1
|
Wang Q, Wang X, Ye H, Yao Y, Li H, Qin X, Zhao Y, Jiang W, Xu M, Zi T, Li X, Chen X, Zhou J, Wu G. VAV2 exists in extrachromosomal circular DNA and contributes Enzalutamide resistance of prostate cancer via stabilization of AR/ARv7. Int J Biol Sci 2025; 21:2843-2863. [PMID: 40303312 PMCID: PMC12035891 DOI: 10.7150/ijbs.109271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 03/19/2025] [Indexed: 05/02/2025] Open
Abstract
Extrachromosomal circular DNAs (eccDNAs) are circular, double-stranded DNA molecules ubiquitously present across various organisms, playing a critical role in tumorigenesis and tumor progression. However, their precise contribution to prostate cancer (PCa) remains incompletely understood. To elucidate the function of eccDNAs in PCa, eccDNA sequencing and annotation were performed in PCa tissues and cell lines using Circle-seq. Amplified genes on eccDNAs were identified by cross-referencing annotated eccDNA-associated genes with those overexpressed in PCa based on TCGA data. Furthermore, eccDNA profiles were compared between Enzalutamide-sensitive and -resistant cell lines to investigate their role in resistance mechanisms. Notably, VAV2 was detected on both linear and circular DNA, as confirmed by PCR and Sanger sequencing. Functional analyses demonstrated that VAV2 overexpression promotes PCa proliferation and metastasis by activating the PAK1/AKT signaling pathway through PAK1 phosphorylation. Additionally, VAV2 contributes to Enzalutamide resistance by enhancing AR/ARv7 protein stability via reduced ubiquitination, mediated through the recruitment of the deubiquitinating enzyme USP48. These findings establish VAV2, identified through eccDNA sequencing, as a potential oncogene and a promising biomarker for PCa diagnosis and prognosis.
Collapse
Affiliation(s)
- Qinghua Wang
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Xin'an Wang
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Hanchu Ye
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Yicong Yao
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Haopeng Li
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Xin Qin
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Yan Zhao
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Wei Jiang
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Mingming Xu
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Tong Zi
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Xilei Li
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Xi Chen
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Juan Zhou
- Department of ICU, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Gang Wu
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| |
Collapse
|
2
|
Pinilla-Macua I, Surve S, Sorkin A. Cell migration signaling through the EGFR-VAV2-Rac1 pathway is sustained in endosomes. J Cell Sci 2025; 138:jcs263541. [PMID: 39744818 PMCID: PMC11828472 DOI: 10.1242/jcs.263541] [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: 09/13/2024] [Accepted: 12/16/2024] [Indexed: 01/28/2025] Open
Abstract
Ligand binding to EGFR activates Rho family GTPases, triggering actin cytoskeleton reorganization, cell migration and invasion. Activated EGFR is also rapidly endocytosed but the role of EGFR endocytosis in cell motility is poorly understood. Hence, we used live-cell microscopy imaging to demonstrate that endogenous fluorescently labeled VAV2, a guanine nucleotide exchange factor for Rho GTPases, is co-endocytosed with EGFR in genome-edited human oral squamous cell carcinoma (HSC3) cells, an in vitro model for head-and-neck cancer where VAV2 is known to promote metastasis and is associated with poor prognosis. Chemotactic migration of HSC3 cells toward an EGF gradient is found to require both VAV2 and clathrin-mediated endocytosis. Moreover, sustained activation of Rac1, a Rho family GTPase promoting cell migration and a major substrate of VAV2, also depends on clathrin. Endogenous fluorescently labeled Rac1 localizes to EGFR-containing endosomes. Altogether, our findings suggest that signaling through the EGFR-VAV2-Rac1 pathway persists in endosomes and that this endosomal signaling is required for EGFR-driven cell migration.
Collapse
Affiliation(s)
- Itziar Pinilla-Macua
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Sachin Surve
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Alexander Sorkin
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| |
Collapse
|
3
|
Zhang W, Zhuang X, Wu C, Jin Y, Xing J, Hou M, Yang W, Feng Q, Wang H. Apigenin inhibits tumor angiogenesis by hindering microvesicle biogenesis via ARHGEF1. Cancer Lett 2024; 596:216961. [PMID: 38823764 DOI: 10.1016/j.canlet.2024.216961] [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: 02/14/2024] [Revised: 05/11/2024] [Accepted: 05/13/2024] [Indexed: 06/03/2024]
Abstract
Extracellular vesicles are essential for intercellular communication and are involved in tumor progression. Inhibiting the direct release of extracellular vesicles seems to be an effective strategy in inhibiting tumor progression, but lacks of investigation. Here, we report a natural flavonoid compound, apigenin, could significantly inhibit the growth of hepatocellular carcinoma by preventing microvesicle secretion. Mechanistically, apigenin primarily targets the guanine nucleotide exchange factor ARHGEF1, inhibiting the activity of small G protein Cdc42, which is essential in regulating the release of microvesicles from tumor cells. In turn, this inhibits tumor angiogenesis related to VEGF90K transported on microvesicles, ultimately impeding tumor progression. Collectively, these findings highlight the therapeutic potential of apigenin and shed light on its anticancer mechanisms through inhibiting microvesicle biogenesis, providing a solid foundation for the refinement and practical application of apigenin.
Collapse
Affiliation(s)
- Wanying Zhang
- Cancer Research Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230036, China
| | - XiangJin Zhuang
- Cancer Research Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230036, China
| | - Chenlong Wu
- Cancer Research Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230036, China
| | - Yong Jin
- Cancer Research Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230036, China
| | - Jiayu Xing
- Cancer Research Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230036, China
| | - Mei Hou
- Cancer Research Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230036, China
| | - Wen Yang
- Cancer Research Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230036, China; International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200438, China; National Center for Liver Cancer, Naval Medical University (Second Military Medical University), Shanghai, 201805, China; Key Laboratory of Signaling Regulation and Targeting Therapy of Liver Cancer, Ministry of Education, Shanghai, 200438, China.
| | - Qiyu Feng
- Cancer Research Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230036, China; Key Laboratory of Signaling Regulation and Targeting Therapy of Liver Cancer, Ministry of Education, Shanghai, 200438, China.
| | - Hongyang Wang
- Cancer Research Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230036, China; International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200438, China; National Center for Liver Cancer, Naval Medical University (Second Military Medical University), Shanghai, 201805, China; Key Laboratory of Signaling Regulation and Targeting Therapy of Liver Cancer, Ministry of Education, Shanghai, 200438, China.
| |
Collapse
|
4
|
Nguyen TPX, Roytrakul S, Buranapraditkun S, Shuangshoti S, Kitkumthorn N, Keelawat S. Proteomics profile in encapsulated follicular patterned thyroid neoplasms. Sci Rep 2024; 14:16343. [PMID: 39013964 PMCID: PMC11252349 DOI: 10.1038/s41598-024-67079-6] [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: 03/12/2024] [Accepted: 07/08/2024] [Indexed: 07/18/2024] Open
Abstract
Diagnosing encapsulated follicular-patterned thyroid tumors like Invasive Encapsulated Follicular Variant of Papillary Thyroid Carcinoma (IEFVPTC), Non-invasive Follicular Thyroid Neoplasm with Papillary-like Nuclear Features (NIFTP), and Well-Differentiated Tumor of Uncertain Malignant Potential (WDT-UMP) remains challenging due to their morphological and molecular similarities. This study aimed to investigate the protein distinctions among these three thyroid tumors and discover biological tumorigenesis through proteomic analysis. We employed total shotgun proteome analysis allowing to discover the quantitative expression of over 1398 proteins from 12 normal thyroid tissues, 13 IEFVPTC, 11 NIFTP, and 10 WDT-UMP. Principal component analysis revealed a distinct separation of IEFVPTC and normal tissue samples, distinguishing them from the low-risk tumor group (NIFTP and WDT-UMP). IEFVPTC exhibited the highest number of differentially expressed proteins (DEPs) compared to the other tumors. No discriminatory proteins between NIFTP and WDT-UMP were identified. Moreover, DEPs in IEFVPTC were significantly associated with thyroid tumor progression pathways. Certain hub genes linked to the response of immune checkpoint inhibitor therapy, revealing the potential predictor of prognosis. In conclusion, the proteomic profile of IEFVPTC differs from that of low-risk tumors. These findings may provide valuable insights into tumor biology and offer a basis for developing novel therapeutic strategies for follicular-patterned thyroid neoplasms.
Collapse
Affiliation(s)
- Truong Phan-Xuan Nguyen
- Department of Pathology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Sittiruk Roytrakul
- Functional Proteomics Technology Laboratory, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani 12120, Thailand
| | - Supranee Buranapraditkun
- Division of Allergy and Clinical Immunology, Department of Medicine, King Chulalongkorn Memorial Hospital, Faculty of Medicine, Chulalongkorn University, Thai Red Cross Society, Bangkok, 10330, Thailand
- Center of Excellence in Thai Pediatric Gastroenterology, Hepatology and Immunology (TPGHAI), Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Shanop Shuangshoti
- Department of Pathology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
- Chulalongkorn GenePRO Center, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Nakarin Kitkumthorn
- Department of Oral Biology, Faculty of Dentistry, Mahidol University, Bangkok, 10330, Thailand.
| | - Somboon Keelawat
- Department of Pathology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand.
- Precision Pathology of Neoplasia Research Group, Department of Pathology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
5
|
Chen A, Wang K, Qi L, Hu W, Zhou B. Development of a novel prognostic signature for colorectal cancer based on angiogenesis-related genes. Heliyon 2024; 10:e33662. [PMID: 39040272 PMCID: PMC11261139 DOI: 10.1016/j.heliyon.2024.e33662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 07/24/2024] Open
Abstract
Background Colorectal cancer (CRC) is the third most common malignant tumor worldwide. Angiogenesis is closely related to tumor metastasis, which is the main cause of cancer death. Although several angiogenesis signatures have been proposed in some cancer types, no angiogenic signature has been developed to predict the prognosis and efficacy of antiangiogenic bevacizumab in CRC patients. Methods We developed a novel CRC angiogenic signature by refining seven publicly available angiogenic gene sets using least absolute shrinkage and selection operator (LASSO). Immune and stromal cells within the tumor microenvironment were compared between the high- and low-risk groups in more than 1000 CRC samples classified by calculating the risk score based on the customized angiogenic signature. The correlation of this new gene set with the efficacy of bevacizumab was also compared. Results A new prognostic-associated angiogenesis signature gene set was constructed that can divide CRC patients into two high- and low-risk groups. The high-risk angiogenic group was significantly associated with extracellular matrix organization, epithelial-mesenchymal transition (EMT), and myogenesis. In addition, the high-risk group had higher infiltration of stromal and immune cells and was more resistant to bevacizumab than the low-risk group. Conclusion Briefly, we constructed a novel angiogenic signature that can predict the prognosis of CRC patients and the efficacy of bevacizumab in treating CRC. Our results provide new insights into the relationships among angiogenesis, metastasis, and medication for CRC.
Collapse
Affiliation(s)
- Aiqin Chen
- Department of Nursing, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Kailai Wang
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Medical Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Lina Qi
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Medical Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Wangxiong Hu
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Biting Zhou
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Medical Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, China
- Department of Radiation Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| |
Collapse
|
6
|
Fernández-Parejo N, Lorenzo-Martín LF, García-Pedrero JM, Rodrigo JP, Dosil M, Bustelo XR. VAV2 orchestrates the interplay between regenerative proliferation and ribogenesis in both keratinocytes and oral squamous cell carcinoma. Sci Rep 2024; 14:4060. [PMID: 38374399 PMCID: PMC10876654 DOI: 10.1038/s41598-024-54808-0] [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: 11/22/2023] [Accepted: 02/16/2024] [Indexed: 02/21/2024] Open
Abstract
VAV2 is an activator of RHO GTPases that promotes and maintains regenerative proliferation-like states in normal keratinocytes and oral squamous cell carcinoma (OSCC) cells. Here, we demonstrate that VAV2 also regulates ribosome biogenesis in those cells, a program associated with poor prognosis of human papilloma virus-negative (HPV-) OSCC patients. Mechanistically, VAV2 regulates this process in a catalysis-dependent manner using a conserved pathway comprising the RAC1 and RHOA GTPases, the PAK and ROCK family kinases, and the c-MYC and YAP/TAZ transcription factors. This pathway directly promotes RNA polymerase I activity and synthesis of 47S pre-rRNA precursors. This process is further consolidated by the upregulation of ribosome biogenesis factors and the acquisition of the YAP/TAZ-dependent undifferentiated cell state. Finally, we show that RNA polymerase I is a therapeutic Achilles' heel for both keratinocytes and OSCC patient-derived cells endowed with high VAV2 catalytic activity. Collectively, these findings highlight the therapeutic potential of modulating VAV2 and the ribosome biogenesis pathways in both preneoplastic and late progression stages of OSCC.
Collapse
Affiliation(s)
- Natalia Fernández-Parejo
- Centro de Investigación del Cáncer and Instituto de Biología Molecular del Cáncer, CSIC and Universidad de Salamanca, 37007, Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer, 28029, Madrid, Spain
| | - L Francisco Lorenzo-Martín
- Centro de Investigación del Cáncer and Instituto de Biología Molecular del Cáncer, CSIC and Universidad de Salamanca, 37007, Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer, 28029, Madrid, Spain
- Laboratory of Stem Cell Bioengineering, École Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland
| | - Juana M García-Pedrero
- Centro de Investigación Biomédica en Red de Cáncer, 28029, Madrid, Spain
- Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, 33011, Oviedo, Spain
| | - Juan P Rodrigo
- Centro de Investigación Biomédica en Red de Cáncer, 28029, Madrid, Spain
- Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, 33011, Oviedo, Spain
| | - Mercedes Dosil
- Centro de Investigación del Cáncer and Instituto de Biología Molecular del Cáncer, CSIC and Universidad de Salamanca, 37007, Salamanca, Spain.
- Centro de Investigación Biomédica en Red de Cáncer, 28029, Madrid, Spain.
| | - Xosé R Bustelo
- Centro de Investigación del Cáncer and Instituto de Biología Molecular del Cáncer, CSIC and Universidad de Salamanca, 37007, Salamanca, Spain.
- Centro de Investigación Biomédica en Red de Cáncer, 28029, Madrid, Spain.
| |
Collapse
|
7
|
Chen Y, Wu Y, Feng W, Luo X, Xiao B, Ding X, Gu Y, Lu Y, Yu Y. Vav2 promotes ductus arteriosus anatomic closure via the remodeling of smooth muscle cells by Rac1 activation. J Mol Med (Berl) 2023; 101:1567-1585. [PMID: 37804474 DOI: 10.1007/s00109-023-02377-6] [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: 03/13/2023] [Revised: 08/16/2023] [Accepted: 09/18/2023] [Indexed: 10/09/2023]
Abstract
The ductus arteriosus (DA), bridging the aorta and pulmonary artery, immediately starts closing after birth. Remodeling of DA leads to anatomic obstruction to prevent repatency. Several histological changes, especially extracellular matrices (ECMs) deposition and smooth muscle cells (SMCs) migration bring to anatomic closure. The genetic etiology and mechanism of DA closure remain elusive. We have previously reported a novel copy number variant containing Vav2 in patent ductus arteriosus (PDA) patients, but its specific role in DA closure remains unknown. The present study revealed that the expression of Vav2 was reduced in human patent DA, and it was less enrichment in the adjacent aorta. Matrigel experiments demonstrated that Vav2 could promote SMC migration from PDA patient explants. Smooth muscle cells with Vav2 overexpression also presented an increased capacity in migration and downregulated contractile-related proteins. Meanwhile, SMCs with Vav2 overexpression exhibited higher expression of collagen III and lessened protein abundance of lysyl oxidase, and both changes are beneficial to DA remodeling. Overexpression of Vav2 resulted in increased activity of Rac1, Cdc42, and RhoA in SMCs. Further investigation noteworthily found that the above alterations caused by Vav2 overexpression were particularly reversed by Rac1 inhibitor. A heterozygous, rare Vav2 variant was identified in PDA patients. Compared with the wild type, this variant attenuated Vav2 protein expression and weakened the activation of downstream Rac1, further impairing its functions in SMCs. In conclusion, Vav2 functions as an activator for Rac1 in SMCs to promote SMCs migration, dedifferentiation, and ECMs production. Deleterious variant potentially induces Vav2 loss of function, further providing possible molecular mechanisms about Vav2 in PDA pathogenesis. These findings enriched the current genetic etiology of PDA, which may provide a novel target for prenatal diagnosis and treatment. KEY MESSAGES: Although we have proposed the potential association between Vav2 and PDA incidence through whole exome sequencing, the molecular mechanisms underlying Vav2 in PDA have never been reported. This work, for the first time, demonstrated that Vav2 was exclusively expressed in closed DAs. Moreover, we found that Vav2 participated in the process of anatomic closure by mediating SMCs migration, dedifferentiation, and ECMs deposition through Rac1 activation. Our findings first identified a deleterious Vav2 c.701C>T variant that affected its function in SMCs by impairing Rac1 activation, which may lead to PDA defect. Vav2 may become an early diagnosis and an effective intervention target for PDA clinical therapy.
Collapse
Affiliation(s)
- Yinghui Chen
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200092, China
| | - Yizhuo Wu
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200092, China
| | - Weiqi Feng
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200092, China
| | - Xueyang Luo
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200092, China
| | - Bing Xiao
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200092, China
| | - Xiaowei Ding
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200092, China
| | - Yongjia Gu
- Department of Stomatology, Shidong Hospital of Yangpu District, Shanghai, 200438, China.
| | - Yanan Lu
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200092, China.
| | - Yu Yu
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200092, China.
| |
Collapse
|
8
|
Pereira M, Glogova A, Haagsma J, Stewart J, Shepherd TG, Petrik J. Mutant p53 murine oviductal epithelial cells induce progression of high-grade serous carcinoma and are most sensitive to simvastatin therapy in vitro and in vivo. J Ovarian Res 2023; 16:218. [PMID: 37986175 PMCID: PMC10662458 DOI: 10.1186/s13048-023-01307-x] [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: 02/14/2023] [Accepted: 11/02/2023] [Indexed: 11/22/2023] Open
Abstract
High-grade serous carcinoma (HGSC) is the most common and aggressive subtype of epithelial ovarian cancer, characterized by gain-of-function TP53 mutations originating in the fallopian tube epithelium. Therapeutic intervention occurs at advanced metastatic disease, due to challenges in early-stage diagnosis, with common disease recurrence and therapy resistance despite initial therapy success. The mevalonate pathway is exploited by many cancers and is potently inhibited by statin drugs. Statins have shown anti-cancer activity in many, but not all cancers. Here, we investigated the role of p53 status in relation to mevalonate pathway signaling in murine oviductal epithelial (OVE) cells and identified OVE cell sensitivity to statin inhibition. We found that p53R175H mutant and Trp53 knockout OVE cells have increased mevalonate pathway signaling compared to p53 wild-type OVE cells. Through orthotopic implantation to replicate the fallopian tube origin of HGSC, p53R175H mutant cells upregulated the mevalonate pathway to drive progression to advanced-stage ovarian cancer, and simvastatin treatment abrogated this effect. Additionally, simvastatin was more efficacious at inhibiting cell metabolic activity in OVE cells than atorvastatin, rosuvastatin and pravastatin. In vitro, simvastatin demonstrated potent effects on cell proliferation, apoptosis, invasion and migration in OVE cells regardless of p53 status. In vivo, simvastatin induced ovarian cancer disease regression through decreased primary ovarian tumor weight and increased apoptosis. Simvastatin also significantly increased cytoplasmic localization of HMG-CoA reductase in ovarian tumors. Downstream of the mevalonate pathway, simvastatin had no effect on YAP or small GTPase activity. This study suggests that simvastatin can induce anti-tumor effects and could be an important inhibitor of ovarian cancer progression.
Collapse
Affiliation(s)
- Madison Pereira
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Alice Glogova
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Jacob Haagsma
- The Mary & John Knight Translational Ovarian Cancer Research Unit, London Regional Cancer Program, London, ON, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Julia Stewart
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Trevor G Shepherd
- The Mary & John Knight Translational Ovarian Cancer Research Unit, London Regional Cancer Program, London, ON, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Department of Oncology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Department of Obstetrics & Gynaecology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Jim Petrik
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, N1G 2W1, Canada.
| |
Collapse
|
9
|
Lorenzo-Martín LF, Bustelo XR. The Rho GTPase exchange factor Vav2 promotes extensive age-dependent rewiring of the hair follicle stem cell transcriptome. Front Cell Dev Biol 2023; 11:1252834. [PMID: 37822868 PMCID: PMC10562702 DOI: 10.3389/fcell.2023.1252834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 09/11/2023] [Indexed: 10/13/2023] Open
Abstract
Both the number and regenerative activity of hair follicle stem cells (HFSCs) are regulated by Vav2, a GDP/GTP exchange factor involved in the catalytic stimulation of the GTPases Rac1 and RhoA. However, whether Vav2 signaling changes in HFSCs over the mouse lifespan is not yet known. Using a mouse knock-in mouse model, we now show that the expression of a catalytically active version of Vav2 (Vav2Onc) promotes an extensive rewiring of the overall transcriptome of HFSCs, the generation of new transcription factor hubs, and the synchronization of many transcriptional programs associated with specific HFSC states and well-defined signaling pathways. Interestingly, this transcriptome rewiring is not fixed in time, as it involves the induction of 15 gene expression waves with diverse distribution patterns during the life of the animals. These expression waves are consistent with the promotion by Vav2Onc of several functional HFSC states that differ from those normally observed in wild-type HFSCs. These results further underscore the role of Vav2 in the regulation of the functional state of HFSCs. They also indicate that, unlike other Vav2-dependent biological processes, the signaling output of this exchange factor is highly contingent on age-dependent intrinsic and/or extrinsic HFSC factors that shape the final biological readouts triggered in this cell type.
Collapse
Affiliation(s)
- L. Francisco Lorenzo-Martín
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC) and University of Salamanca, Salamanca, Spain
- Instituto de Biología Molecular y Celular del Cáncer, CSIC and University of Salamanca, Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Salamanca, Spain
| | - Xosé R. Bustelo
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC) and University of Salamanca, Salamanca, Spain
- Instituto de Biología Molecular y Celular del Cáncer, CSIC and University of Salamanca, Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Salamanca, Spain
| |
Collapse
|
10
|
Clavaín L, Fernández-Pisonero I, Movilla N, Lorenzo-Martín LF, Nieto B, Abad A, García-Navas R, Llorente-González C, Sánchez-Martín M, Vicente-Manzanares M, Santos E, Alarcón B, García-Aznar JM, Dosil M, Bustelo XR. Characterization of mutant versions of the R-RAS2/TC21 GTPase found in tumors. Oncogene 2023; 42:389-405. [PMID: 36476833 PMCID: PMC9883167 DOI: 10.1038/s41388-022-02563-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 11/23/2022] [Accepted: 11/24/2022] [Indexed: 12/12/2022]
Abstract
The R-RAS2 GTP hydrolase (GTPase) (also known as TC21) has been traditionally considered quite similar to classical RAS proteins at the regulatory and signaling levels. Recently, a long-tail hotspot mutation targeting the R-RAS2/TC21 Gln72 residue (Q72L) was identified as a potent oncogenic driver. Additional point mutations were also found in other tumors at low frequencies. Despite this, little information is available regarding the transforming role of these mutant versions and their relevance for the tumorigenic properties of already-transformed cancer cells. Here, we report that many of the RRAS2 mutations found in human cancers are highly transforming when expressed in immortalized cell lines. Moreover, the expression of endogenous R-RAS2Q72L is important for maintaining optimal levels of PI3K and ERK activities as well as for the adhesion, invasiveness, proliferation, and mitochondrial respiration of ovarian and breast cancer cell lines. Endogenous R-RAS2Q72L also regulates gene expression programs linked to both cell adhesion and inflammatory/immune-related responses. Endogenous R-RAS2Q72L is also quite relevant for the in vivo tumorigenic activity of these cells. This dependency is observed even though these cancer cell lines bear concurrent gain-of-function mutations in genes encoding RAS signaling elements. Finally, we show that endogenous R-RAS2, unlike the case of classical RAS proteins, specifically localizes in focal adhesions. Collectively, these results indicate that gain-of-function mutations of R-RAS2/TC21 play roles in tumor initiation and maintenance that are not fully redundant with those regulated by classical RAS oncoproteins.
Collapse
Affiliation(s)
- Laura Clavaín
- grid.11762.330000 0001 2180 1817Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC and University of Salamanca, 37007 Salamanca, Spain ,grid.11762.330000 0001 2180 1817Instituto de Biología Molecular y Celular del Cáncer, CSIC and University of Salamanca, 37007 Salamanca, Spain ,grid.11762.330000 0001 2180 1817Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC and University of Salamanca, 37007 Salamanca, Spain
| | - Isabel Fernández-Pisonero
- grid.11762.330000 0001 2180 1817Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC and University of Salamanca, 37007 Salamanca, Spain ,grid.11762.330000 0001 2180 1817Instituto de Biología Molecular y Celular del Cáncer, CSIC and University of Salamanca, 37007 Salamanca, Spain ,grid.11762.330000 0001 2180 1817Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC and University of Salamanca, 37007 Salamanca, Spain
| | - Nieves Movilla
- grid.11205.370000 0001 2152 8769Aragon Institute of Engineering Research, Department of Mechanical Engineering, University of Zaragoza, 50018 Zaragoza, Spain
| | - L. Francisco Lorenzo-Martín
- grid.11762.330000 0001 2180 1817Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC and University of Salamanca, 37007 Salamanca, Spain ,grid.11762.330000 0001 2180 1817Instituto de Biología Molecular y Celular del Cáncer, CSIC and University of Salamanca, 37007 Salamanca, Spain ,grid.11762.330000 0001 2180 1817Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC and University of Salamanca, 37007 Salamanca, Spain
| | - Blanca Nieto
- grid.11762.330000 0001 2180 1817Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC and University of Salamanca, 37007 Salamanca, Spain ,grid.11762.330000 0001 2180 1817Instituto de Biología Molecular y Celular del Cáncer, CSIC and University of Salamanca, 37007 Salamanca, Spain
| | - Antonio Abad
- grid.11762.330000 0001 2180 1817Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC and University of Salamanca, 37007 Salamanca, Spain ,grid.11762.330000 0001 2180 1817Instituto de Biología Molecular y Celular del Cáncer, CSIC and University of Salamanca, 37007 Salamanca, Spain ,grid.11762.330000 0001 2180 1817Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC and University of Salamanca, 37007 Salamanca, Spain
| | - Rósula García-Navas
- grid.11762.330000 0001 2180 1817Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC and University of Salamanca, 37007 Salamanca, Spain ,grid.11762.330000 0001 2180 1817Instituto de Biología Molecular y Celular del Cáncer, CSIC and University of Salamanca, 37007 Salamanca, Spain ,grid.11762.330000 0001 2180 1817Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC and University of Salamanca, 37007 Salamanca, Spain
| | - Clara Llorente-González
- grid.11762.330000 0001 2180 1817Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC and University of Salamanca, 37007 Salamanca, Spain ,grid.11762.330000 0001 2180 1817Instituto de Biología Molecular y Celular del Cáncer, CSIC and University of Salamanca, 37007 Salamanca, Spain
| | - Manuel Sánchez-Martín
- grid.11762.330000 0001 2180 1817Transgenesis Facility and Nucleus Platform for Research Services, University of Salamanca, 37007 Salamanca, Spain
| | - Miguel Vicente-Manzanares
- grid.11762.330000 0001 2180 1817Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC and University of Salamanca, 37007 Salamanca, Spain ,grid.11762.330000 0001 2180 1817Instituto de Biología Molecular y Celular del Cáncer, CSIC and University of Salamanca, 37007 Salamanca, Spain
| | - Eugenio Santos
- grid.11762.330000 0001 2180 1817Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC and University of Salamanca, 37007 Salamanca, Spain ,grid.11762.330000 0001 2180 1817Instituto de Biología Molecular y Celular del Cáncer, CSIC and University of Salamanca, 37007 Salamanca, Spain ,grid.11762.330000 0001 2180 1817Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC and University of Salamanca, 37007 Salamanca, Spain
| | - Balbino Alarcón
- grid.5515.40000000119578126Centro de Biología Molecular Severo Ochoa, CSIC and Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - José M. García-Aznar
- grid.11205.370000 0001 2152 8769Aragon Institute of Engineering Research, Department of Mechanical Engineering, University of Zaragoza, 50018 Zaragoza, Spain
| | - Mercedes Dosil
- grid.11762.330000 0001 2180 1817Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC and University of Salamanca, 37007 Salamanca, Spain ,grid.11762.330000 0001 2180 1817Instituto de Biología Molecular y Celular del Cáncer, CSIC and University of Salamanca, 37007 Salamanca, Spain ,grid.11762.330000 0001 2180 1817Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC and University of Salamanca, 37007 Salamanca, Spain
| | - Xosé R. Bustelo
- grid.11762.330000 0001 2180 1817Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC and University of Salamanca, 37007 Salamanca, Spain ,grid.11762.330000 0001 2180 1817Instituto de Biología Molecular y Celular del Cáncer, CSIC and University of Salamanca, 37007 Salamanca, Spain ,grid.11762.330000 0001 2180 1817Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC and University of Salamanca, 37007 Salamanca, Spain
| |
Collapse
|
11
|
Correlation between DNA Methylation and Cell Proliferation Identifies New Candidate Predictive Markers in Meningioma. Cancers (Basel) 2022; 14:cancers14246227. [PMID: 36551712 PMCID: PMC9776514 DOI: 10.3390/cancers14246227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/05/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Meningiomas are the most common primary tumors of the central nervous system. Based on the 2021 WHO classification, they are classified into three grades reflecting recurrence risk and aggressiveness. However, the WHO's histopathological criteria defining these grades are somewhat subjective. Together with reliable immunohistochemical proliferation indices, other molecular markers such as those studied with genome-wide epigenetics promise to revamp the current prognostic classification. In this study, 48 meningiomas of various grades were randomly included and explored for DNA methylation with the Infinium MethylationEPIC microarray over 850k CpG sites. We conducted differential and correlative analyses on grade and several proliferation indices and markers, such as mitotic index and Ki-67 or MCM6 immunohistochemistry. We also set up Cox proportional hazard models for extensive associations between CpG methylation and survival. We identified loci highly correlated with cell growth and a targeted methylation signature of regulatory regions persistently associated with proliferation, grade, and survival. Candidate genes under the control of these regions include SMC4, ESRRG, PAX6, DOK7, VAV2, OTX1, and PCDHA-PCDHB-PCDHG, i.e., the protocadherin gene clusters. This study highlights the crucial role played by epigenetic mechanisms in shaping dysregulated cellular proliferation and provides potential biomarkers bearing prognostic and therapeutic value for the clinical management of meningioma.
Collapse
|
12
|
Rapado‐González Ó, Brea‐Iglesias J, Rodríguez‐Casanova A, Bao‐Caamano A, López‐Cedrún J, Triana‐Martínez G, Díaz‐Peña R, Santos MA, López‐López R, Muinelo‐Romay L, Martínez‐Fernández M, Díaz‐Lagares Á, Suárez‐Cunqueiro MM. Somatic mutations in tumor and plasma of locoregional recurrent and/or metastatic head and neck cancer using a next‐generation sequencing panel: A preliminary study. Cancer Med 2022; 12:6615-6622. [PMID: 36420687 PMCID: PMC10067107 DOI: 10.1002/cam4.5436] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 10/13/2022] [Accepted: 11/01/2022] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND We explore the utility of TruSight Tumor 170 panel (TST170) for detecting somatic mutations in tumor and cfDNA from locoregional recurrent and/or metastatic head and neck squamous cell carcinoma (HNSCC). METHODS Targeted NGS of tumor DNA and plasma cfDNA was performed using TST170 panel. In addition, a set of somatic mutations previously described in HNSCC were selected for validating in tumor, plasma, and saliva by digital droplet PCR. RESULTS The TST170 panel identified 13 non-synonymous somatic mutations, of which five were detected in tumoral tissue, other five in plasma cfDNA, and three in both tissue and plasma cfDNA. Of the eight somatic mutations identified in tissue, three were also identified in plasma cfDNA, showing an overall concordance rate of 37.5%. CONCLUSIONS This preliminary study shows the possibility to detect somatic mutations in tumor and plasma of HNSCC patients using a single assay that would facilitate the clinical implementation of personalized medicine in the clinic.
Collapse
Affiliation(s)
- Óscar Rapado‐González
- Department of Surgery and Medical‐Surgical Specialties, Medicine and Dentistry School Universidade de Santiago de Compostela (USC) Santiago de Compostela Spain
- Galician Precision Oncology Research Group (ONCOGAL), Medicine and Dentistry School Universidade de Santiago de Compostela (USC) Santiago de Compostela Spain
- Liquid Biopsy Analysis Unit, Translational Medical Oncology Group (ONCOMET) Health Research Institute of Santiago (IDIS) Santiago de Compostela Spain
- Centro de Investigación Biomédica en Red en Cáncer (CIBERONC) Instituto de Salud Carlos III Madrid Spain
| | - Jenifer Brea‐Iglesias
- Translational Molecular Oncology Unit, Galicia Sur Health Research Institute (IIS Galicia Sur) SERGAS‐UVIGO, Hospital Álvaro Cunqueiro Vigo Spain
| | - Aitor Rodríguez‐Casanova
- Galician Precision Oncology Research Group (ONCOGAL), Medicine and Dentistry School Universidade de Santiago de Compostela (USC) Santiago de Compostela Spain
- Epigenomics Unit, Cancer Epigenomics, Translational Medical Oncology Group (ONCOMET) Health Research Institute of Santiago (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (CHUS,SERGAS) Santiago de Compostela Spain
- Roche‐Chus Joint Unit, Translational Medical Oncology Group (ONCOMET) Health Research Institute of Santiago (IDIS) Santiago de Compostela Spain
- Universidade de Santiago de Compostela (USC) Santiago de Compostela Spain
| | - Aida Bao‐Caamano
- Galician Precision Oncology Research Group (ONCOGAL), Medicine and Dentistry School Universidade de Santiago de Compostela (USC) Santiago de Compostela Spain
- Epigenomics Unit, Cancer Epigenomics, Translational Medical Oncology Group (ONCOMET) Health Research Institute of Santiago (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (CHUS,SERGAS) Santiago de Compostela Spain
- Universidade de Santiago de Compostela (USC) Santiago de Compostela Spain
| | - José‐Luis López‐Cedrún
- Department of Oral and Maxillofacial Surgery Complexo Hospitalario Universitario de A Coruña (CHUAC, SERGAS) A Coruña Spain
| | | | - Roberto Díaz‐Peña
- Fundación Pública Galega de Medicina Xenómica, SERGAS, Grupo de Medicina Xenómica‐USC Health Research Institute of Santiago de Compostela (IDIS) Santiago de Compostela Spain
- Faculty of Health Sciences Universidad Autónoma de Chile Talca Chile
| | - María Arminda Santos
- Department of Surgery and Medical‐Surgical Specialties, Medicine and Dentistry School Universidade de Santiago de Compostela (USC) Santiago de Compostela Spain
- Department of Oral Rehabilitation Instituto Universitario de Ciências da Saúde (IUCS) Gandra Portugal
| | - Rafael López‐López
- Galician Precision Oncology Research Group (ONCOGAL), Medicine and Dentistry School Universidade de Santiago de Compostela (USC) Santiago de Compostela Spain
- Centro de Investigación Biomédica en Red en Cáncer (CIBERONC) Instituto de Salud Carlos III Madrid Spain
- Translational Medical Oncology Group (ONCOMET) Health Research Institute of Santiago (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (CHUS, SERGAS) Santiago de Compostela Spain
| | - Laura Muinelo‐Romay
- Galician Precision Oncology Research Group (ONCOGAL), Medicine and Dentistry School Universidade de Santiago de Compostela (USC) Santiago de Compostela Spain
- Liquid Biopsy Analysis Unit, Translational Medical Oncology Group (ONCOMET) Health Research Institute of Santiago (IDIS) Santiago de Compostela Spain
- Centro de Investigación Biomédica en Red en Cáncer (CIBERONC) Instituto de Salud Carlos III Madrid Spain
| | - Mónica Martínez‐Fernández
- Translational Molecular Oncology Unit, Galicia Sur Health Research Institute (IIS Galicia Sur) SERGAS‐UVIGO, Hospital Álvaro Cunqueiro Vigo Spain
| | - Ángel Díaz‐Lagares
- Galician Precision Oncology Research Group (ONCOGAL), Medicine and Dentistry School Universidade de Santiago de Compostela (USC) Santiago de Compostela Spain
- Centro de Investigación Biomédica en Red en Cáncer (CIBERONC) Instituto de Salud Carlos III Madrid Spain
- Epigenomics Unit, Cancer Epigenomics, Translational Medical Oncology Group (ONCOMET) Health Research Institute of Santiago (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (CHUS,SERGAS) Santiago de Compostela Spain
| | - María Mercedes Suárez‐Cunqueiro
- Department of Surgery and Medical‐Surgical Specialties, Medicine and Dentistry School Universidade de Santiago de Compostela (USC) Santiago de Compostela Spain
- Galician Precision Oncology Research Group (ONCOGAL), Medicine and Dentistry School Universidade de Santiago de Compostela (USC) Santiago de Compostela Spain
- Centro de Investigación Biomédica en Red en Cáncer (CIBERONC) Instituto de Salud Carlos III Madrid Spain
- Translational Medical Oncology Group (ONCOMET) Health Research Institute of Santiago (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (CHUS, SERGAS) Santiago de Compostela Spain
| |
Collapse
|
13
|
Ren J, Wu X, Shang FF, Qi Y, Tang Z, Wen C, Cao W, Cheng Q, Tan L, Chen H, Zhou HH, Zou H. The tRNA-Cys-GCA Derived tsRNAs Suppress Tumor Progression of Gliomas via Regulating VAV2. DISEASE MARKERS 2022; 2022:8708312. [PMID: 36426134 PMCID: PMC9681550 DOI: 10.1155/2022/8708312] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 09/29/2022] [Indexed: 02/03/2024]
Abstract
The tsRNAs (tRNA-derived small RNAs) are new types of small noncoding RNAs derived from tRNAs. Gliomas are well-known malignant brain tumors. The study focused on tsRNA characterizations within gliomas. Datasets processing, bioinformatics analyses, and visualizations were performed with the packages of Python and R. Cell proliferations were demonstrated via CCK8 assays and colony formation assays, and in vivo xenograft experiments. Dual-luciferase reporter assay was performed to confirm the binding of tsRNA with its targets. Via using bioinformatics approaches, the hundreds of tsRNAs with available expression abundance were identified in gliomas dataset, most of them derived from D-loop or T-loop fragments of tRNAs. Among tsRNAs derived from tRNA-Cys-GCA, tRFdb-3003a and tRFdb-3003b (tRFdb-3003a/b) were remarkably down-regulated in gliomas. The survival outcome of gliomas patients with low tRFdb-3003a/b expressions was notably worse than that of high-expression patients. In glioma cells, tRFdb-3003a could suppress cells proliferation and colony formation ability. In vivo, tRFdb-3003a suppressed the tumor growth of xenograft gliomas. Enrichment analyses displayed the tRFdb-3003a-related mRNAs were enriched in the specific GO terms, spliceosome and autophagy pathways, and three GSEA molecular signatures. Mechanically, 3'-UTR regions of VAV2 mRNA were predicted to contain the binding positions of tRFdb-3003a/b, tRFdb-3003a and tRFdb-3003b was effective to reduce the relative luciferase activity of cells with VAV2 wild-type reporter. Overexpression of tRFdb-3003a/b could down-regulated the expression levels of VAV2 protein and mRNA in glioma cells. The tRNA-Cys-GCA derived tRFdb-3003a and tRFdb-3003b might act as key player in tumor progressions of gliomas; tRFdb-3003a/b might directly bind to VAV2 and regulate VAV2 expressions in gliomas.
Collapse
Affiliation(s)
- Jian Ren
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
- Department of Neurology, The Affiliated Hospital of Chongqing Medical and Pharmaceutical College, Chongqing 401331, China
| | - Xiaoling Wu
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
- Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Fei-Fei Shang
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Yingqiang Qi
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Zhurong Tang
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Chunjie Wen
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Weiguo Cao
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Quan Cheng
- Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
| | - Lihong Tan
- Department of Neurology, The Affiliated Hospital of Chongqing Medical and Pharmaceutical College, Chongqing 401331, China
| | - Huan Chen
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Hong-Hao Zhou
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
- Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
| | - Hecun Zou
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
14
|
Ando T, Okamoto K, Shintani T, Yanamoto S, Miyauchi M, Gutkind JS, Kajiya M. Integrating Genetic Alterations and the Hippo Pathway in Head and Neck Squamous Cell Carcinoma for Future Precision Medicine. J Pers Med 2022; 12:jpm12101544. [PMID: 36294681 PMCID: PMC9604790 DOI: 10.3390/jpm12101544] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/14/2022] [Accepted: 09/15/2022] [Indexed: 11/27/2022] Open
Abstract
Genetic alterations and dysregulation of signaling pathways are indispensable for the initiation and progression of cancer. Understanding the genetic, molecular, and signaling diversities in cancer patients has driven a dynamic change in cancer therapy. Patients can select a suitable molecularly targeted therapy or immune checkpoint inhibitor based on the driver gene alterations determined by sequencing of cancer tissue. This “precision medicine” approach requires detailed elucidation of the mechanisms connecting genetic alterations of driver genes and aberrant downstream signaling pathways. The regulatory mechanisms of the Hippo pathway and Yes-associated protein/transcriptional co-activator with PDZ binding motif (YAP/TAZ) that have central roles in cancer cell proliferation are not fully understood, reflecting their recent discovery. Nevertheless, emerging evidence has shown that various genetic alterations dysregulate the Hippo pathway and hyperactivate YAP/TAZ in cancers, including head and neck squamous cell carcinoma (HNSCC). Here, we summarize the latest evidence linking genetic alterations and the Hippo pathway in HNSCC, with the aim of contributing to the continued development of precision medicine.
Collapse
Affiliation(s)
- Toshinori Ando
- Center of Oral Clinical Examination, Hiroshima University Hospital, Hiroshima 734-8551, Japan
- Correspondence: ; Tel.: +81-82-257-5727
| | - Kento Okamoto
- Department of Oral Oncology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Tomoaki Shintani
- Center of Oral Clinical Examination, Hiroshima University Hospital, Hiroshima 734-8551, Japan
| | - Souichi Yanamoto
- Department of Oral Oncology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Mutsumi Miyauchi
- Department of Oral and Maxillofacial Pathobiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - J. Silvio Gutkind
- Moores Cancer Center, University of California, San Diego, CA 92093, USA
- Department of Pharmacology, University of California, San Diego, CA 92093, USA
| | - Mikihito Kajiya
- Center of Oral Clinical Examination, Hiroshima University Hospital, Hiroshima 734-8551, Japan
| |
Collapse
|
15
|
Gu J, Zhang X, Jiang G, Li Q, Wang E, Yu J. ARHGEF40 promotes non-small cell lung cancer proliferation and invasion via the AKT-Wnt axis by binding to RhoA. Mol Carcinog 2022; 61:1016-1030. [PMID: 36000254 DOI: 10.1002/mc.23457] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 05/06/2022] [Accepted: 05/13/2022] [Indexed: 11/11/2022]
Abstract
Rho guanine nucleotide exchange factor 40 (ARHGEF40) is a member of the Dbl-family of guanine nucleotide factor proteins. However, its expression pattern and biological function in malignant tumors, notably in nonsmall cell lung cancer (NSCLC) are currently unknown. The present study demonstrated that ARHGEF40 was highly expressed in NSCLC specimens and that its expression was significantly associated with advanced TNM stage (p < 0.001), lymph node metastasis (p = 0.002), and poor prognosis (p = 0.0056). In addition, ARHGEF40 accelerated nuclear translocation of the key component β-catenin and increased the expression levels of the Wnt signaling pathway targets c-myc, cyclin D1 and MMP7. Moreover, it promoted lung cancer cell proliferation and invasion in vitro and in vivo. To elucidate the underlying molecular mechanism, the current study demonstrated that ARHGEF40 could induce activation of the Wnt signaling pathway by increasing the phosphorylation levels of AKT and GSK3β via interaction with RhoA. Moreover, the Dbl homology (DH)-pleckstrin homology (PH) domain of ARHGEF40 was responsible for this interaction. Its deletion abolished the binding, which blocked the activation of the Wnt signaling. Taken together, the data indicated that ARHGEF40 promoted the malignant phenotype of lung cancer cells by activating the AKT-Wnt axis. This was achieved by its interaction with RhoA via the DH-PH domain. ARHGEF40 may serve as a novel target for NSCLC treatment.
Collapse
Affiliation(s)
- Jian Gu
- Department of Pathology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiupeng Zhang
- Department of Pathology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Guiyang Jiang
- Department of Pathology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Qingchang Li
- Department of Pathology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Enhua Wang
- Department of Pathology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Juanhan Yu
- Department of Pathology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
16
|
Zhang Y, Yang X, Liu Y, Ge L, Wang J, Sun X, Wu B, Wang J. Vav2 is a novel APP-interacting protein that regulates APP protein level. Sci Rep 2022; 12:12752. [PMID: 35882892 PMCID: PMC9325707 DOI: 10.1038/s41598-022-16883-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 07/18/2022] [Indexed: 11/24/2022] Open
Abstract
Amyloid precursor protein (APP) is a transmembrane protein that plays critical role in the pathogenesis of Alzheimer's disease (AD). It is also involved in many types of cancers. Increasing evidence has shown that the tyrosine phosphorylation site Y682 in the intracellular tail of APP is crucial for APP function. Here, we report that Vav2, a guanine nucleotide exchange factor (GEF) for Rho family GTPase, is a novel interaction partner of APP. We found that Vav2-SH2 domain was able to bind directly to the Y682-phosphorylated intracellular tail of APP through isothermal titration calorimetry and NMR titrating experiments. The crystal structure of Vav2-SH2 in complex with an APP-derived phosphopeptide was determined to understand the structural basis of this recognition specificity. The interaction of APP and Vav2 in a full-length manner was further confirmed in cells by GST pull-down, co-immunoprecipitation and immunofluorescence staining experiments. In addition, we found overexpression of Vav2 could inhibit APP degradation and markedly increase the protein levels of APP and its cleavage productions in 20E2 cells, and this function of Vav2 required a functional SH2 domain.
Collapse
Affiliation(s)
- Youjia Zhang
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China.,University of Science and Technology of China, Hefei, Anhui, China
| | - Xiaxin Yang
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, China.,Brain Research Institute, Qilu Hospital of Shandong University, Jinan, China
| | - Yongrui Liu
- University of Science and Technology of China, Hefei, Anhui, China
| | - Liang Ge
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China
| | - Jiarong Wang
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China
| | - Xiulian Sun
- Brain Research Institute, Qilu Hospital of Shandong University, Jinan, China. .,The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission, Qilu Hospital of Shandong University, Jinan, China. .,NHC Key Laboratory of Otorhinolaryngology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
| | - Bo Wu
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China.
| | - Junfeng Wang
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China. .,University of Science and Technology of China, Hefei, Anhui, China. .,Institute of Physical Science and Information Technology, Anhui University, Hefei, Anhui, China.
| |
Collapse
|
17
|
Lorenzo-Martín LF, Menacho-Márquez M, Fernández-Parejo N, Rodríguez-Fdez S, Pascual G, Abad A, Crespo P, Dosil M, Benitah SA, Bustelo XR. The Rho guanosine nucleotide exchange factors Vav2 and Vav3 modulate epidermal stem cell function. Oncogene 2022; 41:3341-3354. [PMID: 35534539 PMCID: PMC9187518 DOI: 10.1038/s41388-022-02341-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 04/26/2022] [Accepted: 04/27/2022] [Indexed: 12/21/2022]
Abstract
It is known that Rho GTPases control different aspects of the biology of skin stem cells (SSCs). However, little information is available on the role of their upstream regulators under normal and tumorigenic conditions in this process. To address this issue, we have used here mouse models in which the activity of guanosine nucleotide exchange factors of the Vav subfamily has been manipulated using both gain- and loss-of-function strategies. These experiments indicate that Vav2 and Vav3 regulate the number, functional status, and responsiveness of hair follicle bulge stem cells. This is linked to gene expression programs related to the reinforcement of the identity and the quiescent state of normal SSCs. By contrast, in the case of cancer stem cells, they promote transcriptomal programs associated with the identity, activation state, and cytoskeletal remodeling. These results underscore the role of these Rho exchange factors in the regulation of normal and tumor epidermal stem cells.
Collapse
Affiliation(s)
- L Francisco Lorenzo-Martín
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, 37007, Salamanca, Spain.,Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, 37007, Salamanca, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 37007, Salamanca, Spain
| | - Mauricio Menacho-Márquez
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, 37007, Salamanca, Spain.,Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, 37007, Salamanca, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 37007, Salamanca, Spain
| | - Natalia Fernández-Parejo
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, 37007, Salamanca, Spain.,Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, 37007, Salamanca, Spain
| | - Sonia Rodríguez-Fdez
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, 37007, Salamanca, Spain.,Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, 37007, Salamanca, Spain
| | | | - Antonio Abad
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, 37007, Salamanca, Spain.,Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, 37007, Salamanca, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 37007, Salamanca, Spain
| | - Piero Crespo
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 37007, Salamanca, Spain.,Instituto de Biomedicina y Biotecnología de Cantabria, CSIC-University of Cantabria, 39011, Santander, Spain
| | - Mercedes Dosil
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, 37007, Salamanca, Spain.,Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, 37007, Salamanca, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 37007, Salamanca, Spain
| | | | - Xosé R Bustelo
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, 37007, Salamanca, Spain. .,Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, 37007, Salamanca, Spain. .,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 37007, Salamanca, Spain.
| |
Collapse
|
18
|
Yokoyama Y, Iioka H, Horii A, Kondo E. Crumbs3 is expressed in oral squamous cell carcinomas and promotes cell migration and proliferation by affecting RhoA activity. Oncol Lett 2022; 23:173. [PMID: 35497937 PMCID: PMC9019858 DOI: 10.3892/ol.2022.13293] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 03/17/2022] [Indexed: 11/23/2022] Open
Abstract
Despite the recent progression of treatments, the 5-year survival rate of patients with oral squamous cell carcinoma (OSCC) is still poor. One of the most critical factors affecting prognosis is tumor metastasis. Developing novel molecular targeted therapies by analyzing the molecular pathway of OSCC metastasis is an urgent issue. The present study aimed to characterize the expression and function of crumbs3 (Crb3) in OSCC cell migration. Immunohistochemistry and immunoblotting revealed that Crb3 was expressed in tissues from patients with OSCC and OSCC cell lines. The motility of OSCC cell lines was decreased by knockdown of Crb3 without affecting proliferation. However, Crb3-knockout (KO) clones exhibited decreases in both cell migration and proliferation. The expression of epithelial-mesenchymal transition markers was not altered in Crb3-KO clones compared with parent cells. A xenograft mouse model of lung metastasis revealed that the metastatic potential of Crb3-KO clones was reduced. As seen with Crb3-KO clones, the motility of OSCC cells was decreased by treatment with inhibitors of RhoA activation. Serum-induced activation of RhoA in OSCC cells was evaluated by comparing the amount of GTP-bound RhoA using affinity matrices, revealing that RhoA activation was decreased in Crb3-KO clones. To the best of our knowledge, the present study was the first to demonstrate that Crb3 was expressed in squamous cell carcinoma tissues and promoted cell migration and proliferation, which was associated with RhoA activation in OSCC cells.
Collapse
Affiliation(s)
- Yusuke Yokoyama
- Division of Molecular and Cellular Pathology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951‑8510, Japan
| | - Hidekazu Iioka
- Division of Molecular and Cellular Pathology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951‑8510, Japan
| | - Arata Horii
- Department of Otolaryngology, Head and Neck Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951‑8510, Japan
| | - Eisaku Kondo
- Division of Molecular and Cellular Pathology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951‑8510, Japan
| |
Collapse
|
19
|
Mu D, Long S, Guo L, Liu W. High Expression of VAV Gene Family Predicts Poor Prognosis of Acute Myeloid Leukemia. Technol Cancer Res Treat 2021; 20:15330338211065877. [PMID: 34894858 PMCID: PMC8679409 DOI: 10.1177/15330338211065877] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Objectives: VAV family genes (VAV1, VAV2, and
VAV3) are associated with prognosis in various cancers;
however, they have not been evaluated in acute myeloid leukemia (AML). In this
study, the prognostic value of VAV expression in AML was evaluated by a
single-center study in combination with bioinformatics analyses.
Methods: The expression and prognostic value of VAVs in
patients with AML were investigated using various databases, including GEPIA,
CCLE, EMBL-EBI, UALCAN, cBioPortal, STRING, and DAVID. Blood samples from 35
patients with AML (non-M3 subtype) and 13 benigh individuals were collected at
our center. VAV expression levels were detected by real-time quantitative PCR
(RT-qPCR) and western blotting. Clinical data were derived from medical records.
Results: Based on data from multiple databases, the expression
levels of VAV1, VAV2, and VAV3 were significantly higher in AML than in control
tissues (P < 0.05). RT-qPCR and western blotting results
showed that VAV expression in mRNA and protein levels were
higher in patients with AML that in the control group (P <
0.05). Complete remission rates were lower and risks were higher in patients
with AML with high VAV1 expression than with low
VAV1 expression (P < 0.05). High levels
of VAV2, VAV3, and VAV1 were related to a poor overall survival, and this
relationship was significant for VAV1 (P < 0.05). High
expression levels of genes correlated with VAV1, such as
SIPA1, SH2D3C, and HMHA1
were also related to a poor prognosis in AML. Functional and pathways enrichment
analyses indicated that the contribution of the VAV family to AML may be
mediated by the NF-κB, cAMP, and other pathways. Conclusion: VAVs
were highly expressed in AML. In particular, VAV1 has prognostic value and is a
promising therapeutic target for AML.
Collapse
Affiliation(s)
- Dan Mu
- 556508Department of Pediatrics Hematology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China.,556508Children Hematological Oncology and Birth Defects Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Sili Long
- 556508Department of Pediatrics Hematology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China.,556508Children Hematological Oncology and Birth Defects Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China.,Sichuan Clinical Research Center for Birth Defects, Luzhou, Sichuan, 646000, China
| | - Ling Guo
- 556508Department of Pediatrics Hematology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China.,556508Children Hematological Oncology and Birth Defects Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China.,Sichuan Clinical Research Center for Birth Defects, Luzhou, Sichuan, 646000, China
| | - Wenjun Liu
- 556508Department of Pediatrics Hematology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China.,556508Children Hematological Oncology and Birth Defects Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China.,Sichuan Clinical Research Center for Birth Defects, Luzhou, Sichuan, 646000, China
| |
Collapse
|
20
|
Robles-Valero J, Fernández-Nevado L, Lorenzo-Martín LF, Cuadrado M, Fernández-Pisonero I, Rodríguez-Fdez S, Astorga-Simón EN, Abad A, Caloto R, Bustelo XR. Cancer-associated mutations in VAV1 trigger variegated signaling outputs and T-cell lymphomagenesis. EMBO J 2021; 40:e108125. [PMID: 34617326 PMCID: PMC8591544 DOI: 10.15252/embj.2021108125] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 09/04/2021] [Accepted: 09/09/2021] [Indexed: 12/25/2022] Open
Abstract
Mutations in VAV1, a gene that encodes a multifunctional protein important for lymphocytes, are found at different frequencies in peripheral T‐cell lymphoma (PTCL), non‐small cell lung cancer, and other tumors. However, their pathobiological significance remains unsettled. After cataloguing 51 cancer‐associated VAV1 mutations, we show here that they can be classified in five subtypes according to functional impact on the three main VAV1 signaling branches, GEF‐dependent activation of RAC1, GEF‐independent adaptor‐like, and tumor suppressor functions. These mutations target new and previously established regulatory layers of the protein, leading to quantitative and qualitative changes in VAV1 signaling output. We also demonstrate that the most frequent VAV1 mutant subtype drives PTCL formation in mice. This process requires the concurrent engagement of two downstream signaling branches that promote the chronic activation and transformation of follicular helper T cells. Collectively, these data reveal the genetic constraints associated with the lymphomagenic potential of VAV1 mutant subsets, similarities with other PTCL driver genes, and potential therapeutic vulnerabilities.
Collapse
Affiliation(s)
- Javier Robles-Valero
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, Salamanca, Spain
| | - Lucía Fernández-Nevado
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, Salamanca, Spain
| | - L Francisco Lorenzo-Martín
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, Salamanca, Spain
| | - Myriam Cuadrado
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, Salamanca, Spain
| | - Isabel Fernández-Pisonero
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, Salamanca, Spain
| | - Sonia Rodríguez-Fdez
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, Salamanca, Spain
| | - Elsa N Astorga-Simón
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, Salamanca, Spain
| | - Antonio Abad
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, Salamanca, Spain
| | - Rubén Caloto
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, Salamanca, Spain
| | - Xosé R Bustelo
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, Salamanca, Spain
| |
Collapse
|
21
|
Crosas-Molist E, Samain R, Kohlhammer L, Orgaz J, George S, Maiques O, Barcelo J, Sanz-Moreno V. RhoGTPase Signalling in Cancer Progression and Dissemination. Physiol Rev 2021; 102:455-510. [PMID: 34541899 DOI: 10.1152/physrev.00045.2020] [Citation(s) in RCA: 134] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Rho GTPases are a family of small G proteins that regulate a wide array of cellular processes related to their key roles controlling the cytoskeleton. On the other hand, cancer is a multi-step disease caused by the accumulation of genetic mutations and epigenetic alterations, from the initial stages of cancer development when cells in normal tissues undergo transformation, to the acquisition of invasive and metastatic traits, responsible for a large number of cancer related deaths. In this review, we discuss the role of Rho GTPase signalling in cancer in every step of disease progression. Rho GTPases contribute to tumour initiation and progression, by regulating proliferation and apoptosis, but also metabolism, senescence and cell stemness. Rho GTPases play a major role in cell migration, and in the metastatic process. They are also involved in interactions with the tumour microenvironment and regulate inflammation, contributing to cancer progression. After years of intensive research, we highlight the importance of relevant models in the Rho GTPase field, and we reflect on the therapeutic opportunities arising for cancer patients.
Collapse
Affiliation(s)
- Eva Crosas-Molist
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Remi Samain
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Leonie Kohlhammer
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Jose Orgaz
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom.,Instituto de Investigaciones Biomédicas 'Alberto Sols', CSIC-UAM, 28029, Madrid, Spain
| | - Samantha George
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Oscar Maiques
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Jaume Barcelo
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | | |
Collapse
|
22
|
Cuadrado M, Robles-Valero J. VAV Proteins as Double Agents in Cancer: Oncogenes with Tumor Suppressor Roles. BIOLOGY 2021; 10:biology10090888. [PMID: 34571765 PMCID: PMC8466051 DOI: 10.3390/biology10090888] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/04/2021] [Accepted: 09/07/2021] [Indexed: 01/02/2023]
Abstract
Simple Summary The role of the VAV family (comprised of VAV1, VAV2, and VAV3) in proactive pathways involved in cell transformation has been historically assumed. Indeed, the discovery of potential gain-of-function VAV1 mutations in specific tumor subtypes reinforced this functional archetype. Contrary to this paradigm, we demonstrated that VAV1 could unexpectedly act as a tumor suppressor in some in vivo contexts. In this review, we discuss recent findings in the field, where the emerging landscape is one in which GTPases and their regulators, such as VAV proteins, can exhibit tumor suppressor functions. Abstract Guanosine nucleotide exchange factors (GEFs) are responsible for catalyzing the transition of small GTPases from the inactive (GDP-bound) to the active (GTP-bound) states. RHO GEFs, including VAV proteins, play essential signaling roles in a wide variety of fundamental cellular processes and in human diseases. Although the most widespread archetype in the field is that RHO GEFs exert proactive functions in cancer, recent studies in mice and humans are providing new insights into the in vivo function of these proteins in cancer. These results suggest a more complex scenario where the role of GEFs is not so clearly defined. For example, VAV1 can unexpectedly play non-catalytic tumor suppressor functions in T-cell acute lymphoblastic leukemia (T-ALL) by controlling the levels of the active form of NOTCH1 (ICN1). This review focuses on emerging work unveiling tumor suppressor roles for these proteins that should prompt a reevaluation of the role of VAV GEF family in tumor biology.
Collapse
Affiliation(s)
- Myriam Cuadrado
- Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain;
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, 37007 Salamanca, Spain
| | - Javier Robles-Valero
- Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain;
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, 37007 Salamanca, Spain
- Correspondence:
| |
Collapse
|
23
|
Rodríguez-Fdez S, Lorenzo-Martín LF, Fabbiano S, Menacho-Márquez M, Sauzeau V, Dosil M, Bustelo XR. New Functions of Vav Family Proteins in Cardiovascular Biology, Skeletal Muscle, and the Nervous System. BIOLOGY 2021; 10:biology10090857. [PMID: 34571735 PMCID: PMC8472352 DOI: 10.3390/biology10090857] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 08/27/2021] [Accepted: 08/29/2021] [Indexed: 11/16/2022]
Abstract
Simple Summary In this review, we provide information on the role of Vav proteins, a group of signaling molecules that act as both Rho GTPase activators and adaptor molecules, in the cardiovascular system, skeletal muscle, and the nervous system. We also describe how these functions impact in other physiological and pathological processes such as sympathoregulation, blood pressure regulation, systemic metabolism, and metabolic syndrome. Abstract Vav proteins act as tyrosine phosphorylation-regulated guanosine nucleotide exchange factors for Rho GTPases and as molecular scaffolds. In mammals, this family of signaling proteins is composed of three members (Vav1, Vav2, Vav3) that work downstream of protein tyrosine kinases in a wide variety of cellular processes. Recent work with genetically modified mouse models has revealed that these proteins play key signaling roles in vascular smooth and skeletal muscle cells, specific neuronal subtypes, and glia cells. These functions, in turn, ensure the proper regulation of blood pressure levels, skeletal muscle mass, axonal wiring, and fiber myelination events as well as systemic metabolic balance. The study of these mice has also led to the discovery of new physiological interconnection among tissues that contribute to the ontogeny and progression of different pathologies such as, for example, hypertension, cardiovascular disease, and metabolic syndrome. Here, we provide an integrated view of all these new Vav family-dependent signaling and physiological functions.
Collapse
Affiliation(s)
- Sonia Rodríguez-Fdez
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain; (S.R.-F.); (L.F.L.-M.); (S.F.); (M.M.-M.); (V.S.); (M.D.)
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, 37007 Salamanca, Spain
| | - L. Francisco Lorenzo-Martín
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain; (S.R.-F.); (L.F.L.-M.); (S.F.); (M.M.-M.); (V.S.); (M.D.)
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, 37007 Salamanca, Spain
| | - Salvatore Fabbiano
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain; (S.R.-F.); (L.F.L.-M.); (S.F.); (M.M.-M.); (V.S.); (M.D.)
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain
| | - Mauricio Menacho-Márquez
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain; (S.R.-F.); (L.F.L.-M.); (S.F.); (M.M.-M.); (V.S.); (M.D.)
- Instituto de Inmunología Clínica y Experimental, CONICET, Rosario 3100, Argentina
| | - Vincent Sauzeau
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain; (S.R.-F.); (L.F.L.-M.); (S.F.); (M.M.-M.); (V.S.); (M.D.)
- Institut du Thorax, UMR1087 CNRS 6291, INSERM, Université de Nantes, 44096 Nantes, France
| | - Mercedes Dosil
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain; (S.R.-F.); (L.F.L.-M.); (S.F.); (M.M.-M.); (V.S.); (M.D.)
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, 37007 Salamanca, Spain
| | - Xosé R. Bustelo
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain; (S.R.-F.); (L.F.L.-M.); (S.F.); (M.M.-M.); (V.S.); (M.D.)
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, 37007 Salamanca, Spain
- Correspondence: ; Tel.: +34-663-194-634
| |
Collapse
|
24
|
VAV2 is required for DNA repair and implicated in cancer radiotherapy resistance. Signal Transduct Target Ther 2021; 6:322. [PMID: 34462423 PMCID: PMC8405816 DOI: 10.1038/s41392-021-00735-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 07/19/2021] [Accepted: 08/10/2021] [Indexed: 02/07/2023] Open
Abstract
Radiotherapy remains the mainstay for treatment of various types of human cancer; however, the clinical efficacy is often limited by radioresistance, in which the underlying mechanism is largely unknown. Here, using esophageal squamous cell carcinoma (ESCC) as a model, we demonstrate that guanine nucleotide exchange factor 2 (VAV2), which is overexpressed in most human cancers, plays an important role in primary and secondary radioresistance. We have discovered for the first time that VAV2 is required for the Ku70/Ku80 complex formation and participates in non-homologous end joining repair of DNA damages caused by ionizing radiation. We show that VAV2 overexpression substantially upregulates signal transducer and activator of transcription 1 (STAT1) and the STAT1 inhibitor Fludarabine can significantly promote the sensitivity of radioresistant patient-derived ESCC xenografts in vivo in mice to radiotherapy. These results shed new light on the mechanism of cancer radioresistance, which may be important for improving clinical radiotherapy.
Collapse
|
25
|
Pecora A, Laprise J, Dahmene M, Laurin M. Skin Cancers and the Contribution of Rho GTPase Signaling Networks to Their Progression. Cancers (Basel) 2021; 13:4362. [PMID: 34503171 PMCID: PMC8431333 DOI: 10.3390/cancers13174362] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/20/2021] [Accepted: 08/26/2021] [Indexed: 02/06/2023] Open
Abstract
Skin cancers are the most common cancers worldwide. Among them, melanoma, basal cell carcinoma of the skin and cutaneous squamous cell carcinoma are the three major subtypes. These cancers are characterized by different genetic perturbations even though they are similarly caused by a lifelong exposure to the sun. The main oncogenic drivers of skin cancer initiation have been known for a while, yet it remains unclear what are the molecular events that mediate their oncogenic functions and that contribute to their progression. Moreover, patients with aggressive skin cancers have been known to develop resistance to currently available treatment, which is urging us to identify new therapeutic opportunities based on a better understanding of skin cancer biology. More recently, the contribution of cytoskeletal dynamics and Rho GTPase signaling networks to the progression of skin cancers has been highlighted by several studies. In this review, we underline the various perturbations in the activity and regulation of Rho GTPase network components that contribute to skin cancer development, and we explore the emerging therapeutic opportunities that are surfacing from these studies.
Collapse
Affiliation(s)
- Alessandra Pecora
- Oncology Division, CHU de Québec–Université Laval Research Center, Québec City, QC G1V 4G2, Canada; (A.P.); (J.L.); (M.D.)
| | - Justine Laprise
- Oncology Division, CHU de Québec–Université Laval Research Center, Québec City, QC G1V 4G2, Canada; (A.P.); (J.L.); (M.D.)
| | - Manel Dahmene
- Oncology Division, CHU de Québec–Université Laval Research Center, Québec City, QC G1V 4G2, Canada; (A.P.); (J.L.); (M.D.)
| | - Mélanie Laurin
- Oncology Division, CHU de Québec–Université Laval Research Center, Québec City, QC G1V 4G2, Canada; (A.P.); (J.L.); (M.D.)
- Université Laval Cancer Research Center, Université Laval, Québec City, QC G1R 3S3, Canada
- Molecular Biology, Medical Biochemistry and Pathology Department, Faculty of Medicine, Université Laval, Québec City, QC G1V OA6, Canada
| |
Collapse
|
26
|
Baltanás FC, Mucientes-Valdivieso C, Lorenzo-Martín LF, Fernández-Parejo N, García-Navas R, Segrelles C, Calzada N, Fuentes-Mateos R, Paramio JM, Bustelo XR, Santos E. Functional Specificity of the Members of the Sos Family of Ras-GEF Activators: Novel Role of Sos2 in Control of Epidermal Stem Cell Homeostasis. Cancers (Basel) 2021; 13:cancers13092152. [PMID: 33946974 PMCID: PMC8124217 DOI: 10.3390/cancers13092152] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/26/2021] [Accepted: 04/27/2021] [Indexed: 12/18/2022] Open
Abstract
Simple Summary The Sos Ras-GEFs are known to participate in a wide range of skin-related diseases including cutaneous cancers, cardio-facio-cutaneous syndromes, or hirsutism. However, the specific functional roles played by the Sos1 and/or Sos2 family members in specific skin compartments remain largely unknown. This report aimed at precisely characterizing the specific functions played by Sos1 and/or Sos2 in keratinocytes, an essential cellular component of the skin. Our data show that Sos1 and Sos2 make overlapping contributions to both keratinocyte proliferation and survival. However, Sos1 seems to have a preferential involvement in regulating the ERK axis, whereas Sos2 seems to control the signaling output from the PI3K axis. We also uncovered an essential role of Sos2 in the control of the population of epidermal stem cells. Abstract Prior reports showed the critical requirement of Sos1 for epithelial carcinogenesis, but the specific functionalities of the homologous Sos1 and Sos2 GEFs in skin homeostasis and tumorigenesis remain unclear. Here, we characterize specific mechanistic roles played by Sos1 or Sos2 in primary mouse keratinocytes (a prevalent skin cell lineage) under different experimental conditions. Functional analyses of actively growing primary keratinocytes of relevant genotypes—WT, Sos1-KO, Sos2-KO, and Sos1/2-DKO—revealed a prevalent role of Sos1 regarding transcriptional regulation and control of RAS activation and mechanistic overlapping of Sos1 and Sos2 regarding cell proliferation and survival, with dominant contribution of Sos1 to the RAS-ERK axis and Sos2 to the RAS-PI3K/AKT axis. Sos1/2-DKO keratinocytes could not grow under 3D culture conditions, but single Sos1-KO and Sos2-KO keratinocytes were able to form pseudoepidermis structures that showed disorganized layer structure, reduced proliferation, and increased apoptosis in comparison with WT 3D cultures. Remarkably, analysis of the skin of both newborn and adult Sos2-KO mice uncovered a significant reduction of the population of stem cells located in hair follicles. These data confirm that Sos1 and Sos2 play specific, cell-autonomous functions in primary keratinocytes and reveal a novel, essential role of Sos2 in control of epidermal stem cell homeostasis.
Collapse
Affiliation(s)
- Fernando C. Baltanás
- Mechanisms of Cancer Program, Centro de Investigación del Cáncer (CIC), Instituto de Biología Molecular y Celular del Cáncer (IBMCC), University of Salamanca-CSIC, E-37007 Salamanca, Spain; (C.M.-V.); (L.F.L.-M.); (N.F.-P.); (R.G.-N.); (N.C.); (R.F.-M.); (X.R.B.)
- Mechanisms of Tumor Progression Program, CIBERONC, University of Salamanca-CSIC, E-37007 Salamanca, Spain; (C.S.); (J.M.P.)
- Correspondence: (F.C.B.); (E.S.)
| | - Cynthia Mucientes-Valdivieso
- Mechanisms of Cancer Program, Centro de Investigación del Cáncer (CIC), Instituto de Biología Molecular y Celular del Cáncer (IBMCC), University of Salamanca-CSIC, E-37007 Salamanca, Spain; (C.M.-V.); (L.F.L.-M.); (N.F.-P.); (R.G.-N.); (N.C.); (R.F.-M.); (X.R.B.)
- Mechanisms of Tumor Progression Program, CIBERONC, University of Salamanca-CSIC, E-37007 Salamanca, Spain; (C.S.); (J.M.P.)
| | - L. Francisco Lorenzo-Martín
- Mechanisms of Cancer Program, Centro de Investigación del Cáncer (CIC), Instituto de Biología Molecular y Celular del Cáncer (IBMCC), University of Salamanca-CSIC, E-37007 Salamanca, Spain; (C.M.-V.); (L.F.L.-M.); (N.F.-P.); (R.G.-N.); (N.C.); (R.F.-M.); (X.R.B.)
- Mechanisms of Tumor Progression Program, CIBERONC, University of Salamanca-CSIC, E-37007 Salamanca, Spain; (C.S.); (J.M.P.)
| | - Natalia Fernández-Parejo
- Mechanisms of Cancer Program, Centro de Investigación del Cáncer (CIC), Instituto de Biología Molecular y Celular del Cáncer (IBMCC), University of Salamanca-CSIC, E-37007 Salamanca, Spain; (C.M.-V.); (L.F.L.-M.); (N.F.-P.); (R.G.-N.); (N.C.); (R.F.-M.); (X.R.B.)
- Mechanisms of Tumor Progression Program, CIBERONC, University of Salamanca-CSIC, E-37007 Salamanca, Spain; (C.S.); (J.M.P.)
| | - Rósula García-Navas
- Mechanisms of Cancer Program, Centro de Investigación del Cáncer (CIC), Instituto de Biología Molecular y Celular del Cáncer (IBMCC), University of Salamanca-CSIC, E-37007 Salamanca, Spain; (C.M.-V.); (L.F.L.-M.); (N.F.-P.); (R.G.-N.); (N.C.); (R.F.-M.); (X.R.B.)
- Mechanisms of Tumor Progression Program, CIBERONC, University of Salamanca-CSIC, E-37007 Salamanca, Spain; (C.S.); (J.M.P.)
| | - Carmen Segrelles
- Mechanisms of Tumor Progression Program, CIBERONC, University of Salamanca-CSIC, E-37007 Salamanca, Spain; (C.S.); (J.M.P.)
- Molecular Oncology Division, CIEMAT and Instituto de Investigación Sanitaria Hospital Universitario 12 de Octubre, E-28040 Madrid, Spain
| | - Nuria Calzada
- Mechanisms of Cancer Program, Centro de Investigación del Cáncer (CIC), Instituto de Biología Molecular y Celular del Cáncer (IBMCC), University of Salamanca-CSIC, E-37007 Salamanca, Spain; (C.M.-V.); (L.F.L.-M.); (N.F.-P.); (R.G.-N.); (N.C.); (R.F.-M.); (X.R.B.)
- Mechanisms of Tumor Progression Program, CIBERONC, University of Salamanca-CSIC, E-37007 Salamanca, Spain; (C.S.); (J.M.P.)
| | - Rocío Fuentes-Mateos
- Mechanisms of Cancer Program, Centro de Investigación del Cáncer (CIC), Instituto de Biología Molecular y Celular del Cáncer (IBMCC), University of Salamanca-CSIC, E-37007 Salamanca, Spain; (C.M.-V.); (L.F.L.-M.); (N.F.-P.); (R.G.-N.); (N.C.); (R.F.-M.); (X.R.B.)
- Mechanisms of Tumor Progression Program, CIBERONC, University of Salamanca-CSIC, E-37007 Salamanca, Spain; (C.S.); (J.M.P.)
| | - Jesús M. Paramio
- Mechanisms of Tumor Progression Program, CIBERONC, University of Salamanca-CSIC, E-37007 Salamanca, Spain; (C.S.); (J.M.P.)
- Molecular Oncology Division, CIEMAT and Instituto de Investigación Sanitaria Hospital Universitario 12 de Octubre, E-28040 Madrid, Spain
| | - Xosé R. Bustelo
- Mechanisms of Cancer Program, Centro de Investigación del Cáncer (CIC), Instituto de Biología Molecular y Celular del Cáncer (IBMCC), University of Salamanca-CSIC, E-37007 Salamanca, Spain; (C.M.-V.); (L.F.L.-M.); (N.F.-P.); (R.G.-N.); (N.C.); (R.F.-M.); (X.R.B.)
- Mechanisms of Tumor Progression Program, CIBERONC, University of Salamanca-CSIC, E-37007 Salamanca, Spain; (C.S.); (J.M.P.)
| | - Eugenio Santos
- Mechanisms of Cancer Program, Centro de Investigación del Cáncer (CIC), Instituto de Biología Molecular y Celular del Cáncer (IBMCC), University of Salamanca-CSIC, E-37007 Salamanca, Spain; (C.M.-V.); (L.F.L.-M.); (N.F.-P.); (R.G.-N.); (N.C.); (R.F.-M.); (X.R.B.)
- Mechanisms of Tumor Progression Program, CIBERONC, University of Salamanca-CSIC, E-37007 Salamanca, Spain; (C.S.); (J.M.P.)
- Correspondence: (F.C.B.); (E.S.)
| |
Collapse
|
27
|
Simoneschi D, Rona G, Zhou N, Jeong YT, Jiang S, Milletti G, Arbini AA, O'Sullivan A, Wang AA, Nithikasem S, Keegan S, Siu Y, Cianfanelli V, Maiani E, Nazio F, Cecconi F, Boccalatte F, Fenyö D, Jones DR, Busino L, Pagano M. CRL4 AMBRA1 is a master regulator of D-type cyclins. Nature 2021; 592:789-793. [PMID: 33854235 DOI: 10.1038/s41586-021-03445-y] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 03/11/2021] [Indexed: 12/20/2022]
Abstract
D-type cyclins are central regulators of the cell division cycle and are among the most frequently deregulated therapeutic targets in human cancer1, but the mechanisms that regulate their turnover are still being debated2,3. Here, by combining biochemical and genetics studies in somatic cells, we identify CRL4AMBRA1 (also known as CRL4DCAF3) as the ubiquitin ligase that targets all three D-type cyclins for degradation. During development, loss of Ambra1 induces the accumulation of D-type cyclins and retinoblastoma (RB) hyperphosphorylation and hyperproliferation, and results in defects of the nervous system that are reduced by treating pregnant mice with the FDA-approved CDK4 and CDK6 (CDK4/6) inhibitor abemaciclib. Moreover, AMBRA1 acts as a tumour suppressor in mouse models and low AMBRA1 mRNA levels are predictive of poor survival in cancer patients. Cancer hotspot mutations in D-type cyclins abrogate their binding to AMBRA1 and induce their stabilization. Finally, a whole-genome, CRISPR-Cas9 screen identified AMBRA1 as a regulator of the response to CDK4/6 inhibition. Loss of AMBRA1 reduces sensitivity to CDK4/6 inhibitors by promoting the formation of complexes of D-type cyclins with CDK2. Collectively, our results reveal the molecular mechanism that controls the stability of D-type cyclins during cell-cycle progression, in development and in human cancer, and implicate AMBRA1 as a critical regulator of the RB pathway.
Collapse
Affiliation(s)
- Daniele Simoneschi
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, USA.,Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, USA
| | - Gergely Rona
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, USA.,Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, USA.,Howard Hughes Medical Institute, NYU Grossman School of Medicine, New York, NY, USA
| | - Nan Zhou
- Department of Cancer Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Yeon-Tae Jeong
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, USA.,Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, USA
| | - Shaowen Jiang
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, USA.,Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, USA
| | - Giacomo Milletti
- Department of Pediatric Hemato-Oncology and Cell and Gene Therapy, IRCCS Bambino Gesù Children's Hospital, Rome, Italy.,Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Arnaldo A Arbini
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, USA.,Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
| | - Alfie O'Sullivan
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, USA.,Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, USA
| | - Andrew A Wang
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, USA.,Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, USA
| | - Sorasicha Nithikasem
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, USA.,Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, USA
| | - Sarah Keegan
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, USA.,Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, USA.,Institute for Systems Genetics, NYU Grossman School of Medicine, New York, NY, USA
| | - Yik Siu
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, USA
| | - Valentina Cianfanelli
- Department of Pediatric Hemato-Oncology and Cell and Gene Therapy, IRCCS Bambino Gesù Children's Hospital, Rome, Italy.,Cell Stress and Survival Unit, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Emiliano Maiani
- Cell Stress and Survival Unit, Danish Cancer Society Research Center, Copenhagen, Denmark.,Computational Biology Laboratory, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Francesca Nazio
- Department of Pediatric Hemato-Oncology and Cell and Gene Therapy, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Francesco Cecconi
- Department of Pediatric Hemato-Oncology and Cell and Gene Therapy, IRCCS Bambino Gesù Children's Hospital, Rome, Italy.,Department of Biology, University of Rome Tor Vergata, Rome, Italy.,Cell Stress and Survival Unit, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Francesco Boccalatte
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, USA.,Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
| | - David Fenyö
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, USA.,Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, USA.,Institute for Systems Genetics, NYU Grossman School of Medicine, New York, NY, USA
| | - Drew R Jones
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, USA
| | - Luca Busino
- Department of Cancer Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| | - Michele Pagano
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, USA. .,Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, USA. .,Howard Hughes Medical Institute, NYU Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
28
|
Lorenzo-Martín LF, Menacho-Márquez M, Bustelo XR. Drug Vulnerabilities and Disease Prognosis Linked to the Stem Cell-Like Gene Expression Program Triggered by the RHO GTPase Activator VAV2 in Hyperplastic Keratinocytes and Head and Neck Cancer. Cancers (Basel) 2020; 12:cancers12092498. [PMID: 32899210 PMCID: PMC7563609 DOI: 10.3390/cancers12092498] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/28/2020] [Accepted: 08/31/2020] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Head and neck squamous cell carcinoma are epithelial tumors with a very poor prognosis. They are also in high need of new targeted and immune-based therapeutics to limit tumor recurrence and improve long-term survival. The poor prognosis of patients with head and neck tumors is usually associated with histological features associated with poor differentiation and high proliferative activity found in their tumor biopsies. Therefore, it is of paramount importance to identify vulnerabilities associated with such pathobiological programs. In this work, the authors utilize a stem cell-like program linked to the deregulated activity of VAV2, a protein frequently overexpressed in this type of tumors, to identify new therapeutic targets that can discriminate tumors from healthy cells. The authors also show that this gene expression program can be used to stratify patients according to long-term prognosis. Abstract We have recently shown that VAV2, a guanosine nucleotide exchange factor that catalyzes the stimulation step of RHO GTPases, is involved in a stem cell-like (SCL) regenerative proliferation program that is important for the development and subsequent maintenance of the tumorigenesis of both cutaneous (cSCC) and head and neck squamous cell carcinomas (hnSCC). In line with this, we have observed that the levels of the VAV2 mRNA and VAV2-regulated gene signatures are associated with poor prognosis in the case of human papillomavirus-negative hnSCC patients. These results suggest that the SCL program elicited by VAV2 in those cells can harbor therapeutically actionable downstream targets. We have addressed this issue using a combination of both in silico and wet-lab approaches. Here, we show that the VAV2-regulated SCL program does harbor a number of cell cycle- and signaling-related kinases that are essential for the viability of undifferentiated keratinocytes and hnSCC patient-derived cells endowed with high levels of VAV2 activity. Our results also show that the VAV2-regulated SCL gene signature is associated with poor hnSCC patient prognosis. Collectively, these data underscore the critical role of this VAV2-regulated SCL program for the viability of both preneoplastic and fully transformed keratinocytes.
Collapse
Affiliation(s)
- Luis Francisco Lorenzo-Martín
- Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain;
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, 37007 Salamanca, Spain
- Correspondence: (L.F.L.-M.); (X.R.B.)
| | - Mauricio Menacho-Márquez
- Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain;
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, 37007 Salamanca, Spain
| | - Xosé R. Bustelo
- Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain;
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, 37007 Salamanca, Spain
- Correspondence: (L.F.L.-M.); (X.R.B.)
| |
Collapse
|