1
|
Koelsch N, Mirshahi F, Aqbi HF, Seneshaw M, Idowu MO, Olex AL, Sanyal AJ, Manjili MH. Effective anti-tumor immune responses are orchestrated by immune cell partnership network that functions through tissue homeostatic pathways, not direct cytotoxicity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.12.598563. [PMID: 38903113 PMCID: PMC11188117 DOI: 10.1101/2024.06.12.598563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
The liver hosts a diverse array of immune cells that play pivotal roles in both maintaining tissue homeostasis and responding to disease. However, the precise contributions of these immune cells in the progression of nonalcoholic fatty liver disease (NAFLD) and hepatocellular carcinoma (HCC) remain unclear. Utilizing a systems immunology approach, we reveal that liver immune responses are governed by a dominant-subdominant hierarchy of ligand-receptor-mediated homeostatic pathways. In healthy individuals, inflammatory immune responses operate within these pathways, challenging the notion of the liver as a purely tolerogenic organ. Chronic consumption of a Western diet (WD) disrupts hepatocyte function and reconfigures immune interactions, resulting in hepatic stellate cells (HSCs), cancer cells, and NKT cells driving 80% of the immune activity during NAFLD. In HCC, 80% of immune response involves NKT cells and monocytes collaborating with hepatocytes and myofibroblasts to restore disrupted homeostasis. Interestingly, dietary correction during NAFLD yields nonlinear outcomes: tumor progression coincides with the failure of mounting homeostatic immune responses, whereas tumor prevention is associated with sustained immune responses, predominantly orchestrated by monocytes. These monocytes actively target fibroblasts and myofibroblasts, creating a tumor-suppressive microenvironment. Notably, only 5% of T cells displayed apoptosis-inducing activity, selectively contributing to the turnover of hepatic stromal cells, particularly myofibroblasts and fibroblasts. Our findings suggest that effective anti-tumor immune responses in the liver are primarily mediated by immune cells sustaining tissue homeostasis, rather than relying on direct cytotoxic mechanisms.
Collapse
Affiliation(s)
- Nicholas Koelsch
- Department of Microbiology & Immunology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA
| | - Faridoddin Mirshahi
- Department of Internal Medicine, VCU School of Medicine, Richmond, VA 23298, USA
- Stravitz-Sanyal Institute for Liver Disease and Metabolic Health, Richmond, VA 23298
| | - Hussein F. Aqbi
- College of Science, Mustansiriyah University, Baghdad, P.O. Box 14022, Iraq
| | - Mulugeta Seneshaw
- Department of Internal Medicine, VCU School of Medicine, Richmond, VA 23298, USA
- Stravitz-Sanyal Institute for Liver Disease and Metabolic Health, Richmond, VA 23298
| | - Michael O. Idowu
- Department of Pathology, VCU School of Medicine, Richmond, VA 23298, USA
- VCU Massey Comprehensive Cancer Center, Richmond, VA 23298, USA
| | - Amy L. Olex
- VCU Massey Comprehensive Cancer Center, Richmond, VA 23298, USA
- C. Kenneth and Dianne Wright Center for Clinical and Translational Research, Virginia Commonwealth University School of Medicine
| | - Arun J. Sanyal
- Department of Internal Medicine, VCU School of Medicine, Richmond, VA 23298, USA
- Stravitz-Sanyal Institute for Liver Disease and Metabolic Health, Richmond, VA 23298
- VCU Massey Comprehensive Cancer Center, Richmond, VA 23298, USA
| | - Masoud H. Manjili
- Department of Microbiology & Immunology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA
- VCU Massey Comprehensive Cancer Center, Richmond, VA 23298, USA
- VCU Institute of Molecular Medicine, Richmond VA 23298
| |
Collapse
|
2
|
Ruiz-Martín V, Marcos T, de Pereda JM, Sánchez-Crespo M, de la Fuente MA, Bayón Y, Alonso A. LYP regulates SLP76 and other adaptor proteins in T cells. Biol Res 2024; 57:69. [PMID: 39342392 PMCID: PMC11438317 DOI: 10.1186/s40659-024-00536-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 08/06/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND The LYP tyrosine phosphatase presents a SNP (1858C > T) that increases the risk of developing autoimmune diseases such as type I diabetes and arthritis. It remains unclear how this SNP affects LYP function and promotes the development of these diseases. The scarce information about LYP substrates is in part responsible for the poor understanding of LYP function. RESULTS In this study, we identify in T lymphocytes several adaptor proteins as potential substrates targeted by LYP, including FYB, SLP-76, HS-1, Vav, SKAP1 and SKAP2. We also show that LYP co-localizes with SLP76 in microclusters, upon TCR engagement. CONCLUSIONS These data indicate that LYP may modulate T cell activation by dephosphorylating several adaptor proteins, such as FYB, SLP-76, HS-1, Vav, SKAP1 and SKAP2 upon TCR engagement.
Collapse
Affiliation(s)
- Virginia Ruiz-Martín
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), CSIC-Universidad de Valladolid, c/ Sanz y Forés 3, 47003, Valladolid, Spain
| | - Tamara Marcos
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), CSIC-Universidad de Valladolid, c/ Sanz y Forés 3, 47003, Valladolid, Spain
| | - José María de Pereda
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC), CSIC-Universidad de Salamanca, Campus Unamuno, 37007, Salamanca, Spain
| | - Mariano Sánchez-Crespo
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), CSIC-Universidad de Valladolid, c/ Sanz y Forés 3, 47003, Valladolid, Spain
| | - Miguel Angel de la Fuente
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), CSIC-Universidad de Valladolid, c/ Sanz y Forés 3, 47003, Valladolid, Spain
| | - Yolanda Bayón
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), CSIC-Universidad de Valladolid, c/ Sanz y Forés 3, 47003, Valladolid, Spain.
| | - Andrés Alonso
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), CSIC-Universidad de Valladolid, c/ Sanz y Forés 3, 47003, Valladolid, Spain.
| |
Collapse
|
3
|
Conde J, Fernández-Pisonero I, Lorenzo-Martín LF, García-Gómez R, Casar B, Crespo P, Bustelo XR. The mevalonate pathway contributes to breast primary tumorigenesis and lung metastasis. Mol Oncol 2024. [PMID: 39119789 DOI: 10.1002/1878-0261.13716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 07/01/2024] [Accepted: 07/30/2024] [Indexed: 08/10/2024] Open
Abstract
The mevalonate pathway plays an important role in breast cancer and other tumor types. However, many issues remain obscure as yet regarding its mechanism of regulation and action. In the present study, we report that the expression of mevalonate pathway enzymes is mediated by the RHO guanosine nucleotide exchange factors VAV2 and VAV3 in a RAC1- and sterol regulatory element-binding factor (SREBF)-dependent manner in breast cancer cells. Furthermore, in vivo tumorigenesis experiments indicated that the two most upstream steps of this metabolic pathway [3-hydroxy-3-methylglutaryl-coenzyme A synthase 1 (HMGCS1) and 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGCR)] are important for primary tumorigenesis, angiogenesis, and cell survival in breast cancer cells. HMGCR, but not HMGCS1, is also important for the extravasation and subsequent fitness of breast cancer cells in the lung parenchyma. Genome-wide expression analyses revealed that HMGCR influences the expression of gene signatures linked to proliferation, metabolism, and immune responses. The HMGCR-regulated gene signature predicts long-term tumor recurrence but not metastasis in cohorts of nonsegregated and chemotherapy-resistant breast cancer patients. These results reveal a hitherto unknown, VAV-catalysis-dependent mechanism involved in the regulation of the mevalonate pathway in breast cancer cells. They also identify specific mevalonate-pathway-dependent processes that contribute to the malignant features of breast cancer cells.
Collapse
Affiliation(s)
- Javier Conde
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC) and Universidad de Salamanca, Spain
- Instituto de Biología Molecular y Celular del Cáncer, CSIC and Universidad de Salamanca, Spain
| | - Isabel Fernández-Pisonero
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC) and Universidad de Salamanca, Spain
- Instituto de Biología Molecular y Celular del Cáncer, CSIC and Universidad de Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - L Francisco Lorenzo-Martín
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC) and Universidad de Salamanca, Spain
- Instituto de Biología Molecular y Celular del Cáncer, CSIC and Universidad de Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Rocío García-Gómez
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), CSIC and Universidad de Cantabria, Santander, Spain
| | - Berta Casar
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), CSIC and Universidad de Cantabria, Santander, Spain
| | - Piero Crespo
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), CSIC and Universidad de Cantabria, Santander, Spain
| | - Xosé R Bustelo
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC) and Universidad de Salamanca, Spain
- Instituto de Biología Molecular y Celular del Cáncer, CSIC and Universidad de Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
4
|
Chang CF, Bao BY, Hsueh YM, Chen PL, Chang LH, Li CY, Geng JH, Lu TL, Huang CY, Huang SP. Prognostic Significance of VAV3 Gene Variants and Expression in Renal Cell Carcinoma. Biomedicines 2024; 12:1694. [PMID: 39200159 PMCID: PMC11351164 DOI: 10.3390/biomedicines12081694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 07/23/2024] [Accepted: 07/28/2024] [Indexed: 09/01/2024] Open
Abstract
Renal cell carcinoma (RCC) is characterized by high mortality and morbidity rates. Vav guanine nucleotide exchange factors (VAVs), crucial for signal transduction between cell membrane receptors and intracellular mediators, have been implicated in carcinogenesis. However, their potential prognostic value in RCC remains unclear. The impact of 150 common VAV polymorphisms on RCC risk and survival was investigated in a cohort of 630 individuals. Publicly available gene expression datasets were utilized to analyze VAV gene expression in relation to patient outcomes. The VAV3 rs17019888 polymorphism was significantly associated with RCC risk and overall survival after adjusting for false discovery rates. Expression quantitative trait loci analysis revealed that the risk allele of rs17019888 is linked to reduced VAV3 expression. Analysis of 19 kidney cancer gene expression datasets revealed lower VAV3 expression in RCC tissues compared to normal tissues, with higher expression correlating with better prognosis. Gene set enrichment analysis demonstrated that VAV3 negatively regulates the ubiquitin-proteasome system, extracellular matrix and membrane receptors, inflammatory responses, matrix metalloproteinases, and cell cycle pathways. Furthermore, elevated VAV3 expression was associated with increased infiltration of B cells, macrophages, and neutrophils into the RCC tumor microenvironment. Our findings suggest that VAV3 gene variants influence RCC risk and survival, contributing to a favorable prognosis in RCC.
Collapse
Affiliation(s)
- Chi-Fen Chang
- Department of Anatomy, School of Medicine, China Medical University, Taichung 406, Taiwan;
| | - Bo-Ying Bao
- Department of Pharmacy, China Medical University, Taichung 406, Taiwan; (B.-Y.B.); (T.-L.L.)
| | - Yu-Mei Hsueh
- Department of Family Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 110, Taiwan;
- Department of Public Health, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Pei-Ling Chen
- Department of Urology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei 100, Taiwan;
| | - Li-Hsin Chang
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Chia-Yang Li
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Jiun-Hung Geng
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan;
- Department of Urology, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung 812, Taiwan
| | - Te-Ling Lu
- Department of Pharmacy, China Medical University, Taichung 406, Taiwan; (B.-Y.B.); (T.-L.L.)
| | - Chao-Yuan Huang
- Department of Urology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei 100, Taiwan;
| | - Shu-Pin Huang
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan;
- Institute of Medical Science and Technology, College of Medicine, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| |
Collapse
|
5
|
Jasim SA, Al-Hawary SIS, Kaur I, Ahmad I, Hjazi A, Petkov I, Ali SHJ, Redhee AH, Shuhata Alubiady MH, Al-Ani AM. Critical role of exosome, exosomal non-coding RNAs and non-coding RNAs in head and neck cancer angiogenesis. Pathol Res Pract 2024; 256:155238. [PMID: 38493725 DOI: 10.1016/j.prp.2024.155238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 02/13/2024] [Accepted: 03/02/2024] [Indexed: 03/19/2024]
Abstract
Head and neck cancer (HNC) refers to the epithelial malignancies of the upper aerodigestive tract. HNCs have a constant yet slow-growing rate with an unsatisfactory overall survival rate globally. The development of new blood vessels from existing blood conduits is regarded as angiogenesis, which is implicated in the growth, progression, and metastasis of cancer. Aberrant angiogenesis is a known contributor to human cancer progression. Representing a promising therapeutic target, the blockade of angiogenesis aids in the reduction of the tumor cells oxygen and nutrient supplies. Despite the promise, the association of existing anti-angiogenic approaches with severe side effects, elevated cancer regrowth rates, and limited survival advantages is incontrovertible. Exosomes appear to have an essential contribution to the support of vascular proliferation, the regulation of tumor growth, tumor invasion, and metastasis, as they are a key mediator of information transfer between cells. In the exocrine region, various types of noncoding RNAs (ncRNAs) identified to be enriched and stable and contribute to the occurrence and progression of cancer. Mounting evidence suggest that exosome-derived ncRNAs are implicated in tumor angiogenesis. In this review, the characteristics of angiogenesis, particularly in HNC, and the impact of ncRNAs on HNC angiogenesis will be outlined. Besides, we aim to provide an insight on the regulatory role of exosomes and exosome-derived ncRNAs in angiogenesis in different types of HNC.
Collapse
Affiliation(s)
| | | | - Irwanjot Kaur
- Department of Biotechnology and Genetics, Jain (Deemed-to-be) University, Bengaluru, Karnataka 560069, India; Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan 303012, India
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Ahmed Hjazi
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia.
| | - Iliya Petkov
- Medical University - Sofia, Department of Neurology, Sofia, Bulgaria
| | - Saad Hayif Jasim Ali
- Department of medical laboratory, College of Health and Medical Technololgy, Al-Ayen University, Thi-Qar, Iraq
| | - Ahmed Huseen Redhee
- Medical laboratory technique college, the Islamic University, Najaf, Iraq; Medical laboratory technique college, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq; Medical laboratory technique college, the Islamic University of Babylon, Babylon, Iraq
| | | | | |
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
|
Cowell E, Jaber H, Kris LP, Fitzgerald MG, Sanders VM, Norbury AJ, Eyre NS, Carr JM. Vav proteins do not influence dengue virus replication but are associated with induction of phospho-ERK, IL-6, and viperin mRNA following DENV infection in vitro. Microbiol Spectr 2024; 12:e0239123. [PMID: 38054722 PMCID: PMC10782993 DOI: 10.1128/spectrum.02391-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 11/03/2023] [Indexed: 12/07/2023] Open
Abstract
IMPORTANCE Dengue disease is characterized by an inflammatory-mediated immunopathology, with elevated levels of circulating factors including TNF-α and IL-6. If the damaging inflammatory pathways could be blocked without loss of antiviral responses or exacerbating viral replication, then this would be of potential therapeutic benefit. The study here has investigated the Vav guanine exchange factors as a potential alternative signaling pathway that may drive dengue virus (DENV)-induced inflammatory responses, with a focus on Vav1 and 2. While Vav proteins were positively associated with mRNA for inflammatory cytokines, blocking Vav signaling didn't affect DENV replication but prevented DENV-induction of p-ERK and enhanced IL-6 (inflammatory) and viperin (antiviral) mRNA. These initial data suggest that Vav proteins could be a target that does not compromise control of viral replication and should be investigated further for broader impact on host inflammatory responses, in settings such as antibody-dependent enhancement of infection and in different cell types.
Collapse
Affiliation(s)
- Evangeline Cowell
- College of Medicine and Public Health and Flinders Health and Medical Research Institute, Flinders University, Bedford Park, Adelaide, South Australia, Australia
| | - Hawraa Jaber
- College of Medicine and Public Health and Flinders Health and Medical Research Institute, Flinders University, Bedford Park, Adelaide, South Australia, Australia
| | - Luke P. Kris
- College of Medicine and Public Health and Flinders Health and Medical Research Institute, Flinders University, Bedford Park, Adelaide, South Australia, Australia
| | - Madeleine G. Fitzgerald
- College of Medicine and Public Health and Flinders Health and Medical Research Institute, Flinders University, Bedford Park, Adelaide, South Australia, Australia
| | - Valeria M. Sanders
- College of Medicine and Public Health and Flinders Health and Medical Research Institute, Flinders University, Bedford Park, Adelaide, South Australia, Australia
| | - Aidan J. Norbury
- Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Nicholas S. Eyre
- College of Medicine and Public Health and Flinders Health and Medical Research Institute, Flinders University, Bedford Park, Adelaide, South Australia, Australia
| | - Jillian M. Carr
- College of Medicine and Public Health and Flinders Health and Medical Research Institute, Flinders University, Bedford Park, Adelaide, South Australia, Australia
| |
Collapse
|
8
|
Stafuzza NB, Freitas ACD, Mioto MB, Silva RMDO, Fragomeni BDO, Pedrosa VB, Costa RLDD, Paz CCPD. Weighted single-step genome-wide association study and functional enrichment analyses for gastrointestinal nematode resistance traits in Santa Ines sheep. Vet Parasitol 2023; 323:110047. [PMID: 37857178 DOI: 10.1016/j.vetpar.2023.110047] [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: 06/27/2023] [Revised: 10/03/2023] [Accepted: 10/05/2023] [Indexed: 10/21/2023]
Abstract
This study aimed to identify genomic regions, pathways, and putative candidate genes associated with resistance to gastrointestinal nematode in Santa Ines sheep. The phenotypic information comprised 5529 records from 1703 naturally infected animals. After genomic data quality control, 37,511 SNPs from 589 animals were available. The weighted single-step approach for genome-wide association study was performed to estimate the SNP effects and variances accounted by 10-SNP sliding windows. Confirming the polygenic nature of the studied traits, 20, 22, 21, and 19 genomic windows that explained more than 0.5% of the additive genetic variance were identified for fecal egg counts (FEC), Famacha© (FAM), packed cell volume (PCV), and total plasma protein (TPP), respectively. A total of 81, 122, 106, and 101 protein-coding genes were found in windows associated with FEC, FAM, PCV, and TPP, respectively. Several protein-coding genes related to the immune system and inflammatory response functions were identified within those genomic regions, such as ADCY9, ADRB2, BRAF, CADM1, CCL20, CD70, CREBBP, FNBP1, HTR4, IL16, IL22, IL26, MAPK8, NDFIP1, NLRC3, PAK5, PLCB1, PLCB4, ROCK1, TEK, TNFRSF12A, and VAV1. Functional enrichment analysis by DAVID tool also revealed many significant (P < 0.05) pathways and Gene Ontology terms that could be related to resistance to gastrointestinal nematode in Santa Ines sheep, such as chemokine signaling pathway (oas04062), cAMP signaling pathway (oas04024), cGMP-PKG signaling pathway (Oas04022), platelet activation (Oas04611), Rap1 signaling pathway (oas04015), and oxidoreductase activity, acting on paired donors, with incorporation or reduction of molecular oxygen (GO:0016705). These results contribute to improving the knowledge of the genetic architecture of resistance to gastrointestinal nematode in Santa Ines sheep.
Collapse
Affiliation(s)
- Nedenia Bonvino Stafuzza
- Sustainable Livestock Research Center, Animal Science Institute, 15130-000 São José do Rio Preto, SP, Brazil.
| | - Ana Claudia de Freitas
- São Paulo Agency of Agribusiness and Technology, Animal Science Institute, 13380-011 Nova Odessa, SP, Brazil; Agricultural Research Agency of the State of Minas Gerais, 38709-899 Patos de Minas, MG, Brazil
| | - Marina B Mioto
- Sustainable Livestock Research Center, Animal Science Institute, 15130-000 São José do Rio Preto, SP, Brazil
| | | | | | - Victor Breno Pedrosa
- Department of Animal Science, State University of Ponta Grossa, 84030-900 Ponta Grossa, PR, Brazil
| | - Ricardo Lopes Dias da Costa
- São Paulo Agency of Agribusiness and Technology, Animal Science Institute, 13380-011 Nova Odessa, SP, Brazil
| | | |
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
|
Mehrabipour M, Jasemi NSK, Dvorsky R, Ahmadian MR. A Systematic Compilation of Human SH3 Domains: A Versatile Superfamily in Cellular Signaling. Cells 2023; 12:2054. [PMID: 37626864 PMCID: PMC10453029 DOI: 10.3390/cells12162054] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 08/02/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023] Open
Abstract
SRC homology 3 (SH3) domains are fundamental modules that enable the assembly of protein complexes through physical interactions with a pool of proline-rich/noncanonical motifs from partner proteins. They are widely studied modular building blocks across all five kingdoms of life and viruses, mediating various biological processes. The SH3 domains are also implicated in the development of human diseases, such as cancer, leukemia, osteoporosis, Alzheimer's disease, and various infections. A database search of the human proteome reveals the existence of 298 SH3 domains in 221 SH3 domain-containing proteins (SH3DCPs), ranging from 13 to 720 kilodaltons. A phylogenetic analysis of human SH3DCPs based on their multi-domain architecture seems to be the most practical way to classify them functionally, with regard to various physiological pathways. This review further summarizes the achievements made in the classification of SH3 domain functions, their binding specificity, and their significance for various diseases when exploiting SH3 protein modular interactions as drug targets.
Collapse
Affiliation(s)
- Mehrnaz Mehrabipour
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (M.M.); (N.S.K.J.)
| | - Neda S. Kazemein Jasemi
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (M.M.); (N.S.K.J.)
| | - Radovan Dvorsky
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (M.M.); (N.S.K.J.)
- Center for Interdisciplinary Biosciences, P. J. Šafárik University, 040 01 Košice, Slovakia
| | - Mohammad R. Ahmadian
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (M.M.); (N.S.K.J.)
| |
Collapse
|
11
|
Al-Hawary SIS, Alsalamy A, Gupta R, Alsaab HO, Hjazi A, Edilboyev U, Ramadan MF, Hussien BM, Ahmed M, Hosseini-Fard SR. VAV3 in human cancers: Mechanism and clinical implication. Pathol Res Pract 2023; 248:154681. [PMID: 37467637 DOI: 10.1016/j.prp.2023.154681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/04/2023] [Accepted: 07/07/2023] [Indexed: 07/21/2023]
Abstract
Guanine nucleotide exchange factors (GEFs) are primarily involved in signal transmission between cell membrane receptors and intracellular mediators. Upon replacing GDP with GTP, GEFs can alter their conformation, resulting in their binding to downstream effectors, such as GTPases like Ras homologous (Rho). VAV GEF family are versatile proteins working as an adaptor mediator and GEF for Rho GTPase. They act as a phosphorylation-dependent molecular switcher, fluctuating between active (tyrosine phosphorylated) and inactive (non-phosphorylated) conformation in cell signaling. Accumulating data showed that VAV3 is implicated in cancer progression. The higher levels of VAV3 in human cancers proposed that it may have an oncogenic role in cancer progression. Available studies demonstrated that VAV3 promoted cell proliferation, epithelial-mesenchymal transition (EMT), colony formation, cell cycle, survival, migration and invasion, and suppressed cell apoptosis. In addition, other studies indicated that VAV3 may have a prognostic value in cancer as well as it may act as a mediator in cancer chemoresistance. Here, we aimed to investigate the underlying molecular mechanism of VAV3 in cancer progression as well as to review its value as a prognostic biomarker and chemoresistance mediator in human cancers.
Collapse
Affiliation(s)
| | - Ali Alsalamy
- College of Engineering, Imam Ja'afar Al-Sadiq University, Al-Muthanna 66002, Iraq
| | - Reena Gupta
- Institute of Pharmaceutical Research, GLA University, District-Mathura, U.P., 281406, India
| | - Hashem O Alsaab
- Department of Pharmaceutics and Pharmaceutical Technology, Taif University, Taif 21944, Saudi Arabia
| | - Ahmed Hjazi
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Unarbek Edilboyev
- Department of Engineering Graphics and Design Theory, Tashkent Institute of Irrigation and Agricultural Mechanization Engineers, National Research University, Tashkent, Uzbekistan
| | | | - Beneen M Hussien
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | - Muhja Ahmed
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| | - Seyed Reza Hosseini-Fard
- Biochemistry Department, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
12
|
Cabral-Dias R, Antonescu CN. Control of phosphatidylinositol-3-kinase signaling by nanoscale membrane compartmentalization. Bioessays 2023; 45:e2200196. [PMID: 36567275 DOI: 10.1002/bies.202200196] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 09/12/2022] [Accepted: 12/13/2022] [Indexed: 12/27/2022]
Abstract
Phosphatidylinositol-3-kinases (PI3Ks) are lipid kinases that produce 3-phosphorylated derivatives of phosphatidylinositol upon activation by various cues. These 3-phosphorylated lipids bind to various protein effectors to control many cellular functions. Lipid phosphatases such as phosphatase and tensin homolog (PTEN) terminate PI3K-derived signals and are critical to ensure appropriate signaling outcomes. Many lines of evidence indicate that PI3Ks and PTEN, as well as some specific lipid effectors are highly compartmentalized, either in plasma membrane nanodomains or in endosomal compartments. We examine the evidence for specific recruitment of PI3Ks, PTEN, and other related enzymes to membrane nanodomains and endocytic compartments. We then examine the hypothesis that scaffolding of the sources (PI3Ks), terminators (PTEN), and effectors of these lipid signals with a common plasma membrane nanodomain may achieve highly localized lipid signaling and ensure selective activation of specific effectors. This highlights the importance of spatial regulation of PI3K signaling in various physiological and disease contexts.
Collapse
Affiliation(s)
- Rebecca Cabral-Dias
- Department of Chemistry and Biology and Graduate Program in Molecular Science, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Costin N Antonescu
- Department of Chemistry and Biology and Graduate Program in Molecular Science, Toronto Metropolitan University, Toronto, Ontario, Canada
| |
Collapse
|
13
|
Wong DCP, Pan CQ, Er SY, Thivakar T, Rachel TZY, Seah SH, Chua PJ, Jiang T, Chew TW, Chaudhuri PK, Mukherjee S, Salim A, Aye TA, Koh CG, Lim CT, Tan PH, Bay BH, Ridley AJ, Low BC. The scaffold RhoGAP protein ARHGAP8/BPGAP1 synchronizes Rac and Rho signaling to facilitate cell migration. Mol Biol Cell 2023; 34:ar13. [PMID: 36598812 PMCID: PMC10011724 DOI: 10.1091/mbc.e21-03-0099] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 12/14/2022] [Accepted: 12/21/2022] [Indexed: 01/05/2023] Open
Abstract
Rho GTPases regulate cell morphogenesis and motility under the tight control of guanine nucleotide exchange factors (GEFs) and GTPase-activating proteins (GAPs). However, the underlying mechanism(s) that coordinate their spatiotemporal activities, whether separately or together, remain unclear. We show that a prometastatic RhoGAP, ARHGAP8/BPGAP1, binds to inactive Rac1 and localizes to lamellipodia. BPGAP1 recruits the RacGEF Vav1 under epidermal growth factor (EGF) stimulation and activates Rac1, leading to polarized cell motility, spreading, invadopodium formation, and cell extravasation and promotes cancer cell migration. Importantly, BPGAP1 down-regulates local RhoA activity, which influences Rac1 binding to BPGAP1 and its subsequent activation by Vav1. Our results highlight the importance of BPGAP1 in recruiting Vav1 and Rac1 to promote Rac1 activation for cell motility. BPGAP1 also serves to control the timing of Rac1 activation with RhoA inactivation via its RhoGAP activity. BPGAP1, therefore, acts as a dual-function scaffold that recruits Vav1 to activate Rac1 while inactivating RhoA to synchronize both Rho and Rac signaling in cell motility. As epidermal growth factor receptor (EGFR), Vav1, RhoA, Rac1, and BPGAP1 are all associated with cancer metastasis, BPGAP1 could provide a crucial checkpoint for the EGFR-BPGAP1-Vav1-Rac1-RhoA signaling axis for cancer intervention.
Collapse
Affiliation(s)
| | | | - Shi Yin Er
- Cell Signaling and Developmental Biology Laboratory, Department of Biological Sciences, National University of Singapore, Singapore 117558
| | - T. Thivakar
- Mechanobiology Institute, National University of Singapore, Singapore 117411
| | - Tan Zi Yi Rachel
- Mechanobiology Institute, National University of Singapore, Singapore 117411
| | - Sock Hong Seah
- Mechanobiology Institute, National University of Singapore, Singapore 117411
| | - Pei Jou Chua
- Department of Anatomy, Yong Loo Lin School of Medicine, National University Health System, Singapore 117594
| | - Tingting Jiang
- Cell Signaling and Developmental Biology Laboratory, Department of Biological Sciences, National University of Singapore, Singapore 117558
| | - Ti Weng Chew
- Mechanobiology Institute, National University of Singapore, Singapore 117411
| | | | - Somsubhro Mukherjee
- Mechanobiology Institute, National University of Singapore, Singapore 117411
| | - Agus Salim
- Melbourne School of Population and Global Health and School of Mathematics and Statistics, The University of Melbourne, Melbourne, Victoria 3052, Australia
| | - Thike Aye Aye
- Department of Pathology, Singapore General Hospital, Singapore 169856
| | - Cheng Gee Koh
- Division of Molecular Genetics & Cell Biology, School of Biological Sciences, Nanyang Technological University, Singapore 637551
| | - Chwee Teck Lim
- Mechanobiology Institute, National University of Singapore, Singapore 117411
- Department of Biomedical Engineering, National University of Singapore, Singapore 117583
| | - Puay Hoon Tan
- Department of Pathology, Singapore General Hospital, Singapore 169856
| | - Boon Huat Bay
- Department of Anatomy, Yong Loo Lin School of Medicine, National University Health System, Singapore 117594
| | - Anne J. Ridley
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, United Kingdom
| | - Boon Chuan Low
- Mechanobiology Institute, National University of Singapore, Singapore 117411
- Cell Signaling and Developmental Biology Laboratory, Department of Biological Sciences, National University of Singapore, Singapore 117558
- NUS College, National University of Singapore, Singapore 138593
| |
Collapse
|
14
|
Badaoui M, Sobolewski C, Luscher A, Bacchetta M, Köhler T, van Delden C, Foti M, Chanson M. Targeting HuR-Vav3 mRNA interaction prevents Pseudomonas aeruginosa adhesion to the cystic fibrosis airway epithelium. JCI Insight 2023; 8:161961. [PMID: 36602863 PMCID: PMC9977432 DOI: 10.1172/jci.insight.161961] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 12/21/2022] [Indexed: 01/06/2023] Open
Abstract
Cystic fibrosis (CF) is characterized by chronic bacterial infections leading to progressive bronchiectasis and respiratory failure. Pseudomonas aeruginosa (Pa) is the predominant opportunistic pathogen infecting the CF airways. The guanine nucleotide exchange factor Vav3 plays a critical role in Pa adhesion to the CF airways by inducing luminal fibronectin deposition that favors bacteria trapping. Here we report that Vav3 overexpression in CF is caused by upregulation of the mRNA-stabilizing protein HuR. We found that HuR accumulates in the cytoplasm of CF airway epithelial cells and that it binds to and stabilizes Vav3 mRNA. Interestingly, disruption of the HuR-Vav3 mRNA interaction improved the CF epithelial integrity, inhibited the formation of the fibronectin-made bacterial docking platforms, and prevented Pa adhesion to the CF airway epithelium. These findings indicate that targeting HuR represents a promising antiadhesive approach in CF that can prevent initial stages of Pa infection in a context of emergence of multidrug-resistant pathogens.
Collapse
Affiliation(s)
| | | | - Alexandre Luscher
- Department of Microbiology & Molecular Medicine, Faculty of Medicine, University of Geneva, Switzerland
| | | | - Thilo Köhler
- Department of Microbiology & Molecular Medicine, Faculty of Medicine, University of Geneva, Switzerland
| | - Christian van Delden
- Department of Microbiology & Molecular Medicine, Faculty of Medicine, University of Geneva, Switzerland
| | | | | |
Collapse
|
15
|
Xia D, Li W, Tang C, Jiang J. Astragaloside IV, as a potential anticancer agent. Front Pharmacol 2023; 14:1065505. [PMID: 36874003 PMCID: PMC9981805 DOI: 10.3389/fphar.2023.1065505] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 02/08/2023] [Indexed: 02/19/2023] Open
Abstract
Cancer is a global intractable disease, and its morbidity and mortality are increasing year by year in developing countries. Surgery and chemotherapy are often used to treat cancer, but they result in unsatisfactory outcomes, such as severe side effects and drug resistance. With the accelerated modernization of traditional Chinese medicine (TCM), an increasing body of evidence has shown that several TCM components have significant anticancer activities. Astragaloside IV (AS-IV) is considered the main active ingredient of the dried root of Astragalus membranaceus. AS-IV exhibits various pharmacological effects, such as anti-inflammatory, hypoglycemic, antifibrotic, and anticancer activities. AS-IV possesses a wide range of activities, such as the modulation of reactive oxygen species-scavenging enzyme activities, participation in cell cycle arrest, induction of apoptosis and autophagy, and suppression of cancer cell proliferation, invasiveness, and metastasis. These effects are involved in the inhibition of different malignant tumors, such as lung, liver, breast, and gastric cancers. This article reviews the bioavailability, anticancer activity, and mechanism of AS-IV and provides suggestions for further research of this TCM.
Collapse
Affiliation(s)
- Dongqin Xia
- Chongqing University Cancer Hospital, Chongqing, China
| | - Wenjie Li
- Affiliated Hospital of Northwest Minzu University, Lanzhou, China
| | - Ce Tang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Juan Jiang
- Chongqing University Cancer Hospital, Chongqing, China
| |
Collapse
|
16
|
Schäfer I, Bauch J, Wegrzyn D, Roll L, van Leeuwen S, Jarocki A, Faissner A. The guanine nucleotide exchange factor Vav3 intervenes in the migration pathway of oligodendrocyte precursor cells on tenascin-C. Front Cell Dev Biol 2022; 10:1042403. [PMID: 36531963 PMCID: PMC9748482 DOI: 10.3389/fcell.2022.1042403] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 11/10/2022] [Indexed: 10/22/2024] Open
Abstract
Oligodendrocyte precursor cells (OPCs) are the exclusive source of myelination in the central nervous system (CNS). Prior to myelination, OPCs migrate to target areas and mature into myelinating oligodendrocytes. This process is underpinned by drastic changes of the cytoskeleton and partially driven by pathways involving small GTPases of the Rho subfamily. In general, the myelination process requires migration, proliferation and differentiation of OPCs. Presently, these processes are only partially understood. In this study, we analyzed the impact of the guanine nucleotide exchange factor (GEF) Vav3 on the migration behavior of OPCs. Vav3 is known to regulate RhoA, Rac1 and RhoG activity and is therefore a promising candidate with regard to a regulatory role concerning the rearrangement of the cytoskeleton. Our study focused on the Vav3 knockout mouse and revealed an enhanced migration capacity of Vav3 -/- OPCs on the extracellular matrix (ECM) glycoprotein tenascin-C (TnC). The migration behavior of individual OPCs on further ECM molecules such as laminin-1 (Ln1), laminin-2 (Ln2) and tenascin-R (TnR) was not affected by the elimination of Vav3. The migration process was further investigated with regard to intracellular signal transmission by pharmacological blockade of downstream pathways of specific Rho GTPases. Our data suggest that activation of RhoA GTPase signaling compromises migration, as inhibition of RhoA-signaling promoted migration behavior. This study provides novel insights into the control of OPC migration, which could be useful for further understanding of the complex differentiation and myelination process.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
17
|
Ben-Mahmoud A, Jun KR, Gupta V, Shastri P, de la Fuente A, Park Y, Shin KC, Kim CA, da Cruz AD, Pinto IP, Minasi LB, Silva da Cruz A, Faivre L, Callier P, Racine C, Layman LC, Kong IK, Kim CH, Kim WY, Kim HG. A rigorous in silico genomic interrogation at 1p13.3 reveals 16 autosomal dominant candidate genes in syndromic neurodevelopmental disorders. Front Mol Neurosci 2022; 15:979061. [PMID: 36277487 PMCID: PMC9582330 DOI: 10.3389/fnmol.2022.979061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 08/30/2022] [Indexed: 11/13/2022] Open
Abstract
Genome-wide chromosomal microarray is extensively used to detect copy number variations (CNVs), which can diagnose microdeletion and microduplication syndromes. These small unbalanced chromosomal structural rearrangements ranging from 1 kb to 10 Mb comprise up to 15% of human mutations leading to monogenic or contiguous genomic disorders. Albeit rare, CNVs at 1p13.3 cause a variety of neurodevelopmental disorders (NDDs) including development delay (DD), intellectual disability (ID), autism, epilepsy, and craniofacial anomalies (CFA). Most of the 1p13.3 CNV cases reported in the pre-microarray era encompassed a large number of genes and lacked the demarcating genomic coordinates, hampering the discovery of positional candidate genes within the boundaries. In this study, we present four subjects with 1p13.3 microdeletions displaying DD, ID, autism, epilepsy, and CFA. In silico comparative genomic mapping with three previously reported subjects with CNVs and 22 unreported DECIPHER CNV cases has resulted in the identification of four different sub-genomic loci harboring five positional candidate genes for DD, ID, and CFA at 1p13.3. Most of these genes have pathogenic variants reported, and their interacting genes are involved in NDDs. RT-qPCR in various human tissues revealed a high expression pattern in the brain and fetal brain, supporting their functional roles in NDDs. Interrogation of variant databases and interacting protein partners led to the identification of another set of 11 potential candidate genes, which might have been dysregulated by the position effect of these CNVs at 1p13.3. Our studies define 1p13.3 as a genomic region harboring 16 NDD candidate genes and underscore the critical roles of small CNVs in in silico comparative genomic mapping for disease gene discovery. Our candidate genes will help accelerate the isolation of pathogenic heterozygous variants from exome/genome sequencing (ES/GS) databases.
Collapse
Affiliation(s)
- Afif Ben-Mahmoud
- Neurological Disorders Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Doha, Qatar
| | - Kyung Ran Jun
- Department of Laboratory Medicine, Inje University Haeundae Paik Hospital, Busan, South Korea
| | - Vijay Gupta
- Neurological Disorders Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Doha, Qatar
| | - Pinang Shastri
- Department of Cardiovascular Medicine, Cape Fear Valley Medical Center, Fayetteville, NC, United States
| | - Alberto de la Fuente
- Diabetes Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Doha, Qatar
| | - Yongsoo Park
- Neurological Disorders Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Doha, Qatar
| | - Kyung Chul Shin
- Neurological Disorders Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Doha, Qatar
| | - Chong Ae Kim
- Faculdade de Medicina, Unidade de Genética do Instituto da Criança – Hospital das Clínicas HCFMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Aparecido Divino da Cruz
- School of Medical and Life Sciences, Genetics Master Program, Replicon Research Group, Pontifical Catholic University of Goiás, Goiânia, Brazil
- Genetics Master Program, Replicon Research Nucleus, School of Agrarian and Biological Sciences, Pontifical Catholic University of Goias, Goiás, Brazil
| | - Irene Plaza Pinto
- School of Medical and Life Sciences, Genetics Master Program, Replicon Research Group, Pontifical Catholic University of Goiás, Goiânia, Brazil
- Genetics Master Program, Replicon Research Nucleus, School of Agrarian and Biological Sciences, Pontifical Catholic University of Goias, Goiás, Brazil
| | - Lysa Bernardes Minasi
- School of Medical and Life Sciences, Genetics Master Program, Replicon Research Group, Pontifical Catholic University of Goiás, Goiânia, Brazil
- Genetics Master Program, Replicon Research Nucleus, School of Agrarian and Biological Sciences, Pontifical Catholic University of Goias, Goiás, Brazil
| | - Alex Silva da Cruz
- School of Medical and Life Sciences, Genetics Master Program, Replicon Research Group, Pontifical Catholic University of Goiás, Goiânia, Brazil
- Genetics Master Program, Replicon Research Nucleus, School of Agrarian and Biological Sciences, Pontifical Catholic University of Goias, Goiás, Brazil
| | - Laurence Faivre
- Inserm UMR 1231 GAD, Genetics of Developmental Disorders, Université de Bourgogne-Franche Comté, Dijon, France
- Centre de Référence Anomalies du Développement et Syndromes Malformatifs, Hôpital d’Enfants, Dijon, France
| | - Patrick Callier
- UMR 1231 GAD, Inserm – Université Bourgogne-Franche Comté, Dijon, France
| | - Caroline Racine
- UMR 1231 GAD, Inserm – Université Bourgogne-Franche Comté, Dijon, France
| | - Lawrence C. Layman
- Section of Reproductive Endocrinology, Infertility and Genetics, Department of Obstetrics and Gynecology, Augusta University, Augusta, GA, United States
- Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, GA, United States
| | - Il-Keun Kong
- Department of Animal Science, Division of Applied Life Science (BK21 Four), Gyeongsang National University, Jinju, South Korea
| | - Cheol-Hee Kim
- Department of Biology, Chungnam National University, Daejeon, South Korea
| | - Woo-Yang Kim
- Department of Biological Sciences, Kent State University, Kent, OH, United States
| | - Hyung-Goo Kim
- Neurological Disorders Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Doha, Qatar
- *Correspondence: Hyung-Goo Kim,
| |
Collapse
|
18
|
Ujcikova H, Roubalova L, Lee YS, Slaninova J, Brejchova J, Svoboda P. The Dose-Dependent Effects of Multifunctional Enkephalin Analogs on the Protein Composition of Rat Spleen Lymphocytes, Cortex, and Hippocampus; Comparison with Changes Induced by Morphine. Biomedicines 2022; 10:biomedicines10081969. [PMID: 36009516 PMCID: PMC9406115 DOI: 10.3390/biomedicines10081969] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/28/2022] [Accepted: 08/12/2022] [Indexed: 11/16/2022] Open
Abstract
This work aimed to test the effect of 7-day exposure of rats to multifunctional enkephalin analogs LYS739 and LYS744 at doses of 3 mg/kg and 10 mg/kg on the protein composition of rat spleen lymphocytes, brain cortex, and hippocampus. Alterations of proteome induced by LYS739 and LYS744 were compared with those elicited by morphine. The changes in rat proteome profiles were analyzed by label-free quantification (MaxLFQ). Proteomic analysis indicated that the treatment with 3 mg/kg of LYS744 caused significant alterations in protein expression levels in spleen lymphocytes (45), rat brain cortex (31), and hippocampus (42). The identified proteins were primarily involved in RNA processing and the regulation of cytoskeletal dynamics. In spleen lymphocytes, the administration of the higher 10 mg/kg dose of both enkephalin analogs caused major, extensive modifications in protein expression levels: LYS739 (119) and LYS744 (182). Among these changes, the number of proteins associated with immune responses and apoptotic processes was increased. LYS739 treatment resulted in the highest number of alterations in the rat brain cortex (152) and hippocampus (45). The altered proteins were functionally related to the regulation of transcription and cytoskeletal reorganization, which plays an essential role in neuronal plasticity. Administration with LYS744 did not increase the number of altered proteins in the brain cortex (26) and hippocampus (26). Our findings demonstrate that the effect of κ-OR full antagonism of LYS744 is opposite in the central nervous system and the peripheral region (spleen lymphocytes).
Collapse
Affiliation(s)
- Hana Ujcikova
- Laboratory of Neurochemistry, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, Czech Republic
- Correspondence:
| | - Lenka Roubalova
- Laboratory of Neurochemistry, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, Czech Republic
| | - Yeon Sun Lee
- Department of Pharmacology, University of Arizona, Tucson, AZ 85724, USA
| | - Jirina Slaninova
- Laboratory of Neurochemistry, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, Czech Republic
| | - Jana Brejchova
- Laboratory of Neurochemistry, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, Czech Republic
| | - Petr Svoboda
- Laboratory of Neurochemistry, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, Czech Republic
| |
Collapse
|
19
|
Robles-Valero J, Fernández-Nevado L, Cuadrado M, Lorenzo-Martín LF, Fernández-Pisonero I, Abad A, Redín E, Montuenga L, Martín-Zanca D, Bigas A, Mallo M, Dosil M, Bustelo XR. Characterization of the spectrum of trivalent VAV1-mutation-driven tumors using a gene-edited mouse model. Mol Oncol 2022; 16:3533-3553. [PMID: 35895495 PMCID: PMC9533688 DOI: 10.1002/1878-0261.13295] [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: 03/23/2022] [Revised: 07/07/2022] [Accepted: 07/26/2022] [Indexed: 11/28/2022] Open
Abstract
Mutations in the VAV1 guanine nucleotide exchange factor 1 have been recently found in peripheral T cell lymphoma and nonsmall‐cell lung cancer (NSCLC). To understand their pathogenic potential, we generated a gene‐edited mouse model that expresses a VAV1 mutant protein that recapitulates the signalling alterations present in the VAV1 mutant subclass most frequently found in tumours. We could not detect any overt tumourigenic process in those mice. However, the concurrent elimination of the Trp53 tumour suppressor gene in them drives T cell lymphomagenesis. This process represents an exacerbation of the normal functions that wild‐type VAV1 plays in follicular helper T cells. We also found that, in combination with the Kras oncogene, the VAV1 mutant version favours progression of NSCLC. These data indicate that VAV1 mutations play critical, although highly cell‐type‐specific, roles in tumourigenesis. They also indicate that such functions are contingent on the mutational landscape of the tumours involved.
Collapse
Affiliation(s)
- Javier Robles-Valero
- Molecular Mechanisms of Cancer Program, 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
| | - Lucía Fernández-Nevado
- Molecular Mechanisms of Cancer Program, 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
| | - Myriam Cuadrado
- Molecular Mechanisms of Cancer Program, 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
| | - L Francisco Lorenzo-Martín
- Molecular Mechanisms of Cancer Program, 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
| | - Isabel Fernández-Pisonero
- Molecular Mechanisms of Cancer Program, 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
| | - Antonio Abad
- Molecular Mechanisms of Cancer Program, 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
| | - Esther Redín
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, 37007, Salamanca, Spain.,Solid Tumors Program, Center of Applied Medical Research, University of Navarra, 31008, Pamplona, Spain
| | - Luis Montuenga
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, 37007, Salamanca, Spain.,Solid Tumors Program, Center of Applied Medical Research, University of Navarra, 31008, Pamplona, Spain
| | - Dionisio Martín-Zanca
- Instituto de Biología Funcional y Genómica, CSIC-University of Salamanca, 37007, Salamanca, Spain
| | - Anna Bigas
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, 37007, Salamanca, Spain.,Institut Hospital del Mar d'Investigacions Médiques, 08003, Barcelona, Spain
| | - Moisés Mallo
- Gulbenkian Institute, 2780-156, Oeiras, Portugal
| | - Mercedes Dosil
- Molecular Mechanisms of Cancer Program, 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
- Molecular Mechanisms of Cancer Program, 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
| |
Collapse
|
20
|
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: 0.5] [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
|
21
|
Tumor-Derived Exosome FGD5-AS1 Promotes Angiogenesis, Vascular Permeability, and Metastasis in Thyroid Cancer by Targeting the miR-6838-5p/VAV2 Axis. JOURNAL OF ONCOLOGY 2022; 2022:4702855. [PMID: 35528244 PMCID: PMC9076303 DOI: 10.1155/2022/4702855] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 03/05/2022] [Accepted: 03/25/2022] [Indexed: 02/06/2023]
Abstract
Exosomes are small vesicles with a diameter of 30~150 nm secreted by cells, which are rich in mRNA, microRNA, and long noncoding RNA (lncRNA). The biological functions of most exosomal lncRNAs are not well understood. Studies have shown that tumor exosome FGD5-AS1 plays an important role in the proliferation, migration, and invasion of tumor cells. In this study, SW1736 and KAT18 TC cells with high expression of FGD5-AS1 were screened. Exosomes with high expression of FGD5-AS1 were collected. The collected exosomes were then added to HUVEC cells. After incubation for 24 h, the effects on the proliferation and migration of HUVEC cells and vascular permeability were detected. The results showed that TC cells SW1736 and KAT18 could secrete a large number of exosomes, which could be taken up by HUVEC cells. Overexpression of FGD5-AS1 enhanced proliferation, migration, angiogenesis, and permeability of HUVEC. This effect is achieved through activation of the miR-6838-5p/VAV2 axis. These results suggest that FGD5-AS1 in tumor-derived exoskeleton promotes angiogenesis, vascular permeability, and metastasis by regulating the endothelial miR-6838-5p/VAV2 axis and ultimately promotes the occurrence and development of TC.
Collapse
|
22
|
Hegde S, Gasilina A, Wunderlich M, Lin Y, Buchholzer M, Krumbach OHF, Akbarzadeh M, Ahmadian MR, Seibel W, Zheng Y, Perentesis JP, Mizukawa BE, Vinnedge LP, Cancelas JA, Nassar NN. Inhibition of the RacGEF VAV3 by the small molecule IODVA1 impedes RAC signaling and overcomes resistance to tyrosine kinase inhibition in acute lymphoblastic leukemia. Leukemia 2022; 36:637-647. [PMID: 34711926 PMCID: PMC8885421 DOI: 10.1038/s41375-021-01455-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 10/01/2021] [Accepted: 10/15/2021] [Indexed: 01/09/2023]
Abstract
Aberrant RHO guanine nucleotide exchange factor (RhoGEF) activation is chief mechanism driving abnormal activation of their GTPase targets in transformation and tumorigenesis. Consequently, a small-molecule inhibitor of RhoGEF can make an anti-cancer drug. We used cellular, mouse, and humanized models of RAC-dependent BCR-ABL1-driven and Ph-like acute lymphoblastic leukemia to identify VAV3, a tyrosine phosphorylation-dependent RacGEF, as the target of the small molecule IODVA1. We show that through binding to VAV3, IODVA1 inhibits RAC activation and signaling and increases pro-apoptotic activity in BCR-ABL1-transformed cells. Consistent with this mechanism of action, cellular and animal models of BCR-ABL1-induced leukemia in Vav3-null background do not respond to IODVA1. By durably decreasing in vivo RAC signaling, IODVA1 eradicates leukemic propagating activity of TKI-resistant BCR-ABL1(T315I) B-ALL cells after treatment withdrawal. Importantly, IODVA1 suppresses the leukemic burden in the treatment refractory pediatric Ph+ and TKI-resistant Ph+ B-ALL patient-derived xenograft models better than standard-of-care dasatinib or ponatinib and provides a more durable response after treatment withdrawal. Pediatric leukemia samples with diverse genetic lesions show high sensitivity to IODVA1 ex vivo and this sensitivity is VAV3 dependent. IODVA1 thus spearheads a novel class of drugs that inhibits a RacGEF and holds promise as an anti-tumor therapy.
Collapse
Affiliation(s)
- Shailaja Hegde
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Research Foundation, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
- Hoxworth Blood Center, University of Cincinnati Academic Health Center, Cincinnati, OH, USA
| | - Anjelika Gasilina
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Research Foundation, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
| | - Mark Wunderlich
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Research Foundation, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
| | - Yuan Lin
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Research Foundation, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
| | - Marcel Buchholzer
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, 40225, Germany
| | - Oliver H F Krumbach
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, 40225, Germany
| | - Mohammad Akbarzadeh
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, 40225, Germany
| | - Mohammad Reza Ahmadian
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, 40225, Germany
| | - William Seibel
- Division of Oncology, Cincinnati Children's Hospital Medical Center, Cancer and Blood Diseases Institute, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
| | - Yi Zheng
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Research Foundation, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Ave, Cincinnati, OH, 45267, USA
| | - John P Perentesis
- Division of Oncology, Cincinnati Children's Hospital Medical Center, Cancer and Blood Diseases Institute, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
| | - Benjamin E Mizukawa
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Research Foundation, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
| | - Lisa Privette Vinnedge
- Division of Oncology, Cincinnati Children's Hospital Medical Center, Cancer and Blood Diseases Institute, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Ave, Cincinnati, OH, 45267, USA
| | - José A Cancelas
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Research Foundation, 3333 Burnet Ave, Cincinnati, OH, 45229, USA
- Hoxworth Blood Center, University of Cincinnati Academic Health Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Ave, Cincinnati, OH, 45267, USA
| | - Nicolas N Nassar
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Research Foundation, 3333 Burnet Ave, Cincinnati, OH, 45229, USA.
- Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Ave, Cincinnati, OH, 45267, USA.
| |
Collapse
|
23
|
Vav Proteins in Development of the Brain: A Potential Relationship to the Pathogenesis of Congenital Zika Syndrome? Viruses 2022; 14:v14020386. [PMID: 35215978 PMCID: PMC8874935 DOI: 10.3390/v14020386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/07/2022] [Accepted: 02/10/2022] [Indexed: 12/07/2022] Open
Abstract
Zika virus (ZIKV) infection during pregnancy can result in a significant impact on the brain and eye of the developing fetus, termed congenital zika syndrome (CZS). At a morphological level, the main serious presentations of CZS are microcephaly and retinal scarring. At a cellular level, many cell types of the brain may be involved, but primarily neuronal progenitor cells (NPC) and developing neurons. Vav proteins have guanine exchange activity in converting GDP to GTP on proteins such as Rac1, Cdc42 and RhoA to stimulate intracellular signaling pathways. These signaling pathways are known to play important roles in maintaining the polarity and self-renewal of NPC pools by coordinating the formation of adherens junctions with cytoskeletal rearrangements. In developing neurons, these same pathways are adopted to control the formation and growth of neurites and mediate axonal guidance and targeting in the brain and retina. This review describes the role of Vavs in these processes and highlights the points of potential ZIKV interaction, such as (i) the binding and entry of ZIKV in cells via TAM receptors, which may activate Vav/Rac/RhoA signaling; (ii) the functional convergence of ZIKV NS2A with Vav in modulating adherens junctions; (iii) ZIKV NS4A/4B protein effects on PI3K/AKT in a regulatory loop via PPI3 to influence Vav/Rac1 signaling in neurite outgrowth; and (iv) the induction of SOCS1 and USP9X following ZIKV infection to regulate Vav protein degradation or activation, respectively, and impact Vav/Rac/RhoA signaling in NPC and neurons. Experiments to define these interactions will further our understanding of the molecular basis of CZS and potentially other developmental disorders stemming from in utero infections. Additionally, Vav/Rac/RhoA signaling pathways may present tractable targets for therapeutic intervention or molecular rationale for disease severity in CZS.
Collapse
|
24
|
Steichen C, Hervé C, Hauet T, Bourmeyster N. Rho GTPases in kidney physiology and diseases. Small GTPases 2022; 13:141-161. [PMID: 34138686 PMCID: PMC9707548 DOI: 10.1080/21541248.2021.1932402] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 05/08/2021] [Accepted: 05/17/2021] [Indexed: 02/06/2023] Open
Abstract
Rho family GTPases are molecular switches best known for their pivotal role in dynamic regulation of the actin cytoskeleton, but also of cellular morphology, motility, adhesion and proliferation. The prototypic members of this family (RhoA, Rac1 and Cdc42) also contribute to the normal kidney function and play important roles in the structure and function of various kidney cells including tubular epithelial cells, mesangial cells and podocytes. The kidney's vital filtration function depends on the structural integrity of the glomerulus, the proximal portion of the nephron. Within the glomerulus, the architecturally actin-based cytoskeleton podocyte forms the final cellular barrier to filtration. The glomerulus appears as a highly dynamic signalling hub that is capable of integrating intracellular cues from its individual structural components. Dynamic regulation of the podocyte cytoskeleton is required for efficient barrier function of the kidney. As master regulators of actin cytoskeletal dynamics, Rho GTPases are therefore of critical importance for sustained kidney barrier function. Dysregulated activities of the Rho GTPases and of their effectors are implicated in the pathogenesis of both hereditary and idiopathic forms of kidney diseases. Diabetic nephropathy is a progressive kidney disease that is caused by injury to kidney glomeruli. High glucose activates RhoA/Rho-kinase in mesangial cells, leading to excessive extracellular matrix production (glomerulosclerosis). This RhoA/Rho-kinase pathway also seems involved in the post-transplant hypertension frequently observed during treatment with calcineurin inhibitors, whereas Rac1 activation was observed in post-transplant ischaemic acute kidney injury.
Collapse
Affiliation(s)
- Clara Steichen
- Inserm UMR-1082 Irtomit, Poitiers, France
- Faculté De Médecine Et De Pharmacie, Université De Poitiers, Poitiers, France
| | | | - Thierry Hauet
- Inserm UMR-1082 Irtomit, Poitiers, France
- Faculté De Médecine Et De Pharmacie, Université De Poitiers, Poitiers, France
- Department of Medical Biology, Service De Biochimie, CHU De Poitiers, Poitiers, France
| | - Nicolas Bourmeyster
- Faculté De Médecine Et De Pharmacie, Université De Poitiers, Poitiers, France
- Department of Medical Biology, Service De Biochimie, CHU De Poitiers, Poitiers, France
- Laboratoire STIM CNRS ERL 7003, Université de Poitiers, Poitiers Cédex, France
| |
Collapse
|
25
|
Kazim N, Yen A. Evidence of off-target effects of bosutinib that promote retinoic acid-induced differentiation of non-APL AML cells. Cell Cycle 2021; 20:2638-2651. [PMID: 34836491 DOI: 10.1080/15384101.2021.2005275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
In the present study, we determined the effects of the Src family kinase (SFK) inhibitor, Bosutinib, and the engineered loss of the Lyn SFK on all-trans retinoic acid-induced leukemic cell differentiation. Retinoic acid (RA) is an embryonic morphogen and dietary factor that demonstrates chemotherapeutic efficacy in inducing differentiation of a non-APL AML cell model, the HL-60 human myeloblastic (FAB-M2) leukemia cell line, via activation of a novel signalsome containing an ensemble of signaling molecules that drive differentiation. Bosutinib is an inhibitor of SFKs used to treat myeloid leukemias where prominent high expression of SFKs, in particular Lyn, has been observed. Using either Bosutinib or loss of Lyn expression due to shRNA promoted RA-induced phenotypic differentiation, G0 arrest, and respiratory burst (functional differentiation) of HL-60 cells. Signaling events putatively seminal to RA-induced differentiation, the expression of Fgr, Cbl, Slp-76 and Vav, and the phosphorylation of c-Raf (pS259), Vav (p-tyr), and Slp76 (p-tyr) were not inhibited by Bosutinib or loss of Lyn. Nor was RA-induced upregulation of p-tyr phosphorylation of p47phox, a member of the NADPH complex that produces ROS, a putative phosphorylation dependent signaling regulator. Surprisingly, Bosutinib still works in the absence of Lyn to enhance RA-induced differentiation and neither compromised RA-induced expression, nor phosphorylation of signaling molecules that drive differentiation. These findings suggested there is a novel, off-target, Lyn-independent effect of Bosutinib that is of therapeutic significance to differentiation therapy.
Collapse
Affiliation(s)
- Noor Kazim
- Department of Biomedical Science, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Andrew Yen
- Department of Biomedical Science, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| |
Collapse
|
26
|
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: 3.3] [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
|
27
|
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: 0.7] [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
|
28
|
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: 2.3] [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
|
29
|
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: 6.0] [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
|
30
|
Tsou YS, Wang CY, Chang MY, Hsu TI, Wu MT, Wu YH, Tsai WL, Chuang JY, Kao TJ. Vav2 is required for Netrin-1 receptor-class-specific spinal motor axon guidance. Dev Dyn 2021; 251:444-458. [PMID: 34374463 DOI: 10.1002/dvdy.409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 08/06/2021] [Accepted: 08/06/2021] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Proper guidance of neuronal axons to their targets is required to assemble neural circuits during the development of the nervous system. However, the mechanism by which the guidance of axonal growth cones is regulated by specific intermediaries activated by receptor signaling pathways to mediate cytoskeleton dynamics is unclear. Vav protein members have been proposed to mediate this process, prompting us to investigate their role in the limb selection of the axon trajectory of spinal lateral motor column (LMC) neurons. RESULTS We found Vav2 and Vav3 expression in LMC neurons when motor axons grew into the limb. Vav2, but not Vav3, loss-of-function perturbed LMC pathfinding, while Vav2 gain-of-function exhibited the opposite effects, demonstrating that Vav2 plays an important role in motor axon growth. Vav2 knockdown also attenuated the redirectional phenotype of LMC axons induced by Dcc, but not EphA4, in vivo and lateral LMC neurite growth preference to Netrin-1 in vitro. This study showed that Vav2 knockdown and ectopic nonphosphorylable Vav2 mutant expression abolished the Src-induced stronger growth preference of lateral LMC neurites to Netrin-1, suggesting that Vav2 is downstream of Src in this context. CONCLUSIONS Vav2 is essential for Netrin-1-regulated LMC motor axon pathfinding through Src interaction.
Collapse
Affiliation(s)
- Yi-Syue Tsou
- The Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei, Taiwan.,Department of Neurosurgery, Taipei Medical University Hospital, Taipei, Taiwan.,Taipei Neuroscience Institute, Taipei Medical University, Taipei, Taiwan
| | - Chih-Yang Wang
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Ming-Yuan Chang
- Division of Neurosurgery, Department of Surgery, Min-Sheng General Hospital, Taoyuan, Taiwan
| | - Tsung-I Hsu
- Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.,Research Center of Neuroscience, Taipei Medical University, Taipei, Taiwan.,TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Meng-Ting Wu
- Department of Neurosurgery, Cheng Hsin General Hospital, Taipei, Taiwan.,Ph.D. Program of Electrical and Communications Engineering, Feng Chia University, Taichung, Taiwan
| | - Yi-Hsin Wu
- Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Wan-Ling Tsai
- The Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei, Taiwan
| | - Jian-Ying Chuang
- Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.,Research Center of Neuroscience, Taipei Medical University, Taipei, Taiwan.,TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Tzu-Jen Kao
- Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.,Research Center of Neuroscience, Taipei Medical University, Taipei, Taiwan.,TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
31
|
Karunakaran U, Elumalai S, Moon JS, Won KC. CD36 Signal Transduction in Metabolic Diseases: Novel Insights and Therapeutic Targeting. Cells 2021; 10:cells10071833. [PMID: 34360006 PMCID: PMC8305429 DOI: 10.3390/cells10071833] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/14/2021] [Accepted: 07/17/2021] [Indexed: 12/24/2022] Open
Abstract
The cluster of differentiation 36 (CD36) is a scavenger receptor present on various types of cells and has multiple biological functions that may be important in inflammation and in the pathogenesis of metabolic diseases, including diabetes. Here, we consider recent insights into how the CD36 response becomes deregulated under metabolic conditions, as well as the therapeutic benefits of CD36 inhibition, which may provide clues for developing strategies aimed at the treatment or prevention of diabetes associated with metabolic diseases. To facilitate this process further, it is important to pinpoint regulatory mechanisms that are relevant under physiological and pathological conditions. In particular, understanding the mechanisms involved in dictating specific CD36 downstream cellular outcomes will aid in the discovery of potent compounds that target specific CD36 downstream signaling cascades.
Collapse
Affiliation(s)
- Udayakumar Karunakaran
- Innovative Center for Aging Research, Yeungnam University Medical Center, Daegu 42415, Korea; (U.K.); (S.E.)
| | - Suma Elumalai
- Innovative Center for Aging Research, Yeungnam University Medical Center, Daegu 42415, Korea; (U.K.); (S.E.)
| | - Jun-Sung Moon
- Innovative Center for Aging Research, Yeungnam University Medical Center, Daegu 42415, Korea; (U.K.); (S.E.)
- Yeungnam University College of Medicine, Daegu 42415, Korea
- Correspondence: (J.-S.M.); (K.-C.W.); Tel.: +82-53-620-3825 (J.-S.M.); +82-53-620-3846 (K.-C.W.)
| | - Kyu-Chang Won
- Innovative Center for Aging Research, Yeungnam University Medical Center, Daegu 42415, Korea; (U.K.); (S.E.)
- Yeungnam University College of Medicine, Daegu 42415, Korea
- Correspondence: (J.-S.M.); (K.-C.W.); Tel.: +82-53-620-3825 (J.-S.M.); +82-53-620-3846 (K.-C.W.)
| |
Collapse
|
32
|
Boudria R, Laurienté V, Oudar A, Harouna-Rachidi S, Dondi E, Le Roy C, Gardano L, Varin-Blank N, Guittat L. Regulatory interplay between Vav1, Syk and β-catenin occurs in lung cancer cells. Cell Signal 2021; 86:110079. [PMID: 34252536 DOI: 10.1016/j.cellsig.2021.110079] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 06/30/2021] [Accepted: 07/05/2021] [Indexed: 01/15/2023]
Abstract
Vav1 exhibits two signal transducing properties as an adaptor protein and a regulator of cytoskeleton organization through its Guanine nucleotide Exchange Factor module. Although the expression of Vav1 is restricted to the hematopoietic lineage, its ectopic expression has been unraveled in a number of solid tumors. In this study, we show that in lung cancer cells, as such in hematopoietic cells, Vav1 interacts with the Spleen Tyrosine Kinase, Syk. Likewise, Syk interacts with β-catenin and, together with Vav1, regulates the phosphorylation status of β-catenin. Depletion of Vav1, Syk or β-catenin inhibits Rac1 activity and decreases cell migration suggesting the interplay of the three effectors to a common signaling pathway. This model is further supported by the finding that in turn, β-catenin regulates the transcription of Syk gene expression. This study highlights the elaborated connection between Vav1, Syk and β-catenin and the contribution of the trio to cell migration.
Collapse
Affiliation(s)
- Rofia Boudria
- INSERM, UMR 978, Bobigny, France; Labex Inflamex, Université Sorbonne Paris Nord, UFR SMBH, Bobigny, France
| | - Vanessa Laurienté
- INSERM, UMR 978, Bobigny, France; Labex Inflamex, Université Sorbonne Paris Nord, UFR SMBH, Bobigny, France
| | - Antonin Oudar
- INSERM, UMR 978, Bobigny, France; Labex Inflamex, Université Sorbonne Paris Nord, UFR SMBH, Bobigny, France
| | - Souleymane Harouna-Rachidi
- INSERM, UMR 978, Bobigny, France; Labex Inflamex, Université Sorbonne Paris Nord, UFR SMBH, Bobigny, France
| | - Elisabetta Dondi
- INSERM, UMR 978, Bobigny, France; Labex Inflamex, Université Sorbonne Paris Nord, UFR SMBH, Bobigny, France
| | - Christine Le Roy
- INSERM, UMR 978, Bobigny, France; Labex Inflamex, Université Sorbonne Paris Nord, UFR SMBH, Bobigny, France
| | - Laura Gardano
- INSERM, UMR 978, Bobigny, France; Labex Inflamex, Université Sorbonne Paris Nord, UFR SMBH, Bobigny, France
| | - Nadine Varin-Blank
- INSERM, UMR 978, Bobigny, France; Labex Inflamex, Université Sorbonne Paris Nord, UFR SMBH, Bobigny, France.
| | - Lionel Guittat
- INSERM, UMR 978, Bobigny, France; Labex Inflamex, Université Sorbonne Paris Nord, UFR SMBH, Bobigny, France.
| |
Collapse
|
33
|
Conde J, Fernández-Pisonero I, Cuadrado M, Abad A, Robles-Valero J, Bustelo XR. Distinct Roles of Vav Family Members in Adaptive and Innate Immune Models of Arthritis. Biomedicines 2021; 9:695. [PMID: 34205377 PMCID: PMC8234068 DOI: 10.3390/biomedicines9060695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/17/2021] [Accepted: 06/18/2021] [Indexed: 12/03/2022] Open
Abstract
Genetic evidence suggests that three members of the VAV family (VAV1, VAV2 and VAV3) of signal transduction proteins could play important roles in rheumatoid arthritis. However, it is not known currently whether the inhibition of these proteins protects against this disease and, if so, the number of family members that must be eliminated to get a therapeutic impact. To address this issue, we have used a collection of single and compound Vav family knockout mice in experimental models for antigen-dependent (methylated bovine serum albumin injections) and neutrophil-dependent (Zymosan A injections) rheumatoid arthritis in mice. We show here that the specific elimination of Vav1 is sufficient to block the development of antigen-induced arthritis. This protection is likely associated with the roles of this Vav family member in the development and selection of immature T cells within the thymus as well as in the subsequent proliferation and differentiation of effector T cells. By contrast, we have found that depletion of Vav2 reduces the number of neutrophils present in the joints of Zymosan A-treated mice. Despite this, the elimination of Vav2 does not protect against the joint degeneration triggered by this experimental model. These findings indicate that Vav1 is the most important pharmacological target within this family, although its main role is limited to the protection against antigen-induced rheumatoid arthritis. They also indicate that the three Vav family proteins do not play redundant roles in these pathobiological processes.
Collapse
Affiliation(s)
- Javier Conde
- Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain; (J.C.); (I.F.-P.); (M.C.); (A.A.); (J.R.-V.)
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain
| | - Isabel Fernández-Pisonero
- Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain; (J.C.); (I.F.-P.); (M.C.); (A.A.); (J.R.-V.)
- 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
| | - Myriam Cuadrado
- Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain; (J.C.); (I.F.-P.); (M.C.); (A.A.); (J.R.-V.)
- 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
| | - Antonio Abad
- Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain; (J.C.); (I.F.-P.); (M.C.); (A.A.); (J.R.-V.)
- 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
| | - Javier Robles-Valero
- Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain; (J.C.); (I.F.-P.); (M.C.); (A.A.); (J.R.-V.)
- 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
- Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain; (J.C.); (I.F.-P.); (M.C.); (A.A.); (J.R.-V.)
- 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
| |
Collapse
|
34
|
Kazim N, Yen A. Role for Fgr and Numb in retinoic acid-induced differentiation and G0 arrest of non-APL AML cells. Oncotarget 2021; 12:1147-1164. [PMID: 34136084 PMCID: PMC8202776 DOI: 10.18632/oncotarget.27969] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 05/03/2021] [Indexed: 12/15/2022] Open
Abstract
Retinoic acid (RA) is a fundamental regulator of cell cycle and cell differentiation. Using a leukemic patient-derived in vitro model of a non-APL AML, we previously found that RA evokes activation of a macromolecular signaling complex, a signalosome, built of numerous MAPK-pathway-related signaling molecules; and this signaling enabled Retinoic-Acid-Response-Elements (RAREs) to regulate gene expression that results in cell differentiation/cell cycle arrest. Toward mechanistic insight into the nature of this novel signaling, we now find that the NUMB cell fate determinant protein is an apparent scaffold for the signalosome. Numb exists in the cell bound to an ensemble of signalosome molecules, including Raf, Lyn, Slp-76, and Vav. Addition of RA induces the expression of Fgr. Fgr binds NUMB, which is associated with (p-tyr)phosphorylation of NUMB and enhanced NUMB-binding and (p-tyr)phosphorylation of select signalosome components, thereby betraying signalosome activation. Signalosome activation is associated with cell differentiation along the myeloid lineage and G1/0 cell cycle arrest. If RA-induced Fgr expression is ablated by a CRISPR-KO; then the RA-induced (p-tyr) phosphorylation of NUMB and enhanced NUMB-binding and (p-tyr)phosphorylation of select signalosome components are lost. The cells now fail to undergo RA-induced differentiation or G1/0 arrest. In sum we find that NUMB acts as a scaffold for a signaling machine that functions to propel RA-induced differentiation and G1/0 arrest, and that Fgr binding to NUMB turns the function on. The Numb fate determinant protein thus appears to regulate the retinoic acid embryonic morphogen using the Fgr Src-Family-Kinase. These mechanistic insights suggest therapeutic targets for a hitherto incurable AML.
Collapse
Affiliation(s)
- Noor Kazim
- Department of Biomedical Science, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Andrew Yen
- Department of Biomedical Science, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
35
|
Raiter A, Zlotnik O, Lipovetsky J, Mugami S, Dar S, Lubin I, Sharon E, Cohen CJ, Yerushalmi R. A novel role for an old target: CD45 for breast cancer immunotherapy. Oncoimmunology 2021; 10:1929725. [PMID: 34104545 PMCID: PMC8158046 DOI: 10.1080/2162402x.2021.1929725] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Breast cancer subtypes have not shown significant response to current immunomodulatory therapies. Although most subtypes are treatable, triple negative breast cancer (TNBC), an aggressive highly metastatic cancer, comprising 10-20% of breast cancers, remains an unmet medical need. New strategies are needed in order to overcome flaws in the responsiveness to current TNBC therapies. Our aims were: first, to determine the efficacy of a novel immunomodulatory peptide, C24D, on TNBC and second, to elucidate the molecular mechanism by which C24D induces immune-modulating tumor killing. Using mass spectrometry analysis, we identified CD45 as the C24D binding receptor. In vitro and in vivo TNBC models were used to assess the efficacy of C24D in reversing TNBC-induced immunosuppression and in triggering immune-modulated tumor cell killing. The CD45 signal transduction pathway was evaluated by western blot and FACS analyses. We revealed that addition of PBMCs from healthy female donors to TNBC cells results in a cascade of suppressive CD45 intracellular signals. On binding to CD45's extra-cellular domain on TNBC-suppressed leukocytes, the C24D peptide re-activates the Src family of tyrosine kinases, resulting in specific tumor immune response. In vitro, immune reactivation by C24D results in an increase of CD69+ T and CD69+ NK cells, triggering specific killing of TNBC cells. In vivo, C24D induced CD8+ and activated CD56+ tumor infiltrated cells, resulting in tumor apoptosis. Our results should renew interest in molecules targeting CD45, such as the C24D peptide, as a novel strategy for TNBC immunotherapy.
Collapse
Affiliation(s)
- Annat Raiter
- Felsenstein Medical Research Center, Sackler School of Medicine, Tel Aviv University, Petach Tikva, Israel
- CONTACT Annat Raiter Felsenstein Medical Research Center, Sackler School of Medicine, Tel Aviv University, Petach Tikva49100, Israel
| | - Oran Zlotnik
- Felsenstein Medical Research Center, Sackler School of Medicine, Tel Aviv University, Petach Tikva, Israel
- Surgery Department, Breast Cancer Unit, Beilinson Hospital, Rabin Medical Center, Petach Tikva, Israel
| | - Julia Lipovetsky
- Felsenstein Medical Research Center, Sackler School of Medicine, Tel Aviv University, Petach Tikva, Israel
| | - Shany Mugami
- Felsenstein Medical Research Center, Sackler School of Medicine, Tel Aviv University, Petach Tikva, Israel
| | - Shira Dar
- Felsenstein Medical Research Center, Sackler School of Medicine, Tel Aviv University, Petach Tikva, Israel
| | - Ido Lubin
- Felsenstein Medical Research Center, Sackler School of Medicine, Tel Aviv University, Petach Tikva, Israel
| | - Eran Sharon
- Surgery Department, Breast Cancer Unit, Beilinson Hospital, Rabin Medical Center, Petach Tikva, Israel
| | - Cyrille J. Cohen
- Laboratory of Tumor Immunotherapy, the Goodman Faculty of Life Sciences, Bar Ilan University, Ramat Gan, Israel
| | - Rinat Yerushalmi
- Felsenstein Medical Research Center, Sackler School of Medicine, Tel Aviv University, Petach Tikva, Israel
- Breast Cancer Unit, Davidoff Cancer Center, Rabin Medical Center, Petach Tikva, Israel
| |
Collapse
|
36
|
Novel Roles of SH2 and SH3 Domains in Lipid Binding. Cells 2021; 10:cells10051191. [PMID: 34068055 PMCID: PMC8152464 DOI: 10.3390/cells10051191] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/10/2021] [Accepted: 05/11/2021] [Indexed: 01/07/2023] Open
Abstract
Signal transduction, the ability of cells to perceive information from the surroundings and alter behavior in response, is an essential property of life. Studies on tyrosine kinase action fundamentally changed our concept of cellular regulation. The induced assembly of subcellular hubs via the recognition of local protein or lipid modifications by modular protein interactions is now a central paradigm in signaling. Such molecular interactions are mediated by specific protein interaction domains. The first such domain identified was the SH2 domain, which was postulated to be a reader capable of finding and binding protein partners displaying phosphorylated tyrosine side chains. The SH3 domain was found to be involved in the formation of stable protein sub-complexes by constitutively attaching to proline-rich surfaces on its binding partners. The SH2 and SH3 domains have thus served as the prototypes for a diverse collection of interaction domains that recognize not only proteins but also lipids, nucleic acids, and small molecules. It has also been found that particular SH2 and SH3 domains themselves might also bind to and rely on lipids to modulate complex assembly. Some lipid-binding properties of SH2 and SH3 domains are reviewed here.
Collapse
|
37
|
Chen Y, André M, Adhikari K, Blin M, Bonfante B, Mendoza-Revilla J, Fuentes-Guajardo M, Palmal S, Chacón-Duque JC, Hurtado M, Villegas V, Granja V, Jaramillo C, Arias W, Lozano RB, Everardo-Martínez P, Gómez-Valdés J, Villamil-Ramírez H, de Cerqueira CCS, Hünemeier T, Ramallo V, Gonzalez-José R, Schüler-Faccini L, Bortolini MC, Acuña-Alonzo V, Canizales-Quinteros S, Gallo C, Poletti G, Bedoya G, Rothhammer F, Balding D, Tobin DJ, Wang S, Faux P, Ruiz-Linares A. A genome-wide association study identifies novel gene associations with facial skin wrinkling and mole count in Latin Americans. Br J Dermatol 2021; 185:988-998. [PMID: 33959940 DOI: 10.1111/bjd.20436] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2021] [Indexed: 12/01/2022]
Abstract
BACKGROUND Genome-wide association studies (GWASs) have identified genes influencing skin ageing and mole count in Europeans, but little is known about the relevance of these (or other genes) in non-Europeans. OBJECTIVES To conduct a GWAS for facial skin ageing and mole count in adults < 40 years old, of mixed European, Native American and African ancestry, recruited in Latin America. METHODS Skin ageing and mole count scores were obtained from facial photographs of over 6000 individuals. After quality control checks, three wrinkling traits and mole count were retained for genetic analyses. DNA samples were genotyped with Illumina's HumanOmniExpress chip. Association testing was performed on around 8 703 729 single-nucleotide polymorphisms (SNPs) across the autosomal genome. RESULTS Genome-wide significant association was observed at four genome regions: two were associated with wrinkling (in 1p13·3 and 21q21·2), one with mole count (in 1q32·3) and one with both wrinkling and mole count (in 5p13·2). Associated SNPs in 5p13·2 and in 1p13·3 are intronic within SLC45A2 and VAV3, respectively, while SNPs in 1q32·3 are near the SLC30A1 gene, and those in 21q21·2 occur in a gene desert. Analyses of SNPs in IRF4 and MC1R are consistent with a role of these genes in skin ageing. CONCLUSIONS We replicate the association of wrinkling with variants in SLC45A2, IRF4 and MC1R reported in Europeans. We identify VAV3 and SLC30A1 as two novel candidate genes impacting on wrinkling and mole count, respectively. We provide the first evidence that SLC45A2 influences mole count, in addition to variants in this gene affecting melanoma risk in Europeans.
Collapse
Affiliation(s)
- Y Chen
- Ministry of Education Key Laboratory of Contemporary Anthropology and Collaborative Innovation Center of Genetics and Development, School of Life Sciences and Human Phenome Institute, Fudan University, Yangpu District, Shanghai, China
| | - M André
- UMR 7268 ADES, CNRS, Aix-Marseille Université, EFS, Faculté de Médecine Timone, Marseille, 13005, France.,Estonian Biocentre, Institute of Genomics, University of Tartu, Tartu, 51010, Estonia
| | - K Adhikari
- School of Mathematics and Statistics, Faculty of Science, Technology, Engineering and Mathematics, The Open University, Milton Keynes, MK7 6AA, UK.,Department of Genetics, Evolution and Environment, and UCL Genetics Institute, University College London, London, WC1E 6BT, UK
| | - M Blin
- UMR 7268 ADES, CNRS, Aix-Marseille Université, EFS, Faculté de Médecine Timone, Marseille, 13005, France
| | - B Bonfante
- UMR 7268 ADES, CNRS, Aix-Marseille Université, EFS, Faculté de Médecine Timone, Marseille, 13005, France
| | - J Mendoza-Revilla
- Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima 31, Perú.,Unit of Human Evolutionary Genetics, Institut Pasteur, Paris, 75015, France
| | - M Fuentes-Guajardo
- Departamento de Tecnología Médica, Facultad de Ciencias de la Salud, Universidad de Tarapacá, Arica, 1000000, Chile
| | - S Palmal
- UMR 7268 ADES, CNRS, Aix-Marseille Université, EFS, Faculté de Médecine Timone, Marseille, 13005, France
| | - J C Chacón-Duque
- Division of Vertebrates and Anthropology, Department of Earth Sciences, Natural History Museum, London, SW7 5BD, UK
| | - M Hurtado
- Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima 31, Perú
| | - V Villegas
- Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima 31, Perú
| | - V Granja
- Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima 31, Perú
| | - C Jaramillo
- GENMOL (Genética Molecular), Universidad de Antioquia, Medellín, 5001000, Colombia
| | - W Arias
- GENMOL (Genética Molecular), Universidad de Antioquia, Medellín, 5001000, Colombia
| | - R B Lozano
- National Institute of Anthropology and History, Mexico City, MC, 6600, Mexico.,Department of Archaeogenetics, Max Planck Institute for the Science of Human History (MPI-SHH), Jena, 07745, Germany
| | - P Everardo-Martínez
- National Institute of Anthropology and History, Mexico City, MC, 6600, Mexico
| | - J Gómez-Valdés
- National Institute of Anthropology and History, Mexico City, MC, 6600, Mexico
| | - H Villamil-Ramírez
- Unidad de Genomica de Poblaciones Aplicada a la Salud, Facultad de Química, UNAM-Instituto Nacional de Medicina Genómica, Mexico City, MC, 4510, Mexico
| | | | - T Hünemeier
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, 05508-090, Brazil
| | - V Ramallo
- Departamento de Genética, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 90040-060, Brazil.,Instituto Patagónico de Ciencias Sociales y Humanas, Centro Nacional Patagónico, CONICET, Puerto Madryn, U9129ACD, Argentina
| | - R Gonzalez-José
- Instituto Patagónico de Ciencias Sociales y Humanas, Centro Nacional Patagónico, CONICET, Puerto Madryn, U9129ACD, Argentina
| | - L Schüler-Faccini
- Departamento de Genética, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 90040-060, Brazil
| | - M-C Bortolini
- Departamento de Genética, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 90040-060, Brazil
| | - V Acuña-Alonzo
- National Institute of Anthropology and History, Mexico City, MC, 6600, Mexico
| | - S Canizales-Quinteros
- Unidad de Genomica de Poblaciones Aplicada a la Salud, Facultad de Química, UNAM-Instituto Nacional de Medicina Genómica, Mexico City, MC, 4510, Mexico
| | - C Gallo
- Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima 31, Perú
| | - G Poletti
- Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima 31, Perú
| | - G Bedoya
- GENMOL (Genética Molecular), Universidad de Antioquia, Medellín, 5001000, Colombia
| | - F Rothhammer
- Instituto de Alta Investigación, Universidad de Tarapacá, Arica, 1000000, Chile
| | - D Balding
- Melbourne Integrative Genomics, Schools of BioSciences and Mathematics & Statistics, University of Melbourne, Melbourne, VIC, 3010, Australia
| | - D J Tobin
- The Charles Institute of Dermatology, University College Dublin, Dublin, Ireland
| | - S Wang
- Chinese Academy of Sciences Key Laboratory of Computational Biology, Shanghai Institutes for Biological Sciences, University of the Chinese Academy of Sciences, Shanghai, 200031, China
| | - P Faux
- UMR 7268 ADES, CNRS, Aix-Marseille Université, EFS, Faculté de Médecine Timone, Marseille, 13005, France
| | - A Ruiz-Linares
- Ministry of Education Key Laboratory of Contemporary Anthropology and Collaborative Innovation Center of Genetics and Development, School of Life Sciences and Human Phenome Institute, Fudan University, Yangpu District, Shanghai, China.,UMR 7268 ADES, CNRS, Aix-Marseille Université, EFS, Faculté de Médecine Timone, Marseille, 13005, France.,Department of Genetics, Evolution and Environment, and UCL Genetics Institute, University College London, London, WC1E 6BT, UK
| |
Collapse
|
38
|
Badaoui M, Zoso A, Idris T, Bacchetta M, Simonin J, Lemeille S, Wehrle-Haller B, Chanson M. Vav3 Mediates Pseudomonas aeruginosa Adhesion to the Cystic Fibrosis Airway Epithelium. Cell Rep 2021; 32:107842. [PMID: 32640241 DOI: 10.1016/j.celrep.2020.107842] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 04/13/2020] [Accepted: 06/09/2020] [Indexed: 01/08/2023] Open
Abstract
Pseudomonas aeruginosa (Pa) represents the leading cause of airway infection in cystic fibrosis (CF). Early airways colonization can be explained by enhanced adhesion of Pa to the respiratory epithelium. RNA sequencing (RNA-seq) on fully differentiated primary cultures of airway epithelial cells from CF and non-CF donors predict that VAV3, β1 INTEGRIN, and FIBRONECTIN genes are significantly enriched in CF. Indeed, Vav3 is apically overexpressed in CF, associates with active β1 integrin luminally exposed, and increases fibronectin deposition. These luminal microdomains, rich in fibronectin and β1 integrin and regulated by Vav3, mediate the increased Pa adhesion to the CF epithelium. Interestingly, Vav3 inhibition normalizes the CF-dependent fibronectin and β1-integrin ectopic expression, improves the CF epithelial integrity, and prevents the enhanced Pa trapping to the CF epithelium. Through its capacity to promote a luminal complex with active β1 integrin and fibronectin that favors bacteria trapping, Vav3 may represent a new target in CF.
Collapse
Affiliation(s)
- Mehdi Badaoui
- Faculty of Medicine, Department of Pediatrics, Gynecology & Obstetrics, University of Geneva, Geneva 1211, Switzerland; Faculty of Medicine, Department of Cell Physiology & Metabolism, University of Geneva, Geneva 1211, Switzerland
| | - Alice Zoso
- Faculty of Medicine, Department of Pediatrics, Gynecology & Obstetrics, University of Geneva, Geneva 1211, Switzerland; Faculty of Medicine, Department of Cell Physiology & Metabolism, University of Geneva, Geneva 1211, Switzerland
| | - Tahir Idris
- Faculty of Medicine, Department of Pediatrics, Gynecology & Obstetrics, University of Geneva, Geneva 1211, Switzerland; Faculty of Medicine, Department of Cell Physiology & Metabolism, University of Geneva, Geneva 1211, Switzerland
| | - Marc Bacchetta
- Faculty of Medicine, Department of Pediatrics, Gynecology & Obstetrics, University of Geneva, Geneva 1211, Switzerland; Faculty of Medicine, Department of Cell Physiology & Metabolism, University of Geneva, Geneva 1211, Switzerland
| | - Juliette Simonin
- Faculty of Medicine, Department of Pediatrics, Gynecology & Obstetrics, University of Geneva, Geneva 1211, Switzerland; Faculty of Medicine, Department of Cell Physiology & Metabolism, University of Geneva, Geneva 1211, Switzerland
| | - Sylvain Lemeille
- Faculty of Medicine, Department of Pathology and Immunology, University of Geneva, Geneva 1211, Switzerland
| | - Bernhard Wehrle-Haller
- Faculty of Medicine, Department of Cell Physiology & Metabolism, University of Geneva, Geneva 1211, Switzerland
| | - Marc Chanson
- Faculty of Medicine, Department of Pediatrics, Gynecology & Obstetrics, University of Geneva, Geneva 1211, Switzerland; Faculty of Medicine, Department of Cell Physiology & Metabolism, University of Geneva, Geneva 1211, Switzerland.
| |
Collapse
|
39
|
Machin PA, Tsonou E, Hornigold DC, Welch HCE. Rho Family GTPases and Rho GEFs in Glucose Homeostasis. Cells 2021; 10:cells10040915. [PMID: 33923452 PMCID: PMC8074089 DOI: 10.3390/cells10040915] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/11/2021] [Accepted: 04/13/2021] [Indexed: 12/17/2022] Open
Abstract
Dysregulation of glucose homeostasis leading to metabolic syndrome and type 2 diabetes is the cause of an increasing world health crisis. New intriguing roles have emerged for Rho family GTPases and their Rho guanine nucleotide exchange factor (GEF) activators in the regulation of glucose homeostasis. This review summates the current knowledge, focusing in particular on the roles of Rho GEFs in the processes of glucose-stimulated insulin secretion by pancreatic β cells and insulin-stimulated glucose uptake into skeletal muscle and adipose tissues. We discuss the ten Rho GEFs that are known so far to regulate glucose homeostasis, nine of which are in mammals, and one is in yeast. Among the mammalian Rho GEFs, P-Rex1, Vav2, Vav3, Tiam1, Kalirin and Plekhg4 were shown to mediate the insulin-stimulated translocation of the glucose transporter GLUT4 to the plasma membrane and/or insulin-stimulated glucose uptake in skeletal muscle or adipose tissue. The Rho GEFs P-Rex1, Vav2, Tiam1 and β-PIX were found to control the glucose-stimulated release of insulin by pancreatic β cells. In vivo studies demonstrated the involvement of the Rho GEFs P-Rex2, Vav2, Vav3 and PDZ-RhoGEF in glucose tolerance and/or insulin sensitivity, with deletion of these GEFs either contributing to the development of metabolic syndrome or protecting from it. This research is in its infancy. Considering that over 80 Rho GEFs exist, it is likely that future research will identify more roles for Rho GEFs in glucose homeostasis.
Collapse
Affiliation(s)
- Polly A. Machin
- Signalling Programme, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK; (P.A.M.); (E.T.)
| | - Elpida Tsonou
- Signalling Programme, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK; (P.A.M.); (E.T.)
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Cambridge CB22 3AT, UK;
| | - David C. Hornigold
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Cambridge CB22 3AT, UK;
| | - Heidi C. E. Welch
- Signalling Programme, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK; (P.A.M.); (E.T.)
- Correspondence: ; Tel.: +44-(0)1223-496-596
| |
Collapse
|
40
|
Benito-Vicente A, Jebari-Benslaiman S, Galicia-Garcia U, Larrea-Sebal A, Uribe KB, Martin C. Molecular mechanisms of lipotoxicity-induced pancreatic β-cell dysfunction. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021; 359:357-402. [PMID: 33832653 DOI: 10.1016/bs.ircmb.2021.02.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Type 2 diabetes (T2D), a heterogeneous disorder derived from metabolic dysfunctions, leads to a glucose overflow in the circulation due to both defective insulin secretion and peripheral insulin resistance. One of the critical risk factor for T2D is obesity, which represents a global epidemic that has nearly tripled since 1975. Obesity is characterized by chronically elevated free fatty acid (FFA) levels, which cause deleterious effects on glucose homeostasis referred to as lipotoxicity. Here, we review the physiological FFA roles onto glucose-stimulated insulin secretion (GSIS) and the pathological ones affecting many steps of the mechanisms and modulation of GSIS. We also describe in vitro and in vivo experimental evidences addressing lipotoxicity in β-cells and the role of saturation and chain length of FFA on the potency of GSIS stimulation. The molecular mechanisms underpinning lipotoxic-β-cell dysfunction are also reviewed. Among them, endoplasmic reticulum stress, oxidative stress and mitochondrial dysfunction, inflammation, impaired autophagy and β-cell dedifferentiation. Finally therapeutic strategies for the β-cells dysfunctions such as the use of metformin, glucagon-like peptide 1, thiazolidinediones, anti-inflammatory drugs, chemical chaperones and weight are discussed.
Collapse
Affiliation(s)
- Asier Benito-Vicente
- Department of Molecular Biophysics, Biofisika Institute (University of Basque Country and Consejo Superior de Investigaciones Científicas (UPV/EHU, CSIC)), Leioa, Spain; Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Shifa Jebari-Benslaiman
- Department of Molecular Biophysics, Biofisika Institute (University of Basque Country and Consejo Superior de Investigaciones Científicas (UPV/EHU, CSIC)), Leioa, Spain; Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Unai Galicia-Garcia
- Department of Molecular Biophysics, Biofisika Institute (University of Basque Country and Consejo Superior de Investigaciones Científicas (UPV/EHU, CSIC)), Leioa, Spain; Department of Molecular Biophysics, Fundación Biofísica Bizkaia, Leioa, Spain
| | - Asier Larrea-Sebal
- Department of Molecular Biophysics, Biofisika Institute (University of Basque Country and Consejo Superior de Investigaciones Científicas (UPV/EHU, CSIC)), Leioa, Spain; Department of Molecular Biophysics, Fundación Biofísica Bizkaia, Leioa, Spain
| | - Kepa B Uribe
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia San Sebastián, Spain
| | - Cesar Martin
- Department of Molecular Biophysics, Biofisika Institute (University of Basque Country and Consejo Superior de Investigaciones Científicas (UPV/EHU, CSIC)), Leioa, Spain; Department of Biochemistry and Molecular Biology, University of the Basque Country (UPV/EHU), Leioa, Spain.
| |
Collapse
|
41
|
Biochemical and NMR characterization of the interactions of Vav2-SH2 domain with lipids and the EphA2 juxtamembrane region on membrane. Biochem J 2021; 477:3791-3801. [PMID: 32897354 DOI: 10.1042/bcj20200300] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 09/05/2020] [Accepted: 09/07/2020] [Indexed: 12/21/2022]
Abstract
Vav2 is a ubiquitous guanine nucleotide exchange factor (GEF) for Rho family GTPases that is involved in regulating a wide range of biological processes. It interacts with several tyrosine-phosphorylated cell surface receptors, including the Eph family receptors, through its SH2 domain. The interaction of Vav2 with EphA2 is crucial for EphA2-mediated tumor angiogenesis. Here we show that Vav2-SH2 domain is a lipid-binding module that can recognize PI(4,5)P2 and PI(3,4,5)P3 lipids weakly but specifically. The specific lipid-binding site in Vav2-SH2 domain was identified by NMR chemical shift perturbation experiments using the head groups of PI(4,5)P2 and PI(3,4,5)P3, both of which bind to Vav2-SH2 with millimolar binding affinities. In addition, the interaction between Vav2-SH2 and the phosphorylated juxtamembrane region (JM) of EphA2 (Y594 phosphorylated) was investigated using NMR techniques. Furthermore, by using a nickel-lipid containing peptide-based nanodiscs system, we studied the binding of Vav2-SH2 to the phosphorylated JM region of EphA2 on lipid membrane and uncovered a role of membrane environment in modulating this protein-protein recognition.
Collapse
|
42
|
Mineo C. Lipoprotein receptor signalling in atherosclerosis. Cardiovasc Res 2021; 116:1254-1274. [PMID: 31834409 DOI: 10.1093/cvr/cvz338] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 11/01/2019] [Accepted: 12/10/2019] [Indexed: 12/11/2022] Open
Abstract
The founding member of the lipoprotein receptor family, low-density lipoprotein receptor (LDLR) plays a major role in the atherogenesis through the receptor-mediated endocytosis of LDL particles and regulation of cholesterol homeostasis. Since the discovery of the LDLR, many other structurally and functionally related receptors have been identified, which include low-density lipoprotein receptor-related protein (LRP)1, LRP5, LRP6, very low-density lipoprotein receptor, and apolipoprotein E receptor 2. The scavenger receptor family members, on the other hand, constitute a family of pattern recognition proteins that are structurally diverse and recognize a wide array of ligands, including oxidized LDL. Among these are cluster of differentiation 36, scavenger receptor class B type I and lectin-like oxidized low-density lipoprotein receptor-1. In addition to the initially assigned role as a mediator of the uptake of macromolecules into the cell, a large number of studies in cultured cells and in in vivo animal models have revealed that these lipoprotein receptors participate in signal transduction to modulate cellular functions. This review highlights the signalling pathways by which these receptors influence the process of atherosclerosis development, focusing on their roles in the vascular cells, such as macrophages, endothelial cells, smooth muscle cells, and platelets. Human genetics of the receptors is also discussed to further provide the relevance to cardiovascular disease risks in humans. Further knowledge of the vascular biology of the lipoprotein receptors and their ligands will potentially enhance our ability to harness the mechanism to develop novel prophylactic and therapeutic strategies against cardiovascular diseases.
Collapse
Affiliation(s)
- Chieko Mineo
- Department of Pediatrics and Cell Biology, Center for Pulmonary and Vascular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9063, USA
| |
Collapse
|
43
|
Large-Scale Proteomic Analysis of Follicular Lymphoma Reveals Extensive Remodeling of Cell Adhesion Pathway and Identifies Hub Proteins Related to the Lymphomagenesis. Cancers (Basel) 2021; 13:cancers13040630. [PMID: 33562532 PMCID: PMC7915278 DOI: 10.3390/cancers13040630] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/27/2021] [Accepted: 02/01/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Follicular lymphoma represents the major subtype of indolent B-cell non-Hodgkin lymphomas, ranging from about 20 to 30% of all B-NHLs cases in western countries. Yet, the global proteome profile of follicular lymphoma remains largely undocumented; thus, we aimed to employ for the first time a comprehensive proteomic analysis to outline its molecular landscape. A total of 15 lymphoma fine-needle aspiration biopsy samples and 14 controls were evaluated by label-free quantitative proteomics. Among the 7673 proteins identified in our dataset, 1186 proteins were differentially expressed between lymphoma and control samples. Importantly, dysregulated proteins were enriched in biological processes such as B-cell receptor signaling pathway, cellular adhesion molecules pathway, or membrane trafficking. Additionally, we identified several novel hub proteins related to lymphomagenesis. To summarize, we have determined the molecular characteristics of follicular lymphoma and discovered proteins which may hold potential for biomarkers or therapeutic targets. Abstract Follicular lymphoma (FL) represents the major subtype of indolent B-cell non-Hodgkin lymphomas (B-NHLs) and results from the malignant transformation of mature B-cells in lymphoid organs. Although gene expression and genomic studies have identified multiple disease driving gene aberrations, only a few proteomic studies focused on the protein level. The present work aimed to examine the proteomic profiles of follicular lymphoma vs. normal B-cells obtained by fine-needle aspiration biopsy (FNAB) to gain deep insight into the most perturbed pathway of FL. The cells of interest were purified by magnetic-activated cell sorting (MACS). High-throughput proteomic profiling was performed using liquid chromatography-tandem mass spectrometry (LC-MS/MS) and allowed to identify of 6724 proteins in at least 75% of each group of samples. The ‘Total Protein Approach’ (TPA) was applied to the absolute quantification of proteins in this study. We identified 1186 differentially abundant proteins (DAPs) between FL and control samples, causing an extensive remodeling of several molecular pathways, including the B-cell receptor signaling pathway, cellular adhesion molecules, and PPAR pathway. Additionally, the construction of protein–protein interactions networks (PPINs) and identification of hub proteins allowed us to indicate the key player proteins for FL pathology. Finally, ICAM1, CD9, and CD79B protein expression was validated in an independent cohort by flow cytometry (FCM), and the results were consistent with the mass spectrometry (MS) data.
Collapse
|
44
|
Hawkins MB, Henke K, Harris MP. Latent developmental potential to form limb-like skeletal structures in zebrafish. Cell 2021; 184:899-911.e13. [PMID: 33545089 DOI: 10.1016/j.cell.2021.01.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 10/28/2020] [Accepted: 01/04/2021] [Indexed: 12/13/2022]
Abstract
Changes in appendage structure underlie key transitions in vertebrate evolution. Addition of skeletal elements along the proximal-distal axis facilitated critical transformations, including the fin-to-limb transition that permitted generation of diverse modes of locomotion. Here, we identify zebrafish mutants that form supernumerary long bones in their pectoral fins. These new bones integrate into musculature, form joints, and articulate with neighboring elements. This phenotype is caused by activating mutations in previously unrecognized regulators of appendage patterning, vav2 and waslb, that function in a common pathway. This pathway is required for appendage development across vertebrates, and loss of Wasl in mice causes defects similar to those seen in murine Hox mutants. Concordantly, formation of supernumerary bones requires Hox11 function, and mutations in the vav2/wasl pathway drive enhanced expression of hoxa11b, indicating developmental homology with the forearm. Our findings reveal a latent, limb-like pattern ability in fins that is activated by simple genetic perturbation.
Collapse
Affiliation(s)
- M Brent Hawkins
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Department of Orthopedic Research, Boston Children's Hospital, Boston, MA 02115, USA; Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA; Museum of Comparative Zoology, Harvard University, Cambridge, MA 02138, USA
| | - Katrin Henke
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Department of Orthopedic Research, Boston Children's Hospital, Boston, MA 02115, USA
| | - Matthew P Harris
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Department of Orthopedic Research, Boston Children's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
45
|
Pickering KA, Gilroy K, Cassidy JW, Fey SK, Najumudeen AK, Zeiger LB, Vincent DF, Gay DM, Johansson J, Fordham RP, Miller B, Clark W, Hedley A, Unal EB, Kiel C, McGhee E, Machesky LM, Nixon C, Johnsson AE, Bain M, Strathdee D, van Hoof SR, Medema JP, Anderson KI, Brachmann SM, Stucke VM, Malliri A, Drysdale M, Turner M, Serrano L, Myant K, Campbell AD, Sansom OJ. A RAC-GEF network critical for early intestinal tumourigenesis. Nat Commun 2021; 12:56. [PMID: 33397922 PMCID: PMC7782582 DOI: 10.1038/s41467-020-20255-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 11/17/2020] [Indexed: 01/29/2023] Open
Abstract
RAC1 activity is critical for intestinal homeostasis, and is required for hyperproliferation driven by loss of the tumour suppressor gene Apc in the murine intestine. To avoid the impact of direct targeting upon homeostasis, we reasoned that indirect targeting of RAC1 via RAC-GEFs might be effective. Transcriptional profiling of Apc deficient intestinal tissue identified Vav3 and Tiam1 as key targets. Deletion of these indicated that while TIAM1 deficiency could suppress Apc-driven hyperproliferation, it had no impact upon tumourigenesis, while VAV3 deficiency had no effect. Intriguingly, deletion of either gene resulted in upregulation of Vav2, with subsequent targeting of all three (Vav2-/- Vav3-/- Tiam1-/-), profoundly suppressing hyperproliferation, tumourigenesis and RAC1 activity, without impacting normal homeostasis. Critically, the observed RAC-GEF dependency was negated by oncogenic KRAS mutation. Together, these data demonstrate that while targeting RAC-GEF molecules may have therapeutic impact at early stages, this benefit may be lost in late stage disease.
Collapse
Affiliation(s)
- K A Pickering
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - K Gilroy
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - J W Cassidy
- CRUK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 ORE, UK
| | - S K Fey
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1QH, UK
| | - A K Najumudeen
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - L B Zeiger
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1QH, UK
| | - D F Vincent
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - D M Gay
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - J Johansson
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - R P Fordham
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - B Miller
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - W Clark
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - A Hedley
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - E B Unal
- EMBL/CRG Systems Biology Research Unit, Centre for Genomic Regulation (CRC), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), 08003, Barcelona, Spain
- Institute for Theoretical Biology, Humboldt Universität zu Berlin, Berlin, Germany
| | - C Kiel
- EMBL/CRG Systems Biology Research Unit, Centre for Genomic Regulation (CRC), Barcelona, Spain
| | - E McGhee
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - L M Machesky
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1QH, UK
| | - C Nixon
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - A E Johnsson
- The Babraham Institute, Babraham Hall, Babraham, Cambridge, CB22 3AT, UK
| | - M Bain
- IBAHCM and School of Veterinary Medicine, 464 Bearsden Road, Bearsden, Glasgow, G61 1QH, UK
| | - D Strathdee
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - S R van Hoof
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental Molecular Medicine (CEMM) and Cancer Center Amsterdam, Academic Medical Center, Amsterdam, The Netherlands
- Oncode Institute, Academic Medical Center, Amsterdam, The Netherlands
| | - J P Medema
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental Molecular Medicine (CEMM) and Cancer Center Amsterdam, Academic Medical Center, Amsterdam, The Netherlands
- Oncode Institute, Academic Medical Center, Amsterdam, The Netherlands
| | - K I Anderson
- The Francis Crick Institute, Mill Hill Laboratory, London, NW7 1AA, UK
| | - S M Brachmann
- Novartis Institutes for BioMedical Research, Klybeckstrasse, 141, 4002, Basel, Switzerland
| | - V M Stucke
- Novartis Institutes for BioMedical Research, Klybeckstrasse, 141, 4002, Basel, Switzerland
| | - A Malliri
- CRUK Manchester Institute, 553 Wilmslow Road, Manchester, M20 4BX, UK
| | - M Drysdale
- Broad Institute, 415 Main St, Cambridge, MA, 02142, United States
| | - M Turner
- The Babraham Institute, Babraham Hall, Babraham, Cambridge, CB22 3AT, UK
| | - L Serrano
- EMBL/CRG Systems Biology Research Unit, Centre for Genomic Regulation (CRC), Barcelona, Spain
| | - K Myant
- Edinburgh Research Centre, The Institute of Genetics and Molecular Medicine, Crewe Road South, Edinburgh, EH4 2XR, UK.
| | - A D Campbell
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK.
| | - O J Sansom
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK.
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1QH, UK.
| |
Collapse
|
46
|
Vav2 catalysis-dependent pathways contribute to skeletal muscle growth and metabolic homeostasis. Nat Commun 2020; 11:5808. [PMID: 33199701 PMCID: PMC7669868 DOI: 10.1038/s41467-020-19489-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 10/16/2020] [Indexed: 12/17/2022] Open
Abstract
Skeletal muscle promotes metabolic balance by regulating glucose uptake and the stimulation of multiple interorgan crosstalk. We show here that the catalytic activity of Vav2, a Rho GTPase activator, modulates the signaling output of the IGF1- and insulin-stimulated phosphatidylinositol 3-kinase pathway in that tissue. Consistent with this, mice bearing a Vav2 protein with decreased catalytic activity exhibit reduced muscle mass, lack of proper insulin responsiveness and, at much later times, a metabolic syndrome-like condition. Conversely, mice expressing a catalytically hyperactive Vav2 develop muscle hypertrophy and increased insulin responsiveness. Of note, while hypoactive Vav2 predisposes to, hyperactive Vav2 protects against high fat diet-induced metabolic imbalance. These data unveil a regulatory layer affecting the signaling output of insulin family factors in muscle. Skeletal muscle plays a key role in regulating systemic glucose and metabolic homeostasis. Here, the authors show that the catalytic activity of Vav2, an activator of Rho GTPases, modulates those processes by favoring the responsiveness of this tissue to insulin and related factors.
Collapse
|
47
|
Yamamoto R, Ohnishi H, Omori K, Yamamoto N. In silico analysis of inner ear development using public whole embryonic body single-cell RNA-sequencing data. Dev Biol 2020; 469:160-171. [PMID: 33131705 DOI: 10.1016/j.ydbio.2020.10.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 10/19/2020] [Accepted: 10/20/2020] [Indexed: 02/02/2023]
Abstract
The inner ear comprises four epithelial domains: the cochlea, vestibule, semicircular canals, and endolymphatic duct/sac. These structures are segregated at embryonic day 13.5 (E13.5). However, these four anatomical structures remain undefined at E10.5. Here, we aimed to identify lineage-specific genes in the early developing inner ear using published data obtained from single-cell RNA-sequencing (scRNA-seq) of embryonic mice. We downloaded 5000 single-cell transcriptome data, named 'auditory epithelial trajectory', from the Mouse Organogenesis Cell Atlas. The dataset was supposed to include otic epithelial cells at E9.5-13.5. We projected the 5000 cells onto a two-dimensional space encoding the transcriptional state and visualised the pattern of otic epithelial cell differentiation. We identified 15 clusters, which were annotated as one of the four components of the inner ear epithelium using known genes that characterise the four different tissues. Additionally, we classified 15 clusters into sub-regions of the four inner ear components. By comparing transcriptomes between these 15 clusters, we identified several candidates of lineage-specific genes. Characterising these new candidate genes will help future studies about inner ear development.
Collapse
Affiliation(s)
- Ryosuke Yamamoto
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, 54, Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 6068507, Japan.
| | - Hiroe Ohnishi
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, 54, Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 6068507, Japan.
| | - Koichi Omori
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, 54, Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 6068507, Japan.
| | - Norio Yamamoto
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, 54, Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 6068507, Japan.
| |
Collapse
|
48
|
Lorenzo-Martín LF, Fernández-Parejo N, Menacho-Márquez M, Rodríguez-Fdez S, Robles-Valero J, Zumalave S, Fabbiano S, Pascual G, García-Pedrero JM, Abad A, García-Macías MC, González N, Lorenzano-Menna P, Pavón MA, González-Sarmiento R, Segrelles C, Paramio JM, Tubío JMC, Rodrigo JP, Benitah SA, Cuadrado M, Bustelo XR. VAV2 signaling promotes regenerative proliferation in both cutaneous and head and neck squamous cell carcinoma. Nat Commun 2020; 11:4788. [PMID: 32963234 PMCID: PMC7508832 DOI: 10.1038/s41467-020-18524-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 08/27/2020] [Indexed: 12/30/2022] Open
Abstract
Regenerative proliferation capacity and poor differentiation are histological features usually linked to poor prognosis in head and neck squamous cell carcinoma (hnSCC). However, the pathways that regulate them remain ill-characterized. Here, we show that those traits can be triggered by the RHO GTPase activator VAV2 in keratinocytes present in the skin and oral mucosa. VAV2 is also required to maintain those traits in hnSCC patient-derived cells. This function, which is both catalysis- and RHO GTPase-dependent, is mediated by c-Myc- and YAP/TAZ-dependent transcriptomal programs associated with regenerative proliferation and cell undifferentiation, respectively. High levels of VAV2 transcripts and VAV2-regulated gene signatures are both associated with poor hnSCC patient prognosis. These results unveil a druggable pathway linked to the malignancy of specific SCC subtypes. The Rho signalling pathway is frequently activated in squamous carcinomas. Here, the authors find that the Rho GEF VAV2 is over expressed in both cutaneous and head and neck squamous cell carcinomas and that at the molecular level VAV2 promotes a pro-tumorigenic stem cell-like signalling programme.
Collapse
Affiliation(s)
- L 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
| | - Natalia Fernández-Parejo
- 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
| | - 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.,Instituto de Inmunología Clínica y Experimental de Rosario (IDICER, CONICET-UNR). Facultad de Ciencias Médicas Universidad Nacional de Rosario (M.M.-M.) and CellPress editorial office (S.F.), S2000LRJ, Rosario, Argentina
| | - Sonia Rodríguez-Fdez
- 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
| | - Javier Robles-Valero
- 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
| | - Sonia Zumalave
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Salvatore Fabbiano
- 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.,Instituto de Inmunología Clínica y Experimental de Rosario (IDICER, CONICET-UNR). Facultad de Ciencias Médicas Universidad Nacional de Rosario (M.M.-M.) and CellPress editorial office (S.F.), S2000LRJ, Rosario, Argentina
| | - Gloria Pascual
- Institute for Research in Biomedicine, 33011, Barcelona, Spain.,The Barcelona Institute of Science and Technology, Barcelona, 33011, Spain
| | - Juana M García-Pedrero
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, 37007, Salamanca, Spain.,Hospital Universitario Central de Asturias, Oviedo University, 33011, Oviedo, Spain
| | - Antonio Abad
- 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
| | - María C García-Macías
- 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
| | - Nazareno González
- 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
| | - Pablo Lorenzano-Menna
- Laboratory of Molecular Oncology and National University of Quilmes, Buenos Aires, B1876BXD, Argentina.,National Council of Scientific and Technical Research (CONICET), National University of Quilmes, Buenos Aires, B1876BXD, Argentina
| | - Miguel A Pavón
- Institut Català d'Oncologia, 08908, L'Hospitalet de Llobregat, Spain.,Centro Biomédica de Investigación en Red de Enfermedades Respiratorias (CIBERESP), 08908, L'Hospitalet de Llobregat, Spain
| | - Rogelio González-Sarmiento
- 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.,Instituto de Investigación Biomédica de Salamanca, 37007, Salamanca, Spain
| | - Carmen Segrelles
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, 37007, Salamanca, Spain.,Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas, 28040, Madrid, Spain
| | - Jesús M Paramio
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, 37007, Salamanca, Spain.,Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas, 28040, Madrid, Spain
| | - José M C Tubío
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Juan P Rodrigo
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, 37007, Salamanca, Spain.,Hospital Universitario Central de Asturias, Oviedo University, 33011, Oviedo, Spain
| | - Salvador A Benitah
- Institute for Research in Biomedicine, 33011, Barcelona, Spain.,The Barcelona Institute of Science and Technology, Barcelona, 33011, Spain.,Catalan Institution for Research and Advanced Studies (ICREA), 33011, Barcelona, Spain
| | - Myriam Cuadrado
- 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.
| |
Collapse
|
49
|
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: 1.0] [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
|
50
|
Löber J, Hitzing C, Münchhalfen M, Engels N. Vav family proteins constitute disparate branching points for distinct BCR signaling pathways. Eur J Immunol 2020; 50:1912-1928. [PMID: 32671844 DOI: 10.1002/eji.202048621] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 05/18/2020] [Accepted: 07/10/2020] [Indexed: 01/16/2023]
Abstract
Antigen recognition by B-cell antigen receptors (BCRs) activates distinct intracellular signaling pathways that control the differentiation fate of activated B lymphocytes. BCR-proximal signaling enzymes comprise protein tyrosine kinases, phosphatases, and plasma membrane lipid-modifying enzymes, whose function is furthermore coordinated by catalytically inert adaptor proteins. Here, we show that an additional class of enzymatic activity provided by guanine-nucleotide exchange factors (GEFs) of the Vav family controls BCR-proximal Ca2+ mobilization, cytoskeletal actin reorganization, and activation of the PI3 kinase/Akt pathway. Whereas Vav1 and Vav3 supported all of those signaling processes to different extents in a human B-cell model system, Vav2 facilitated Actin remodeling, and activation of Akt but did not promote Ca2+ signaling. On BCR activation, Vav1 was directly recruited to the phosphorylated BCR and to the central adaptor protein SLP65 via its Src homology 2 domain. Pharmacological inhibition or genetic inactivation of the substrates of Vav GEFs, small G proteins of the Rho/Rac family, impaired BCR-induced Ca2+ mobilization, probably because phospholipase Cγ2 requires activated Rac proteins for optimal activity. Our findings show that Vav family members are key relays of the BCR signalosome that differentially control distinct signaling pathways both in a catalysis-dependent and -independent manner.
Collapse
Affiliation(s)
- Jens Löber
- Institute of Cellular & Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Christoffer Hitzing
- Institute of Cellular & Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Matthias Münchhalfen
- Institute of Cellular & Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Niklas Engels
- Institute of Cellular & Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|