1
|
Pasculli E, Gadaleta RM, Arconzo M, Cariello M, Moschetta A. The Role of Exogenous microRNAs on Human Health: The Plant-Human Trans-Kingdom Hypothesis. Nutrients 2024; 16:3658. [PMID: 39519491 PMCID: PMC11547593 DOI: 10.3390/nu16213658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 10/25/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
MicroRNAs (miRNAs) are small, endogenous, single-stranded RNAs that act on gene silencing at the post-transcriptional level by binding to a target messenger RNA (mRNA), leading to its degradation or inhibiting translation into functional proteins. The key role of miRNAs in development, proliferation, differentiation andapoptosis has been deeply investigated, revealing that deregulation in their expression is critical in various diseases, such as metabolic disorders and cancer. Since these small molecules initially evolved as a mechanism of protection against viruses and transposable elements, the fascinating hypothesis that they can move between organisms both of the same or different species has been postulated. Trans-kingdom is the term used to define the migration that occurs between species. This mechanism has been well analyzed between plants and their pests, in order to boost defense and increase pathogenicity, respectively. Intriguingly, in the last decades, the plant-human trans-kingdom migration via food intake hypothesis arose. In particular, various studies highlighted the ability of exogenous miRNAs, abundant in the mainly consumed plant-derived food, to enter the human body affecting gene expression. Notably, plant miRNAs can resist the strict conditions of the gastrointestinal tract through a methylation step that occurs during miRNA maturation, conferring high stability to these small molecules. Recent studies observed the anti-tumoral, immune modulator and anti-inflammatory abilities of trans-kingdom interaction between plant and human. Here, we depict the existing knowledge and discuss the fascinating plant-human trans-kingdom interaction, highlighting first the eventual role of plant miRNAs from foods on our somatic gene identity card and then the potential impact of using plant miRNAs as novel therapeutic avenues.
Collapse
Affiliation(s)
- Emanuela Pasculli
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (E.P.); (R.M.G.); (M.A.)
| | - Raffaella Maria Gadaleta
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (E.P.); (R.M.G.); (M.A.)
- INBB National Institute for Biostructure and Biosystems, Viale delle Medaglie d’Oro 305, 00136 Rome, Italy
| | - Maria Arconzo
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (E.P.); (R.M.G.); (M.A.)
| | - Marica Cariello
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (E.P.); (R.M.G.); (M.A.)
- INBB National Institute for Biostructure and Biosystems, Viale delle Medaglie d’Oro 305, 00136 Rome, Italy
| | - Antonio Moschetta
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (E.P.); (R.M.G.); (M.A.)
- INBB National Institute for Biostructure and Biosystems, Viale delle Medaglie d’Oro 305, 00136 Rome, Italy
| |
Collapse
|
2
|
Ruiz-Martín V, Marcos T, de Pereda JM, Sánchez-Crespo M, de la Fuente MA, Bayón Y, Alonso A. LYP regulates SLP76 and other adaptor proteins in T cells. Biol Res 2024; 57:69. [PMID: 39342392 PMCID: PMC11438317 DOI: 10.1186/s40659-024-00536-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 08/06/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND The LYP tyrosine phosphatase presents a SNP (1858C > T) that increases the risk of developing autoimmune diseases such as type I diabetes and arthritis. It remains unclear how this SNP affects LYP function and promotes the development of these diseases. The scarce information about LYP substrates is in part responsible for the poor understanding of LYP function. RESULTS In this study, we identify in T lymphocytes several adaptor proteins as potential substrates targeted by LYP, including FYB, SLP-76, HS-1, Vav, SKAP1 and SKAP2. We also show that LYP co-localizes with SLP76 in microclusters, upon TCR engagement. CONCLUSIONS These data indicate that LYP may modulate T cell activation by dephosphorylating several adaptor proteins, such as FYB, SLP-76, HS-1, Vav, SKAP1 and SKAP2 upon TCR engagement.
Collapse
Affiliation(s)
- Virginia Ruiz-Martín
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), CSIC-Universidad de Valladolid, c/ Sanz y Forés 3, 47003, Valladolid, Spain
| | - Tamara Marcos
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), CSIC-Universidad de Valladolid, c/ Sanz y Forés 3, 47003, Valladolid, Spain
| | - José María de Pereda
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC), CSIC-Universidad de Salamanca, Campus Unamuno, 37007, Salamanca, Spain
| | - Mariano Sánchez-Crespo
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), CSIC-Universidad de Valladolid, c/ Sanz y Forés 3, 47003, Valladolid, Spain
| | - Miguel Angel de la Fuente
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), CSIC-Universidad de Valladolid, c/ Sanz y Forés 3, 47003, Valladolid, Spain
| | - Yolanda Bayón
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), CSIC-Universidad de Valladolid, c/ Sanz y Forés 3, 47003, Valladolid, Spain.
| | - Andrés Alonso
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), CSIC-Universidad de Valladolid, c/ Sanz y Forés 3, 47003, Valladolid, Spain.
| |
Collapse
|
3
|
Gunasinghe SD, Peres NG, Goyette J, Gaus K. Biomechanics of T Cell Dysfunctions in Chronic Diseases. Front Immunol 2021; 12:600829. [PMID: 33717081 PMCID: PMC7948521 DOI: 10.3389/fimmu.2021.600829] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 01/12/2021] [Indexed: 12/12/2022] Open
Abstract
Understanding the mechanisms behind T cell dysfunctions during chronic diseases is critical in developing effective immunotherapies. As demonstrated by several animal models and human studies, T cell dysfunctions are induced during chronic diseases, spanning from infections to cancer. Although factors governing the onset and the extent of the functional impairment of T cells can differ during infections and cancer, most dysfunctional phenotypes share common phenotypic traits in their immune receptor and biophysical landscape. Through the latest developments in biophysical techniques applied to explore cell membrane and receptor-ligand dynamics, we are able to dissect and gain further insights into the driving mechanisms behind T cell dysfunctions. These insights may prove useful in developing immunotherapies aimed at reinvigorating our immune system to fight off infections and malignancies more effectively. The recent success with checkpoint inhibitors in treating cancer opens new avenues to develop more effective, targeted immunotherapies. Here, we highlight the studies focused on the transformation of the biophysical landscape during infections and cancer, and how T cell biomechanics shaped the immunopathology associated with chronic diseases.
Collapse
Affiliation(s)
- Sachith D Gunasinghe
- EMBL Australia Node in Single Molecule Science, University of New South Wales, Sydney, NSW, Australia.,ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, NSW, Australia
| | - Newton G Peres
- EMBL Australia Node in Single Molecule Science, University of New South Wales, Sydney, NSW, Australia.,ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, NSW, Australia
| | - Jesse Goyette
- EMBL Australia Node in Single Molecule Science, University of New South Wales, Sydney, NSW, Australia.,ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, NSW, Australia
| | - Katharina Gaus
- EMBL Australia Node in Single Molecule Science, University of New South Wales, Sydney, NSW, Australia.,ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
4
|
Minutolo A, Potestà M, Roglia V, Cirilli M, Iacovelli F, Cerva C, Fokam J, Desideri A, Andreoni M, Grelli S, Colizzi V, Muleo R, Montesano C. Plant microRNAs from Moringa oleifera Regulate Immune Response and HIV Infection. Front Pharmacol 2021; 11:620038. [PMID: 33643043 PMCID: PMC7905167 DOI: 10.3389/fphar.2020.620038] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 12/03/2020] [Indexed: 11/24/2022] Open
Abstract
Traditional medicine is often chosen due to its affordability, its familiarity with patient’s cultural practices, and its wider access to the local community. Plants play an important role in providing indispensable nutrients, while specific small RNAs can regulate human gene expression in a cross-kingdom manner. The aim of the study was to evaluate the effects of plant-enriched purified extract microRNAs from Moringa oleifera seeds (MO) on the immune response and on HIV infection. Bioinformatic analysis shows that plant microRNAs (p-miRs) from MO belonging to 18 conserved families, including p-miR160h, p-miR166, p-miR482b, p-miR159c, p-miR395d, p-miR2118a, p-miR393a, p-miR167f-3p, and p-miR858b are predicted to target with high affinity BCL2, IL2RA, TNF, and VAV1, all these being involved in the cell cycle, apoptosis, immune response and also in the regulation of HIV pathogenesis. The effects of MO p-miRs transfected into HIV+ PBMCs were analyzed and revealed a decrease in viability associated with an increase of apoptosis; an increase of T helper cells expressing Fas and a decrease of intracellular Bcl2 protein expression. Meanwhile no effects were detected in PBMCs from healthy donors. In CD4+ T cells, transfection significantly reduced cell activation and modified the T cell differentiation, thereby decreasing both central and effector memory cells while increasing terminal effector memory cells. Interestingly, the p-miRs transfection induces a reduction of intracellular HIV p24 protein and a reduction of viral DNA integration. Finally, we evaluated the effect of synthetic (mimic) p-miR858b whose sequence is present in the MO p-miR pool and predicted to target VAV1, a protein involved in HIV-Nef binding. This protein plays a pivotal role in T cell antigen receptor (TCR) signaling, so triggering the activation of various pathways. The transfection of HIV+ PBMCs with the synthetic p-miR858b showed a reduced expression of VAV1 and HIV p24 proteins. Overall, our evidence defines putative mechanisms underlying a supplementary benefit of traditional medicine, alongside current antiretroviral therapy, in managing HIV infection in resource-limited settings where MO remains widely available.
Collapse
Affiliation(s)
| | - Marina Potestà
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Valentina Roglia
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Marco Cirilli
- Department of Agricultural and Forestry Science, University of Tuscia, Viterbo, Italy.,Department of Agricultural and Environmental Sciences, University of Milan, Milan, Italy
| | | | - Carlotta Cerva
- Department of System Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Joseph Fokam
- Chantal BIYA International Reference Centre for Research on HIV/AIDS Prevention and Management, Yaoundé, Cameroon
| | | | - Massimo Andreoni
- Department of System Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Sandro Grelli
- Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Vittorio Colizzi
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy.,Faculty of Sciences and Technology, Evangelic University of Cameroon, Bandjoun, Cameroon
| | - Rosario Muleo
- Department of Agricultural and Forestry Science, University of Tuscia, Viterbo, Italy
| | - Carla Montesano
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| |
Collapse
|
5
|
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
|
6
|
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
|
7
|
Rodríguez-Fdez S, Fernández-Nevado L, Lorenzo-Martín LF, Bustelo XR. Lysine Acetylation Reshapes the Downstream Signaling Landscape of Vav1 in Lymphocytes. Cells 2020; 9:cells9030609. [PMID: 32143292 PMCID: PMC7140538 DOI: 10.3390/cells9030609] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 02/27/2020] [Accepted: 03/02/2020] [Indexed: 12/16/2022] Open
Abstract
Vav1 works both as a catalytic Rho GTPase activator and an adaptor molecule. These functions, which are critical for T cell development and antigenic responses, are tyrosine phosphorylation-dependent. However, it is not known whether other posttranslational modifications can contribute to the regulation of the biological activity of this protein. Here, we show that Vav1 becomes acetylated on lysine residues in a stimulation- and SH2 domain-dependent manner. Using a collection of both acetylation- and deacetylation-mimicking mutants, we show that the acetylation of four lysine residues (Lys222, Lys252, Lys587, and Lys716) leads to the downmodulation of the adaptor function of Vav1 that triggers the stimulation of the nuclear factor of activated T cells (NFAT). These sites belong to two functional subclasses according to mechanistic criteria. We have also unveiled additional acetylation sites potentially involved in either the stimulation (Lys782) or the downmodulation (Lys335, Lys374) of specific Vav1-dependent downstream responses. Collectively, these results indicate that Nε-lysine acetylation can play variegated roles in the regulation of Vav1 signaling. Unlike the case of the tyrosine phosphorylation step, this new regulatory layer is not conserved in other Vav family paralogs.
Collapse
Affiliation(s)
- Sonia Rodríguez-Fdez
- Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain; (S.R.-F.); (L.F.-N.); (L.F.L.-M.)
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, 37007 Salamanca, Spain
| | - Lucía Fernández-Nevado
- Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain; (S.R.-F.); (L.F.-N.); (L.F.L.-M.)
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain
| | - L. Francisco Lorenzo-Martín
- Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain; (S.R.-F.); (L.F.-N.); (L.F.L.-M.)
- 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; (S.R.-F.); (L.F.-N.); (L.F.L.-M.)
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, 37007 Salamanca, Spain
- Correspondence: ; Tel.: +34-663194634
| |
Collapse
|
8
|
Abstract
Advances in academic and clinical studies during the last several years have resulted in practical outcomes in adoptive immune therapy of cancer. Immune cells can be programmed with molecular modules that increase their therapeutic potency and specificity. It has become obvious that successful immunotherapy must take into account the full complexity of the immune system and, when possible, include the use of multifactor cell reprogramming that allows fast adjustment during the treatment. Today, practically all immune cells can be stably or transiently reprogrammed against cancer. Here, we review works related to T cell reprogramming, as the most developed field in immunotherapy. We discuss factors that determine the specific roles of αβ and γδ T cells in the immune system and the structure and function of T cell receptors in relation to other structures involved in T cell target recognition and immune response. We also discuss the aspects of T cell engineering, specifically the construction of synthetic T cell receptors (synTCRs) and chimeric antigen receptors (CARs) and the use of engineered T cells in integrative multifactor therapy of cancer.
Collapse
Affiliation(s)
- Samuel G Katz
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | | |
Collapse
|
9
|
Rodríguez-Fdez S, Bustelo XR. The Vav GEF Family: An Evolutionary and Functional Perspective. Cells 2019; 8:E465. [PMID: 31100928 PMCID: PMC6562523 DOI: 10.3390/cells8050465] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 05/10/2019] [Accepted: 05/15/2019] [Indexed: 02/07/2023] Open
Abstract
Vav proteins play roles as guanosine nucleotide exchange factors for Rho GTPases and signaling adaptors downstream of protein tyrosine kinases. The recent sequencing of the genomes of many species has revealed that this protein family originated in choanozoans, a group of unicellular organisms from which animal metazoans are believed to have originated from. Since then, the Vav family underwent expansions and reductions in its members during the evolutionary transitions that originated the agnates, chondrichthyes, some teleost fish, and some neoaves. Exotic members of the family harboring atypical structural domains can be also found in some invertebrate species. In this review, we will provide a phylogenetic perspective of the evolution of the Vav family. We will also pay attention to the structure, signaling properties, regulatory layers, and functions of Vav proteins in both invertebrate and vertebrate species.
Collapse
Affiliation(s)
- Sonia Rodríguez-Fdez
- Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC) and University of Salamanca, Campus Unamuno, E37007 Salamanca, Spain.
- Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC) and University of Salamanca, Campus Unamuno, E37007 Salamanca, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Consejo Superior de Investigaciones Científicas (CSIC) and University of Salamanca, Campus Unamuno, E37007 Salamanca, Spain.
| | - Xosé R Bustelo
- Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC) and University of Salamanca, Campus Unamuno, E37007 Salamanca, Spain.
- Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC) and University of Salamanca, Campus Unamuno, E37007 Salamanca, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Consejo Superior de Investigaciones Científicas (CSIC) and University of Salamanca, Campus Unamuno, E37007 Salamanca, Spain.
| |
Collapse
|
10
|
Del Río-Iñiguez I, Vázquez-Chávez E, Cuche C, Di Bartolo V, Bouchet J, Alcover A. HIV-1 Nef Hijacks Lck and Rac1 Endosomal Traffic To Dually Modulate Signaling-Mediated and Actin Cytoskeleton-Mediated T Cell Functions. THE JOURNAL OF IMMUNOLOGY 2018; 201:2624-2640. [PMID: 30282749 DOI: 10.4049/jimmunol.1800372] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 09/03/2018] [Indexed: 01/03/2023]
Abstract
Endosomal traffic of TCR and signaling molecules regulates immunological synapse formation and T cell activation. We recently showed that Rab11 endosomes regulate the subcellular localization of the tyrosine kinase Lck and of the GTPase Rac1 and control their functions in TCR signaling and actin cytoskeleton remodeling. HIV-1 infection of T cells alters their endosomal traffic, activation capacity, and actin cytoskeleton organization. The viral protein Nef is pivotal for these modifications. We hypothesized that HIV-1 Nef could jointly alter Lck and Rac1 endosomal traffic and concomitantly modulate their functions. In this study, we show that HIV-1 infection of human T cells sequesters both Lck and Rac1 in a pericentrosomal compartment in an Nef-dependent manner. Strikingly, the Nef-induced Lck compartment contains signaling-competent forms (phosphorylated on key Tyr residues) of Lck and some of its downstream effectors, TCRζ, ZAP70, SLP76, and Vav1, avoiding the proximal LAT adaptor. Importantly, Nef-induced concentration of signaling molecules was concomitant with the upregulation of several early and late T cell activation genes. Moreover, preventing the concentration of the Nef-induced Lck compartment by depleting the Rab11 effector FIP3 counteracted Nef-induced gene expression upregulation. In addition, Nef extensively sequesters Rac1 and downregulates Rac1-dependent actin cytoskeleton remodeling, thus reducing T cell spreading. Therefore, by modifying their endosomal traffic, Nef hijacks signaling and actin cytoskeleton regulators to dually modulate their functional outputs. Our data shed new light into the molecular mechanisms that modify T cell physiology during HIV-1 infection.
Collapse
Affiliation(s)
- Iratxe Del Río-Iñiguez
- Lymphocyte Cell Biology Unit, Department of Immunology, Institut Pasteur, 75724 Paris, France.,INSERM U1221, 75015 Paris, France; and.,Collège Doctoral, Sorbonne Université, 75014 Paris, France
| | - Elena Vázquez-Chávez
- Lymphocyte Cell Biology Unit, Department of Immunology, Institut Pasteur, 75724 Paris, France.,INSERM U1221, 75015 Paris, France; and
| | - Céline Cuche
- Lymphocyte Cell Biology Unit, Department of Immunology, Institut Pasteur, 75724 Paris, France.,INSERM U1221, 75015 Paris, France; and
| | - Vincenzo Di Bartolo
- Lymphocyte Cell Biology Unit, Department of Immunology, Institut Pasteur, 75724 Paris, France.,INSERM U1221, 75015 Paris, France; and
| | - Jérôme Bouchet
- Lymphocyte Cell Biology Unit, Department of Immunology, Institut Pasteur, 75724 Paris, France; .,INSERM U1221, 75015 Paris, France; and
| | - Andrés Alcover
- Lymphocyte Cell Biology Unit, Department of Immunology, Institut Pasteur, 75724 Paris, France; .,INSERM U1221, 75015 Paris, France; and
| |
Collapse
|
11
|
Barreira M, Rodríguez-Fdez S, Bustelo XR. New insights into the Vav1 activation cycle in lymphocytes. Cell Signal 2018; 45:132-144. [PMID: 29410283 PMCID: PMC7615736 DOI: 10.1016/j.cellsig.2018.01.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Revised: 01/28/2018] [Accepted: 01/30/2018] [Indexed: 10/18/2022]
Abstract
Vav1 is a hematopoietic-specific Rho GDP/GTP exchange factor and signaling adaptor. Although these activities are known to be stimulated by direct Vav1 phosphorylation, little information still exists regarding the regulatory layers that influence the overall Vav1 activation cycle. Using a collection of cell models and activation-mimetic Vav1 mutants, we show here that the dephosphorylated state of Vav1 in nonstimulated T cells requires the presence of a noncatalytic, phospholipase Cγ1-Slp76-mediated inhibitory pathway. Upon T cell stimulation, Vav1 becomes rapidly phosphorylated via the engagement of Lck and, to a much lesser extent, other Src family kinases and Zap70. In this process, Lck, Zap70 and the adaptor protein Lat contribute differently to the dynamics and amplitude of the Vav1 phosphorylated pool. Consistent with a multiphosphosite activation mechanism, the optimal stimulation of Vav1 can only be recapitulated by the combination of several activation-mimetic phosphosite mutants. The analysis of these mutants has also unveiled the presence of different Vav1 signaling competent states that are influenced by phosphosites present in the N- and C-terminal domains of the protein.
Collapse
Affiliation(s)
- María Barreira
- Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC), University of Salamanca, 37007 Salamanca, Spain; Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC), University of Salamanca, 37007 Salamanca, Spain
| | - Sonia Rodríguez-Fdez
- Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC), University of Salamanca, 37007 Salamanca, Spain; Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC), University of Salamanca, 37007 Salamanca, Spain
| | - Xosé R Bustelo
- Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC), University of Salamanca, 37007 Salamanca, Spain; Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC), University of Salamanca, 37007 Salamanca, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Consejo Superior de Investigaciones Científicas (CSIC), University of Salamanca, 37007 Salamanca, Spain.
| |
Collapse
|
12
|
Abstract
CD28 superagonistic antibodies (CD28SAb) can preferentially activate and expand immunosuppressive regulatory T cells (Treg) in mice. However, pre-clinical trials assessing CD28SAbs for the therapy of autoimmune diseases reveal severe systemic inflammatory response syndrome in humans, thereby implying the existence of distinct signalling abilities between human and mouse CD28. Here, we show that a single amino acid variant within the C-terminal proline-rich motif of human and mouse CD28 (P212 in human vs. A210 in mouse) regulates CD28-induced NF-κB activation and pro-inflammatory cytokine gene expression. Moreover, this Y209APP212 sequence in humans is crucial for the association of CD28 with the Nck adaptor protein for actin cytoskeleton reorganisation events necessary for CD28 autonomous signalling. This study thus unveils different outcomes between human and mouse CD28 signalling to underscore the importance of species difference when transferring results from preclinical models to the bedside. CD28 transmits co-stimulatory signals for the activation of both mouse and human T cells, but in vivo hyperactivation of CD28 has opposite effects on system immunity. Here, the authors show that a single amino acid difference between mouse and human CD28 dictates this function distinction via differential recruitment of Nck.
Collapse
|
13
|
Mutzbauer G, Maurus K, Buszello C, Pischimarov J, Roth S, Rosenwald A, Chott A, Geissinger E. SYK expression in monomorphic epitheliotropic intestinal T-cell lymphoma. Mod Pathol 2018; 31:505-516. [PMID: 29052597 DOI: 10.1038/modpathol.2017.145] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Revised: 09/02/2017] [Accepted: 09/11/2017] [Indexed: 01/02/2023]
Abstract
Monomorphic epitheliotropic intestinal T-cell lymphoma (MEITL), formerly known as type II enteropathy associated T-cell lymphoma (type II EATL), is a rare, aggressive primary intestinal T-cell lymphoma with a poor prognosis and an incompletely understood pathogenesis. We collected 40 cases of MEITL and 27 cases of EATL, formerly known as type I EATL, and comparatively investigated the T-cell receptor (TCR) itself and associated signaling molecules using immunohistochemistry, amplicon deep sequencing and bisulfite pyrosequencing. The TCR showed both an αβ-T-cell origin (30%) and a γδ-T-cell derivation (55%) resulting in a predominant positive TCR phenotype in MEITL compared with the mainly silent TCR phenotype in EATL (65%). The immunohistochemical expression of the spleen tyrosine kinase (SYK) turned out to be a distinctive feature of MEITL (95%) compared with EATL (0%). Aberrant SYK overexpression in MEITL is likely caused by hypomethylation of the SYK promoter, while no common mutations in the SYK gene or in its promoter could be detected. Using amplicon deep sequencing, mutations in DNMT3A, IDH2, and TET2 were infrequent events in MEITL and EATL. Immunohistochemical expression of linker for activation of T-cells (LAT) subdivided MEITL into a LAT expressing subset (33%) and a LAT silent subset (67%) with a potentially earlier disease onset in LAT-positive MEITL.
Collapse
Affiliation(s)
- Grit Mutzbauer
- Institute of Pathology, University of Wuerzburg, Wuerzburg, Germany
| | - Katja Maurus
- Institute of Pathology, University of Wuerzburg, Wuerzburg, Germany
| | - Clara Buszello
- Institute of Pathology, University of Wuerzburg, Wuerzburg, Germany
| | | | - Sabine Roth
- Institute of Pathology, University of Wuerzburg, Wuerzburg, Germany
| | - Andreas Rosenwald
- Institute of Pathology, University of Wuerzburg, Wuerzburg, Germany.,Comprehensive Cancer Center Mainfranken, University and University Hospital, Wuerzburg, Germany
| | - Andreas Chott
- Institute of Pathology and Microbiology, Wilhelminenspital, Vienna, Austria
| | - Eva Geissinger
- Institute of Pathology, University of Wuerzburg, Wuerzburg, Germany.,Comprehensive Cancer Center Mainfranken, University and University Hospital, Wuerzburg, Germany
| |
Collapse
|
14
|
Adamek M, Döhler B, Hasan KK, Fiedler G, Scherer S, Opelz G, Tran TH. Assessing the impact of FoxP3 and Vav1 gene polymorphisms on kidney allograft survival. HLA 2017; 90:102-105. [DOI: 10.1111/tan.13049] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Revised: 04/01/2017] [Accepted: 04/11/2017] [Indexed: 01/14/2023]
Affiliation(s)
- M. Adamek
- Transplantation Immunology, Institute of Immunology; University of Heidelberg; Heidelberg Germany
| | - B. Döhler
- Transplantation Immunology, Institute of Immunology; University of Heidelberg; Heidelberg Germany
| | - K. K. Hasan
- Department of Biology, College of Science; University of Baghdad; Baghdad Iraq
| | - G. Fiedler
- Transplantation Immunology, Institute of Immunology; University of Heidelberg; Heidelberg Germany
| | - S. Scherer
- Transplantation Immunology, Institute of Immunology; University of Heidelberg; Heidelberg Germany
| | - G. Opelz
- Transplantation Immunology, Institute of Immunology; University of Heidelberg; Heidelberg Germany
| | - T. H. Tran
- Transplantation Immunology, Institute of Immunology; University of Heidelberg; Heidelberg Germany
| |
Collapse
|
15
|
Abstract
Three-dimensional live cell imaging of the interaction of T cells with antigen-presenting cells (APCs) visualizes the subcellular distributions of signaling intermediates during T cell activation at thousands of resolved positions within a cell. These information-rich maps of local protein concentrations are a valuable resource in understanding T cell signaling. Here, we describe a protocol for the efficient acquisition of such imaging data and their computational processing to create four-dimensional maps of local concentrations. This protocol allows quantitative analysis of T cell signaling as it occurs inside live cells with resolution in time and space across thousands of cells.
Collapse
Affiliation(s)
- Rachel Ambler
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
| | - Xiangtao Ruan
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, 7723 Gates-Hillman Center, Pittsburgh, PA, 15213, USA
| | - Robert F Murphy
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, 7723 Gates-Hillman Center, Pittsburgh, PA, 15213, USA.
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, 15213, USA.
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, USA.
- Department of Machine Learning, Carnegie Mellon University, Pittsburgh, PA, 15213, USA.
| | - Christoph Wülfing
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK.
| |
Collapse
|
16
|
TGFβ promotes mesenchymal phenotype of pancreatic cancer cells, in part, through epigenetic activation of VAV1. Oncogene 2016; 36:2202-2214. [PMID: 27893715 DOI: 10.1038/onc.2016.378] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 08/14/2016] [Accepted: 08/29/2016] [Indexed: 02/06/2023]
Abstract
The highly homeostasis-resistant nature of cancer cells leads to their escape from treatment and to liver metastasis, which in turn makes pancreatic ductal adenocarcinoma (PDAC) difficult to treat, especially the squamous/epithelial-to-mesenchymal transition (EMT)-like subtype. As the molecular mechanisms underlying tumour heterogeneity remain elusive, we investigated whether epigenetic regulation might explain inter-individual differences in the progression of specific subtypes. DNA methylation profiling performed on cancer tissues prior to chemo/radiotherapy identified one hypermethylated CpG site (CpG6882469) in the VAV1 gene body that was correlated with demethylation of two promoter CpGs (CpG6772370/CpG6772811) in both PDAC and peripheral blood. Transforming growth factor β treatment induced gene-body hypermethylation, dissociation of DNMT1 from the promoter, and VAV1 expression via SMAD4 and mutant KrasG12D. Pharmacological inhibition of TGFβ-VAV1 signalling decreased the squamous/EMT-like cancer cells, promoted nuclear VAV1 localization, and enhanced the efficacy of gemcitabine in prolonging the survival of KPfl/flC mice. Together, the three VAV1 CpGs serve as biomarkers for prognosis and early detection, and the TGFβ-VAV1 axis represents a therapeutic target.
Collapse
|
17
|
A Natural Variant of the T Cell Receptor-Signaling Molecule Vav1 Reduces Both Effector T Cell Functions and Susceptibility to Neuroinflammation. PLoS Genet 2016; 12:e1006185. [PMID: 27438086 PMCID: PMC4954684 DOI: 10.1371/journal.pgen.1006185] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 06/22/2016] [Indexed: 12/16/2022] Open
Abstract
The guanine nucleotide exchange factor Vav1 is essential for transducing T cell antigen receptor signals and therefore plays an important role in T cell development and activation. Our previous genetic studies identified a locus on rat chromosome 9 that controls the susceptibility to neuroinflammation and contains a non-synonymous polymorphism in the major candidate gene Vav1. To formally demonstrate the causal implication of this polymorphism, we generated a knock-in mouse bearing this polymorphism (Vav1R63W). Using this model, we show that Vav1R63W mice display reduced susceptibility to experimental autoimmune encephalomyelitis (EAE) induced by MOG35-55 peptide immunization. This is associated with a lower production of effector cytokines (IFN-γ, IL-17 and GM-CSF) by autoreactive CD4 T cells. Despite increased proportion of Foxp3+ regulatory T cells in Vav1R63W mice, we show that this lowered cytokine production is intrinsic to effector CD4 T cells and that Treg depletion has no impact on EAE development. Finally, we provide a mechanism for the above phenotype by showing that the Vav1R63W variant has normal enzymatic activity but reduced adaptor functions. Together, these data highlight the importance of Vav1 adaptor functions in the production of inflammatory cytokines by effector T cells and in the susceptibility to neuroinflammation. The understanding of the physiological role of Vav1, a key regulator of T cell receptor signaling, was primarily inferred from studies using Vav1-deficient mice. Such models, however, provide little insight on how polymorphisms leading to quantitative changes in Vav1 activity could affect immune system functions. In the present study, we focused on a recently identified Vav1R63W natural variant that has been supposed to play a central role in the susceptibility to neuroinflammation. Using a Vav1R63W knock-in mouse model, we show that Vav1R63W leads to defects in adaptor functions and reduces the susceptibility to experimental autoimmune encephalomyelitis, together with an intrinsic defect in the production of Th1/Th17 cytokines by autoreactive effector CD4 T cells. Thus, our study highlights the importance of Vav1 adaptor functions in CD4 T cells differentiation and suggests that genetic or acquired alterations of this Vav1 function could play a major role in susceptibility to Th1/Th17 mediated diseases.
Collapse
|
18
|
Palmer E, Drobek A, Stepanek O. Opposing effects of actin signaling and LFA-1 on establishing the affinity threshold for inducing effector T-cell responses in mice. Eur J Immunol 2016; 46:1887-901. [PMID: 27188212 DOI: 10.1002/eji.201545909] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 05/02/2016] [Accepted: 05/13/2016] [Indexed: 11/10/2022]
Abstract
Mature CD8(+) T cells use a narrow antigen affinity threshold to generate tissue-infiltrating cytotoxic effector T cells and induce autoimmune pathology, but the mechanisms that establish this antigen affinity threshold are poorly understood. Only antigens with affinities above the threshold induce stable contacts with APCs, polarization of a T cell, and asymmetric T-cell division. Previously published data indicate that LFA-1 inside-out signaling might be involved in establishing the antigen affinity threshold. Here, we show that subthreshold antigens weakly activate all major distal TCR signaling pathways. Low-affinity antigens are more dependent on LFA-1 than suprathreshold antigens. Moreover, augmenting the inside-out signaling by hyperactive Rap1 does not increase responses to the subthreshold antigens. Thus, LFA-1 signaling does not contribute to the affinity-based antigen discrimination. However, we found that subthreshold antigens do not induce actin rearrangement toward an APC, mediated by Rho-family GTPases, Cdc42, and Rac. Our data suggest that Rac and Cdc42 contribute to the establishment of the antigen affinity threshold in CD8(+) T cells by enhancing responses to high-affinity antigens, or by reducing the responses to low-affinity antigens.
Collapse
Affiliation(s)
- Ed Palmer
- Departments of Biomedicine and Nephrology, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Ales Drobek
- Laboratory of Adaptive Immunity, Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czech Republic
| | - Ondrej Stepanek
- Departments of Biomedicine and Nephrology, University Hospital Basel and University of Basel, Basel, Switzerland.,Laboratory of Adaptive Immunity, Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
19
|
Zvezdova E, Mikolajczak J, Garreau A, Marcellin M, Rigal L, Lee J, Choi S, Blaize G, Argenty J, Familiades J, Li L, Gonzalez de Peredo A, Burlet-Schiltz O, Love PE, Lesourne R. Themis1 enhances T cell receptor signaling during thymocyte development by promoting Vav1 activity and Grb2 stability. Sci Signal 2016; 9:ra51. [PMID: 27188442 DOI: 10.1126/scisignal.aad1576] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The T cell signaling protein Themis1 is essential for the positive and negative selection of thymocytes in the thymus. Although the developmental defect that results from the loss of Themis1 suggests that it enhances T cell receptor (TCR) signaling, Themis1 also recruits Src homology 2 domain-containing phosphatase-1 (SHP-1) to the vicinity of TCR signaling complexes, suggesting that it has an inhibitory role in TCR signaling. We used TCR signaling reporter mice and quantitative proteomics to explore the role of Themis1 in developing T cells. We found that Themis1 acted mostly as a positive regulator of TCR signaling in vivo when receptors were activated by positively selecting ligands. Proteomic analysis of the Themis1 interactome identified SHP-1, the TCR-associated adaptor protein Grb2, and the guanine nucleotide exchange factor Vav1 as the principal interacting partners of Themis1 in isolated mouse thymocytes. Analysis of TCR signaling in Themis1-deficient and Themis1-overexpressing mouse thymocytes demonstrated that Themis1 promoted Vav1 activity both in vitro and in vivo. The reduced activity of Vav1 and the impaired T cell development in Themis1(-/-) mice were due in part to increased degradation of Grb2, which suggests that Themis1 is required to maintain the steady-state abundance of Grb2 in thymocytes. Together, these data suggest that Themis1 acts as a positive regulator of TCR signaling in developing T cells, and identify a mechanism by which Themis1 regulates thymic selection.
Collapse
Affiliation(s)
- Ekaterina Zvezdova
- Section on Cellular and Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Judith Mikolajczak
- Centre de Physiopathologie de Toulouse Purpan, Toulouse F-31300, France. Institut National de la Santé et de la Recherche Médicale, U1043, Toulouse F-31300, France. Centre National de la Recherche Scientifique, U5282, Toulouse F-31300, France. Université de Toulouse, Université Paul Sabatier, Toulouse F-31300, France
| | - Anne Garreau
- Centre de Physiopathologie de Toulouse Purpan, Toulouse F-31300, France. Institut National de la Santé et de la Recherche Médicale, U1043, Toulouse F-31300, France. Centre National de la Recherche Scientifique, U5282, Toulouse F-31300, France. Université de Toulouse, Université Paul Sabatier, Toulouse F-31300, France
| | - Marlène Marcellin
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse F-31077, France
| | - Lise Rigal
- Centre de Physiopathologie de Toulouse Purpan, Toulouse F-31300, France. Institut National de la Santé et de la Recherche Médicale, U1043, Toulouse F-31300, France. Centre National de la Recherche Scientifique, U5282, Toulouse F-31300, France. Université de Toulouse, Université Paul Sabatier, Toulouse F-31300, France
| | - Jan Lee
- Section on Cellular and Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Seeyoung Choi
- Section on Cellular and Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Gaëtan Blaize
- Centre de Physiopathologie de Toulouse Purpan, Toulouse F-31300, France. Institut National de la Santé et de la Recherche Médicale, U1043, Toulouse F-31300, France. Centre National de la Recherche Scientifique, U5282, Toulouse F-31300, France. Université de Toulouse, Université Paul Sabatier, Toulouse F-31300, France
| | - Jérémy Argenty
- Centre de Physiopathologie de Toulouse Purpan, Toulouse F-31300, France. Institut National de la Santé et de la Recherche Médicale, U1043, Toulouse F-31300, France. Centre National de la Recherche Scientifique, U5282, Toulouse F-31300, France. Université de Toulouse, Université Paul Sabatier, Toulouse F-31300, France
| | - Julien Familiades
- Centre de Physiopathologie de Toulouse Purpan, Toulouse F-31300, France. Institut National de la Santé et de la Recherche Médicale, U1043, Toulouse F-31300, France. Centre National de la Recherche Scientifique, U5282, Toulouse F-31300, France. Université de Toulouse, Université Paul Sabatier, Toulouse F-31300, France
| | - Liqi Li
- Section on Cellular and Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Anne Gonzalez de Peredo
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse F-31077, France
| | - Odile Burlet-Schiltz
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse F-31077, France
| | - Paul E Love
- Section on Cellular and Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Renaud Lesourne
- Centre de Physiopathologie de Toulouse Purpan, Toulouse F-31300, France. Institut National de la Santé et de la Recherche Médicale, U1043, Toulouse F-31300, France. Centre National de la Recherche Scientifique, U5282, Toulouse F-31300, France. Université de Toulouse, Université Paul Sabatier, Toulouse F-31300, France.
| |
Collapse
|
20
|
Abstract
The Vav family is a group of tyrosine phosphorylation-regulated signal transduction molecules hierarchically located downstream of protein tyrosine kinases. The main function of these proteins is to work as guanosine nucleotide exchange factors (GEFs) for members of the Rho GTPase family. In addition, they can exhibit a variety of catalysis-independent roles in specific signaling contexts. Vav proteins play essential signaling roles for both the development and/or effector functions of a large variety of cell lineages, including those belonging to the immune, nervous, and cardiovascular systems. They also contribute to pathological states such as cancer, immune-related dysfunctions, and atherosclerosis. Here, I will provide an integrated view about the evolution, regulation, and effector properties of these signaling molecules. In addition, I will discuss the pros and cons for their potential consideration as therapeutic targets.
Collapse
Key Words
- Ac, acidic
- Ahr, aryl hydrocarbon receptor
- CH, calponin homology
- CSH3, most C-terminal SH3 domain of Vav proteins
- DAG, diacylglycerol
- DH, Dbl-homology domain
- Dbl-homology
- GDP/GTP exchange factors
- GEF, guanosine nucleotide exchange factor
- HIV, human immunodeficiency virus
- IP3, inositoltriphosphate
- NFAT, nuclear factor of activated T-cells
- NSH3, most N-terminal SH3 domain of Vav proteins
- PH, plekstrin-homology domain
- PI3K, phosphatidylinositol-3 kinase
- PIP3, phosphatidylinositol (3,4,5)-triphosphate
- PKC, protein kinase C
- PKD, protein kinase D
- PLC-g, phospholipase C-g
- PRR, proline-rich region
- PTK, protein tyrosine kinase
- Phox, phagocyte oxidase
- Rho GTPases
- SH2, Src homology 2
- SH3, Src homology 3
- SNP, single nucleotide polymorphism
- TCR, T-cell receptor
- Vav
- ZF, zinc finger region
- cGMP, cyclic guanosine monophosphate
- cancer
- cardiovascular biology
- disease
- immunology
- nervous system
- signaling
- therapies
Collapse
Affiliation(s)
- Xosé R Bustelo
- a Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer ; Consejo Superior de Investigaciones Científicas (CSIC) and University of Salamanca ; Campus Unamuno; Salamanca , Spain
| |
Collapse
|
21
|
Roybal KT, Sinai P, Verkade P, Murphy RF, Wülfing C. The actin-driven spatiotemporal organization of T-cell signaling at the system scale. Immunol Rev 2014; 256:133-47. [PMID: 24117818 DOI: 10.1111/imr.12103] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
T cells are activated through interaction with antigen-presenting cells (APCs). During activation, receptors and signaling intermediates accumulate in diverse spatiotemporal distributions. These distributions control the probability of signaling interactions and thus govern information flow through the signaling system. Spatiotemporally resolved system-scale investigation of signaling can extract the regulatory information thus encoded, allowing unique insight into the control of T-cell function. Substantial technical challenges exist, and these are briefly discussed herein. While much of the work assessing T-cell spatiotemporal organization uses planar APC substitutes, we focus here on B-cell APCs with often stark differences. Spatiotemporal signaling distributions are driven by cell biologically distinct structures, a large protein assembly at the interface center, a large invagination, the actin-supported interface periphery as extended by smaller individual lamella, and a newly discovered whole-interface actin-driven lamellum. The more than 60 elements of T-cell activation studied to date are dynamically distributed between these structures, generating a complex organization of the signaling system. Signal initiation and core signaling prefer the interface center, while signal amplification is localized in the transient lamellum. Actin dynamics control signaling distributions through regulation of the underlying structures and drive a highly undulating T-cell/APC interface that imposes substantial constraints on T-cell organization. We suggest that the regulation of actin dynamics, by controlling signaling distributions and membrane topology, is an important rheostat of T-cell signaling.
Collapse
Affiliation(s)
- Kole T Roybal
- Department of Immunology, UT Southwestern Medical Center, Dallas, TX, USA
| | | | | | | | | |
Collapse
|
22
|
Barreira M, Fabbiano S, Couceiro JR, Torreira E, Martínez-Torrecuadrada JL, Montoya G, Llorca O, Bustelo XR. The C-terminal SH3 domain contributes to the intramolecular inhibition of Vav family proteins. Sci Signal 2014; 7:ra35. [PMID: 24736456 DOI: 10.1126/scisignal.2004993] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Vav proteins are phosphorylation-dependent guanine nucleotide exchange factors (GEFs) that catalyze the activation of members of the Rho family of guanosine triphosphatases (GTPases). The current regulatory model holds that the nonphosphorylated, catalytically inactive state of these GEFs is maintained by intramolecular interactions among the amino-terminal domains and the central catalytic core, which block the binding of Vav proteins to GTPases. We showed that this autoinhibition is mechanistically more complex, also involving the bivalent association of the carboxyl-terminal Src homology 3 (SH3) region of Vav with its catalytic and pleckstrin homology (PH) domains. Such interactions occurred through proline-rich region-independent mechanisms. Full release from this double-locked state required synergistic weakening effects from multiple phosphorylated tyrosine residues, thus providing an optimized system to generate gradients of Vav GEF activity depending on upstream signaling inputs. This mechanism is shared by mammalian and Drosophila melanogaster Vav proteins, suggesting that it may be a common regulatory feature for this protein family.
Collapse
Affiliation(s)
- María Barreira
- 1Centro de Investigación del Cáncer, Campus Unamuno, E37007 Salamanca, Spain
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Zenker S, Panteleev-Ivlev J, Wirtz S, Kishimoto T, Waldner MJ, Ksionda O, Tybulewicz VLJ, Neurath MF, Atreya I. A key regulatory role for Vav1 in controlling lipopolysaccharide endotoxemia via macrophage-derived IL-6. THE JOURNAL OF IMMUNOLOGY 2014; 192:2830-2836. [PMID: 24532586 DOI: 10.4049/jimmunol.1300157] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Macrophages are centrally involved in the pathogenesis of acute inflammatory diseases, peritonitis, endotoxemia, and septic shock. However, the molecular mechanisms controlling such macrophage activation are incompletely understood. In this article, we provide evidence that Vav1, a member of the RhoGEF family, plays a crucial role in macrophage activation and septic endotoxemia. Vav1-deficient mice demonstrated a significantly increased susceptibility for LPS endotoxemia that could be abrogated by anti-IL-6R Ab treatment. Subsequent studies showed that Vav1-deficient macrophages display augmented production of the proinflammatory cytokine IL-6. Nuclear Vav1 was identified as a key negative regulator of macrophage-derived IL-6 production. In fact, Vav1 formed a nuclear DNA-binding complex with heat shock transcription factor 1 at the HSE2 region of the IL-6 promoter to suppress IL-6 gene transcription in macrophages. These findings provide new insights into the pathogenesis of endotoxemia and suggest new avenues for therapy.
Collapse
Affiliation(s)
- Stefanie Zenker
- Medical Clinic 1, Friedrich-Alexander University of Erlangen-Nürnberg, University Hospital of Erlangen, Germany
| | - Julia Panteleev-Ivlev
- Medical Clinic 1, Friedrich-Alexander University of Erlangen-Nürnberg, University Hospital of Erlangen, Germany
| | - Stefan Wirtz
- Medical Clinic 1, Friedrich-Alexander University of Erlangen-Nürnberg, University Hospital of Erlangen, Germany
| | | | - Maximilian J Waldner
- Medical Clinic 1, Friedrich-Alexander University of Erlangen-Nürnberg, University Hospital of Erlangen, Germany
| | - Olga Ksionda
- MRC National Institute for Medical Research, London, United Kingdom
| | | | - Markus F Neurath
- Medical Clinic 1, Friedrich-Alexander University of Erlangen-Nürnberg, University Hospital of Erlangen, Germany
| | - Imke Atreya
- Medical Clinic 1, Friedrich-Alexander University of Erlangen-Nürnberg, University Hospital of Erlangen, Germany
| |
Collapse
|
24
|
Majkut P, Claußnitzer I, Merk H, Freund C, Hackenberger CPR, Gerrits M. Completion of proteomic data sets by Kd measurement using cell-free synthesis of site-specifically labeled proteins. PLoS One 2013; 8:e82352. [PMID: 24340019 PMCID: PMC3858276 DOI: 10.1371/journal.pone.0082352] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Accepted: 10/16/2013] [Indexed: 11/20/2022] Open
Abstract
The characterization of phosphotyrosine mediated protein-protein interactions is vital for the interpretation of downstream pathways of transmembrane signaling processes. Currently however, there is a gap between the initial identification and characterization of cellular binding events by proteomic methods and the in vitro generation of quantitative binding information in the form of equilibrium rate constants (Kd values). In this work we present a systematic, accelerated and simplified approach to fill this gap: using cell-free protein synthesis with site-specific labeling for pull-down and microscale thermophoresis (MST) we were able to validate interactions and to establish a binding hierarchy based on Kd values as a completion of existing proteomic data sets. As a model system we analyzed SH2-mediated interactions of the human T-cell phosphoprotein ADAP. Putative SH2 domain-containing binding partners were synthesized from a cDNA library using Expression-PCR with site-specific biotinylation in order to analyze their interaction with fluorescently labeled and in vitro phosphorylated ADAP by pull-down. On the basis of the pull-down results, selected SH2’s were subjected to MST to determine Kd values. In particular, we could identify an unexpectedly strong binding of ADAP to the previously found binding partner Rasa1 of about 100 nM, while no evidence of interaction was found for the also predicted SH2D1A. Moreover, Kd values between ADAP and its known binding partners SLP-76 and Fyn were determined. Next to expanding data on ADAP suggesting promising candidates for further analysis in vivo, this work marks the first Kd values for phosphotyrosine/SH2 interactions on a phosphoprotein level.
Collapse
Affiliation(s)
- Paul Majkut
- Department Chemical Biology II, Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | | | | | - Christian Freund
- Institut für Chemie und Biochemie, Freie Universität Berlin, Berlin, Germany
| | - Christian P. R. Hackenberger
- Department Chemical Biology II, Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany
- Institut für Chemie, Humboldt-Universität zu Berlin, Berlin, Germany
- * E-mail: (MG); (CH)
| | | |
Collapse
|