1
|
Neurath MF, Berg LJ. VAV1 as a putative therapeutic target in autoimmune and chronic inflammatory diseases. Trends Immunol 2024; 45:580-596. [PMID: 39060140 DOI: 10.1016/j.it.2024.06.004] [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: 06/07/2024] [Revised: 06/24/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024]
Abstract
The guanine nucleotide exchange factor (GEF) VAV1, a previously 'undruggable' protein integral to T/B lymphocyte antigen-receptor signaling, promotes actin polymerization, immunological synapse formation, T cell activation and differentiation, and cytokine production. With the development of novel modalities for targeting proteins, we hypothesize that interventions targeting VAV1 will have therapeutic potential in T and T/B cell-mediated autoimmune and chronic inflammatory diseases. This opinion is supported by recent CRISPR-Cas9 studies showing VAV1 as a key positive regulator of T cell receptor (TCR) activation and cytokine production in primary human CD4+ and CD8+ T cells; data demonstrating that loss/suppression of VAV1 regulates autoimmunity and inflammation; and promising preclinical data from T and T/B cell-mediated disease models of arthritis and colitis showing the effectiveness of selective VAV1 targeting via protein degradation.
Collapse
Affiliation(s)
- Markus F Neurath
- Department of Medicine, 1 & Deutsches Zentrum Immuntherapie, University of Erlangen-Nürnberg, Kussmaul Campus for Medical Research, 91054 Erlangen, Germany
| | - Leslie J Berg
- Department of Immunology & Microbiology, University of Colorado Anschutz School of Medicine, Aurora, CO 80045, USA.
| |
Collapse
|
2
|
Wu M, Chen Y, Chiu I, Wu M. Genetic Insight into Primary Glomerulonephritis. Nephrology (Carlton) 2022; 27:649-657. [DOI: 10.1111/nep.14074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 04/18/2022] [Accepted: 06/06/2022] [Indexed: 11/28/2022]
Affiliation(s)
- Mei‐Yi Wu
- Division of Nephrology, Department of Internal Medicine Taipei Medical University‐Shuang Ho Hospital New Taipei City Taiwan
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine Taipei Medical University Taipei Taiwan
- Institute of Epidemiology and Preventive Medicine, College of Public Health National Taiwan University Taipei Taiwan
- TMU Research Center of Urology and Kidney Taipei Medical University Taipei Taiwan
| | - Ying‐Chun Chen
- Division of Nephrology, Department of Internal Medicine Taipei Medical University‐Shuang Ho Hospital New Taipei City Taiwan
| | - I‐Jen Chiu
- Division of Nephrology, Department of Internal Medicine Taipei Medical University‐Shuang Ho Hospital New Taipei City Taiwan
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine Taipei Medical University Taipei Taiwan
- TMU Research Center of Urology and Kidney Taipei Medical University Taipei Taiwan
| | - Mai‐Szu Wu
- Division of Nephrology, Department of Internal Medicine Taipei Medical University‐Shuang Ho Hospital New Taipei City Taiwan
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine Taipei Medical University Taipei Taiwan
- TMU Research Center of Urology and Kidney Taipei Medical University Taipei Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine Taipei Medical University Taipei Taiwan
| |
Collapse
|
3
|
Nuclear Vav3 is required for polycomb repression complex-1 activity in B-cell lymphoblastic leukemogenesis. Nat Commun 2022; 13:3056. [PMID: 35650206 PMCID: PMC9160250 DOI: 10.1038/s41467-022-30651-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 05/10/2022] [Indexed: 12/23/2022] Open
Abstract
Acute B-cell lymphoblastic leukemia (B-ALL) results from oligo-clonal evolution of B-cell progenitors endowed with initiating and propagating leukemia properties. The activation of both the Rac guanine nucleotide exchange factor (Rac GEF) Vav3 and Rac GTPases is required for leukemogenesis mediated by the oncogenic fusion protein BCR-ABL. Vav3 expression becomes predominantly nuclear upon expression of BCR-ABL signature. In the nucleus, Vav3 interacts with BCR-ABL, Rac, and the polycomb repression complex (PRC) proteins Bmi1, Ring1b and Ezh2. The GEF activity of Vav3 is required for the proliferation, Bmi1-dependent B-cell progenitor self-renewal, nuclear Rac activation, protein interaction with Bmi1, mono-ubiquitination of H2A(K119) (H2AK119Ub) and repression of PRC-1 (PRC1) downstream target loci, of leukemic B-cell progenitors. Vav3 deficiency results in de-repression of negative regulators of cell proliferation and repression of oncogenic transcriptional factors. Mechanistically, we show that Vav3 prevents the Phlpp2-sensitive and Akt (S473)-dependent phosphorylation of Bmi1 on the regulatory residue S314 that, in turn, promotes the transcriptional factor reprogramming of leukemic B-cell progenitors. These results highlight the importance of non-canonical nuclear Rho GTPase signaling in leukemogenesis. Ph+ and Ph-like B-ALL remain poor prognosis leukemias. VAV3, a guanine nucleotide exchange factor, is activated and overexpressed in these leukemias. Here the authors reveal that leukemic VAV3 is predominantly nuclear. Nuclear VAV3, through its guanine nucleotide exchange factor and its effector nuclear RAC2, controls the repressive transcriptional activity of the polycomb repression complex-1 via nuclear AKT/PHLPP2 regulated BMI1.
Collapse
|
4
|
Link between the EZH2 noncanonical pathway and microtubule organization center polarization during early T lymphopoiesis. Sci Rep 2022; 12:3655. [PMID: 35256668 PMCID: PMC8901749 DOI: 10.1038/s41598-022-07684-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 02/22/2022] [Indexed: 12/11/2022] Open
Abstract
EZH2 plays an essential role at the β-selection checkpoint of T lymphopoiesis by regulating histone H3 lysine 27 trimethylation (H3K27me3) via its canonical mode of action. Increasing data suggest that EZH2 could also regulate other cellular functions, such as cytoskeletal reorganization, via its noncanonical pathway. Consequently, we investigated whether the EZH2 noncanonical pathway could be involved in early T-cell maturation, which requires cell polarization. We observed that EZH2 localization is tightly regulated during the early stages of T-cell development and that EZH2 relocalizes in the nucleus of double-negative thymocytes enduring TCRβ recombination and β-selection processes. Furthermore, we observed that EZH2 and EED, but not Suz12, colocalize with the microtubule organization center (MTOC), which might prevent its inappropriate polarization in double negative cells. In accordance with these results, we evidenced the existence of direct or indirect interaction between EED and α-tubulin. Taken together, these results suggest that the EZH2 noncanonical pathway, in association with EED, is involved in the early stages of T-cell maturation.
Collapse
|
5
|
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: 1.7] [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
|
6
|
Betzler AC, Kieser S, Fiedler K, Laban S, Theodoraki MN, Schuler PJ, Wirth T, Tedford K, Fischer KD, Hoffmann TK, Brunner C. Differential Requirement of Vav Proteins for Btk-dependent and –Independent Signaling During B Cell Development. Front Cell Dev Biol 2022; 10:654181. [PMID: 35281114 PMCID: PMC8904969 DOI: 10.3389/fcell.2022.654181] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 02/09/2022] [Indexed: 11/13/2022] Open
Abstract
Btk and Vav proteins are all components of the signalosome that builds upon B cell receptor (BCR) activation. However, the role of Vav proteins within the signalosome is quite complex and not yet fully understood. Until now, studies of these have focused predominantly on a deficiency of Vav proteins alone or in combination with other Vav protein family members. Since a physical association of Btk with Vav was shown previously, we asked whether these molecules lie in the same or independent signaling pathways. By analyzing Vav1 and Vav3 single knock-out mice and generating double-knock-out animals deficient for either Vav1 or Vav3 and Btk, we observed, in line with previous publications, no severe B cell developmental defects when either Vav1 or Vav3 alone are not expressed. However, a simultaneous deficiency of Btk together with either Vav1 or Vav3 leads to a severe reduction of splenic B cells, which exhibit an immature phenotype. B cell developmental defects of Btk/Vav1-double deficient mice in the periphery were more severe than those observed in Btk-single-deficient animals. Additionally, morphological changes in splenic microarchitecture were observed in double- but also in single-knock-out mutants. These observations were accompanied by reduced BCR-induced Ca2+ mobilization, proliferation, germinal center formation and immunoglobulin secretion. Although deletion of Btk alone impaired Ca2+ mobilization upon BCR activation, the defect was even more severe when Vav1 or Vav3 were also mutated, indicating that Btk and the Vav proteins act in separate pathways that converge on Ca2+ signaling. In vitro ASC differentiation suggests that both B and T cells contribute to the observed phenotype of a Btk/Vav-double deficiency. Our results show that Vav proteins and Btk are both components of the BCR-activated signalosome but control separate signaling pathways important for B cell development.
Collapse
Affiliation(s)
- Annika C. Betzler
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
| | - Sebastian Kieser
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
| | - Katja Fiedler
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
- Institute for Physiological Chemistry, Ulm University, Ulm, Germany
| | - Simon Laban
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
| | - Marie-Nicole Theodoraki
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
| | - Patrick J. Schuler
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
| | - Thomas Wirth
- Institute for Physiological Chemistry, Ulm University, Ulm, Germany
| | - Kerry Tedford
- Institute of Biochemistry and Cell Biology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Klaus-Dieter Fischer
- Institute of Biochemistry and Cell Biology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Thomas K. Hoffmann
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
| | - Cornelia Brunner
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
- *Correspondence: Cornelia Brunner,
| |
Collapse
|
7
|
Abstract
Cell migration, a crucial step in numerous biological processes, is tightly regulated in space and time. Cells employ Rho GTPases, primarily Rho, Rac, and Cdc42, to regulate their motility. Like other small G proteins, Rho GTPases function as biomolecular switches in regulating cell migration by operating between GDP bound 'OFF' and GTP bound 'ON' states. Guanine nucleotide exchange factors (GEFs) catalyse the shuttling of GTPases from OFF to ON state. G protein-coupled receptors (GPCRs) are the largest family of cell surface receptors that are involved in many signalling phenomena including cell survival and cell migration events. In this review, we summarize signalling mechanisms, involving GPCRs, leading to the activation of RhoGEFs. GPCRs exhibit diverse GEF activation modes that include the interaction of heterotrimeric G protein subunits with different domains of GEFs, phosphorylation, protein-protein interaction, protein-lipid interaction, and/or a combination of these processes.
Collapse
Affiliation(s)
- Aishwarya Omble
- Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Pune, India,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Kiran Kulkarni
- Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Pune, India,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India,CONTACT Kiran Kulkarni Academy of Scientific and Innovative Research (Acsir), Ghaziabad 201002, India
| |
Collapse
|
8
|
Robles-Valero J, Fernández-Nevado L, Lorenzo-Martín LF, Cuadrado M, Fernández-Pisonero I, Rodríguez-Fdez S, Astorga-Simón EN, Abad A, Caloto R, Bustelo XR. Cancer-associated mutations in VAV1 trigger variegated signaling outputs and T-cell lymphomagenesis. EMBO J 2021; 40:e108125. [PMID: 34617326 PMCID: PMC8591544 DOI: 10.15252/embj.2021108125] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 09/04/2021] [Accepted: 09/09/2021] [Indexed: 12/25/2022] Open
Abstract
Mutations in VAV1, a gene that encodes a multifunctional protein important for lymphocytes, are found at different frequencies in peripheral T‐cell lymphoma (PTCL), non‐small cell lung cancer, and other tumors. However, their pathobiological significance remains unsettled. After cataloguing 51 cancer‐associated VAV1 mutations, we show here that they can be classified in five subtypes according to functional impact on the three main VAV1 signaling branches, GEF‐dependent activation of RAC1, GEF‐independent adaptor‐like, and tumor suppressor functions. These mutations target new and previously established regulatory layers of the protein, leading to quantitative and qualitative changes in VAV1 signaling output. We also demonstrate that the most frequent VAV1 mutant subtype drives PTCL formation in mice. This process requires the concurrent engagement of two downstream signaling branches that promote the chronic activation and transformation of follicular helper T cells. Collectively, these data reveal the genetic constraints associated with the lymphomagenic potential of VAV1 mutant subsets, similarities with other PTCL driver genes, and potential therapeutic vulnerabilities.
Collapse
Affiliation(s)
- Javier Robles-Valero
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, Salamanca, Spain
| | - Lucía Fernández-Nevado
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, Salamanca, Spain
| | - L Francisco Lorenzo-Martín
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, Salamanca, Spain
| | - Myriam Cuadrado
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, Salamanca, Spain
| | - Isabel Fernández-Pisonero
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, Salamanca, Spain
| | - Sonia Rodríguez-Fdez
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, Salamanca, Spain
| | - Elsa N Astorga-Simón
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, Salamanca, Spain
| | - Antonio Abad
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, Salamanca, Spain
| | - Rubén Caloto
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, Salamanca, Spain
| | - Xosé R Bustelo
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, Salamanca, Spain
| |
Collapse
|
9
|
Cuadrado M, Robles-Valero J. VAV Proteins as Double Agents in Cancer: Oncogenes with Tumor Suppressor Roles. BIOLOGY 2021; 10:biology10090888. [PMID: 34571765 PMCID: PMC8466051 DOI: 10.3390/biology10090888] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/04/2021] [Accepted: 09/07/2021] [Indexed: 01/02/2023]
Abstract
Simple Summary The role of the VAV family (comprised of VAV1, VAV2, and VAV3) in proactive pathways involved in cell transformation has been historically assumed. Indeed, the discovery of potential gain-of-function VAV1 mutations in specific tumor subtypes reinforced this functional archetype. Contrary to this paradigm, we demonstrated that VAV1 could unexpectedly act as a tumor suppressor in some in vivo contexts. In this review, we discuss recent findings in the field, where the emerging landscape is one in which GTPases and their regulators, such as VAV proteins, can exhibit tumor suppressor functions. Abstract Guanosine nucleotide exchange factors (GEFs) are responsible for catalyzing the transition of small GTPases from the inactive (GDP-bound) to the active (GTP-bound) states. RHO GEFs, including VAV proteins, play essential signaling roles in a wide variety of fundamental cellular processes and in human diseases. Although the most widespread archetype in the field is that RHO GEFs exert proactive functions in cancer, recent studies in mice and humans are providing new insights into the in vivo function of these proteins in cancer. These results suggest a more complex scenario where the role of GEFs is not so clearly defined. For example, VAV1 can unexpectedly play non-catalytic tumor suppressor functions in T-cell acute lymphoblastic leukemia (T-ALL) by controlling the levels of the active form of NOTCH1 (ICN1). This review focuses on emerging work unveiling tumor suppressor roles for these proteins that should prompt a reevaluation of the role of VAV GEF family in tumor biology.
Collapse
Affiliation(s)
- Myriam Cuadrado
- Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain;
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, 37007 Salamanca, Spain
| | - Javier Robles-Valero
- Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain;
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, 37007 Salamanca, Spain
- Correspondence:
| |
Collapse
|
10
|
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
|
11
|
Wisniewska M, Banach B, Malinowski D, Domanski L, Sroczynski T, Dziedziejko V, Safranow K, Pawlik A. VAV1 Gene Polymorphism is Associated With Kidney Allograft Rejection. Transplant Proc 2021; 53:1528-1531. [PMID: 33994185 DOI: 10.1016/j.transproceed.2021.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 04/05/2021] [Indexed: 11/17/2022]
Abstract
BACKGROUND VAV1 is an intracellular signal transduction protein that plays a significant role in signal transduction in T cells. Several studies suggest that VAV1 signaling plays significant roles in allograft rejection. The aim of this study was to examine the association between VAV1 gene polymorphisms and renal allograft function. METHODS The study included 270 patients after allograft renal transplantation. We examined the associations between VAV1 gene polymorphisms and complications after transplantation, such as delayed graft function, acute rejection, and chronic allograft dysfunction. RESULTS There were no statistically significant associations between VAV1 genotypes and delayed graft function and chronic allograft dysfunction. Among patients with acute allograft rejection, we observed decreased frequencies of VAV1 rs2546133 TT and CT genotypes (P = .03) and T allele (P = .02), as well as VAV1 rs2617822 GG and AG genotypes (P = .05) and G allele (P = 0.04). In the multivariate regression analysis, the higher number of VAV1 rs2546133 T alleles showed a protective effect against the acute rejection in kidney allograft recipients. CONCLUSIONS The results of our study suggest that polymorphisms in the VAV1 gene are associated with kidney allograft rejection.
Collapse
Affiliation(s)
- Magda Wisniewska
- Department of Nephrology, Transplantology and Internal Medicine, Pomeranian Medical University, Szczecin, Poland
| | - Bolesław Banach
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - Damian Malinowski
- Department of Pharmacology, Pomeranian Medical University, Szczecin, Poland
| | - Leszek Domanski
- Department of Nephrology, Transplantology and Internal Medicine, Pomeranian Medical University, Szczecin, Poland
| | - Tomasz Sroczynski
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - Violetta Dziedziejko
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Szczecin, Poland
| | - Krzysztof Safranow
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Szczecin, Poland
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland.
| |
Collapse
|
12
|
VAV1 mutations contribute to development of T-cell neoplasms in mice. Blood 2021; 136:3018-3032. [PMID: 32992343 DOI: 10.1182/blood.2020006513] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 09/16/2020] [Indexed: 01/23/2023] Open
Abstract
Activating mutations in the Vav guanine nucleotide exchange factor 1 (VAV1) gene are reported in various subtypes of mature T-cell neoplasms (TCNs). However, oncogenic activities associated with VAV1 mutations in TCNs remain unclear. To define them, we established transgenic mice expressing VAV1 mutants cloned from human TCNs. Although we observed no tumors in these mice for up to a year, tumors did develop in comparably aged mice on a p53-null background (p53-/-VAV1-Tg), and p53-/-VAV1-Tg mice died with shorter latencies than did p53-null (p53-/-) mice. Notably, various TCNs with tendency of maturation developed in p53-/-VAV1-Tg mice, whereas p53-/- mice exhibited only immature TCNs. Mature TCNs in p53-/-VAV1-Tg mice mimicked a subtype of human peripheral T-cell lymphoma (PTCL-GATA3) and exhibited features of type 2 T helper (Th2) cells. Phenotypes seen following transplantation of either p53-/-VAV1 or p53-/- tumor cells into nude mice were comparable, indicating cell-autonomous tumor-initiating capacity. Whole-transcriptome analysis showed enrichment of multiple Myc-related pathways in TCNs from p53-/-VAV1-Tg mice relative to p53-/- or wild-type T cells. Remarkably, amplification of the Myc locus was found recurrently in TCNs of p53-/-VAV1-Tg mice. Finally, treatment of nude mice transplanted with p53-/-VAV1-Tg tumor cells with JQ1, a bromodomain inhibitor that targets the Myc pathway, prolonged survival of mice. We conclude that VAV1 mutations function in malignant transformation of T cells in vivo and that VAV1-mutant-expressing mice could provide an efficient tool for screening new therapeutic targets in TCNs harboring these mutations.
Collapse
|
13
|
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: 40] [Impact Index Per Article: 10.0] [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
|
14
|
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: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 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
|
15
|
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: 5.2] [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
|
16
|
He JW, Zhou XJ, Lv JC, Zhang H. Perspectives on how mucosal immune responses, infections and gut microbiome shape IgA nephropathy and future therapies. Am J Cancer Res 2020; 10:11462-11478. [PMID: 33052226 PMCID: PMC7545987 DOI: 10.7150/thno.49778] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 08/30/2020] [Indexed: 02/06/2023] Open
Abstract
Infections have been considered to play a critical role in the pathogenesis of IgA nephropathy (IgAN) because synpharyngitic hematuria is a common feature in IgAN. However, how infections participate in this process is still debated. More recent studies have also revealed that the alteration of the gut microbiome exerts a profound effect on host immune responses, contributing to the etiology or progression of autoimmunity. Considering IgA as the first line of defense against bacterial and viral antigens, this review evaluates the relationships among intestinal infections, gut microbiome, and IgA for a better understanding of the pathogenesis of IgAN. Moreover, as a prototype of IgA immunity, we provide detailed clarification of IgAN pathogenesis to shed light on other diseases in which IgA plays a role. Finally, we discuss potential therapies focusing on microbes and mucosal immune responses in IgAN.
Collapse
|
17
|
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: 0.2] [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
|
18
|
Younes N, Zhou L, Amatullah H, Mei SHJ, Herrero R, Lorente JA, Stewart DJ, Marsden P, Liles WC, Hu P, Dos Santos CC. Mesenchymal stromal/stem cells modulate response to experimental sepsis-induced lung injury via regulation of miR-27a-5p in recipient mice. Thorax 2020; 75:556-567. [PMID: 32546573 PMCID: PMC7361025 DOI: 10.1136/thoraxjnl-2019-213561] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 01/08/2020] [Accepted: 03/13/2020] [Indexed: 01/11/2023]
Abstract
Introduction Mesenchymal stromal cell (MSC) therapy mitigates lung injury and improves survival in murine models of sepsis. Precise mechanisms of therapeutic benefit remain poorly understood. Objectives To identify host-derived regulatory elements that may contribute to the therapeutic effects of MSCs, we profiled the microRNAome (miRNAome) and transcriptome of lungs from mice randomised to experimental polymicrobial sepsis-induced lung injury treated with either placebo or MSCs. Methods and results A total of 11 997 genes and 357 microRNAs (miRNAs) expressed in lungs were used to generate a statistical estimate of association between miRNAs and their putative mRNA targets; 1395 miRNA:mRNA significant association pairs were found to be differentially expressed (false discovery rate ≤0.05). MSC administration resulted in the downregulation of miR-27a-5p and upregulation of its putative target gene VAV3 (adjusted p=1.272E-161) in septic lungs. In human pulmonary microvascular endothelial cells, miR-27a-5p expression levels were increased while VAV3 was decreased following lipopolysaccharide (LPS) or tumour necrosis factor (TNF) stimulation. Transfection of miR-27a-5p mimic or inhibitor resulted in increased or decreased VAV3 message, respectively. Luciferase reporter assay demonstrated specific binding of miR-27a-5p to the 3′UTR of VAV3. miR27a-5p inhibition mitigated TNF-induced (1) delayed wound closure, increased (2) adhesion and (3) transendothelial migration but did not alter permeability. In vivo, cell infiltration was attenuated by intratracheal coinstillation of the miR-27a-5p inhibitor, but this did not protect against endotoxin-induced oedema formation. Conclusions Our data support involvement of miR-27a-5p and VAV3 in cellular adhesion and infiltration during acute lung injury and a potential role for miR-27a-based therapeutics for acute respiratory distress syndrome.
Collapse
Affiliation(s)
- Nadim Younes
- Critical Care Medicine, The Keenan Research Centre for Biomedical Science of Saint Michael's Hospital, Toronto, Ontario, Canada
| | - Louis Zhou
- Critical Care Medicine, The Keenan Research Centre for Biomedical Science of Saint Michael's Hospital, Toronto, Ontario, Canada.,Institute of Medical Sciences and Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Hajera Amatullah
- Critical Care Medicine, The Keenan Research Centre for Biomedical Science of Saint Michael's Hospital, Toronto, Ontario, Canada.,Institute of Medical Sciences and Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Shirley H J Mei
- Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Raquel Herrero
- Critical Care Service, Hospital Universitario de Getafe-CIBER de Enfermedades Respiratorias (CIBERES), Getafe, Spain
| | - Jose Angel Lorente
- Critical Care Service, Hospital Universitario de Getafe-CIBER de Enfermedades Respiratorias (CIBERES), Getafe, Spain
| | - Duncan J Stewart
- Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Philip Marsden
- Critical Care Medicine, The Keenan Research Centre for Biomedical Science of Saint Michael's Hospital, Toronto, Ontario, Canada
| | - W Conrad Liles
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Pingzhao Hu
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Claudia C Dos Santos
- Critical Care Medicine, The Keenan Research Centre for Biomedical Science of Saint Michael's Hospital, Toronto, Ontario, Canada .,Institute of Medical Sciences and Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
19
|
Lindner SE, Egelston CA, Huard SM, Lee PP, Wang LD. Arhgap25 Deficiency Leads to Decreased Numbers of Peripheral Blood B Cells and Defective Germinal Center Reactions. Immunohorizons 2020; 4:274-281. [PMID: 32434881 DOI: 10.4049/immunohorizons.2000021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 05/05/2020] [Indexed: 02/03/2023] Open
Abstract
Rho family GTPases are critical for normal B cell development and function, and their activity is regulated by a large and complex network of guanine nucleotide exchange factors (GEFs) and GTPase-activating proteins (GAPs). However, the role of GAPs in B cell development is poorly understood. In this study, we show that the novel Rac-GAP ARHGAP25 is important for B cell development in mice in a CXCR4-dependent manner. We show that Arhgap25 deficiency in mice leads to a significant decrease in peripheral blood B cell numbers as well as defects in mature B cell differentiation. Arhgap25-/- B cells respond to Ag stimulation in vitro and in vivo but have impaired germinal center formation and decreased IgG1 class switching. Additionally, Arhgap25-/- B cells show evidence of increased baseline motility and augmented chemotaxis to CXCL12. Taken together, these studies demonstrate an important role for Arhgap25 in peripheral B cell development and Ag response.
Collapse
Affiliation(s)
- Silke E Lindner
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010; and
| | - Colt A Egelston
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010; and
| | - Stephanie M Huard
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010; and
| | - Peter P Lee
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010; and
| | - Leo D Wang
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010; and .,Department of Pediatrics, City of Hope National Medical Center, Duarte, CA 91010
| |
Collapse
|
20
|
Pawlik A, Malinowski D, Paradowska-Gorycka A, Safranow K, Dziedziejko V. VAV1 Gene Polymorphisms in Patients with Rheumatoid Arthritis. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17093214. [PMID: 32380774 PMCID: PMC7246862 DOI: 10.3390/ijerph17093214] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 04/30/2020] [Accepted: 05/02/2020] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Rheumatoid arthritis (RA) is an important public health problem because this disease often causes disability. RA is a chronic, destructive autoimmune disease that leads to joint destruction and the development of extraarticular manifestations. VAV1 is an intracellular signal transduction protein that plays a significant role in signal transduction in T cells and affects T cell development, proliferation and activation. The VAV1 gene contains 27 exons and is located on chromosome 19. In this study, we examined the association between VAV1 rs2546133 and rs2617822 polymorphisms and RA. METHODS We examined 422 patients with RA and 338 healthy subjects as the control group. RESULTS Among RA patients, there was a statistically significant increase in the frequency of VAV1 rs2546133 polymorphism in T allele carriers (TT + CT versus CC, odds ratio: 1.69, 95% confidence interval 1.05-2.73, p = 0.035). There was no statistically significant difference in the distribution of the rs2617822 genotypes and alleles between RA patients and the control group. Additionally, patients who carried the VAV1 rs2546133 T and rs2617822 G allele presented an increased frequency of extraarticular manifestations: vasculitis, amyloidosis and Sjogren syndrome. CONCLUSIONS The results suggest an association between VAV1 gene rs2617822 polymorphism and RA.
Collapse
Affiliation(s)
- Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
- Correspondence:
| | - Damian Malinowski
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University, 70-111 Szczecin, Poland;
| | - Agnieszka Paradowska-Gorycka
- Department of Molecular Biology, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland;
| | - Krzysztof Safranow
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.S.); (V.D.)
| | - Violetta Dziedziejko
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.S.); (V.D.)
| |
Collapse
|
21
|
Fray MA, Charpentier JC, Sylvain NR, Seminario MC, Bunnell SC. Vav2 lacks Ca 2+ entry-promoting scaffolding functions unique to Vav1 and inhibits T cell activation via Cdc42. J Cell Sci 2020; 133:jcs238337. [PMID: 31974114 PMCID: PMC7075049 DOI: 10.1242/jcs.238337] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 01/06/2020] [Indexed: 12/16/2022] Open
Abstract
Vav family guanine nucleotide exchange factors (GEFs) are essential regulators of immune function. Despite their structural similarity, Vav1 promotes and Vav2 opposes T cell receptor (TCR)-induced Ca2+ entry. By using a Vav1-deficient Jurkat T cell line, we find that Vav1 facilitates Ca2+ entry via non-catalytic scaffolding functions that are encoded by the catalytic core of Vav1 and flanking linker regions. We implicate, in this scaffolding function, a previously undescribed polybasic motif that is strictly conserved in Vav1 and absent from Vav2 in tetrapods. Conversely, the catalytic activity of Vav2 contributes to the suppression of TCR-mediated Ca2+ entry. By performing an in vivo 'GEF trapping' assay in intact cells, we demonstrate that Cdc42 interacts with the catalytic surface of Vav2 but not Vav1, and that Vav1 discriminates Cdc42 from Rac1 via F56 (W56 in Rac1). Finally, the Cdc42-specific inhibitor ZCL278 and the shRNA-mediated suppression of Cdc42 each prevent the inhibition of TCR-induced Ca2+ entry by Vav2. These findings define stark differences in the functions of Vav1 and Vav2, and provide an explanation for the differential usage of these Vav isoforms by immune subpopulations.
Collapse
Affiliation(s)
- Michael A Fray
- Program in Immunology, Department of Immunology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - John C Charpentier
- Department of Biology, University of Massachusetts Boston, Boston, MA 02125, USA
| | - Nicholas R Sylvain
- Program in Immunology, Department of Immunology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Maria-Cristina Seminario
- Program in Immunology, Department of Immunology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Stephen C Bunnell
- Program in Immunology, Department of Immunology, Tufts University School of Medicine, Boston, MA 02111, USA
| |
Collapse
|
22
|
He M, Westerberg LS. Congenital Defects in Actin Dynamics of Germinal Center B Cells. Front Immunol 2019; 10:296. [PMID: 30894852 PMCID: PMC6414452 DOI: 10.3389/fimmu.2019.00296] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 02/05/2019] [Indexed: 01/02/2023] Open
Abstract
The germinal center (GC) is a transient anatomical structure formed during the adaptive immune response that leads to antibody affinity maturation and serological memory. Recent works using two-photon microscopy reveals that the GC is a highly dynamic structure and GC B cells are highly motile. An efficient selection of high affinity B cells clones within the GC crucially relies on the interplay of proliferation, genome editing, cell-cell interaction, and migration. All these processes require actin cytoskeleton rearrangement to be well-coordinated. Dysregulated actin dynamics may impede on multiple stages during B cell affinity maturation, which could lead to aberrant GC response and result in autoimmunity and B cell malignancy. This review mainly focuses on the recent works that investigate the role of actin regulators during the GC response.
Collapse
Affiliation(s)
- Minghui He
- Department of Microbiology Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Lisa S. Westerberg
- Department of Microbiology Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
23
|
Zinc Finger Protein 521 Regulates Early Hematopoiesis through Cell-Extrinsic Mechanisms in the Bone Marrow Microenvironment. Mol Cell Biol 2018; 38:MCB.00603-17. [PMID: 29915154 DOI: 10.1128/mcb.00603-17] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Accepted: 06/11/2018] [Indexed: 01/06/2023] Open
Abstract
Zinc finger protein 521 (ZFP521), a DNA-binding protein containing 30 Krüppel-like zinc fingers, has been implicated in the differentiation of multiple cell types, including hematopoietic stem and progenitor cells (HSPC) and B lymphocytes. Here, we report a novel role for ZFP521 in regulating the earliest stages of hematopoiesis and lymphoid cell development via a cell-extrinsic mechanism. Mice with inactivated Zfp521 genes (Zfp521-/-) possess reduced frequencies and numbers of hematopoietic stem and progenitor cells, common lymphoid progenitors, and B and T cell precursors. Notably, ZFP521 deficiency changes bone marrow microenvironment cytokine levels and gene expression within resident HSPC, consistent with a skewing of hematopoiesis away from lymphopoiesis. These results advance our understanding of ZFP521's role in normal hematopoiesis, justifying further research to assess its potential as a target for cancer therapies.
Collapse
|
24
|
Kant S, Standen CL, Morel C, Jung DY, Kim JK, Swat W, Flavell RA, Davis RJ. A Protein Scaffold Coordinates SRC-Mediated JNK Activation in Response to Metabolic Stress. Cell Rep 2018; 20:2775-2783. [PMID: 28930674 DOI: 10.1016/j.celrep.2017.08.025] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 06/30/2017] [Accepted: 07/29/2017] [Indexed: 01/25/2023] Open
Abstract
Obesity is a major risk factor for the development of metabolic syndrome and type 2 diabetes. How obesity contributes to metabolic syndrome is unclear. Free fatty acid (FFA) activation of a non-receptor tyrosine kinase (SRC)-dependent cJun NH2-terminal kinase (JNK) signaling pathway is implicated in this process. However, the mechanism that mediates SRC-dependent JNK activation is unclear. Here, we identify a role for the scaffold protein JIP1 in SRC-dependent JNK activation. SRC phosphorylation of JIP1 creates phosphotyrosine interaction motifs that bind the SH2 domains of SRC and the guanine nucleotide exchange factor VAV. These interactions are required for SRC-induced activation of VAV and the subsequent engagement of a JIP1-tethered JNK signaling module. The JIP1 scaffold protein, therefore, plays a dual role in FFA signaling by coordinating upstream SRC functions together with downstream effector signaling by the JNK pathway.
Collapse
Affiliation(s)
- Shashi Kant
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA; Division of Cardiovascular Medicine, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| | - Claire L Standen
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Caroline Morel
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Dae Young Jung
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Jason K Kim
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA; Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Wojciech Swat
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Richard A Flavell
- Howard Hughes Medical Institute and Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Roger J Davis
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA; Howard Hughes Medical Institute, Worcester, MA 01605, USA.
| |
Collapse
|
25
|
Burbage M, Gasparrini F, Aggarwal S, Gaya M, Arnold J, Nair U, Way M, Bruckbauer A, Batista FD. Tuning of in vivo cognate B-T cell interactions by Intersectin 2 is required for effective anti-viral B cell immunity. eLife 2018; 7. [PMID: 29337666 PMCID: PMC5770159 DOI: 10.7554/elife.26556] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 01/01/2018] [Indexed: 12/13/2022] Open
Abstract
Wiskott-Aldrich syndrome (WAS) is an immune pathology associated with mutations in WAS protein (WASp) or in WASp interacting protein (WIP). Together with the small GTPase Cdc42 and other effectors, these proteins participate in the remodelling of the actin network downstream of BCR engagement. Here we show that mice lacking the adaptor protein ITSN2, a G-nucleotide exchange factor (GEF) for Cdc42 that also interacts with WASp and WIP, exhibited increased mortality during primary infection, incomplete protection after Flu vaccination, reduced germinal centre formation and impaired antibody responses to vaccination. These defects were found, at least in part, to be intrinsic to the B cell compartment. In vivo, ITSN2 deficient B cells show a reduction in the expression of SLAM, CD84 or ICOSL that correlates with a diminished ability to form long term conjugates with T cells, to proliferate in vivo, and to differentiate into germinal centre cells. In conclusion, our study not only revealed a key role for ITSN2 as an important regulator of adaptive immune-response during vaccination and viral infection but it is also likely to contribute to a better understanding of human immune pathologies.
Collapse
Affiliation(s)
- Marianne Burbage
- Lymphocyte Biology Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Francesca Gasparrini
- Lymphocyte Biology Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Shweta Aggarwal
- Lymphocyte Biology Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Mauro Gaya
- Lymphocyte Biology Laboratory, The Francis Crick Institute, London, United Kingdom.,Ragon Institute of MGH, MIT and Harvard, Cambridge, United States
| | - Johan Arnold
- Ragon Institute of MGH, MIT and Harvard, Cambridge, United States
| | - Usha Nair
- Ragon Institute of MGH, MIT and Harvard, Cambridge, United States
| | - Michael Way
- Cellular Signalling and Cytoskeletal Function Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Andreas Bruckbauer
- Lymphocyte Biology Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Facundo D Batista
- Lymphocyte Biology Laboratory, The Francis Crick Institute, London, United Kingdom.,Ragon Institute of MGH, MIT and Harvard, Cambridge, United States
| |
Collapse
|
26
|
Roth S, Bergmann H, Jaeger M, Yeroslaviz A, Neumann K, Koenig PA, Prazeres da Costa C, Vanes L, Kumar V, Johnson M, Menacho-Márquez M, Habermann B, Tybulewicz VL, Netea M, Bustelo XR, Ruland J. Vav Proteins Are Key Regulators of Card9 Signaling for Innate Antifungal Immunity. Cell Rep 2017; 17:2572-2583. [PMID: 27926862 PMCID: PMC5177621 DOI: 10.1016/j.celrep.2016.11.018] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 09/26/2016] [Accepted: 11/01/2016] [Indexed: 02/03/2023] Open
Abstract
Fungal infections are major causes of morbidity and mortality, especially in immunocompromised individuals. The innate immune system senses fungal pathogens through Syk-coupled C-type lectin receptors (CLRs), which signal through the conserved immune adaptor Card9. Although Card9 is essential for antifungal defense, the mechanisms that couple CLR-proximal events to Card9 control are not well defined. Here, we identify Vav proteins as key activators of the Card9 pathway. Vav1, Vav2, and Vav3 cooperate downstream of Dectin-1, Dectin-2, and Mincle to engage Card9 for NF-κB control and proinflammatory gene transcription. Although Vav family members show functional redundancy, Vav1/2/3−/− mice phenocopy Card9−/− animals with extreme susceptibility to fungi. In this context, Vav3 is the single most important Vav in mice, and a polymorphism in human VAV3 is associated with susceptibility to candidemia in patients. Our results reveal a molecular mechanism for CLR-mediated Card9 regulation that controls innate immunity to fungal infections. Vav proteins control CLR-mediated inflammatory responses CLR-induced NF-κB activation is regulated by Vav proteins Vav/Card9 signaling is critical for antifungal host defense
Collapse
Affiliation(s)
- Susanne Roth
- Institut für Klinische Chemie und Pathobiochemie, Klinikum rechts der Isar, Technische Universität München, 81675 München, Germany; Chirurgische Klinik, Universitätsklinikum Heidelberg, Ruprecht-Karls-Universität, 69120 Heidelberg, Germany
| | - Hanna Bergmann
- Institut für Klinische Chemie und Pathobiochemie, Klinikum rechts der Isar, Technische Universität München, 81675 München, Germany
| | - Martin Jaeger
- Department of Medicine, Radboud University, Medical Centre, 6500 HB Nijmegen, the Netherlands; Radboud Center for Infectious Diseases, 6500 HB Nijmegen, the Netherlands
| | - Assa Yeroslaviz
- Max Planck Institute of Biochemistry, Research Group Computational Biology, 82152 Martinsried, Germany
| | - Konstantin Neumann
- Institut für Klinische Chemie und Pathobiochemie, Klinikum rechts der Isar, Technische Universität München, 81675 München, Germany
| | - Paul-Albert Koenig
- Institut für Klinische Chemie und Pathobiochemie, Klinikum rechts der Isar, Technische Universität München, 81675 München, Germany
| | - Clarissa Prazeres da Costa
- Institut für Medizinische Mikrobiologie, Immunologie, und Hygiene, Klinikum rechts der Isar, Technische Universität München, 81675 München, Germany
| | | | - Vinod Kumar
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, 9700 RB, the Netherlands
| | - Melissa Johnson
- Duke University Medical Center, Duke Box 102359, Durham, NC 27710, USA
| | | | - Bianca Habermann
- Max Planck Institute of Biochemistry, Research Group Computational Biology, 82152 Martinsried, Germany
| | - Victor L Tybulewicz
- Francis Crick Institute, London NW1 1AT, UK; Department of Medicine, Imperial College, London W12 0NN, UK
| | - Mihai Netea
- Department of Medicine, Radboud University, Medical Centre, 6500 HB Nijmegen, the Netherlands
| | - Xosé R Bustelo
- Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain; Centro de Investigacion Biomedica en Red-Oncologia, Carlos III Health Institute, Spain
| | - Jürgen Ruland
- Institut für Klinische Chemie und Pathobiochemie, Klinikum rechts der Isar, Technische Universität München, 81675 München, Germany; German Cancer Consortium (DKTK), 69120 Heidelberg, Germany; German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany.
| |
Collapse
|
27
|
A Novel Vav3 Homolog Identified in Lamprey, Lampetra japonica, with Roles in Lipopolysaccharide-Mediated Immune Response. Int J Mol Sci 2017; 18:ijms18102035. [PMID: 28937614 PMCID: PMC5666717 DOI: 10.3390/ijms18102035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 09/19/2017] [Accepted: 09/20/2017] [Indexed: 11/16/2022] Open
Abstract
Vav guanine nucleotide exchange factor 3 (Vav3), a Rho family GTPase, regulates multiple cell signaling pathways including those of T- and B-cell receptors in vertebrates through mediating the activities of the Rho family members. Whether the lamprey possesses Vav3 homolog and what role it plays in immune response remain unknown. Gene cloning, recombinant expression, antibody production and expression pattern analyses were performed to characterize the lamprey Vav3 in the current study. The lamprey Vav3 is closer to jawed vertebrates' Vav3 molecules (about 53% identities in general) than to Vav2 molecules of jawless and jawed vertebrates (about 51% identities in general) in sequence similarity. Conserved motif analysis showed that the most distinguished parts between Vav3 and Vav2 proteins are their two Src-homology 3 domains. The relative expression levels of lamprey vav3 mRNA and protein were significantly up-regulated in lamprey lymphocytes and supraneural myeloid bodies after mixed-antigens stimulation, respectively. In addition, lamprey Vav3 were up-regulated drastically in lymphocytes and supraneural myeloid bodies after lipopolysaccharide (LPS) rather than phytohemagglutinin (PHA) stimulation. Lamprey Vav3 distributed in the cytoplasm of variable lymphocyte receptor B positive (VLRB⁺) lymphocytes, and the number of plasmacytes (VLRB and lamprey Vav3 double positive) in blood lymphocytes also increased after LPS stimulation. Our results proved that lamprey Vav3 was involved in the LPS-mediated immune reaction of lamprey and provided a clue for the further study of the precise role lamprey Vav3 played in the signaling pathway of lamprey VLRB⁺ lymphocytes.
Collapse
|
28
|
Shukla A, Shukla V, Joshi SS. Regulation of MAPK signaling and implications in chronic lymphocytic leukemia. Leuk Lymphoma 2017; 59:1565-1573. [PMID: 28882083 DOI: 10.1080/10428194.2017.1370548] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Chronic lymphocytic leukemia (CLL) is a heterogeneous B cell malignancy that still remains incurable. Recent studies have highlighted cellular and non-cellular components of the tissue microenvironment in CLL that help nurture the growth of leukemic cells by providing the necessary stimuli for their proliferation and survival. The diverse stimuli in the specialized tissue microenvironment of CLL lead to constitutive activation of several signaling pathways that includes B cell receptor signaling and the associated mitogen-activated protein kinase (MAPK) signaling. Recent findings have described aberrant activation of MAPK signaling and its interactions with other cellular signaling pathways in the pathogenesis of CLL. These studies have shed light on the deregulated molecular mechanisms contributing to hyperactivation of MAPK signaling and provided avenues for therapeutic options for aggressive CLL. In this review, we describe and discuss the current status of our understanding into the role of MAPK signaling in the pathogenesis of CLL.
Collapse
Affiliation(s)
- Ashima Shukla
- a Sanford Burnham Prebys Medical Discovery Institute , La Jolla , CA , USA
| | - Vipul Shukla
- b La Jolla Institute for Allergy and Immunology , La Jolla , CA , USA
| | - Shantaram S Joshi
- c Department of Genetics Cell Biology and Anatomy , University of Nebraska Medical Centre , Omaha , NE , USA
| |
Collapse
|
29
|
Lang MJ, Mori M, Ruer-Laventie J, Pieters J. A Coronin 1–Dependent Decision Switch in Juvenile Mice Determines the Population of the Peripheral Naive T Cell Compartment. THE JOURNAL OF IMMUNOLOGY 2017; 199:2421-2431. [DOI: 10.4049/jimmunol.1700438] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 07/31/2017] [Indexed: 11/19/2022]
|
30
|
Huang L, Zhang Y, Xu C, Gu X, Niu L, Wang J, Sun X, Bai X, Xuan X, Li Q, Shi C, Yu B, Miller H, Yang G, Westerberg LS, Liu W, Song W, Zhao X, Liu C. Rictor positively regulates B cell receptor signaling by modulating actin reorganization via ezrin. PLoS Biol 2017; 15:e2001750. [PMID: 28821013 PMCID: PMC5562439 DOI: 10.1371/journal.pbio.2001750] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 07/25/2017] [Indexed: 01/13/2023] Open
Abstract
As the central hub of the metabolism machinery, the mammalian target of rapamycin complex 2 (mTORC2) has been well studied in lymphocytes. As an obligatory component of mTORC2, the role of Rictor in T cells is well established. However, the role of Rictor in B cells still remains elusive. Rictor is involved in B cell development, especially the peripheral development. However, the role of Rictor on B cell receptor (BCR) signaling as well as the underlying cellular and molecular mechanism is still unknown. This study used B cell-specfic Rictor knockout (KO) mice to investigate how Rictor regulates BCR signaling. We found that the key positive and negative BCR signaling molecules, phosphorylated Brutons tyrosine kinase (pBtk) and phosphorylated SH2-containing inositol phosphatase (pSHIP), are reduced and enhanced, respectively, in Rictor KO B cells. This suggests that Rictor positively regulates the early events of BCR signaling. We found that the cellular filamentous actin (F-actin) is drastically increased in Rictor KO B cells after BCR stimulation through dysregulating the dephosphorylation of ezrin. The high actin-ezrin intensity area restricts the lateral movement of BCRs upon stimulation, consequently reducing BCR clustering and BCR signaling. The reduction in the initiation of BCR signaling caused by actin alteration is associated with a decreased humoral immune response in Rictor KO mice. The inhibition of actin polymerization with latrunculin in Rictor KO B cells rescues the defects of BCR signaling and B cell differentiation. Overall, our study provides a new pathway linking cell metablism to BCR activation, in which Rictor regulates BCR signaling via actin reorganization.
Collapse
Affiliation(s)
- Lu Huang
- Chongqing Key Laboratory of Child Infection and Immunity, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Department of Pediatric Research Institute, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Yongjie Zhang
- Chongqing Key Laboratory of Child Infection and Immunity, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Department of Pediatric Research Institute, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Chenguang Xu
- MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Institute for Immunology, Tsinghua University, Beijing, China
| | - Xiaomei Gu
- MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Institute for Immunology, Tsinghua University, Beijing, China
| | - Linlin Niu
- Chongqing Key Laboratory of Child Infection and Immunity, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Department of Pediatric Research Institute, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Jinzhi Wang
- Chongqing Key Laboratory of Child Infection and Immunity, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Department of Pediatric Research Institute, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoyu Sun
- Chongqing Key Laboratory of Child Infection and Immunity, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Department of Pediatric Research Institute, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoming Bai
- Chongqing Key Laboratory of Child Infection and Immunity, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Department of Pediatric Research Institute, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Xingtian Xuan
- Chongqing Key Laboratory of Child Infection and Immunity, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Department of Pediatric Research Institute, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Qubei Li
- Children’s Hospital Respiratory Center of Chongqing Medical University, Chongqing, China
| | - Chunwei Shi
- Department of Pathogen Biology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - Bing Yu
- Department of Pathogen Biology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - Heather Miller
- Department of Intracellular Pathogens, National Institute of Allergy and Infectious Diseases, Hamilton, Montana, United States of America
| | - Gangyi Yang
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Lisa S. Westerberg
- Department of Microbiology Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Wanli Liu
- MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Institute for Immunology, Tsinghua University, Beijing, China
| | - Wenxia Song
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, United States of America
| | - Xiaodong Zhao
- Chongqing Key Laboratory of Child Infection and Immunity, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Department of Pediatric Research Institute, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Chaohong Liu
- Department of Pathogen Biology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
31
|
Burbage M, Keppler SJ, Montaner B, Mattila PK, Batista FD. The Small Rho GTPase TC10 Modulates B Cell Immune Responses. THE JOURNAL OF IMMUNOLOGY 2017; 199:1682-1695. [PMID: 28747344 PMCID: PMC5563166 DOI: 10.4049/jimmunol.1602167] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 06/27/2017] [Indexed: 11/28/2022]
Abstract
Rho family GTPases regulate diverse cellular events, such as cell motility, polarity, and vesicle traffic. Although a wealth of data exists on the canonical Rho GTPases RhoA, Rac1, and Cdc42, several other family members remain poorly studied. In B cells, we recently demonstrated a critical role for Cdc42 in plasma cell differentiation. In this study, we focus on a close homolog of Cdc42, TC10 (also known as RhoQ), and investigate its physiological role in B cells. By generating a TC10-deficient mouse model, we show that despite reduced total B cell numbers, B cell development in these mice occurs normally through distinct developmental stages. Upon immunization, IgM levels were reduced and, upon viral infection, germinal center responses were defective in TC10-deficient mice. BCR signaling was mildly affected, whereas cell migration remained normal in TC10-deficient B cells. Furthermore, by generating a TC10/Cdc42 double knockout mouse model, we found that TC10 can compensate for the lack of Cdc42 in TLR-induced cell activation and proliferation, so the two proteins play partly redundant roles. Taken together, by combining in vivo and in vitro analysis using TC10-deficient mice, we define the poorly studied Rho GTPase TC10 as an immunomodulatory molecule playing a role in physiological B cell responses.
Collapse
Affiliation(s)
- Marianne Burbage
- Lymphocyte Interaction Laboratory, Francis Crick Institute, London NW1 1AT, United Kingdom
| | - Selina J Keppler
- Lymphocyte Interaction Laboratory, Francis Crick Institute, London NW1 1AT, United Kingdom
| | - Beatriz Montaner
- Lymphocyte Interaction Laboratory, Francis Crick Institute, London NW1 1AT, United Kingdom
| | - Pieta K Mattila
- Lymphocyte Interaction Laboratory, Francis Crick Institute, London NW1 1AT, United Kingdom; .,Institute of Biomedicine, Unit of Pathology and MediCity Research Laboratories, University of Turku, BioCity, 20520 Turku, Finland; and
| | - Facundo D Batista
- Lymphocyte Interaction Laboratory, Francis Crick Institute, London NW1 1AT, United Kingdom.,Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139
| |
Collapse
|
32
|
Rinke J, Müller JP, Blaess MF, Chase A, Meggendorfer M, Schäfer V, Winkelmann N, Haferlach C, Cross NCP, Hochhaus A, Ernst T. Molecular characterization of EZH2 mutant patients with myelodysplastic/myeloproliferative neoplasms. Leukemia 2017. [DOI: 10.1038/leu.2017.190] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
33
|
Ulc A, Gottschling C, Schäfer I, Wegrzyn D, van Leeuwen S, Luft V, Reinhard J, Faissner A. Involvement of the guanine nucleotide exchange factor Vav3 in central nervous system development and plasticity. Biol Chem 2017; 398:663-675. [DOI: 10.1515/hsz-2016-0275] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 02/10/2017] [Indexed: 12/13/2022]
Abstract
Abstract
Small GTP-hydrolyzing enzymes (GTPases) of the RhoA family play manifold roles in cell biology and are regulated by upstream guanine nucleotide exchange factors (GEFs). Herein, we focus on the GEFs of the Vav subfamily. Vav1 was originally described as a proto-oncogene of the hematopoietic lineage. The GEFs Vav2 and Vav3 are more broadly expressed in various tissues. In particular, the GEF Vav3 may play important roles in the developing nervous system during the differentiation of neural stem cells into the major lineages, namely neurons, oligodendrocytes and astrocytes. We discuss its putative regulatory roles for progenitor differentiation in the developing retina, polarization of neurons and formation of synapses, migration of oligodendrocyte progenitors and establishment of myelin sheaths. We propose that Vav3 mediates the response of various neural cell types to environmental cues.
Collapse
|
34
|
Association of VAV2 and VAV3 polymorphisms with cardiovascular risk factors. Sci Rep 2017; 7:41875. [PMID: 28157227 PMCID: PMC5291103 DOI: 10.1038/srep41875] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 01/03/2017] [Indexed: 02/07/2023] Open
Abstract
Hypertension, diabetes and obesity are cardiovascular risk factors closely associated to the development of renal and cardiovascular target organ damage. VAV2 and VAV3, members of the VAV family proto-oncogenes, are guanosine nucleotide exchange factors for the Rho and Rac GTPase family, which is related with cardiovascular homeostasis. We have analyzed the relationship between the presence of VAV2 rs602990 and VAV3 rs7528153 polymorphisms with cardiovascular risk factors and target organ damage (heart, vessels and kidney) in 411 subjects. Our results show that being carrier of the T allele in VAV2 rs602990 polymorphism is associated with an increased risk of obesity, reduced levels of ankle-brachial index and diastolic blood pressure and reduced retinal artery caliber. In addition, being carrier of T allele is associated with increased risk of target organ damage in males. On the other hand, being carrier of the T allele in VAV3 rs7528153 polymorphism is associated with a decreased susceptibility of developing a pathologic state composed by the presence of hypertension, diabetes, obesity or cardiovascular damage, and with an increased risk of developing altered basal glycaemia. This is the first report showing an association between VAV2 and VAV3 polymorphisms with cardiovascular risk factors and target organ damage.
Collapse
|
35
|
Guerreiro-Cacais AO, Norin U, Gyllenberg A, Berglund R, Beyeen AD, Petit-Teixeira E, Cornélis F, Saoudi A, Fournié GJ, Holmdahl R, Alfredsson L, Klareskog L, Jagodic M, Olsson T, Kockum I, Padyukov L. VAV1 regulates experimental autoimmune arthritis and is associated with anti-CCP negative rheumatoid arthritis. Genes Immun 2017; 18:48-56. [PMID: 28053322 DOI: 10.1038/gene.2016.49] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 11/19/2016] [Accepted: 11/28/2016] [Indexed: 12/19/2022]
Abstract
Rheumatoid arthritis (RA) patients can be stratified into two subgroups defined by the presence or absence of antibodies against citrullinated circular peptides (anti-CCP) with most of the genetic association found in anti-CCP positive RA. Here we addressed the role of VAV1, previously associated to multiple sclerosis (MS), in the pathogenesis of RA in experimental models and in a genetic association study. Experimental arthritis triggered by pristane or collagen type II was induced in DA rats and in the DA.BN-R25 congenic line that carries a polymorphism in Vav1. Difference in arthritis severity was observed only after immunization with pristane. In a case-control study, 34 SNPs from VAV1 locus were analyzed by Immunochip genotyping in 11475 RA patients (7573 anti-CCP positive and 3902 negative) and 15,870 controls in six cohorts of European Caucasians. A combination of the previous MS-associated haplotype and two additional SNPs was associated with anti-CCP negative RA (alleles G-G-A-A of rs682626-rs2546133-rs2617822-rs12979659, OR=1.13, P=1.27 × 10-5). The same markers also contributed to activity of RA at baseline with the strongest association in the anti-CCP negative group for the rs682626-rs12979659 G-A haplotype (β=-0.283, P=0.0048). Our study suggests a role for VAV1 and T-cell signaling in the pathology of anti-CCP-negative RA.
Collapse
Affiliation(s)
- A O Guerreiro-Cacais
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - U Norin
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - A Gyllenberg
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - R Berglund
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - A D Beyeen
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | - E Petit-Teixeira
- GenHotel-EA3886, Evry-Val d'Essonne University, Evry-Genopole, France
| | - F Cornélis
- GenHotel-Auvergne, CHU de Clermont-Ferrand, Auvergne University, France
| | - A Saoudi
- Inserm, U1043, Toulouse, France.,CNRS, U5282, Toulouse, France.,Centre de Physiopathologie de Toulouse Purpan, Université de Toulouse, Toulouse, France
| | - G J Fournié
- Inserm, U1043, Toulouse, France.,CNRS, U5282, Toulouse, France.,Centre de Physiopathologie de Toulouse Purpan, Université de Toulouse, Toulouse, France
| | - R Holmdahl
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - L Alfredsson
- Department of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - L Klareskog
- Rheumatology Unit, Department of Medicine, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - M Jagodic
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - T Olsson
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - I Kockum
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - L Padyukov
- Rheumatology Unit, Department of Medicine, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
36
|
Chen Y, Xu Q, Li Y, Liu R, Huang Z, Wang B, Chen G. Gene expression profile after activation of RIG-I in 5'ppp-dsRNA challenged DF1. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 65:191-200. [PMID: 27450445 DOI: 10.1016/j.dci.2016.07.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 07/14/2016] [Accepted: 07/15/2016] [Indexed: 06/06/2023]
Abstract
Retinoic acid inducible gene I (RIG-I) can recognize influenza viruses and evoke the innate immune response. RIG-I is absent in the chicken genome, but is conserved in the genome of ducks. Lack of RIG-I renders chickens more susceptible to avian influenza infection, and the clinical symptoms are more prominent than in other poultry. It is unknown whether introduction of duck RIG-I into chicken cells can establish the immunity as is seen in ducks and the role of RIG-I in established immunity is unknown. In this study, a chicken cell strain with stable expression of duRIG-I was established by lentiviral infection, giving DF1/LV5-RIG-I, and a control strain DF1/LV5 was established in parallel. To verify stable, high level expression of duRIG-I in DF1 cells, the levels of duRIG-I mRNA and protein were determined by real-time RT-PCR and Western blot, respectively. Further, 5'triphosphate double stranded RNA (5'ppp-dsRNA) was used to mimic an RNA virus infection and the infected DF1/LV5-RIG-I and DF1/LV5 cells were subjected to high-throughput RNA-sequencing, which yielded 193.46 M reads and 39.07 G bases. A total of 278 differentially expressed genes (DEGs), i.e., duRIG-I-mediated responsive genes, were identified by RNA-seq. Among the 278 genes, 120 DEGs are annotated in the KEGG database, and the most reliable KEGG pathways are likely to be the signaling pathways of RIG-I like receptors. Functional analysis by Gene ontology (GO) indicates that the functions of these DEGs are primarily related to Type I interferon (IFN) signaling, IFN-β-mediated cellular responses and up-regulation of the RIG-I signaling pathway. Based on the shared genes among different pathways, a network representing crosstalk between RIG-I and other signaling pathways was constructed using Cytoscape software. The network suggests that RIG-mediated pathway may crosstalk with the Jak-STAT signaling pathway, Toll-like receptor signaling pathway, Wnt signaling pathway, ubiquitin-mediated proteolysis and MAPK signaling pathway during the transduction of antiviral signals. After screening, a group of key responsive genes in RIG-I-mediated signaling pathways, such as ISG12-2, Mx1, IFIT5, TRIM25, USP18, STAT1, STAT2, IRF1, IRF7 and IRF8, were tested for differential expression by real-time RT-PCR. In summary, by combining our results and the current literature, we propose a RIG-I-mediated signaling network in chickens.
Collapse
Affiliation(s)
- Yang Chen
- The Key Laboratory of Animal Genetics and Breeding and Molecular Design of Jiangsu Province, Yangzhou University, Yangzhou 225009, PR China
| | - Qi Xu
- The Key Laboratory of Animal Genetics and Breeding and Molecular Design of Jiangsu Province, Yangzhou University, Yangzhou 225009, PR China
| | - Yang Li
- The Key Laboratory of Animal Genetics and Breeding and Molecular Design of Jiangsu Province, Yangzhou University, Yangzhou 225009, PR China
| | - Ran Liu
- The Key Laboratory of Animal Genetics and Breeding and Molecular Design of Jiangsu Province, Yangzhou University, Yangzhou 225009, PR China
| | - Zhengyang Huang
- The Key Laboratory of Animal Genetics and Breeding and Molecular Design of Jiangsu Province, Yangzhou University, Yangzhou 225009, PR China
| | - Bin Wang
- The Key Laboratory of Animal Genetics and Breeding and Molecular Design of Jiangsu Province, Yangzhou University, Yangzhou 225009, PR China
| | - Guohong Chen
- The Key Laboratory of Animal Genetics and Breeding and Molecular Design of Jiangsu Province, Yangzhou University, Yangzhou 225009, PR China.
| |
Collapse
|
37
|
Walliser C, Hermkes E, Schade A, Wiese S, Deinzer J, Zapatka M, Désiré L, Mertens D, Stilgenbauer S, Gierschik P. The Phospholipase Cγ2 Mutants R665W and L845F Identified in Ibrutinib-resistant Chronic Lymphocytic Leukemia Patients Are Hypersensitive to the Rho GTPase Rac2 Protein. J Biol Chem 2016; 291:22136-22148. [PMID: 27542411 PMCID: PMC5063995 DOI: 10.1074/jbc.m116.746842] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 08/18/2016] [Indexed: 12/29/2022] Open
Abstract
Mutations in the gene encoding phospholipase C-γ2 (PLCγ2) have been shown to be associated with resistance to targeted therapy of chronic lymphocytic leukemia (CLL) with the Bruton's tyrosine kinase inhibitor ibrutinib. The fact that two of these mutations, R665W and L845F, imparted upon PLCγ2 an ∼2-3-fold ibrutinib-insensitive increase in the concentration of cytosolic Ca2+ following ligation of the B cell antigen receptor (BCR) led to the assumption that the two mutants exhibit constitutively enhanced intrinsic activity. Here, we show that the two PLCγ2 mutants are strikingly hypersensitive to activation by Rac2 such that even wild-type Rac2 suffices to activate the mutant enzymes upon its introduction into intact cells. Enhanced "basal" activity of PLCγ2 in intact cells is shown using the pharmacologic Rac inhibitor EHT 1864 and the PLCγ2F897Q mutation mediating Rac resistance to be caused by Rac-stimulated rather than by constitutively enhanced PLCγ2 activity. We suggest that R665W and L845F be referred to as allomorphic rather than hypermorphic mutations of PLCG2 Rerouting of the transmembrane signals emanating from BCR and converging on PLCγ2 through Rac in ibrutinib-resistant CLL cells may provide novel drug treatment strategies to overcome ibrutinib resistance mediated by PLCG2 mutations or to prevent its development in ibrutinib-treated CLL patients.
Collapse
MESH Headings
- Adenine/analogs & derivatives
- Amino Acid Substitution
- Animals
- COS Cells
- Chlorocebus aethiops
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/enzymology
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Mutation, Missense
- Neoplasm Proteins/antagonists & inhibitors
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- Phospholipase C gamma/antagonists & inhibitors
- Phospholipase C gamma/genetics
- Phospholipase C gamma/metabolism
- Piperidines
- Pyrazoles/pharmacology
- Pyrimidines/pharmacology
- Pyrones/pharmacology
- Quinolines/pharmacology
- Receptors, Antigen, B-Cell/genetics
- Receptors, Antigen, B-Cell/metabolism
- Signal Transduction/drug effects
- Signal Transduction/genetics
- rac GTP-Binding Proteins/genetics
- rac GTP-Binding Proteins/metabolism
- RAC2 GTP-Binding Protein
Collapse
Affiliation(s)
| | | | - Anja Schade
- From the Institute of Pharmacology and Toxicology and
| | - Sebastian Wiese
- the Core Unit Mass Spectrometry and Proteomics, Medical Faculty, Ulm University, 89081 Ulm, Germany
| | - Julia Deinzer
- From the Institute of Pharmacology and Toxicology and
| | - Marc Zapatka
- the Division of Molecular Genetics, German Cancer Research Center (DKFZ), 69121 Heidelberg, Germany, and
| | - Laurent Désiré
- the Diaxonhit, 63-65 Boulevard Masséna, 75013 Paris, France
| | - Daniel Mertens
- Department of Internal Medicine III, Ulm University Medical Center, 89070 Ulm, Germany
| | - Stephan Stilgenbauer
- Department of Internal Medicine III, Ulm University Medical Center, 89070 Ulm, Germany
| | | |
Collapse
|
38
|
Vav1: A Dr. Jekyll and Mr. Hyde protein--good for the hematopoietic system, bad for cancer. Oncotarget 2016; 6:28731-42. [PMID: 26353933 PMCID: PMC4745688 DOI: 10.18632/oncotarget.5086] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2015] [Accepted: 08/07/2015] [Indexed: 01/10/2023] Open
Abstract
Many deregulated signal transducer proteins are involved in various cancers at numerous stages of tumor development. One of these, Vav1, is normally expressed exclusively in the hematopoietic system, where it functions as a specific GDP/GTP nucleotide exchange factor (GEF), strictly regulated by tyrosine phosphorylation. Vav was first identified in an NIH3T3 screen for oncogenes. Although the oncogenic form of Vav1 identified in the screen has not been detected in clinical human tumors, its wild-type form has recently been implicated in mammalian malignancies, including neuroblastoma, melanoma, pancreatic, lung and breast cancers, and B-cell chronic lymphocytic leukemia. In addition, it was recently identified as a mutated gene in human cancers of various origins. However, the activity and contribution to cancer of these Vav1 mutants is still unclear. This review addresses the physiological function of wild-type Vav1 and its activity as an oncogene in human cancer. It also discusses the novel mutations identified in Vav1 in various cancers and their potential contribution to cancer development as oncogenes or tumor suppressor genes.
Collapse
|
39
|
Yan W, Song H, Jiang J, Xu W, Gong Z, Duan Q, Li C, Xie Y, Wang L. Characteristics of B cell‑associated gene expression in patients with coronary artery disease. Mol Med Rep 2016; 13:4113-21. [PMID: 27035867 DOI: 10.3892/mmr.2016.5029] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 02/12/2016] [Indexed: 11/05/2022] Open
Abstract
The current study aimed to identify differentially expressed B cell‑associated genes in peripheral blood mononuclear cells and observe the changes in B cell activation at different stages of coronary artery disease. Groups of patients with acute myocardial infarction (AMI) and stable angina (SA), as well as healthy volunteers, were recruited into the study (n=20 per group). Whole human genome microarray analysis was performed to examine the expression of B cell‑associated genes among these three groups. The mRNA expression levels of 60 genes associated with B cell activity and regulation were measured using reverse transcription‑quantitative polymerase chain reaction. The mRNA expression of the B cell antigen receptor (BCR)‑associated genes, CD45, NFAM, SYK and LYN, were significantly upregulated in patients with AMI; however, FCRL3, CD79B, CD19, CD81, FYN, BLK, CD22 and CD5 mRNA expression levels were significantly downregulated, compared with patients in the SA and control group. The mRNA levels of the T‑independent B cell‑associated genes, CD16, CD32, LILRA1 and TLR9, were significantly increased in AMI patients compared with SA and control patients. The mRNA expression of genes associated with T‑dependent B cells were also measured: EMR2 and CD97 were statistically upregulated, whereas SLAMF1, LY9, CD28, CD43, CD72, ICOSL, PD1, CD40 and CD20 mRNAs were significantly downregulated in AMI group patients compared with the two other groups. Additionally the gene expression levels of B cell regulatory genes were measured. In patients with AMI, CR1, LILRB2, LILRB3 and VAV1 mRNA expression levels were statistically increased, whereas, CS1 and IL4I1 mRNAs were significantly reduced compared with the SA and control groups. There was no statistically significant difference in B cell‑associated gene expression levels between patients with SA and the control group. The present study identified the downregulation of genes associated with BCRs, B2 cells and B cell regulators in patients with AMI, indicating a weakened T cell‑B cell interaction and reduced B2 cell activation during AMI. Thus, improving B2 cell‑mediated humoral immunity may be a potential target for medical intervention in patients with AMI.
Collapse
Affiliation(s)
- Wenwen Yan
- Department of Internal Medicine, Division of Cardiology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R. China
| | - Haoming Song
- Department of Internal Medicine, Division of Cardiology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R. China
| | - Jinfa Jiang
- Department of Internal Medicine, Division of Cardiology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R. China
| | - Wenjun Xu
- Department of Internal Medicine, Division of Cardiology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R. China
| | - Zhu Gong
- Department of Internal Medicine, Division of Cardiology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R. China
| | - Qianglin Duan
- Department of Internal Medicine, Division of Cardiology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R. China
| | - Chuangrong Li
- Department of Internal Medicine, Division of Cardiology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R. China
| | - Yuan Xie
- Department of Internal Medicine, Division of Cardiology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R. China
| | - Lemin Wang
- Department of Internal Medicine, Division of Cardiology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R. China
| |
Collapse
|
40
|
Pandzic T, Larsson J, He L, Kundu S, Ban K, Akhtar-Ali M, Hellström AR, Schuh A, Clifford R, Blakemore SJ, Strefford JC, Baumann T, Lopez-Guillermo A, Campo E, Ljungström V, Mansouri L, Rosenquist R, Sjöblom T, Hellström M. Transposon Mutagenesis Reveals Fludarabine Resistance Mechanisms in Chronic Lymphocytic Leukemia. Clin Cancer Res 2016; 22:6217-6227. [PMID: 26957556 DOI: 10.1158/1078-0432.ccr-15-2903] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 02/16/2016] [Accepted: 02/17/2016] [Indexed: 11/16/2022]
Abstract
PURPOSE To identify resistance mechanisms for the chemotherapeutic drug fludarabine in chronic lymphocytic leukemia (CLL), as innate and acquired resistance to fludarabine-based chemotherapy represents a major challenge for long-term disease control. EXPERIMENTAL DESIGN We used piggyBac transposon-mediated mutagenesis, combined with next-generation sequencing, to identify genes that confer resistance to fludarabine in a human CLL cell line. RESULTS In total, this screen identified 782 genes with transposon integrations in fludarabine-resistant pools of cells. One of the identified genes is a known resistance mediator DCK (deoxycytidine kinase), which encodes an enzyme that is essential for the phosphorylation of the prodrug to the active metabolite. BMP2K, a gene not previously linked to CLL, was also identified as a modulator of response to fludarabine. In addition, 10 of 782 transposon-targeted genes had previously been implicated in treatment resistance based on somatic mutations seen in patients refractory to fludarabine-based therapy. Functional characterization of these genes supported a significant role for ARID5B and BRAF in fludarabine sensitivity. Finally, pathway analysis of transposon-targeted genes and RNA-seq profiling of fludarabine-resistant cells suggested deregulated MAPK signaling as involved in mediating drug resistance in CLL. CONCLUSIONS To our knowledge, this is the first forward genetic screen for chemotherapy resistance in CLL. The screen pinpointed novel genes and pathways involved in fludarabine resistance along with previously known resistance mechanisms. Transposon screens can therefore aid interpretation of cancer genome sequencing data in the identification of genes modifying sensitivity to chemotherapy. Clin Cancer Res; 22(24); 6217-27. ©2016 AACR.
Collapse
Affiliation(s)
- Tatjana Pandzic
- Department of Immunology, Genetics and Pathology, Science for Life laboratory, Uppsala University, Uppsala, Sweden
| | - Jimmy Larsson
- Department of Immunology, Genetics and Pathology, Science for Life laboratory, Uppsala University, Uppsala, Sweden
| | - Liqun He
- Department of Immunology, Genetics and Pathology, Science for Life laboratory, Uppsala University, Uppsala, Sweden
| | - Snehangshu Kundu
- Department of Immunology, Genetics and Pathology, Science for Life laboratory, Uppsala University, Uppsala, Sweden
| | - Kenneth Ban
- Department of Immunology, Genetics and Pathology, Science for Life laboratory, Uppsala University, Uppsala, Sweden.,Department of Biochemistry, Yong Loo Lin School of Medicine, NUS, Institute of Molecular and Cell Biology, A*STAR, Singapore
| | - Muhammad Akhtar-Ali
- Department of Immunology, Genetics and Pathology, Science for Life laboratory, Uppsala University, Uppsala, Sweden
| | - Anders R Hellström
- Department of Immunology, Genetics and Pathology, Science for Life laboratory, Uppsala University, Uppsala, Sweden
| | - Anna Schuh
- Radcliffe Department of Medicine, Oxford University, Oxford, United Kingdom
| | - Ruth Clifford
- Radcliffe Department of Medicine, Oxford University, Oxford, United Kingdom
| | - Stuart J Blakemore
- Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Jonathan C Strefford
- Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Tycho Baumann
- Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | | | - Elias Campo
- Unitat de Hematología, Hospital Clíınic, IDIBAPS, Universitat de Barcelona, Barcelona, Spain
| | - Viktor Ljungström
- Department of Immunology, Genetics and Pathology, Science for Life laboratory, Uppsala University, Uppsala, Sweden
| | - Larry Mansouri
- Department of Immunology, Genetics and Pathology, Science for Life laboratory, Uppsala University, Uppsala, Sweden
| | - Richard Rosenquist
- Department of Immunology, Genetics and Pathology, Science for Life laboratory, Uppsala University, Uppsala, Sweden
| | - Tobias Sjöblom
- Department of Immunology, Genetics and Pathology, Science for Life laboratory, Uppsala University, Uppsala, Sweden
| | - Mats Hellström
- Department of Immunology, Genetics and Pathology, Science for Life laboratory, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
41
|
Yong CSM, Westwood JA, Schröder J, Papenfuss AT, von Scheidt B, Moeller M, Devaud C, Darcy PK, Kershaw MH. Expression of a Chimeric Antigen Receptor in Multiple Leukocyte Lineages in Transgenic Mice. PLoS One 2015; 10:e0140543. [PMID: 26505904 PMCID: PMC4624721 DOI: 10.1371/journal.pone.0140543] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 08/24/2015] [Indexed: 12/25/2022] Open
Abstract
Genetically modified CD8+ T lymphocytes have shown significant anti-tumor effects in the adoptive immunotherapy of cancer, with recent studies highlighting a potential role for a combination of other immune subsets to enhance these results. However, limitations in present genetic modification techniques impose difficulties in our ability to fully explore the potential of various T cell subsets and assess the potential of other leukocytes armed with chimeric antigen receptors (CARs). To address this issue, we generated a transgenic mouse model using a pan-hematopoietic promoter (vav) to drive the expression of a CAR specific for a tumor antigen. Here we present a characterization of the immune cell compartment in two unique vav-CAR transgenic mice models, Founder 9 (F9) and Founder 38 (F38). We demonstrate the vav promoter is indeed capable of driving the expression of a CAR in cells from both myeloid and lymphoid lineage, however the highest level of expression was observed in T lymphocytes from F38 mice. Lymphoid organs in vav-CAR mice were smaller and had reduced cell numbers compared to the wild type (WT) controls. Furthermore, the immune composition of F9 mice differed greatly with a significant reduction in lymphocytes found in the thymus, lymph node and spleen of these mice. To gain insight into the altered immune phenotype of F9 mice, we determined the chromosomal integration site of the transgene in both mouse strains using whole genome sequencing (WGS). We demonstrated that compared to the 7 copies found in F38 mice, F9 mice harbored almost 270 copies. These novel vav-CAR models provide a ready source of CAR expressing myeloid and lymphoid cells and will aid in facilitating future experiments to delineate the role for other leukocytes for adoptive immunotherapy against cancer.
Collapse
Affiliation(s)
- Carmen S. M. Yong
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Jennifer A. Westwood
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Jan Schröder
- Bioinformatics Division, The Walter & Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia
- Bioinformatics and Cancer Genomics, Peter MacCallum Cancer Centre, East Melbourne, Victoria, 3002, Australia
- Department of Computing and Information Systems, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Anthony T. Papenfuss
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria 3010, Australia
- Bioinformatics Division, The Walter & Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia
- Bioinformatics and Cancer Genomics, Peter MacCallum Cancer Centre, East Melbourne, Victoria, 3002, Australia
- Department of Computing and Information Systems, The University of Melbourne, Parkville, Victoria, 3010, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Bianca von Scheidt
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Maria Moeller
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Christel Devaud
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria 3010, Australia
- INSERM U1043 Centre de Physiopathologie Toulouse Purpan (CPTP), Toulouse, France
- * E-mail: (MK); (PD); (CD)
| | - Phillip K. Darcy
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria 3010, Australia
- Department of Immunology, Monash University, Prahran Victoria 3181 Australia
- * E-mail: (MK); (PD); (CD)
| | - Michael H. Kershaw
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria 3010, Australia
- Department of Immunology, Monash University, Prahran Victoria 3181 Australia
- * E-mail: (MK); (PD); (CD)
| |
Collapse
|
42
|
Immunosuppression-Independent Role of Regulatory T Cells against Hypertension-Driven Renal Dysfunctions. Mol Cell Biol 2015; 35:3528-46. [PMID: 26240279 DOI: 10.1128/mcb.00518-15] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 07/24/2015] [Indexed: 01/11/2023] Open
Abstract
Hypertension-associated cardiorenal diseases represent one of the heaviest burdens for current health systems. In addition to hemodynamic damage, recent results have revealed that hematopoietic cells contribute to the development of these diseases by generating proinflammatory and profibrotic environments in the heart and kidney. However, the cell subtypes involved remain poorly characterized. Here we report that CD39(+) regulatory T (TREG) cells utilize an immunosuppression-independent mechanism to counteract renal and possibly cardiac damage during angiotensin II (AngII)-dependent hypertension. This mechanism relies on the direct apoptosis of tissue-resident neutrophils by the ecto-ATP diphosphohydrolase activity of CD39. In agreement with this, experimental and genetic alterations in TREG/TH cell ratios have a direct impact on tissue-resident neutrophil numbers, cardiomyocyte hypertrophy, cardiorenal fibrosis, and, to a lesser extent, arterial pressure elevation during AngII-driven hypertension. These results indicate that TREG cells constitute a first protective barrier against hypertension-driven tissue fibrosis and, in addition, suggest new therapeutic avenues to prevent hypertension-linked cardiorenal diseases.
Collapse
|
43
|
Abstract
INTRODUCTION Rho GTPases are master regulators of actomyosin structure and dynamics and play pivotal roles in a variety of cellular processes including cell morphology, gene transcription, cell cycle progression, and cell adhesion. Because aberrant Rho GTPase signaling activities are widely associated with human cancer, key components of Rho GTPase signaling pathways have attracted increasing interest as potential therapeutic targets. Similar to Ras, Rho GTPases themselves were, until recently, deemed "undruggable" because of structure-function considerations. Several approaches to interfere with Rho GTPase signaling have been explored and show promise as new ways for tackling cancer cells. AREAS COVERED This review focuses on the recent progress in targeting the signaling activities of three prototypical Rho GTPases, that is, RhoA, Rac1, and Cdc42. The authors describe the involvement of these Rho GTPases, their key regulators and effectors in cancer. Furthermore, the authors discuss the current approaches for rationally targeting aberrant Rho GTPases along their signaling cascades, upstream and downstream of Rho GTPases, and posttranslational modifications at a molecular level. EXPERT OPINION To date, while no clinically effective drugs targeting Rho GTPase signaling for cancer treatment are available, tool compounds and lead drugs that pharmacologically inhibit Rho GTPase pathways have shown promise. Small-molecule inhibitors targeting Rho GTPase signaling may add new treatment options for future precision cancer therapy, particularly in combination with other anti-cancer agents.
Collapse
Affiliation(s)
- Yuan Lin
- Division of Experimental Hematology and Cancer Biology, Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio 45229, USA
| | - Yi Zheng
- Division of Experimental Hematology and Cancer Biology, Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio 45229, USA
| |
Collapse
|
44
|
Abstract
Rac and PI3Ks are intracellular signal transducers able to regulate multiple signaling pathways fundamental for cell behavior. PI3Ks are lipid kinases that produce phosphorylated lipids which, in turn, transduce extracellular cues within the cell, while Rac is a small G protein that impacts on actin organization. Compelling evidence indicates that in multiple circumstances the 2 signaling pathways appear intermingled. For instance, phosphorylated lipids produced by PI3Ks recruit and activate GEF and GAP proteins, key modulators of Rac function. Conversely, PI3Ks interact with activated Rac, leading to Rac signaling amplification. This review summarizes the molecular mechanisms underlying the cross-talk between Rac and PI3K signaling in 2 different processes, cell migration and ROS production.
Collapse
Affiliation(s)
- Carlo C Campa
- a Molecular Biotechnology Center; Department of Molecular Biotechnology and Health Sciences; University of Torino ; Torino , Italy
| | | | | | | | | |
Collapse
|
45
|
Walliser C, Tron K, Clauss K, Gutman O, Kobitski AY, Retlich M, Schade A, Röcker C, Henis YI, Nienhaus GU, Gierschik P. Rac-mediated Stimulation of Phospholipase Cγ2 Amplifies B Cell Receptor-induced Calcium Signaling. J Biol Chem 2015; 290:17056-72. [PMID: 25903139 DOI: 10.1074/jbc.m115.645739] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Indexed: 12/21/2022] Open
Abstract
The Rho GTPase Rac is crucially involved in controlling multiple B cell functions, including those regulated by the B cell receptor (BCR) through increased cytosolic Ca(2+). The underlying molecular mechanisms and their relevance to the functions of intact B cells have thus far remained unknown. We have previously shown that the activity of phospholipase Cγ2 (PLCγ2), a key constituent of the BCR signalosome, is stimulated by activated Rac through direct protein-protein interaction. Here, we use a Rac-resistant mutant of PLCγ2 to functionally reconstitute cultured PLCγ2-deficient DT40 B cells and to examine the effects of the Rac-PLCγ2 interaction on BCR-mediated changes of intracellular Ca(2+) and regulation of Ca(2+)-regulated and nuclear-factor-of-activated-T-cell-regulated gene transcription at the level of single, intact B cells. The results show that the functional Rac-PLCγ2 interaction causes marked increases in the following: (i) sensitivity of B cells to BCR ligation; (ii) BCR-mediated Ca(2+) release from intracellular stores; (iii) Ca(2+) entry from the extracellular compartment; and (iv) nuclear translocation of the Ca(2+)-regulated nuclear factor of activated T cells. Hence, Rac-mediated stimulation of PLCγ2 activity serves to amplify B cell receptor-induced Ca(2+) signaling.
Collapse
Affiliation(s)
- Claudia Walliser
- From the Institute of Pharmacology and Toxicology, University of Ulm Medical Center, 89070 Ulm, Germany
| | - Kyrylo Tron
- the Institute of Biophysics, University of Ulm, 89069 Ulm, Germany
| | - Karen Clauss
- the Institute of Biophysics, University of Ulm, 89069 Ulm, Germany
| | - Orit Gutman
- the Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Andrei Yu Kobitski
- the Institute of Applied Physics and Center for Functional Nanostructures, Karlsruhe Institute of Technology, 76131 Karlsruhe, Germany
| | - Michael Retlich
- From the Institute of Pharmacology and Toxicology, University of Ulm Medical Center, 89070 Ulm, Germany
| | - Anja Schade
- From the Institute of Pharmacology and Toxicology, University of Ulm Medical Center, 89070 Ulm, Germany
| | - Carlheinz Röcker
- the Institute of Biophysics, University of Ulm, 89069 Ulm, Germany
| | - Yoav I Henis
- the Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - G Ulrich Nienhaus
- the Institute of Applied Physics and Center for Functional Nanostructures, Karlsruhe Institute of Technology, 76131 Karlsruhe, Germany, the Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, 76344 Eggenstein-Leopoldshafen, Germany, and the Department of Physics, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801
| | - Peter Gierschik
- From the Institute of Pharmacology and Toxicology, University of Ulm Medical Center, 89070 Ulm, Germany,
| |
Collapse
|
46
|
Medici M, Visser WE, Visser TJ, Peeters RP. Genetic determination of the hypothalamic-pituitary-thyroid axis: where do we stand? Endocr Rev 2015; 36:214-44. [PMID: 25751422 DOI: 10.1210/er.2014-1081] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
For a long time it has been known that both hypo- and hyperthyroidism are associated with an increased risk of morbidity and mortality. In recent years, it has also become clear that minor variations in thyroid function, including subclinical dysfunction and variation in thyroid function within the reference range, can have important effects on clinical endpoints, such as bone mineral density, depression, metabolic syndrome, and cardiovascular mortality. Serum thyroid parameters show substantial interindividual variability, whereas the intraindividual variability lies within a narrow range. This suggests that every individual has a unique hypothalamus-pituitary-thyroid axis setpoint that is mainly determined by genetic factors, and this heritability has been estimated to be 40-60%. Various mutations in thyroid hormone pathway genes have been identified in persons with thyroid dysfunction or altered thyroid function tests. Because these causes are rare, many candidate gene and linkage studies have been performed over the years to identify more common variants (polymorphisms) associated with thyroid (dys)function, but only a limited number of consistent associations have been found. However, in the past 5 years, advances in genetic research have led to the identification of a large number of new candidate genes. In this review, we provide an overview of the current knowledge about the polygenic basis of thyroid (dys)function. This includes new candidate genes identified by genome-wide approaches, what insights these genes provide into the genetic basis of thyroid (dys)function, and which new techniques will help to further decipher the genetic basis of thyroid (dys)function in the near future.
Collapse
Affiliation(s)
- Marco Medici
- Rotterdam Thyroid Center, Department of Internal Medicine, Erasmus Medical Center, 3015 GE Rotterdam, The Netherlands
| | | | | | | |
Collapse
|
47
|
Burbage M, Keppler SJ, Gasparrini F, Martínez-Martín N, Gaya M, Feest C, Domart MC, Brakebusch C, Collinson L, Bruckbauer A, Batista FD. Cdc42 is a key regulator of B cell differentiation and is required for antiviral humoral immunity. J Exp Med 2015; 212:53-72. [PMID: 25547673 PMCID: PMC4291523 DOI: 10.1084/jem.20141143] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 11/25/2014] [Indexed: 12/22/2022] Open
Abstract
The small Rho GTPase Cdc42, known to interact with Wiskott-Aldrich syndrome (WAS) protein, is an important regulator of actin remodeling. Here, we show that genetic ablation of Cdc42 exclusively in the B cell lineage is sufficient to render mice unable to mount antibody responses. Indeed Cdc42-deficient mice are incapable of forming germinal centers or generating plasma B cells upon either viral infection or immunization. Such severe immune deficiency is caused by multiple and profound B cell abnormalities, including early blocks during B cell development; impaired antigen-driven BCR signaling and actin remodeling; defective antigen presentation and in vivo interaction with T cells; and a severe B cell-intrinsic block in plasma cell differentiation. Thus, our study presents a new perspective on Cdc42 as key regulator of B cell physiology.
Collapse
Affiliation(s)
- Marianne Burbage
- Lymphocyte Interaction Laboratory, Electron Microscopy Unit, London Research Institute, Cancer Research UK, London WC2A 3LY, England, UK
| | - Selina J Keppler
- Lymphocyte Interaction Laboratory, Electron Microscopy Unit, London Research Institute, Cancer Research UK, London WC2A 3LY, England, UK
| | - Francesca Gasparrini
- Lymphocyte Interaction Laboratory, Electron Microscopy Unit, London Research Institute, Cancer Research UK, London WC2A 3LY, England, UK
| | - Nuria Martínez-Martín
- Lymphocyte Interaction Laboratory, Electron Microscopy Unit, London Research Institute, Cancer Research UK, London WC2A 3LY, England, UK
| | - Mauro Gaya
- Lymphocyte Interaction Laboratory, Electron Microscopy Unit, London Research Institute, Cancer Research UK, London WC2A 3LY, England, UK
| | - Christoph Feest
- Lymphocyte Interaction Laboratory, Electron Microscopy Unit, London Research Institute, Cancer Research UK, London WC2A 3LY, England, UK
| | - Marie-Charlotte Domart
- Lymphocyte Interaction Laboratory, Electron Microscopy Unit, London Research Institute, Cancer Research UK, London WC2A 3LY, England, UK
| | - Cord Brakebusch
- Biomedical Institute, Biotech Research and Innovation Centre, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Lucy Collinson
- Lymphocyte Interaction Laboratory, Electron Microscopy Unit, London Research Institute, Cancer Research UK, London WC2A 3LY, England, UK
| | - Andreas Bruckbauer
- Lymphocyte Interaction Laboratory, Electron Microscopy Unit, London Research Institute, Cancer Research UK, London WC2A 3LY, England, UK
| | - Facundo D Batista
- Lymphocyte Interaction Laboratory, Electron Microscopy Unit, London Research Institute, Cancer Research UK, London WC2A 3LY, England, UK
| |
Collapse
|
48
|
Kurgonaite K, Gandhi H, Kurth T, Pautot S, Schwille P, Weidemann T, Bökel C. Essential role of endocytosis for Interleukin-4 receptor mediated JAK/STAT signalling. J Cell Sci 2015; 128:3781-95. [DOI: 10.1242/jcs.170969] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 08/21/2015] [Indexed: 01/15/2023] Open
Abstract
Many important signalling cascades operate through specialized signalling endosomes, but a corresponding mechanism has as yet not been described for hematopoietic cytokine receptors. Based on live cell affinity measurements we recently proposed that ligand induced Interleukin-4 receptor (IL-4R) complex formation and thus JAK/STAT pathway activation requires a local, subcellular increase in receptor density. Here we show that this concentration step is provided by the internalization of IL-4R subunits through a constitutive, Rac1/Pak and actin mediated endocytosis route that causes IL-4R subunits to become enriched by about two orders of magnitude within a population of cortical endosomes. Consistently, ligand induced receptor dimers are preferentially detected within these endosomes. IL-4 signalling can be blocked by pharmacological inhibitors targeting the actin polymerization machinery driving receptor internalization, placing endocytosis unambigously upstream of receptor activation. Together these observations demonstrate a role for endocytosis that is mechanistically distinct from the scaffolding function of signalling endosomes in other pathways.
Collapse
Affiliation(s)
- Kristina Kurgonaite
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Fetscherstr. 105, 01307 Dresden, Germany
| | - Hetvi Gandhi
- BIOTEC/Biophysics, Technische Universität Dresden, Tatzberg 47-51, 01307 Dresden, Germany
| | - Thomas Kurth
- BIOTEC/Biophysics, Technische Universität Dresden, Tatzberg 47-51, 01307 Dresden, Germany
| | - Sophie Pautot
- BIOTEC/Biophysics, Technische Universität Dresden, Tatzberg 47-51, 01307 Dresden, Germany
| | - Petra Schwille
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Fetscherstr. 105, 01307 Dresden, Germany
| | - Thomas Weidemann
- BIOTEC/Biophysics, Technische Universität Dresden, Tatzberg 47-51, 01307 Dresden, Germany
| | - Christian Bökel
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Fetscherstr. 105, 01307 Dresden, Germany
| |
Collapse
|
49
|
The Role of p110δ in the Development and Activation of B Lymphocytes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 850:119-35. [DOI: 10.1007/978-3-319-15774-0_9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
50
|
The guanine nucleotide exchange factor Vav3 regulates differentiation of progenitor cells in the developing mouse retina. Cell Tissue Res 2014; 359:423-440. [PMID: 25501893 DOI: 10.1007/s00441-014-2050-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 11/03/2014] [Indexed: 10/24/2022]
Abstract
The seven main cell types in the mammalian retina arise from multipotent retinal progenitor cells, a process that is tightly regulated by intrinsic and extrinsic signals. However, the molecular mechanisms that control proliferation, differentiation and cell-fate decisions of retinal progenitor cells are not fully understood yet. Here, we report that the guanine nucleotide exchange factor Vav3, a regulator of Rho-GTPases, is involved in retinal development. We demonstrate that Vav3 is expressed in the mouse retina during the embryonic period. In order to study the role of Vav3 in the developing retina, we generate Vav3-deficient mice. The loss of Vav3 results in an accelerated differentiation of retinal ganglion cells and cone photoreceptors during early and late embryonic development. We provide evidence that more retinal progenitor cells express the late progenitor marker Sox9 in Vav3-deficient mice than in wild-types. This premature differentiation is compensated during the postnatal period and late-born cell types such as bipolar cells and Müller glia display normal numbers. Taken together, our data imply that Vav3 is a regulator of retinal progenitor cell differentiation, thus highlighting a novel role for guanine nucleotide exchange factors in retinogenesis.
Collapse
|