Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Cell ; 156(6): 1340-1340.e1, 2014 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-24630731

RESUMO

Integrin αß transmembrane heterodimers play central roles in metazoan development and physiology by mediating adhesion and by transmitting forces and biochemical signals across the plasma membrane. In this SnapShot, we present a simplified "modular" view of the integrin adhesome, centered on the talin-integrin interaction, and provide examples of how this view can help to unravel the adhesome's remarkable functional diversity and plasticity.


Assuntos
Integrinas/metabolismo , Talina/metabolismo , Sequência de Aminoácidos , Animais , Adesão Celular , Integrinas/química , Dados de Sequência Molecular , Talina/química
2.
J Immunol ; 208(6): 1378-1388, 2022 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-35197328

RESUMO

Agonist-induced Rap1 GTP loading results in integrin activation involved in T cell trafficking and functions. MRL proteins Rap1-interacting adapter molecule (RIAM) and lamellipodin (LPD) are Rap1 effectors that can recruit talin1 to integrins, resulting in integrin activation. Recent work also implicates direct Rap1-talin1 interaction in integrin activation. Here, we analyze in mice the connections between Rap1 and talin1 that support integrin activation in conventional CD4+ T (Tconv) and CD25HiFoxp3+CD4+ regulatory T (Treg) cells. Talin1(R35E, R118E) mutation that disrupts both Rap1 binding sites results in a partial defect in αLß2, α4ß1, and α4ß7 integrin activation in both Tconv and Treg cells with resulting defects in T cell homing. Talin1(R35E,R118E) Tconv manifested reduced capacity to induce colitis in an adoptive transfer mouse model. Loss of RIAM exacerbates the defects in Treg cell function caused by the talin1(R35E,R118E) mutation, and deleting both MRL proteins in combination with talin1(R35E,R118E) phenocopy the complete lack of integrin activation observed in Rap1a/b-null Treg cells. In sum, these data reveal the functionally significant connections between Rap1 and talin1 that enable αLß2, α4ß1, and α4ß7 integrin activation in CD4+ T cells.


Assuntos
Talina , Proteínas rap1 de Ligação ao GTP , Animais , Sítios de Ligação , Linfócitos T CD4-Positivos/metabolismo , Integrinas/metabolismo , Camundongos , Talina/genética , Talina/metabolismo , Proteínas rap1 de Ligação ao GTP/metabolismo
3.
J Biol Chem ; 296: 100675, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33865854

RESUMO

Interaction of talin with the cytoplasmic tails of integrin ß triggers integrin activation, leading to an increase of integrin affinity/avidity for extracellular ligands. In talin KO mice, loss of talin interaction with platelet integrin αIIbß3 causes a severe hemostatic defect, and loss of talin interaction with endothelial cell integrin αVß3 affects angiogenesis. In normal cells, talin is autoinhibited and localized in the cytoplasm. Here, we used an optogenetic platform to assess whether recruitment of full-length talin to the plasma membrane was sufficient to induce integrin activation. A dimerization module (Arabidopsis cryptochrome 2 fused to the N terminus of talin; N-terminal of cryptochrome-interacting basic helix-loop-helix domain ended with a CAAX box protein [C: cysteine; A: aliphatic amino acid; X: any C-terminal amino acid]) responsive to 450 nm (blue) light was inserted into Chinese hamster ovary cells and endothelial cells also expressing αIIbß3 or αVß3, respectively. Thus, exposure of the cells to blue light caused a rapid and reversible recruitment of Arabidopsis cryptochrome 2-talin to the N-terminal of cryptochrome-interacting basic helix-loop-helix domain ended with a CAAX box protein [C: cysteine; A: aliphatic amino acid; X: any C-terminal amino acid]-decorated plasma membrane. This resulted in ß3 integrin activation in both cell types, as well as increasing migration of the endothelial cells. However, membrane recruitment of talin was not sufficient for integrin activation, as membrane-associated Ras-related protein 1 (Rap1)-GTP was also required. Moreover, talin mutations that interfered with its direct binding to Rap1 abrogated ß3 integrin activation. Altogether, these results define a role for the plasma membrane recruitment of talin in ß3 integrin activation, and they suggest a nuanced sequence of events thereafter involving Rap1-GTP.


Assuntos
Membrana Celular/metabolismo , Citoplasma/metabolismo , Células Endoteliais/metabolismo , Optogenética , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Talina/metabolismo , Proteínas rap1 de Ligação ao GTP/metabolismo , Animais , Células CHO , Cricetinae , Cricetulus , Camundongos , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/genética , Ligação Proteica , Talina/genética , Proteínas rap1 de Ligação ao GTP/genética
4.
Blood ; 136(10): 1180-1190, 2020 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-32518959

RESUMO

Ras-related protein 1 (Rap1) is a major convergence point of the platelet-signaling pathways that result in talin-1 binding to the integrin ß cytoplasmic domain and consequent integrin activation, platelet aggregation, and effective hemostasis. The nature of the connection between Rap1 and talin-1 in integrin activation is an important remaining gap in our understanding of this process. Previous work identified a low-affinity Rap1-binding site in the talin-1 F0 domain that makes a small contribution to integrin activation in platelets. We recently identified an additional Rap1-binding site in the talin-1 F1 domain that makes a greater contribution than F0 in model systems. Here we generated mice bearing point mutations, which block Rap1 binding without affecting talin-1 expression, in either the talin-1 F1 domain (R118E) alone, which were viable, or in both the F0 and F1 domains (R35E,R118E), which were embryonic lethal. Loss of the Rap1-talin-1 F1 interaction in platelets markedly decreases talin-1-mediated activation of platelet ß1- and ß3-integrins. Integrin activation and platelet aggregation in mice whose platelets express only talin-1(R35E, R118E) are even more impaired, resembling the defect seen in platelets lacking both Rap1a and Rap1b. Although Rap1 is important in thrombopoiesis, platelet secretion, and surface exposure of phosphatidylserine, loss of the Rap1-talin-1 interaction in talin-1(R35E, R118E) platelets had little effect on these processes. These findings show that talin-1 is the principal direct effector of Rap1 GTPases that regulates platelet integrin activation in hemostasis.


Assuntos
Integrina beta1/metabolismo , Integrina beta3/metabolismo , Mutação Puntual , Talina/fisiologia , Trombopoese , Proteínas rap de Ligação ao GTP/fisiologia , Proteínas rap1 de Ligação ao GTP/fisiologia , Animais , Feminino , Integrina beta1/genética , Integrina beta3/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ativação Plaquetária , Agregação Plaquetária , Domínios Proteicos , Transdução de Sinais
5.
J Immunol ; 205(10): 2883-2892, 2020 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-33077644

RESUMO

CD98, which is required for the rapid proliferation of both normal and cancer cells, and MET, the hepatocyte growth factor receptor, are potential targets for therapeutic antitumor Abs. In this study, we report that the antiproliferative activity of a prototype anti-CD98 Ab, UM7F8, is due to Ab-induced membrane-associated ring CH (MARCH) E3 ubiquitin ligase-mediated ubiquitination and downregulation of cell surface CD98. MARCH1-mediated ubiquitination of CD98 is required for UM7F8's capacity to reduce CD98 surface expression and its capacity to inhibit the proliferation of murine T cells. Similarly, CD98 ubiquitination is required for UM7F8's capacity to block the colony-forming ability of murine leukemia-initiating cells. To test the potential generality of the paradigm that MARCH E3 ligases can mediate the antiproliferative response to antitumor Abs, we examined the potential effects of MARCH proteins on responses to emibetuzumab, an anti-MET Ab currently in clinical trials for various cancers. We report that MET surface expression is reduced by MARCH1, 4, or 8-mediated ubiquitination and that emibetuzumab-induced MET ubiquitination contributes to its capacity to downregulate MET and inhibit human tumor cell proliferation. Thus, MARCH E3 ligases can act as cofactors for antitumor Abs that target cell surface proteins, suggesting that the MARCH protein repertoire of cells is a determinant of their response to such Abs.


Assuntos
Antígenos de Neoplasias/metabolismo , Antineoplásicos Imunológicos/farmacologia , Cadeia Pesada da Proteína-1 Reguladora de Fusão/metabolismo , Neoplasias/tratamento farmacológico , Ubiquitina-Proteína Ligases/metabolismo , Animais , Anticorpos Monoclonais Humanizados/farmacologia , Anticorpos Monoclonais Humanizados/uso terapêutico , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Antineoplásicos Imunológicos/uso terapêutico , Proliferação de Células/efeitos dos fármacos , Cadeia Pesada da Proteína-1 Reguladora de Fusão/antagonistas & inibidores , Cadeia Pesada da Proteína-1 Reguladora de Fusão/genética , Cadeia Pesada da Proteína-1 Reguladora de Fusão/imunologia , Técnicas de Inativação de Genes , Células HeLa , Humanos , Células Jurkat , Camundongos , Camundongos Knockout , Neoplasias/imunologia , Neoplasias/patologia , Proteólise , Proteínas Proto-Oncogênicas c-met/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-met/genética , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Ubiquitina-Proteína Ligases/genética , Ubiquitinação/imunologia
6.
Blood ; 133(3): 193-204, 2019 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-30442679

RESUMO

Cerebral cavernous malformations (CCMs) are common brain vascular dysplasias that are prone to acute and chronic hemorrhage with significant clinical sequelae. The pathogenesis of recurrent bleeding in CCM is incompletely understood. Here, we show that central nervous system hemorrhage in CCMs is associated with locally elevated expression of the anticoagulant endothelial receptors thrombomodulin (TM) and endothelial protein C receptor (EPCR). TM levels are increased in human CCM lesions, as well as in the plasma of patients with CCMs. In mice, endothelial-specific genetic inactivation of Krit1 (Krit1 ECKO ) or Pdcd10 (Pdcd10 ECKO ), which cause CCM formation, results in increased levels of vascular TM and EPCR, as well as in enhanced generation of activated protein C (APC) on endothelial cells. Increased TM expression is due to upregulation of transcription factors KLF2 and KLF4 consequent to the loss of KRIT1 or PDCD10. Increased TM expression contributes to CCM hemorrhage, because genetic inactivation of 1 or 2 copies of the Thbd gene decreases brain hemorrhage in Pdcd10 ECKO mice. Moreover, administration of blocking antibodies against TM and EPCR significantly reduced CCM hemorrhage in Pdcd10 ECKO mice. Thus, a local increase in the endothelial cofactors that generate anticoagulant APC can contribute to bleeding in CCMs, and plasma soluble TM may represent a biomarker for hemorrhagic risk in CCMs.


Assuntos
Anticoagulantes/metabolismo , Proteínas Reguladoras de Apoptose/fisiologia , Hemorragia Cerebral/diagnóstico , Endotélio Vascular/patologia , Hemangioma Cavernoso do Sistema Nervoso Central/complicações , Proteína KRIT1/fisiologia , Proteínas de Membrana/fisiologia , Proteína C/metabolismo , Proteínas Proto-Oncogênicas/fisiologia , Trombomodulina/sangue , Adulto , Animais , Coagulação Sanguínea , Estudos de Casos e Controles , Hemorragia Cerebral/sangue , Hemorragia Cerebral/etiologia , Receptor de Proteína C Endotelial/metabolismo , Endotélio Vascular/metabolismo , Hemangioma Cavernoso do Sistema Nervoso Central/metabolismo , Hemangioma Cavernoso do Sistema Nervoso Central/fisiopatologia , Humanos , Fator 4 Semelhante a Kruppel , Camundongos , Camundongos Knockout , Transdução de Sinais , Adulto Jovem
7.
J Immunol ; 198(9): 3410-3415, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28348273

RESUMO

Rap1-interacting adaptor molecule (RIAM) is a Rap1 effector that mediates the recruitment of talin to integrins, thereby supporting their activation. In this study, we investigated the role of RIAM in an adoptive transfer model for type I diabetes and report that RIAM expression in T cells is necessary for diabetes development. Loss of RIAM did not prevent lymphocyte recruitment to draining lymph nodes 24 h after transfer, but it was required for Ag-driven proliferation and cytotoxic killing. RIAM is recruited to immune synapses along with talin and LFA-1, and loss of RIAM profoundly suppresses Ag-dependent conjugate formation in primary naive and effector T cells. These data identify the requirement of RIAM for formation of immunological synapses and in resulting T cell functions in autoimmunity. Moreover, because RIAM-null mice are healthy, fertile, and display no bleeding abnormalities, our results identify RIAM and its regulators as potential targets for therapies of T cell-mediated autoimmunity.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Células Apresentadoras de Antígenos/imunologia , Diabetes Mellitus Tipo 1/imunologia , Antígeno-1 Associado à Função Linfocitária/metabolismo , Proteínas de Membrana/metabolismo , Linfócitos T/imunologia , Talina/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Transferência Adotiva , Animais , Proliferação de Células/genética , Células Cultivadas , Citotoxicidade Imunológica/genética , Humanos , Sinapses Imunológicas/imunologia , Proteínas de Membrana/genética , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Linfócitos T/transplante
8.
Blood ; 128(4): 479-87, 2016 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-27207789

RESUMO

Integrin adhesion receptors mediate the adhesion of blood cells, such as leukocytes, to other cells, such as endothelial cells. Integrins also are critical for anchorage of hematopoietic precursors to the extracellular matrix. Blood cells can dynamically regulate the affinities of integrins for their ligands ("activation"), an event central to their functions. Here we review recent progress in understanding the mechanisms of integrin activation with a focus on the functions of blood cells. We discuss how talin binding to the integrin ß cytoplasmic domain, in conjunction with the plasma membrane, induces long-range allosteric rearrangements that lead to integrin activation. Second, we review our understanding of how signaling events, particularly those involving Rap1 small guanosine triphosphate (GTP)hydrolases, can regulate the talin-integrin interaction and resulting activation. Third, we review recent findings that highlight the role of the Rap1-GTP-interacting adapter molecule (RIAM), encoded by the APBB1IP gene, in leukocyte integrin activation and consequently in leukocyte trafficking.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Movimento Celular/fisiologia , Integrinas/metabolismo , Leucócitos/metabolismo , Proteínas de Membrana/metabolismo , Talina/metabolismo , Proteínas de Ligação a Telômeros/metabolismo , Humanos , Leucócitos/citologia , Domínios Proteicos , Complexo Shelterina
9.
Blood ; 119(20): 4698-707, 2012 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-22394598

RESUMO

Anaplastic large-cell lymphomas (ALCLs) bearing the t(2;5) translocation (ALK(+)ALCLs) are frequently characterized by skin colonization and associated with a poor prognosis. Using conditional transgenic models of anaplastic lymphoma kinase-positive (ALK(+)) lymphomas and human ALK(+)ALCL cell lines, in the present study, we show that high-mobility-group box-1 (HMGB-1), a proinflammatory cytokine, is released by ALK(+) cells, and demonstrate extracellular HMGB-1-stimulated secretion of the IL-8 chemokine by HaCaT keratinocytes through the involvement of MMP-9, PAR-2, and the NF-κB pathway. Furthermore, we demonstrate that, in vitro, IL-8 is able to induce the invasiveness of ALK(+) cells, which express the IL-8 receptors CXCR1 and CXCR2. In vitro and in vivo, HMGB-1 inhibition achieved by glycyrrhizin treatment led to a drastic reduction in ALK(+) cell invasiveness. The pathophysiological relevance of our observations was confirmed by demonstrating that the HMGB-1 and IL-8 receptors are expressed in ALK(+)ALCL biopsies. We have also shown that IL-8 secretion is correlated with leukemic dissemination of ALK(+) cells in a significant number of patients. The results of the present study demonstrate for the first time a relationship among the pro-inflammatory mediators HMGB-1, MMP-9, PAR-2, and IL-8. We propose that these mediators create a premetastatic niche within the skin, thereby participating in ALK(+) lymphoma epidermotropism.


Assuntos
Proteína HMGB1/fisiologia , Interleucina-8/metabolismo , Queratinócitos/metabolismo , Linfoma Anaplásico de Células Grandes/patologia , NF-kappa B/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Pele/metabolismo , Quinase do Linfoma Anaplásico , Animais , Células Cultivadas , Feminino , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Humanos , Queratinócitos/patologia , Infiltração Leucêmica/genética , Infiltração Leucêmica/metabolismo , Infiltração Leucêmica/patologia , Linfoma Anaplásico de Células Grandes/genética , Linfoma Anaplásico de Células Grandes/metabolismo , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Camundongos Transgênicos , NF-kappa B/genética , Receptores Proteína Tirosina Quinases/genética , Receptor PAR-2/genética , Receptor PAR-2/metabolismo , Receptor PAR-2/fisiologia , Transdução de Sinais/fisiologia , Pele/patologia , Nicho de Células-Tronco/genética , Nicho de Células-Tronco/imunologia
10.
bioRxiv ; 2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38798563

RESUMO

Osteoclasts are multinucleated cells unique in their ability to resorb bone. Osteoclastogenesis involves several steps of actin-driven rearrangements that participate not only in the cell-cell fusion process, but also in the formation of the sealing zone, the adhesive structure determining the resorption area. Despite the importance of these actin cytoskeleton-based processes, their precise mechanisms of regulation are still poorly characterized. Here, we found that moesin, a member of the Ezrin/Radixin/Moesin (ERM) protein family, is activated during osteoclast maturation and plays an instrumental role for both osteoclast fusion and function. In mouse and human osteoclast precursors, moesin is negatively regulated to potentiate their ability to fuse and degrade bone. Accordingly, we demonstrated that moesin depletion decreases membrane-to-cortex attachment and enhances formation of tunneling nanotubes (TNTs), F-actin-containing intercellular bridges that we revealed to trigger osteoclast fusion. In addition, via a ß3-integrin/RhoA/SLK pathway and independently of its role in fusion, moesin regulates the number and organization of sealing zones in mature osteoclast, and thus participates in the control of bone resorption. Supporting these findings, we found that moesin-deficient mice are osteopenic with a reduced density of trabecular bones and increased osteoclast abundance and activity. These findings provide a better understanding of the regulation of osteoclast biology, and open new opportunities to specifically target osteoclast activity in bone disease therapy.

11.
Cell Rep ; 42(8): 112996, 2023 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-37598341

RESUMO

Canonical interleukin-2 (IL-2) signaling via the high-affinity CD25-containing IL-2 receptor-Janus kinase (JAK)1,3-signal transducer and activator of transcription 5 (STAT5) pathway is essential for development and maintenance of CD4+CD25HiFoxp3+ regulatory T cells (Tregs) that support immune homeostasis. Here, we report that IL-2 signaling via an alternative CD25-chemokine receptor pathway promotes the suppressive function of Tregs. Using an antibody against CD25 that biases IL-2 signaling toward this alternative pathway, we establish that this pathway increases the suppressive activity of Tregs and ameliorates murine experimental autoimmune encephalomyelitis (EAE). Furthermore, heparan sulfate, an IL-2-binding element of cell surfaces and extracellular matrix, or an engineered IL-2 immunocytokine can also direct IL-2 signaling toward this alternative pathway. Overall, these data reveal a non-canonical mechanism for IL-2 signaling that promotes suppressive functions of Tregs, further elucidates how IL-2 supports immune homeostasis, and suggests approaches to promote or suppress Treg functions.


Assuntos
Encefalomielite Autoimune Experimental , Linfócitos T Reguladores , Camundongos , Animais , Interleucina-2/metabolismo , Receptores de Quimiocinas/metabolismo , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Receptores de Interleucina-2/metabolismo , Transdução de Sinais , Fatores de Transcrição Forkhead/metabolismo
12.
J Biol Chem ; 286(37): 32105-14, 2011 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-21737449

RESUMO

NPM-ALK is a chimeric tyrosine kinase detected in most anaplastic large cell lymphomas that results from the reciprocal translocation t(2,5)(p23;q35) that fuses the N-terminal domain of nucleophosmin (NPM) to the catalytic domain of the anaplastic lymphoma kinase (ALK) receptor. The constitutive activity of the kinase is responsible for its oncogenicity through the stimulation of several downstream signaling pathways, leading to cell proliferation, migration, and survival. We demonstrated previously that the high level of phosphatidylinositol 5-phosphate measured in NPM-ALK-expressing cells is controlled by the phosphoinositide kinase PIKfyve, a lipid kinase known for its role in vesicular trafficking. Here, we show that PIKfyve associates with NPM-ALK and that the interaction involves the 181-300 region of the oncogene. Moreover, we demonstrate that the tyrosine kinase activity of the oncogene controls PIKfyve lipid kinase activity but is dispensable for the formation of the complex. Silencing or inhibition of PIKfyve using siRNA or the PIKfyve inhibitor YM201636 have no effect on NPM-ALK-mediated proliferation and migration but strongly reduce invasive capacities of NPM-ALK-expressing cells and their capacity to degrade the extracellular matrix. Accordingly, immunofluorescence studies confirm a perturbation of matrix metalloproteinase 9 localization at the cell surface and defect in maturation. Altogether, these results suggest a role for PIKfyve in NPM-ALK-mediated invasion.


Assuntos
Proliferação de Células , Proteínas de Fusão Oncogênica/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Animais , Linhagem Celular Tumoral , Movimento Celular , Inativação Gênica , Humanos , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Células NIH 3T3 , Invasividade Neoplásica , Proteínas de Fusão Oncogênica/genética , Fosfatidilinositol 3-Quinases/genética , Proteínas Tirosina Quinases/genética
13.
Front Cell Dev Biol ; 10: 908622, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35721481

RESUMO

Integrins regulate the adhesion and migration of blood cells to ensure the proper positioning of these cells in the environment. Integrins detect physical and chemical stimuli in the extracellular matrix and regulate signaling pathways in blood cells that mediate their functions. Integrins are usually in a resting state in blood cells until agonist stimulation results in a high-affinity conformation ("integrin activation"), which is central to integrins' contribution to blood cells' trafficking and functions. In this review, we summarize the mechanisms of integrin activation in blood cells with a focus on recent advances understanding of mechanisms whereby Rap1 regulates talin1-integrin interaction to trigger integrin activation in lymphocytes, platelets, and neutrophils.

14.
J Exp Med ; 219(8)2022 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-35766979

RESUMO

Rap1 GTPase drives assembly of the Mig-10/RIAM/Lamellipodin (MRL protein)-integrin-talin (MIT) complex that enables integrin-dependent lymphocyte functions. Here we used tandem affinity tag-based proteomics to isolate and analyze the MIT complex and reveal that Phostensin (Ptsn), a regulatory subunit of protein phosphatase 1, is a component of the complex. Ptsn mediates dephosphorylation of Rap1, thereby preserving the activity and membrane localization of Rap1 to stabilize the MIT complex. CRISPR/Cas9-induced deletion of PPP1R18, which encodes Ptsn, markedly suppresses integrin activation in Jurkat human T cells. We generated apparently healthy Ppp1r18-/- mice that manifest lymphocytosis and reduced population of peripheral lymphoid tissues ascribable, in part, to defective activation of integrins αLß2 and α4ß7. Ppp1r18-/- T cells exhibit reduced capacity to induce colitis in a murine adoptive transfer model. Thus, Ptsn enables lymphocyte integrin-mediated functions by dephosphorylating Rap1 to stabilize the MIT complex. As a consequence, loss of Ptsn ameliorates T cell-mediated colitis.


Assuntos
Integrinas , Tecido Linfoide , Proteína Fosfatase 1 , Linfócitos T , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Adesão Celular/fisiologia , Colite/imunologia , Colite/metabolismo , Integrinas/imunologia , Integrinas/metabolismo , Tecido Linfoide/imunologia , Tecido Linfoide/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Proteína Fosfatase 1/imunologia , Proteína Fosfatase 1/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Talina/metabolismo , Proteínas rap1 de Ligação ao GTP/imunologia , Proteínas rap1 de Ligação ao GTP/metabolismo
15.
Structure ; 29(4): 305-307, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33798425

RESUMO

In this issue of Structure, Cho et al. (2020) identified an intermolecular interaction between two RIAM pleckstrin homology (PH) domains that masks the phosphoinositide-binding site, and that phosphorylation by Src unmasks the PH domain. This provides an explanation of how RIAM plasma membrane translocation is regulated to promote integrin activation.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Domínios de Homologia à Plecstrina , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Integrinas/metabolismo , Proteínas de Membrana/metabolismo , Fosforilação
16.
J Exp Med ; 218(2)2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33104169

RESUMO

Integrin activation mediates lymphocyte trafficking and immune functions. Conventional T cell (Tconv cell) integrin activation requires Rap1-interacting adaptor molecule (RIAM). Here, we report that Apbb1ip-/- (RIAM-null) mice are protected from spontaneous colitis due to IL-10 deficiency, a model of inflammatory bowel disease (IBD). Protection is ascribable to reduced accumulation and homing of Tconv cells in gut-associated lymphoid tissue (GALT). Surprisingly, there are abundant RIAM-null regulatory T cells (T reg cells) in the GALT. RIAM-null T reg cells exhibit normal homing to GALT and lymph nodes due to preserved activation of integrins αLß2, α4ß1, and α4ß7. Similar to Tconv cells, T reg cell integrin activation and immune function require Rap1; however, lamellipodin (Raph1), a RIAM paralogue, compensates for RIAM deficiency. Thus, in contrast to Tconv cells, RIAM is dispensable for T reg cell integrin activation and suppressive function. In consequence, inhibition of RIAM can inhibit spontaneous Tconv cell-mediated autoimmune colitis while preserving T reg cell trafficking and function.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/imunologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Integrinas/imunologia , Integrinas/metabolismo , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Animais , Doenças Autoimunes/imunologia , Doenças Autoimunes/metabolismo , Colite/metabolismo , Linfonodos/imunologia , Linfonodos/metabolismo , Ativação Linfocitária/imunologia , Tecido Linfoide/imunologia , Tecido Linfoide/metabolismo , Camundongos , Camundongos Endogâmicos C57BL
17.
J Clin Invest ; 131(13)2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-34043589

RESUMO

Cerebral cavernous malformations (CCMs) are common neurovascular lesions caused by loss-of-function mutations in 1 of 3 genes, including KRIT1 (CCM1), CCM2, and PDCD10 (CCM3), and generally regarded as an endothelial cell-autonomous disease. Here we reported that proliferative astrocytes played a critical role in CCM pathogenesis by serving as a major source of VEGF during CCM lesion formation. An increase in astrocyte VEGF synthesis is driven by endothelial nitric oxide (NO) generated as a consequence of KLF2- and KLF4-dependent elevation of eNOS in CCM endothelium. The increased brain endothelial production of NO stabilized HIF-1α in astrocytes, resulting in increased VEGF production and expression of a "hypoxic" program under normoxic conditions. We showed that the upregulation of cyclooxygenase-2 (COX-2), a direct HIF-1α target gene and a known component of the hypoxic program, contributed to the development of CCM lesions because the administration of a COX-2 inhibitor significantly prevented the progression of CCM lesions. Thus, non-cell-autonomous crosstalk between CCM endothelium and astrocytes propels vascular lesion development, and components of the hypoxic program represent potential therapeutic targets for CCMs.


Assuntos
Astrócitos/fisiologia , Hemangioma Cavernoso do Sistema Nervoso Central/fisiopatologia , Animais , Proteínas Reguladoras de Apoptose/deficiência , Proteínas Reguladoras de Apoptose/genética , Astrócitos/patologia , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Células Endoteliais/metabolismo , Hemangioma Cavernoso do Sistema Nervoso Central/etiologia , Hemangioma Cavernoso do Sistema Nervoso Central/patologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Camundongos Knockout , Modelos Neurológicos , Mutação , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fator A de Crescimento do Endotélio Vascular/biossíntese
18.
Biochem Biophys Res Commun ; 387(1): 127-31, 2009 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-19576174

RESUMO

Phosphatidylinositol 5-phosphate (PtdIns5P), the most recently discovered phosphoinositide, has been proposed to play a role as a lipid mediator of intracellular signaling. We have previously shown that PtdIns5P generated by IpgD, an effector of the causative agent of dysentery Shigella flexneri, activates the PI 3-kinase/Akt pathway. Here, we demonstrate that PtdIns5P is able to protect Akt from dephosphorylation. This effect is not due to inhibition of the phosphoinositide phosphatase regulating PtdIns(3,4,5)P(3) levels PTEN but rather to PtdIns5P-induced phosphorylation and subsequent inhibition of the catalytic subunit of PP2A phosphatases. These data shed light on a new mechanism used by S. flexneri bacteria to sustain Akt activation to increase survival of the host cells during bacterial replication.


Assuntos
PTEN Fosfo-Hidrolase/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Proteína Fosfatase 2/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Células HeLa , Humanos , PTEN Fosfo-Hidrolase/antagonistas & inibidores , Fosfatos de Fosfatidilinositol/farmacologia , Fosforilação , Proteína Fosfatase 2/antagonistas & inibidores , Shigella flexneri/metabolismo
19.
J Cell Biol ; 218(6): 1799-1809, 2019 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-30988001

RESUMO

Rap1 GTPases bind effectors, such as RIAM, to enable talin1 to induce integrin activation. In addition, Rap1 binds directly to the talin1 F0 domain (F0); however, this interaction makes a limited contribution to integrin activation in CHO cells or platelets. Here, we show that talin1 F1 domain (F1) contains a previously undetected Rap1-binding site of similar affinity to that in F0. A structure-guided point mutant (R118E) in F1, which blocks Rap1 binding, abolishes the capacity of Rap1 to potentiate talin1-induced integrin activation. The capacity of F1 to mediate Rap1-dependent integrin activation depends on a unique loop in F1 that has a propensity to form a helix upon binding to membrane lipids. Basic membrane-facing residues of this helix are critical, as charge-reversal mutations led to dramatic suppression of talin1-dependent activation. Thus, a novel Rap1-binding site and a transient lipid-dependent helix in F1 work in tandem to enable a direct Rap1-talin1 interaction to cause integrin activation.


Assuntos
Integrinas/metabolismo , Lipídeos/fisiologia , Talina/metabolismo , Proteínas de Ligação a Telômeros/metabolismo , Animais , Sítios de Ligação , Células CHO , Cricetinae , Cricetulus , Humanos , Mutação , Ligação Proteica , Conformação Proteica , Domínios Proteicos , Complexo Shelterina , Talina/química , Talina/genética , Proteínas de Ligação a Telômeros/genética
20.
J Cell Biol ; 217(4): 1453-1465, 2018 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-29535192

RESUMO

Integrin activation regulates adhesion, extracellular matrix assembly, and cell migration, thereby playing an indispensable role in development and in many pathological processes. A proline mutation in the central integrin ß3 transmembrane domain (TMD) creates a flexible kink that uncouples the topology of the inner half of the TMD from the outer half. In this study, using leukocyte integrin α4ß7, which enables development of gut-associated lymphoid tissue (GALT), we examined the biological effect of such a proline mutation and report that it impairs agonist-induced talin-mediated activation of integrin α4ß7, thereby inhibiting rolling lymphocyte arrest, a key step in transmigration. Furthermore, the α4ß7(L721P) mutation blocks lymphocyte homing to and development of the GALT. These studies show that impairing the ability of an integrin ß TMD to transmit talin-induced TMD topology inhibits agonist-induced physiological integrin activation and biological function in development.


Assuntos
Trato Gastrointestinal/metabolismo , Cadeias beta de Integrinas/metabolismo , Integrinas/metabolismo , Linfócitos/metabolismo , Tecido Linfoide/metabolismo , Animais , Adesão Celular , Feminino , Trato Gastrointestinal/citologia , Trato Gastrointestinal/imunologia , Células HEK293 , Humanos , Cadeias beta de Integrinas/química , Cadeias beta de Integrinas/genética , Integrinas/química , Integrinas/genética , Células Jurkat , Migração e Rolagem de Leucócitos , Ativação Linfocitária , Linfócitos/imunologia , Tecido Linfoide/citologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Transdução de Sinais , Relação Estrutura-Atividade , Talina/genética , Talina/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa