Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 260
Filtrar
1.
Nat Immunol ; 23(4): 518-531, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35354953

RESUMO

Internal organs heal injuries with new connective tissue, but the cellular and molecular events of this process remain obscure. By tagging extracellular matrix around the mesothelium lining in mouse peritoneum, liver and cecum, here we show that preexisting matrix was transferred across organs into wounds in various injury models. Using proteomics, genetic lineage-tracing and selective injury in juxtaposed organs, we found that the tissue of origin for the transferred matrix likely dictated the scarring or regeneration of the healing tissue. Single-cell RNA sequencing and genetic and chemical screens indicated that the preexisting matrix was transferred by neutrophils dependent on the HSF-integrin AM/B2-kindlin3 cascade. Pharmacologic inhibition of this axis prevented matrix transfer and the formation of peritoneal adhesions. Matrix transfer was thus an early event of wound repair and provides a therapeutic window to dampen scaring across a range of conditions.


Assuntos
Neutrófilos , Peritônio , Animais , Epitélio , Matriz Extracelular , Camundongos , Peritônio/lesões , Cicatrização
2.
Cell ; 179(1): 120-131.e13, 2019 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-31539492

RESUMO

Focal adhesions (FAs) are protein machineries essential for cell adhesion, migration, and differentiation. Talin is an integrin-activating and tension-sensing FA component directly connecting integrins in the plasma membrane with the actomyosin cytoskeleton. To understand how talin function is regulated, we determined a cryoelectron microscopy (cryo-EM) structure of full-length talin1 revealing a two-way mode of autoinhibition. The actin-binding rod domains fold into a 15-nm globular arrangement that is interlocked by the integrin-binding FERM head. In turn, the rod domains R9 and R12 shield access of the FERM domain to integrin and the phospholipid PIP2 at the membrane. This mechanism likely ensures synchronous inhibition of integrin, membrane, and cytoskeleton binding. We also demonstrate that compacted talin1 reversibly unfolds to an ∼60-nm string-like conformation, revealing interaction sites for vinculin and actin. Our data explain how fast switching between active and inactive conformations of talin could regulate FA turnover, a process critical for cell adhesion and signaling.


Assuntos
Adesões Focais/metabolismo , Domínios e Motivos de Interação entre Proteínas , Talina/química , Talina/metabolismo , Actinas/metabolismo , Actomiosina/metabolismo , Sítios de Ligação , Adesão Celular/fisiologia , Microscopia Crioeletrônica , Citoesqueleto/metabolismo , Dimerização , Escherichia coli/metabolismo , Humanos , Integrinas/metabolismo , Modelos Moleculares , Ligação Proteica , Transdução de Sinais/fisiologia , Vinculina/metabolismo
3.
J Cell Sci ; 137(9)2024 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-38587458

RESUMO

Talin (herein referring collectively to talin 1 and 2) couples the actomyosin cytoskeleton to integrins and transmits tension to the extracellular matrix. Talin also interacts with numerous additional proteins capable of modulating the actin-integrin linkage and thus downstream mechanosignaling cascades. Here, we demonstrate that the scaffold protein Caskin2 interacts directly with the R8 domain of talin through its C-terminal LD motif. Caskin2 also associates with the WAVE regulatory complex to promote cell migration in an Abi1-dependent manner. Furthermore, we demonstrate that the Caskin2-Abi1 interaction is regulated by growth factor-induced phosphorylation of Caskin2 on serine 878. In MCF7 and UACC893 cells, which contain an amplification of CASKIN2, Caskin2 localizes in plasma membrane-associated plaques and around focal adhesions in cortical microtubule stabilization complexes. Taken together, our results identify Caskin2 as a novel talin-binding protein that might not only connect integrin-mediated adhesion to actin polymerization but could also play a role in crosstalk between integrins and microtubules.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Movimento Celular , Proteínas do Citoesqueleto , Ligação Proteica , Talina , Humanos , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas do Citoesqueleto/metabolismo , Proteínas do Citoesqueleto/genética , Adesões Focais/metabolismo , Integrinas/metabolismo , Células MCF-7 , Microtúbulos/metabolismo , Fosforilação , Talina/metabolismo
4.
Nat Rev Mol Cell Biol ; 15(4): 273-88, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24651544

RESUMO

The adhesive interactions of cells with their environment through the integrin family of transmembrane receptors have key roles in regulating multiple aspects of cellular physiology, including cell proliferation, viability, differentiation and migration. Consequently, failure to establish functional cell adhesions, and thus the assembly of associated cytoplasmic scaffolding and signalling networks, can have severe pathological effects. The roles of specific constituents of integrin-mediated adhesions, which are collectively known as the 'integrin adhesome', in diverse pathological states are becoming clear. Indeed, the prominence of mutations in specific adhesome molecules in various human diseases is now appreciated, and experimental as well as in silico approaches provide insights into the molecular mechanisms underlying these pathological conditions.


Assuntos
Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Adesão Celular/fisiologia , Modelos Animais de Doenças , Integrinas/metabolismo , Transdução de Sinais , Animais , Junções Célula-Matriz/fisiologia , Humanos
5.
Proc Natl Acad Sci U S A ; 120(31): e2301881120, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37494400

RESUMO

Integrin adhesion complexes are essential membrane-associated cellular compartments for metazoan life. The formation of initial integrin adhesion complexes is a dynamic process involving focal adhesion proteins assembled at the integrin cytoplasmic tails and the inner leaflet of the plasma membrane. The weak multivalent protein interactions within the complex and with the plasma membrane suggest that liquid-liquid phase separation could play a role in the nascent adhesion assembly. Here, we report that solid-supported lipid membranes supplemented with phosphoinositides induce the phase separation of minimal integrin adhesion condensates composed of integrin ß1 tails, kindlin, talin, paxillin, and FAK at physiological ionic strengths and protein concentrations. We show that the presence of phosphoinositides is key to enriching kindlin and talin on the lipid membrane, which is necessary to further induce the phase separation of paxillin and FAK at the membrane. Our data demonstrate that lipid membrane surfaces set the local solvent conditions for steering the membrane-localized phase separation even in a regime where no condensate formation of proteins occurs in bulk solution.


Assuntos
Integrinas , Talina , Animais , Integrinas/metabolismo , Paxilina/metabolismo , Talina/metabolismo , Membrana Celular/metabolismo , Integrina beta1/metabolismo , Fosfatidilinositóis , Adesão Celular/fisiologia
6.
Proc Natl Acad Sci U S A ; 120(26): e2218116120, 2023 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-37339195

RESUMO

Integrin-mediated adhesion is essential for metazoan life. Integrin binding to ligand requires an activation step prior to binding ligand that depends on direct binding of talin and kindlin to the ß-integrin cytoplasmic tail and the transmission of force from the actomyosin via talin to the integrin-ligand bonds. However, the affinity of talin for integrin tails is low. It is therefore still unclear how such low-affinity bonds are reinforced to transmit forces up to 10 to 40 pN. In this study, we use single-molecule force spectroscopy by optical tweezers to investigate the mechanical stability of the talin•integrin bond in the presence and absence of kindlin. While talin and integrin alone form a weak and highly dynamic slip bond, the addition of kindlin-2 induces a force-independent, ideal talin•integrin bond, which relies on the steric proximity of and the intervening amino acid sequences between the talin- and kindlin-binding sites in the ß-integrin tail. Our findings show how kindlin cooperates with talin to enable transmission of high forces required to stabilize cell adhesion.


Assuntos
Integrinas , Talina , Animais , Talina/metabolismo , Ligantes , Proteínas de Membrana/metabolismo , Adesão Celular
7.
Development ; 149(13)2022 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-35616334

RESUMO

The extensive morphological changes of oligodendrocytes during axon ensheathment and myelination involve assembly of the Ilk-Parvin-Pinch (IPP) heterotrimeric complex of proteins to relay essential mechanical and biochemical signals between integrins and the actin cytoskeleton. Binding of Pinch1 and Pinch2 isoforms to Ilk is mutually exclusive and allows the formation of distinct IPP complexes with specific signaling properties. Using tissue-specific conditional gene ablation in mice, we reveal an essential role for Pinch2 during central nervous system myelination. Unlike Pinch1 gene ablation, loss of Pinch2 in oligodendrocytes results in hypermyelination and in the formation of pathological myelin outfoldings in white matter regions. These structural changes concur with inhibition of Rho GTPase RhoA and Cdc42 activities and phenocopy aspects of myelin pathology observed in corresponding mouse mutants. We propose a dual role for Pinch2 in preventing an excess of myelin wraps through RhoA-dependent control of membrane growth and in fostering myelin stability via Cdc42-dependent organization of cytoskeletal septins. Together, these findings indicate that IPP complexes containing Pinch2 act as a crucial cell-autonomous molecular hub ensuring synchronous control of key signaling networks during developmental myelination.


Assuntos
Proteínas Serina-Treonina Quinases , Transdução de Sinais , Animais , Sistema Nervoso Central , Citoesqueleto , Camundongos , Bainha de Mielina , Oligodendroglia , Transdução de Sinais/genética
8.
J Allergy Clin Immunol ; 154(3): 745-753, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38636606

RESUMO

BACKGROUND: IgE-mediated degranulation of mast cells (MCs) provides rapid protection against environmental hazards, including animal venoms. A fraction of tissue-resident MCs intimately associates with blood vessels. These perivascular MCs were reported to extend projections into the vessel lumen and to be the first MCs to acquire intravenously injected IgE, suggesting that IgE loading of MCs depends on their vascular association. OBJECTIVE: We sought to elucidate the molecular basis of the MC-blood vessel interaction and to determine its relevance for IgE-mediated immune responses. METHODS: We selectively inactivated the Itgb1 gene, encoding the ß1 chain of integrin adhesion molecules (ITGB1), in MCs by conditional gene targeting in mice. We analyzed skin MCs for blood vessel association, surface IgE density, and capability to bind circulating antibody specific for MC surface molecules, as well as in vivo responses to antigen administered via different routes. RESULTS: Lack of ITGB1 expression severely compromised MC-blood vessel association. ITGB1-deficient MCs showed normal densities of surface IgE but reduced binding of intravenously injected antibodies. While their capacity to degranulate in response to IgE ligation in vivo was unimpaired, anaphylactic responses to antigen circulating in the vasculature were largely abolished. CONCLUSIONS: ITGB1-mediated association of MCs with blood vessels is key for MC immune surveillance of blood vessel content, but is dispensable for slow steady-state loading of endogenous IgE onto tissue-resident MCs.


Assuntos
Imunoglobulina E , Integrina beta1 , Mastócitos , Animais , Mastócitos/imunologia , Integrina beta1/imunologia , Integrina beta1/metabolismo , Integrina beta1/genética , Imunoglobulina E/imunologia , Camundongos , Vasos Sanguíneos/imunologia , Camundongos Knockout , Anafilaxia/imunologia , Degranulação Celular/imunologia , Camundongos Endogâmicos C57BL , Pele/imunologia , Pele/irrigação sanguínea
9.
EMBO J ; 39(19): e106234, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32865270

RESUMO

Focal adhesion kinase (FAK) is a central mediator of cell adhesion, acting both as a scaffold and as catalytically active kinase. Acebrón et al (2020) use cryo-electron microscopy (cryo-EM) to visualize the dramatic structural changes that occur upon FAK recruitment to the plasma membrane, which releases FAK autoinhibition and induces its oligomerization. Since activity control via autoinhibition and protein clustering are features also utilized by other focal adhesion (FA) proteins, they have moved center stage in the endeavor to understand the complex process of cell adhesion regulation.


Assuntos
Lipídeos , Transdução de Sinais , Adesão Celular , Membrana Celular , Microscopia Crioeletrônica , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Fosforilação
10.
J Cell Sci ; 135(11)2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35532004

RESUMO

The vitronectin receptor integrin αVß5 can reside in two distinct adhesion structures - focal adhesions (FAs) and flat clathrin lattices (FCLs). Here, we investigate the mechanism that regulates the subcellular distribution of ß5 in keratinocytes and show that ß5 has approximately 7- and 5-fold higher affinity for the clathrin adaptors ARH (also known as LDLRAP1) and Numb, respectively, than for the talin 1 (TLN1); all proteins that bind to the membrane-proximal NPxY motif of the ß5 cytoplasmic domain. Using mass spectrometry, we identified ß5 interactors, including the Rho GEFs p115Rho-GEF and GEF-H1 (also known as ARHGEF1 and ARHGEF2, respectively), and the serine protein kinase MARK2, depletion of which diminishes the clustering of ß5 in FCLs. Replacement of two serine residues (S759 and S762) in the ß5 cytoplasmic domain with phospho-mimetic glutamate residues causes a shift in the localization of ß5 from FAs into FCLs without affecting the interactions with MARK2, p115Rho-GEF or GEF-H1. Instead, we demonstrate that changes in the actomyosin-based cellular contractility by ectopic expression of activated Rho or disruption of microtubules regulates ß5 localization. Finally, we present evidence that ß5 in either FAs or FCLs functions to promote adhesion to vitronectin, cell spreading, and proliferation.


Assuntos
Clatrina , Receptores de Vitronectina , Adesão Celular/fisiologia , Proliferação de Células , Clatrina/metabolismo , Adesões Focais/metabolismo , Receptores de Vitronectina/metabolismo , Serina/metabolismo
11.
Eur J Immunol ; 52(8): 1228-1242, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35491946

RESUMO

ICAP-1 regulates ß1-integrin activation and cell adhesion. Here, we used ICAP-1-null mice to study ICAP-1 potential involvement during immune cell development and function. Integrin α4ß1-dependent adhesion was comparable between ICAP-1-null and control thymocytes, but lack of ICAP-1 caused a defective single-positive (SP) CD8+ cell generation, thus, unveiling an ICAP-1 involvement in SP thymocyte development. ICAP-1 bears a nuclear localization signal and we found it displayed a strong nuclear distribution in thymocytes. Interestingly, there was a direct correlation between the lack of ICAP-1 and reduced levels in SP CD8+ thymocytes of Runx3, a transcription factor required for CD8+ thymocyte generation. In the spleen, ICAP-1 was found evenly distributed between cytoplasm and nuclear fractions, and ICAP-1-/- spleen T and B cells displayed upregulation of α4ß1-mediated adhesion, indicating that ICAP-1 negatively controls their attachment. Furthermore, CD3+ - and CD19+ -selected spleen cells from ICAP-1-null mice showed reduced proliferation in response to T- and B-cell stimuli, respectively. Finally, loss of ICAP-1 caused a remarkable decrease in marginal zone B- cell frequencies and a moderate increase in follicular B cells. Together, these data unravel an ICAP-1 involvement in the generation of SP CD8+ thymocytes and in the control of marginal zone B-cell numbers.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Linfócitos B , Linfócitos T CD8-Positivos , Ativação Linfocitária , Timócitos , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Linfócitos B/citologia , Linfócitos T CD8-Positivos/citologia , Diferenciação Celular , Integrina beta1/metabolismo , Camundongos , Camundongos Knockout , Baço/citologia , Timócitos/citologia , Timo/citologia
12.
Development ; 147(22)2020 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-33051257

RESUMO

The notochord drives longitudinal growth of the body axis by convergent extension, a highly conserved developmental process that depends on non-canonical Wnt/planar cell polarity (PCP) signaling. However, the role of cell-matrix interactions mediated by integrins in the development of the notochord is unclear. We developed transgenic Cre mice, in which the ß1 integrin gene (Itgb1) is ablated at E8.0 in the notochord only or in the notochord and tail bud. These Itgb1 conditional mutants display misaligned, malformed vertebral bodies, hemi-vertebrae and truncated tails. From early somite stages, the notochord was interrupted and displaced in these mutants. Convergent extension of the notochord was impaired with defective cell movement. Treatment of E7.25 wild-type embryos with anti-ß1 integrin blocking antibodies, to target node pit cells, disrupted asymmetric localization of VANGL2. Our study implicates pivotal roles of ß1 integrin for the establishment of PCP and convergent extension of the developing notochord, its structural integrity and positioning, thereby ensuring development of the nucleus pulposus and the proper alignment of vertebral bodies and intervertebral discs. Failure of this control may contribute to human congenital spine malformations.


Assuntos
Movimento Celular , Integrina beta1/metabolismo , Disco Intervertebral/embriologia , Notocorda/embriologia , Coluna Vertebral/embriologia , Via de Sinalização Wnt , Animais , Integrina beta1/genética , Disco Intervertebral/citologia , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Notocorda/citologia , Coluna Vertebral/citologia
13.
Cell ; 134(2): 353-64, 2008 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-18662549

RESUMO

Stable isotope labeling by amino acids in cell culture (SILAC) has become a versatile tool for quantitative, mass spectrometry (MS)-based proteomics. Here, we completely label mice with a diet containing either the natural or the (13)C(6)-substituted version of lysine. Mice were labeled over four generations with the heavy diet, and development, growth, and behavior were not affected. MS analysis of incorporation levels allowed for the determination of incorporation rates of proteins from blood cells and organs. The F2 generation was completely labeled in all organs tested. SILAC analysis from various organs lacking expression of beta1 integrin, beta-Parvin, or the integrin tail-binding protein Kindlin-3 confirmed their absence and disclosed a structural defect of the red blood cell membrane skeleton in Kindlin-3-deficient erythrocytes. The SILAC-mouse approach is a versatile tool by which to quantitatively compare proteomes from knockout mice and thereby determine protein functions under complex in vivo conditions.


Assuntos
Proteínas do Citoesqueleto/metabolismo , Eritrócitos/metabolismo , Proteômica/métodos , Actinina/metabolismo , Ração Animal , Animais , Plaquetas/metabolismo , Membrana Celular/química , Proteínas do Citoesqueleto/análise , Eritrócitos/química , Feminino , Integrina beta1/metabolismo , Marcação por Isótopo , Masculino , Espectrometria de Massas , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Miocárdio/metabolismo
14.
Proc Natl Acad Sci U S A ; 117(39): 24326-24335, 2020 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-32929022

RESUMO

Kindlin-3 (K3)-mediated integrin adhesion controls homing and bone marrow (BM) retention of normal hematopoietic cells. However, the role of K3 in leukemic stem cell (LSC) retention and growth in the remodeled tumor-promoting BM is unclear. We report that loss of K3 in a mouse model of chronic myeloid leukemia (CML) triggers the release of LSCs from the BM into the circulation and impairs their retention, proliferation, and survival in secondary organs, which curbs CML development, progression, and metastatic dissemination. We found de novo expression of cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) on CML-LSCs but not normal hematopoietic stem cells and this enabled us to specifically deplete K3 with a CTLA-4-binding RNA aptamer linked to a K3-siRNA (small interfering RNA) in CTLA-4+ LSCs in vivo, which mobilized LSCs in the BM, induced disease remission, and prolonged survival of mice with CML. Thus, disrupting interactions of LSCs with the BM environment is a promising strategy to halt the disease-inducing and relapse potential of LSCs.


Assuntos
Medula Óssea/metabolismo , Proteínas do Citoesqueleto/deficiência , Leucemia Mieloide/metabolismo , Células-Tronco Neoplásicas/metabolismo , Animais , Antígeno CTLA-4/genética , Antígeno CTLA-4/metabolismo , Movimento Celular , Proteínas do Citoesqueleto/genética , Proteínas de Fusão bcr-abl/genética , Proteínas de Fusão bcr-abl/metabolismo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Leucemia Mieloide/genética , Leucemia Mieloide/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células-Tronco Neoplásicas/citologia , Nicho de Células-Tronco , Microambiente Tumoral
15.
Mol Microbiol ; 116(5): 1249-1267, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34519119

RESUMO

Staphylococcus aureus, a Gram-positive pathogen, invades cells mainly in an integrin-dependent manner. As the activity or conformation of several integrin-associated proteins can be regulated by phosphatidylinositol-4,5-bisphosphate (PI-4,5-P2 ), we investigated the roles of PI-4,5-P2 and PI-4,5-P2 -producing enzymes in cellular invasion by S. aureus. PI-4,5-P2 accumulated upon contact of S. aureus with the host cell, and targeting of an active PI-4,5-P2 phosphatase to the plasma membrane reduced bacterial invasion. Knockdown of individual phosphatidylinositol-4-phosphate 5-kinases revealed that phosphatidylinositol-4-phosphate 5-kinase γ (PIP5KIγ) plays an important role in bacterial internalization. Specific ablation of the talin and FAK-binding motif in PIP5KIγ90 reduced bacterial invasion, which could be rescued by reexpression of an active, but not inactive PIP5KIγ90. Furthermore, PIP5KIγ90-deficient cells showed normal basal PI-4,5-P2 levels in the plasma membrane but reduced the accumulation of PI-4,5-P2 and talin at sites of S. aureus attachment and overall lower levels of FAK phosphorylation. These results highlight the importance of local synthesis of PI-4,5-P2 by a focal adhesion-associated lipid kinase for integrin-mediated internalization of S. aureus.


Assuntos
Aderência Bacteriana , Interações Hospedeiro-Patógeno , Integrinas/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/metabolismo , Fenômenos Fisiológicos Bacterianos , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Membrana Celular/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Células HEK293 , Humanos , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Ligação Proteica , Transdução de Sinais
16.
J Cell Sci ; 133(18)2020 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-32843574

RESUMO

Integrin function depends on the continuous internalization of integrins and their subsequent endosomal recycling to the plasma membrane to drive adhesion dynamics, cell migration and invasion. Here we assign a pivotal role for Rabgap1 (GAPCenA) in the recycling of endocytosed active ß1 integrins to the plasma membrane. The phosphotyrosine-binding (PTB) domain of Rabgap1 binds to the membrane-proximal NPxY motif in the cytoplasmic domain of ß1 integrin subunits on endosomes. Silencing Rabgap1 in mouse fibroblasts leads to the intracellular accumulation of active ß1 integrins, alters focal adhesion formation, and decreases cell migration and cancer cell invasion. Functionally, Rabgap1 facilitates active ß1 integrin recycling to the plasma membrane through attenuation of Rab11 activity. Taken together, our results identify Rabgap1 as an important factor for conformation-specific integrin trafficking and define the role of Rabgap1 in ß1-integrin-mediated cell migration in mouse fibroblasts and breast cancer cells.


Assuntos
Endossomos , Integrina beta1 , Animais , Adesão Celular , Membrana Celular , Movimento Celular , Proteínas Ativadoras de GTPase , Integrina beta1/genética , Integrinas , Camundongos , Proteínas Associadas aos Microtúbulos
17.
Development ; 146(2)2019 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-30651296

RESUMO

Organ growth and tissue homeostasis rely on the proliferation and differentiation of progenitor cell populations. In the developing lung, localized Fgf10 expression maintains distal Sox9-expressing epithelial progenitors and promotes basal cell differentiation in the cartilaginous airways. Mesenchymal Fgf10 expression is induced by Wnt signaling but inhibited by Shh signaling, and epithelial Fgf10 signaling activates ß-catenin signaling. The Hippo pathway is a well-conserved signaling cascade that regulates organ size and stem/progenitor cell behavior. Here, we show that Hippo signaling promotes lineage commitment of lung epithelial progenitors by curbing Fgf10 and ß-catenin signaling. Our findings show that both inactivation of the Hippo pathway (nuclear Yap) or ablation of Yap result in increased ß-catenin and Fgf10 signaling, suggesting a cytoplasmic role for Yap in epithelial lineage commitment. We further demonstrate redundant and non-redundant functions for the two nuclear effectors of the Hippo pathway, Yap and Taz, during lung development.


Assuntos
Linhagem da Célula , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Fator 10 de Crescimento de Fibroblastos/metabolismo , Pulmão/citologia , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , beta Catenina/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteínas de Ciclo Celular , Diferenciação Celular , Citoplasma/metabolismo , Feminino , Via de Sinalização Hippo , Pulmão/embriologia , Masculino , Camundongos , Modelos Biológicos , Organogênese , Fenótipo , Fosfoproteínas/metabolismo , Alvéolos Pulmonares/embriologia , Transativadores , Proteínas de Sinalização YAP
18.
Exp Cell Res ; 398(1): 112391, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33253712

RESUMO

Kidney Ankyrin Repeat-containing Proteins (KANKs) comprise a family of four evolutionary conserved proteins (KANK1 to 4) that localize to the belt of mature focal adhesions (FAs) where they regulate integrin-mediated adhesion, actomyosin contractility, and link FAs to the cortical microtubule stabilization complex (CMSC). The human KANK proteins were first identified in kidney and have been associated with kidney cancer and nephrotic syndrome. Here, we report the distributions and subcellular localizations of the four Kank mRNAs and proteins in mouse tissues. We found that the KANK family members display distinct and rarely overlapping expression patterns. Whereas KANK1 is expressed at the basal side of epithelial cells of all tissues tested, KANK2 expression is mainly observed at the plasma membrane and/or cytoplasm of mesenchymal cells and KANK3 exclusively in vascular and lymphatic endothelial cells. KANK4 shows the least widespread expression pattern and when present, overlaps with KANK2 in contractile cells, such as smooth muscle cells and pericytes. Our findings show that KANKs are widely expressed in a cell type-specific manner, which suggests that they have cell- and tissue-specific functions.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas do Citoesqueleto/química , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Células Cultivadas , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Distribuição Tecidual
19.
Nat Mater ; 19(2): 218-226, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31959953

RESUMO

Haematopoietic cells and platelets employ G-protein-coupled receptors (GPCRs) to sense extracellular information and respond by initiating integrin-mediated adhesion. So far, such processes have not been demonstrated in non-haematopoietic cells. Here, we report that the activation of protease-activated receptors PAR1 and PAR2 induce multiple signalling pathways to establish α5ß1-integrin-mediated adhesion. First, PARs signal via Gßγ and PI3K to α5ß1-integrins to adopt a talin- and kindlin-dependent high-affinity conformation, which triggers fibronectin binding and initiates cell adhesion. Then, within 60 s, PARs signal via Gα13, Gαi, ROCK and Src to strengthen the α5ß1-integrin-mediated adhesion. Furthermore, PAR signalling changes the abundance of numerous proteins in the adhesome assembled by α5ß1-integrins, including Gα13, vacuolar protein-sorting-associated protein 36, and band 4.1-like protein 4B or 5, and accelerates cell adhesion maturation, spreading and migration. The mechanistic insights describe how agonist binding to PAR employs GPCR and integrin-signalling pathways to initiate and regulate adhesion and to guide physiological responses of non-haematopoietic cells.


Assuntos
Adesão Celular , Integrina alfa5beta1/metabolismo , Receptor PAR-1/metabolismo , Receptor PAR-2/metabolismo , Transdução de Sinais , Células HEK293 , Humanos , Talina/metabolismo
20.
Nat Immunol ; 10(4): 412-9, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19270713

RESUMO

The proliferation of antigen-specific lymphocytes and resulting clonal expansion are essential for adaptive immunity. We report here that B cell-specific deletion of the heavy chain of CD98 (CD98hc) resulted in lower antibody responses due to total suppression of B cell proliferation and subsequent plasma cell formation. Deletion of CD98hc did not impair early B cell activation but did inhibit later activation of the mitogen-activated protein kinase Erk1/2 and downregulation of the cell cycle inhibitor p27. Reconstitution of CD98hc-deficient B cells with CD98hc mutants showed that the integrin-binding domain of CD98hc was required for B cell proliferation but that the amino acid-transport function of CD98hc was dispensable for this. Thus, CD98hc supports integrin-dependent rapid proliferation of B cells. We propose that the advantage of adaptive immunity favored the appearance of CD98hc in vertebrates.


Assuntos
Formação de Anticorpos , Linfócitos B/imunologia , Proliferação de Células , Cadeia Pesada da Proteína-1 Reguladora de Fusão/imunologia , Animais , Linfócitos B/citologia , Transporte Biológico Ativo , Diferenciação Celular/fisiologia , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Integrinas/metabolismo , Ativação Linfocitária , Camundongos , Camundongos Transgênicos , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Plasmócitos/citologia , Plasmócitos/imunologia , Ligação Proteica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA