filament pointed end to destabilize the interface between terminal actin subunits, and subsequently recycles the newly-depolymerized actin monomer for the next round of filament assembly. These findings establish CAP as a molecular machine promoting rapid actin filament depolymerization and monomer recycling, and explain why CAP is critical for actin-dependent processes in all eukaryotes. The cofilin family proteins are actin disassembly factors but the disassembly mechanism is poorly understood. Here authors show that cyclase-associated-protein (CAP) works in synergy with cofilin to accelerate actin filament depolymerization by nearly 100-fold and reveal how CAP destabilizes the interface between terminal actin subunits.
Collapse
|
12
|
Mihajlovic M, Ninic A, Sopic M, Miljkovic M, Stefanovic A, Vekic J, Spasojevic-Kalimanovska V, Zeljkovic D, Trifunovic B, Stjepanovic Z, Zeljkovic A. Association among resistin, adenylate cyclase-associated protein 1 and high-density lipoprotein cholesterol in patients with colorectal cancer: a multi-marker approach, as a hallmark of innovative predictive, preventive, and personalized medicine. EPMA J 2019; 10:307-316. [PMID: 31462946 DOI: 10.1007/s13167-019-00178-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 07/10/2019] [Indexed: 12/13/2022]
Abstract
Background Elevated concentrations of resistin have been reported in colorectal cancer (CRC), but its interactions with adenylate cyclase-associated protein 1 (CAP-1) are largely unexplored. We investigated resistin plasma concentration, peripheral blood mononuclear cells (PBMCs) resistin messenger ribonucleic acid (mRNA), and CAP-1 mRNA levels in CRC patients, as well as the impact of resistin gene polymorphism rs1862513 on the examined markers. We also explored associations of resistin with high-density lipoprotein cholesterol (HDL-C) and predictive potential of our parameters for CRC. Methods Eighty-six patients with CRC and 75 healthy adults were included. Commercial ELISA kit was used for obtaining resistin's concentrations, while polymerase chain reaction (PCR) method was applied for evaluation of resistin and CAP-1 mRNA levels and rs1862513 polymorphism. Results Plasma resistin and CAP-1 mRNA levels were higher in CRC patients (p < 0.001 and p < 0.05, respectively), while resistin mRNA levels were lower (p < 0.001). Negative association existed among plasma resistin and HDL-C concentrations (ρ = - 0.280; p < 0.05). A model including age, body-mass index, HDL-C, low-density lipoprotein cholesterol (LDL-C), and plasma resistin concentrations as independent predictors of CRC showed very good diagnostic accuracy (AUC = 0.898). We found no associations of rs1862513 with the examined markers. Conclusions Our study demonstrated increased plasma resistin and CAP-1 mRNA levels, implying their possible interaction in CRC. The association among plasma resistin and HDL-C might indicate that HDL-C is involved in alterations of resistin's secretion process. As a hallmark of personalized medicine, multi-marker approach in determination of resistin-related parameters might be useful for prediction and prevention of CRC development.
Collapse
Affiliation(s)
- Marija Mihajlovic
- 1Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, POB 146, Belgrade, 11000 Serbia
| | - Ana Ninic
- 1Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, POB 146, Belgrade, 11000 Serbia
| | - Miron Sopic
- 1Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, POB 146, Belgrade, 11000 Serbia
| | - Milica Miljkovic
- 1Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, POB 146, Belgrade, 11000 Serbia
| | - Aleksandra Stefanovic
- 1Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, POB 146, Belgrade, 11000 Serbia
| | - Jelena Vekic
- 1Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, POB 146, Belgrade, 11000 Serbia
| | | | - Dejan Zeljkovic
- 2Clinic of General Surgery, Military Medical Academy, Belgrade, Serbia
| | - Bratislav Trifunovic
- 2Clinic of General Surgery, Military Medical Academy, Belgrade, Serbia
- 3Faculty of Medicine of the Military Medical Academy, University of Defence, Belgrade, Serbia
| | | | - Aleksandra Zeljkovic
- 1Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, POB 146, Belgrade, 11000 Serbia
| |
Collapse
|
13
|
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: 13] [Impact Index Per Article: 2.6] [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
|
14
|
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: 7.6] [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
|
15
|
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: 1.6] [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
|
16
|
Liu B, Xing X, Li X, Guo Q, Xu T, Xu K. ZNF259 promotes breast cancer cells invasion and migration via ERK/GSK3β/snail signaling. Cancer Manag Res 2018; 10:3159-3168. [PMID: 30214308 PMCID: PMC6126507 DOI: 10.2147/cmar.s174745] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Purpose Zinc finger protein 259 (ZNF259), also known as ZPR1, is a zinc finger-containing protein that can bind the intracellular tyrosine kinase domain of EGFR. At present, our knowledge on ZNF259 in cancers is limited. Here, we aimed to explore the biological functions of ZNF259 in breast cancer and reveal their mechanisms. Patients and methods The expression of ZNF259 was measured in 133 cases of breast cancer by immunohistochemistry. The online database Kaplan–Meier (KM) Plotter Online Tool was used to analyze the relationship between ZNF259 expression and breast cancer patient survival prognosis. Plasmid transfection and small interfering RNA and inhibitor treatments were carried out to explore the functions of ZNF259 in breast cancer cell lines and its potential mechanism. Matrigel invasion and wound healing assays were performed to detect the invasion and migration ability of cancer cells. In addition, protein expressions in tissues and cells were determined by Western blotting. Results ZNF259 expression was much higher in breast cancer cells than in the adjacent normal breast duct glandular epithelial cells (75.94% vs 7.52%, P<0.001) and was closely related to the breast cancer patients’ TNM stages (P=0.013) and lymph node metastasis (P=0.021). Knockdown of ZNF259 could downregulate p-ERK, p-GSK3β, and Snail expression, and upregulate the expression of E-cadherin and ZO-1, and then it also inhibited invasion and migration by the breast cancer cell lines MCF-7 and MDA-MB-231. Correspondingly, ZNF259 transfection could upregulate p-ERK, p-GSK3β, and Snail expression, and downregulate E-cadherin and ZO-1 expression, which led to stronger invasion and migration abilities of cancer cells. Furthermore, the ERK inhibitor U0126 could reverse all these effects induced by ZNF259 transfection. Conclusion ZNF259 could promote breast cancer cell invasion and migration by activating the ERK/GSK3β/Snail signaling pathway.
Collapse
Affiliation(s)
- Bin Liu
- Medical Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning Province, People's Republic of China
| | - Xiaojing Xing
- Medical Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning Province, People's Republic of China
| | - Xiang Li
- Breast Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning Province, People's Republic of China
| | - Qianxue Guo
- Medical Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning Province, People's Republic of China
| | - Tonghong Xu
- Medical Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning Province, People's Republic of China
| | - Ke Xu
- Thoracic Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning Province, People's Republic of China,
| |
Collapse
|
17
|
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: 1.0] [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
|
18
|
Zhang X, Cao S, Barila G, Edreira MM, Hong K, Wankhede M, Naim N, Buck M, Altschuler DL. Cyclase-associated protein 1 (CAP1) is a prenyl-binding partner of Rap1 GTPase. J Biol Chem 2018; 293:7659-7673. [PMID: 29618512 PMCID: PMC5961064 DOI: 10.1074/jbc.ra118.001779] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 02/22/2018] [Indexed: 12/20/2022] Open
Abstract
Rap1 proteins are members of the Ras subfamily of small GTPases involved in many biological responses, including adhesion, cell proliferation, and differentiation. Like all small GTPases, they work as molecular allosteric units that are active in signaling only when associated with the proper membrane compartment. Prenylation, occurring in the cytosol, is an enzymatic posttranslational event that anchors small GTPases at the membrane, and prenyl-binding proteins are needed to mask the cytoplasm-exposed lipid during transit to the target membrane. However, several of these proteins still await discovery. In this study, we report that cyclase-associated protein 1 (CAP1) binds Rap1. We found that this binding is GTP-independent, does not involve Rap1's effector domain, and is fully contained in its C-terminal hypervariable region (HVR). Furthermore, Rap1 prenylation was required for high-affinity interactions with CAP1 in a geranylgeranyl-specific manner. The prenyl binding specifically involved CAP1's C-terminal hydrophobic β-sheet domain. We present a combination of experimental and computational approaches, yielding a model whereby the high-affinity binding between Rap1 and CAP1 involves electrostatic and nonpolar side-chain interactions between Rap1's HVR residues, lipid, and CAP1 β-sheet domain. The binding was stabilized by the lipid insertion into the β-solenoid whose interior was occupied by nonpolar side chains. This model was reminiscent of the recently solved structure of the PDEδ-K-Ras complex; accordingly, disruptors of this complex, e.g. deltarasin, blocked the Rap1-CAP1 interaction. These findings indicate that CAP1 is a geranylgeranyl-binding partner of Rap1.
Collapse
Affiliation(s)
- Xuefeng Zhang
- From the Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261 and
| | - Shufen Cao
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44116
| | - Guillermo Barila
- From the Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261 and
| | - Martin M Edreira
- From the Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261 and
| | - Kyoungja Hong
- From the Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261 and
| | - Mamta Wankhede
- From the Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261 and
| | - Nyla Naim
- From the Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261 and
| | - Matthias Buck
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44116
| | - Daniel L Altschuler
- From the Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261 and
| |
Collapse
|
19
|
Muñoz-Palomeque A, Guerrero-Ramirez MA, Rubio-Chavez LA, Rosales-Gomez RC, Lopez-Cardona MG, Barajas-Avila VH, Delgadillo-Barrera A, Canton-Romero JC, Montoya-Fuentes H, Garcia-Cobian TA, Gutierrez-Rubio SA. Association of RETN and CAP1 SNPs, Expression and Serum Resistin Levels with Breast Cancer in Mexican Women. Genet Test Mol Biomarkers 2018; 22:209-217. [PMID: 29641286 DOI: 10.1089/gtmb.2017.0212] [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: 12/31/2022] Open
Abstract
BACKGROUND Breast cancer is the most common cancer in women worldwide. Approximately 70% of female breast cancer patients have a body mass index (BMI) >25. In obesity, adipose tissue secretes additional resistin, which prompts a proinflammatory effect through its action on adenylate cyclase-associated protein 1 (CAP1). Several studies have associated the RETN gene single nucleotide polymorphism (SNP) rs1862513 (-420C<G) with serum resistin levels and breast cancer. The CAP1 gene SNP rs35749351 (missense, Arg294His), located in the extracellular domain, has not previously been studied in cancer. These two SNPs, the mRNA expression levels of the two alleles for each of the cognate genes, and the serum resistin levels were compared between patients and controls to determine their association with breast cancer in Mexican women in this study. MATERIALS AND METHODS This study included 308 controls and 100 female patients with breast cancer. SNPs were detected by PCR-RFLP from DNA isolated from peripheral blood. Gene expression was performed with hydrolysis probes in tumor tissue. Resistin levels were quantified from serum samples by ELISA. RESULTS The RETN rs1862513CG/GG and CAP1 rs35749351GA/AA genotypes were associated with 1.61 and 2.193-fold increased risks of breast cancer, respectively, compared with the CC and GG genotypes. Similarly, carriers of the G allele of rs1862513 and the A allele of rs35749351, had 1.51 and 2.217-fold increased risks of breast cancer compared with the C and G alleles, respectively. The rs1862513GG/rs35749351AA genotype combination increased breast cancer risk by twofold. Serum resistin levels in postmenopausal breast cancer women were higher compared with postmenopausal controls. Tissue CAP1 expression showed differences with regard to molecular subtypes and metastases. CONCLUSION The RETN and CAP1 polymorphisms and gene expression may be potential biomarkers for breast cancer risk.
Collapse
Affiliation(s)
- Alejandrina Muñoz-Palomeque
- 1 Departamento de Fisiologia, Centro Universitario de Ciencias de la Salud , Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - Miguel Angel Guerrero-Ramirez
- 2 Unidad de Medicina Genomica y Genetica, Hospital Dr. Valentin Gomez Farias, Instituto de Seguridad y Servicios Sociales de los Trabajadores del Estado , Zapopan (ISSSTE), Jalisco, Mexico
| | - Lidia Ariadna Rubio-Chavez
- 1 Departamento de Fisiologia, Centro Universitario de Ciencias de la Salud , Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - Roberto Carlos Rosales-Gomez
- 3 Departamento de Ciencias Biomedicas, Centro Universitario de Tonala , Universidad de Guadalajara, Tonala, Jalisco, Mexico .,4 División de Medicina Molecular, Centro de Investigacion Biomedica del Occidente, IMSS Instituto Mexicano del Seguro Social , Guadalajara, Mexico
| | - Maria Guadalupe Lopez-Cardona
- 1 Departamento de Fisiologia, Centro Universitario de Ciencias de la Salud , Universidad de Guadalajara, Guadalajara, Jalisco, Mexico .,2 Unidad de Medicina Genomica y Genetica, Hospital Dr. Valentin Gomez Farias, Instituto de Seguridad y Servicios Sociales de los Trabajadores del Estado , Zapopan (ISSSTE), Jalisco, Mexico
| | - Victor Hugo Barajas-Avila
- 5 Unidad Medica de Alta Especialidad, Hospital de Ginecoobstetricia, Dr. Luis Ignacio Tellez, Centro Medico Nacional de Occidente, Instituto Mexicano del Seguro Social , Guadalajara, Mexico
| | - Alfredo Delgadillo-Barrera
- 5 Unidad Medica de Alta Especialidad, Hospital de Ginecoobstetricia, Dr. Luis Ignacio Tellez, Centro Medico Nacional de Occidente, Instituto Mexicano del Seguro Social , Guadalajara, Mexico
| | - Juan Carlos Canton-Romero
- 5 Unidad Medica de Alta Especialidad, Hospital de Ginecoobstetricia, Dr. Luis Ignacio Tellez, Centro Medico Nacional de Occidente, Instituto Mexicano del Seguro Social , Guadalajara, Mexico
| | - Hector Montoya-Fuentes
- 4 División de Medicina Molecular, Centro de Investigacion Biomedica del Occidente, IMSS Instituto Mexicano del Seguro Social , Guadalajara, Mexico
| | - Teresa Arcelia Garcia-Cobian
- 1 Departamento de Fisiologia, Centro Universitario de Ciencias de la Salud , Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - Susan Andrea Gutierrez-Rubio
- 1 Departamento de Fisiologia, Centro Universitario de Ciencias de la Salud , Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| |
Collapse
|
20
|
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: 4.7] [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
|
21
|
Rosendahl AH, Bergqvist M, Lettiero B, Kimbung S, Borgquist S. Adipocytes and Obesity-Related Conditions Jointly Promote Breast Cancer Cell Growth and Motility: Associations With CAP1 for Prognosis. Front Endocrinol (Lausanne) 2018; 9:689. [PMID: 30524378 PMCID: PMC6262006 DOI: 10.3389/fendo.2018.00689] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 11/02/2018] [Indexed: 12/04/2022] Open
Abstract
The global increase in overweight and obesity rates represent pressing public health concerns associated with severe comorbidities, amongst a rising incidence and impaired outcome of breast cancer. Yet, biological explanations for how obesity affects breast cancer are incompletely mapped. Herein, the joint impact by differentiated 3T3-L1 adipocytes and obesity-related metabolic conditions on breast cancer cells was evaluated in vitro and adipocyte-derived mediators assessed. Adipokine receptor expression was explored among breast cancer cell lines (n = 47) and primary breast tumors (n = 1,881), where associations with survival outcomes were investigated. Adipocytes and metabolic complications jointly stimulated breast cancer cell proliferation and motility, with phenotype-specific differences. Resistin was among the top modulated adipokines secreted by 3T3-L1 adipocytes under obesity-associated metabolic conditions compared with normal physiology. The newly identified resistin receptor, CAP1, was expressed across a large panel of breast cancer cell lines and primary breast tumors. CAP1 was associated with poor tumor characteristics with higher CAP1 expression among estrogen receptor (ER)-negative tumors, relative to ER-positive tumors (P = 0.025), and higher histological grades (P = 0.016). High CAP1 tumor expression was associated with shorter overall survival (adjusted hazard ratio [HRadj] 1.54; 95% confidence interval [CI], 1.11-2.13) and relapse-free survival (HRadj 1.47; 95% CI, 1.10-1.96), compared with low or intermediate CAP1 expression, particularly among ER-positive tumors or lymph node positive tumors. Together, these translational data demonstrate that the adipocyte secretome promote breast cancer cell proliferation and motility and highlight a potential role of CAP1 regarding breast cancer outcome-results that warrant further investigation to elucidate the obesity-breast cancer link in human pathology.
Collapse
Affiliation(s)
- Ann H. Rosendahl
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Lund University and Skåne University Hospital, Lund, Sweden
- *Correspondence: Ann H. Rosendahl
| | - Malin Bergqvist
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Lund University and Skåne University Hospital, Lund, Sweden
| | - Barbara Lettiero
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Lund University and Skåne University Hospital, Lund, Sweden
| | - Siker Kimbung
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Lund University and Skåne University Hospital, Lund, Sweden
| | - Signe Borgquist
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Lund University and Skåne University Hospital, Lund, Sweden
- Departments of Clinical Medicine/Oncology, Aarhus University and Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
22
|
Iwase S, Ono S. Conserved hydrophobic residues in the CARP/β-sheet domain of cyclase-associated protein are involved in actin monomer regulation. Cytoskeleton (Hoboken) 2017; 74:343-355. [PMID: 28696540 DOI: 10.1002/cm.21385] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 07/05/2017] [Accepted: 07/07/2017] [Indexed: 01/12/2023]
Abstract
Cyclase-associated protein (CAP) is a multidomain protein that promotes actin filament dynamics. The C-terminal region of CAP contains a CAP and X-linked retinitis pigmentosa 2 protein (CARP) domain (or a β-sheet domain), which binds to actin monomer and is essential for enhancing exchange of actin-bound nucleotides. However, how the CARP domain binds to actin is not clearly understood. Here, we report that conserved hydrophobic residues in the CARP domain play important roles in the function of CAP to regulate actin dynamics. Single mutations of three conserved surface-exposed hydrophobic residues in the CARP domain of CAS-2, a Caenorhabditis elegans CAP, significantly reduce its binding to actin monomers and suppress its nucleotide exchange activity on actin. As a result, these mutants are weaker than wild-type to compete with ADF/cofilin to promote recycling of actin monomers for polymerization. A double mutation (V367A/I373A) eliminates these actin-regulatory functions of CAS-2. These hydrophobic residues and previously identified functional residues are scattered on a concave β-sheet of the CARP domain, suggesting that a wide area of the β-sheet is involved in binding to actin. These observations suggest that the CARP domain of CAP binds to actin in a distinct manner from other known actin-binding proteins.
Collapse
Affiliation(s)
- Shohei Iwase
- Department of Pathology, Winship Cancer Institute, Emory University, Atlanta, Georgia.,Department of Cell Biology, Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Shoichiro Ono
- Department of Pathology, Winship Cancer Institute, Emory University, Atlanta, Georgia.,Department of Cell Biology, Winship Cancer Institute, Emory University, Atlanta, Georgia
| |
Collapse
|
23
|
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: 1.1] [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
|
24
|
Zhu C, Zhu Q, Wu Z, Yin Y, Kang D, Lu S, Liu P. Isorhapontigenin induced cell growth inhibition and apoptosis by targeting EGFR-related pathways in prostate cancer. J Cell Physiol 2017; 233:1104-1119. [PMID: 28422286 DOI: 10.1002/jcp.25968] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Accepted: 04/18/2017] [Indexed: 12/30/2022]
Abstract
Isorhapontigenin (ISO), a naturally phytopolyphenol compound existing in Chinese herb, apples, and various vegetables, has attracted extensive interest in recent years for its diverse pharmacological characteristics. Increasing evidences reveal that ISO can inhibit cancer cell growth by induced apoptosis, however, the molecular mechanisms is not fully understood. In this study, we found for the first time that ISO apparently induced cell growth inhibition and apoptosis by targeting EGFR and its downstream signal pathways in prostate cancer (PCa) cells both in vitro and in vivo, whereas no obviously effect on normal prostate cells. From the results, we found that ISO competitively targeted EGFR with EGF and inhibited EGFR auto-phosphorylation, and then decreased the levels of p-Erk1/2, p-PI3 K, and p-AKT, and further induced down-regulation of p-FOXO1 and promoted FOXO1 nuclear translocation; and finally resulted in a significantly up-regulation of Bim/p21/27/Bax/cleaved Caspase-3/cleaved PARP-1 and a markedly down-regulation of Sp1/Bcl-2/XIAP/Cyclin D1. Moreover, our experimental data demonstrated that treatment of ISO decreased protein level of AR via both inhibiting the expression of AR gene and promoting the ubiquitination/degradation of AR proteins in proteasome. In vivo, we also found that ISO inhibited the growth of subcutaneous xenotransplanted tumor in nude mice by inducing PCa cell growth inhibition and apoptosis. Taken together, all findings here clearly implicated that EGFR-related signal pathways, including EGFR-PI3K-Akt and EGFR-Erk1/2 pathways, were involved in ISO-induced cell growth inhibition and apoptosis in PCa cells, providing a more solid theoretical basis for the application of ISO to treat patients with prostate cancer in clinic.
Collapse
Affiliation(s)
- Cuicui Zhu
- Jiangsu Province Key Laboratory for Molecular and Medicine Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, Jiangsu, China
| | - Qingyi Zhu
- Laboratory of Molecular Biology, Jiangsu Province Hospital of TCM, Nanjing, Jiangsu, China
| | - Zhaomeng Wu
- Jiangsu Province Key Laboratory for Molecular and Medicine Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, Jiangsu, China
| | - Yingying Yin
- Jiangsu Province Key Laboratory for Molecular and Medicine Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, Jiangsu, China
| | - Dan Kang
- Jiangsu Province Key Laboratory for Molecular and Medicine Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, Jiangsu, China
| | - Shan Lu
- Jiangsu Province Key Laboratory for Molecular and Medicine Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, Jiangsu, China
| | - Ping Liu
- Jiangsu Province Key Laboratory for Molecular and Medicine Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, Jiangsu, China
| |
Collapse
|
25
|
Wilop S, Chou WC, Jost E, Crysandt M, Panse J, Chuang MK, Brümmendorf TH, Wagner W, Tien HF, Kharabi Masouleh B. A three-gene expression-based risk score can refine the European LeukemiaNet AML classification. J Hematol Oncol 2016; 9:78. [PMID: 27585840 PMCID: PMC5009640 DOI: 10.1186/s13045-016-0308-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 08/24/2016] [Indexed: 12/18/2022] Open
Abstract
Background Risk stratification based on cytogenetics of acute myeloid leukemia (AML) remains imprecise. The introduction of novel genetic and epigenetic markers has helped to close this gap and increased the specificity of risk stratification, although most studies have been conducted in specific AML subpopulations. In order to overcome this limitation, we used a genome-wide approach in multiple AML populations to develop a robust prediction model for AML survival. Methods We conducted a genome-wide expression analysis of two data sets from AML patients enrolled into the AMLCG-1999 trial and from the Tumor Cancer Genome Atlas (TCGA) to develop a prognostic score to refine current risk classification and performed a validation on two data sets of the National Taiwan University Hospital (NTUH) and an independent AMLCG cohort. Results In our training set, using a stringent multi-step approach, we identified a small three-gene prognostic scoring system, named Tri-AML score (TriAS) which highly correlated with overall survival (OS). Multivariate analysis revealed TriAS to be an independent prognostic factor in all tested training and additional validation sets, even including age, current cytogenetic-based risk stratification, and three other recently developed expression-based scoring models for AML. Conclusions The Tri-AML score allows robust and clinically practical risk stratification for the outcome of AML patients. TriAS substantially refined current ELN risk stratification assigning 44.5 % of the patients into a different risk category. Electronic supplementary material The online version of this article (doi:10.1186/s13045-016-0308-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Stefan Wilop
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Wen-Chien Chou
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Edgar Jost
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Martina Crysandt
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Jens Panse
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Ming-Kai Chuang
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Tim H Brümmendorf
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Wolfgang Wagner
- Helmholtz-Institute for Biomedical Engineering, Stem Cell Biology and Cellular, Engineering, University Hospital of the RWTH Aachen, Aachen, Germany.,Institute for Biomedical Engineering - Cell Biology, University Hospital of the RWTH, Aachen, Germany
| | - Hwei-Fang Tien
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Behzad Kharabi Masouleh
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Medical Faculty, RWTH Aachen University, Aachen, Germany.
| |
Collapse
|