Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Cell Rep ; 42(6): 112667, 2023 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-37330909

RESUMO

Dynamic regulation of integrin activation and inactivation is critical for precisely controlled cell adhesion and migration in physiological and pathological processes. The molecular basis for integrin activation has been intensively studied; however, the understanding of integrin inactivation is still limited. Here, we identify LRP12 as an endogenous transmembrane inhibitor for α4 integrin activation. The LRP12 cytoplasmic domain directly binds to the integrin α4 cytoplasmic tail and inhibits talin binding to the ß subunit, thus keeping integrin inactive. In migrating cells, LRP12-α4 interaction induces nascent adhesion (NA) turnover at the leading-edge protrusion. Knockdown of LRP12 leads to increased NAs and enhanced cell migration. Consistently, LRP12-deficient T cells show an enhanced homing capability in mice and lead to aggravated chronic colitis in a T cell-transfer colitis model. Altogether, LRP12 is a transmembrane inactivator for integrins that inhibits α4 integrin activation and controls cell migration by maintaining balanced NA dynamics.


Assuntos
Integrina alfa4 , Integrinas , Proteínas Relacionadas a Receptor de LDL , Animais , Cricetinae , Camundongos , Adesão Celular/fisiologia , Movimento Celular/fisiologia , Células CHO , Integrina alfa4/metabolismo , Integrinas/metabolismo , Ligação Proteica , Humanos , Proteínas Relacionadas a Receptor de LDL/metabolismo
3.
Oncogene ; 41(34): 4091-4103, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35854065

RESUMO

Tumor metastasis is the leading cause of cancer-associated mortality. Unfortunately, the underlying mechanism of metastasis is poorly understood. Expression of legumain (LGMN), an endo-lysosomal cysteine protease, positively correlates with breast cancer metastatic progression and poor prognosis. Here, we report that LGMN is secreted in the zymogen form by motile breast cancer cells. Through binding to cell surface integrin αvß3 via an RGD motif, the autocrine pro-LGMN activates FAK-Src-RhoA signaling in cancer cells and promotes cancer cell migration and invasion independent of LGMN protease activity. Either silencing LGMN expression or mutationally abolishing pro-LGMN‒αvß3 interaction significantly inhibits cancer cell migration and invasion in vitro and breast cancer metastasis in vivo. Finally, we developed a monoclonal antibody against LGMN RGD motif, which blocks pro-LGMN‒αvß3 binding, and effectively suppresses cancer cell migration and invasion in vitro and breast cancer metastasis in vivo. Thus, disruption of pro-LGMN‒integrin αvß3 interaction may be a potentially promising strategy for treating breast cancer metastasis.


Assuntos
Neoplasias da Mama , Integrina alfaVbeta3 , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células , Cisteína Endopeptidases , Feminino , Humanos , Integrina alfaVbeta3/genética , Integrina alfaVbeta3/metabolismo , Metástase Neoplásica , Oligopeptídeos
4.
Cell Rep ; 38(12): 110548, 2022 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-35320720

RESUMO

The protein C receptor (Procr) has been implicated as a stem cell surface marker in several tissues. It is unknown whether Procr acts as a functional signaling receptor in stem cells. Here, by conditional knockout in mammary stem cells (MaSCs), we demonstrate that Procr is essential for mammary gland development and homeostasis. Through proteomics profiling, we identify that, upon stimulation by the ligand protein C, Procr interacts with heat shock protein 90 (HSP90AA1) via its short cytoplasmic tail, recruiting Src and IGF1R to the complex at the plasma membrane. We show that Procr acts as a signaling receptor of protein C in regulation of MaSCs through HSP90, Src, and IGF1R in vitro. In vivo, IGF1R deletion in MaSCs displays similar phenotypes to Procr deletion. These findings illustrate the essential role of Procr signaling in MaSC maintenance, shedding light onto the molecular regulation by Procr in tissue stem cells.


Assuntos
Proteína C , Células-Tronco , Animais , Receptor de Proteína C Endotelial/genética , Receptor de Proteína C Endotelial/metabolismo , Glândulas Mamárias Animais/metabolismo , Proteína C/metabolismo , Transdução de Sinais , Células-Tronco/metabolismo
5.
Cell Res ; 31(12): 1291-1307, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34518654

RESUMO

Intestinal stem cell (ISC) differentiation is regulated precisely by a niche in the crypt, where lymphocytes may interact with stem and transient amplifying (TA) cells. However, whether and how lymphocyte-stem/TA cell contact affects ISC differentiation is largely unknown. Here, we uncover a novel role of T cell-stem/TA cell contact in ISC fate decisions. We show that intestinal lymphocyte depletion results in skewed ISC differentiation in mice, which can be rescued by T cell transfer. Mechanistically, integrin αEß7 expressed on T cells binds to E-cadherin on ISCs and TA cells, triggering E-cadherin endocytosis and the consequent Wnt and Notch signaling alterations. Blocking αEß7-E-cadherin adhesion suppresses Wnt signaling and promotes Notch signaling in ISCs and TA cells, leading to defective ISC differentiation. Thus, αEß7+ T cells regulate ISC differentiation at single-cell level through cell-cell contact-mediated αEß7-E-cadherin adhesion signaling, highlighting a critical role of the T cell-stem/TA cell contact in maintaining intestinal homeostasis.


Assuntos
Células-Tronco , Linfócitos T , Animais , Adesão Celular , Diferenciação Celular , Linhagem da Célula , Integrinas , Mucosa Intestinal , Camundongos , Células-Tronco/citologia , Linfócitos T/citologia , Via de Sinalização Wnt
6.
STAR Protoc ; 2(3): 100720, 2021 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-34401786

RESUMO

Fever is a complex physiological response enhancing immune surveillance during infection and inflammation. Fever-range whole-body hyperthermia (WBH) treatment can experimentally mimic the febrile condition in mice. Here, we describe a protocol for the treatment of mice with WBH and normothermia. We describe the isolation of T cells from mouse spleen followed by the evaluation of T-cell adhesion and transmigration. This animal model can be applied to studying the dysfunction of the immune system induced by fever. For complete details on the use and execution of this protocol, please refer to Lin et al. (2019).


Assuntos
Febre/diagnóstico , Hipertermia Induzida/métodos , Animais , Adesão Celular , Modelos Animais de Doenças , Hipertermia/fisiopatologia , Inflamação , Camundongos , Linfócitos T/metabolismo
9.
Cancer Immunol Res ; 9(8): 967-980, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34131019

RESUMO

Immune cell infiltration is important for predicting the clinical outcomes of colorectal cancer. Integrin ß7 (ITGB7), which is expressed on the surface of leukocytes, plays an essential role in the homing of immune cells to gut-associated lymphoid tissue and facilitating the retention of lymphocytes in gut epithelium; however, its role in colorectal cancer pathogenesis is poorly explored. Here, we found that the number of ß7+ cells decreased significantly in tumor tissue compared with adjacent normal tissue. ß7 expression decreased in tumor-derived compared with normal tissue-derived CD8+ T cells. With bulk RNA expression data from public platforms, we demonstrated that higher ITGB7 expression correlated with longer patient survival, higher cytotoxic immune cell infiltration, lower somatic copy-number alterations, decreased mutation frequency of APC and TP53, and better response to immunotherapy. The possible cell-cell interactions mediated by ITGB7 and its ligands MAdCAM-1, VCAM-1, and CDH1 were investigated using public single-cell RNA sequencing data. ITGB7 deficiency led to exaggerated tumorigenesis and progression in both Apcmin /+ spontaneous and MC38 orthotopic models of colorectal cancer, which could be due to a reduced infiltration of activated CD8+ T cells, effector memory CD8+ T cells, IFNγ+ CD8+ T cells, IFNγ+ natural killer cells, CD103+ dendritic cells, and other immune cell subsets that are essential players in antitumor immunity. In conclusion, our data revealed that ITGB7 could inhibit the tumorigenesis and progression of colorectal cancer by maintaining antitumor immunity.


Assuntos
Neoplasias Colorretais/imunologia , Imunoterapia/métodos , Cadeias beta de Integrinas/uso terapêutico , Neoplasias Colorretais/mortalidade , Humanos , Cadeias beta de Integrinas/farmacologia , Análise de Sobrevida
12.
Proc Natl Acad Sci U S A ; 118(16)2021 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-33846258

RESUMO

Macrophages are the key regulator of T-cell responses depending on their activation state. C-C motif chemokine receptor-like 2 (CCRL2), a nonsignaling atypical receptor originally cloned from LPS-activated macrophages, has recently been shown to regulate immune responses under several inflammatory conditions. However, whether CCRL2 influences macrophage function and regulates tumor immunity remains unknown. Here, we found that tumoral CCRL2 expression is a predictive indicator of robust antitumor T-cell responses in human cancers. CCRL2 is selectively expressed in tumor-associated macrophages (TAM) with immunostimulatory phenotype in humans and mice. Conditioned media from tumor cells could induce CCRL2 expression in macrophages primarily via TLR4, which is negated by immunosuppressive factors. Ccrl2-/- mice exhibit accelerated melanoma growth and impaired antitumor immunity characterized by significant reductions in immunostimulatory macrophages and T-cell responses in tumor. Depletion of CD8+ T cells or macrophages eliminates the difference in tumor growth between WT and Ccrl2-/- mice. Moreover, CCRL2 deficiency impairs immunogenic activation of macrophages, resulting in attenuated antitumor T-cell responses and aggravated tumor growth in a coinjection tumor model. Mechanically, CCRL2 interacts with TLR4 on the cell surface to retain membrane TLR4 expression and further enhance its downstream Myd88-NF-κB inflammatory signaling in macrophages. Similarly, Tlr4-/- mice exhibit reduced CCRL2 expression in TAM and accelerated melanoma growth. Collectively, our study reveals a functional role of CCRL2 in activating immunostimulatory macrophages, thereby potentiating antitumor T-cell response and tumor rejection, and suggests CCLR2 as a potential biomarker candidate and therapeutic target for cancer immunotherapy.


Assuntos
Ativação de Macrófagos/imunologia , Neoplasias/imunologia , Receptores CCR/metabolismo , Animais , Linfócitos T CD8-Positivos/imunologia , China , Feminino , Imunização , Ativação de Macrófagos/fisiologia , Masculino , Melanoma/metabolismo , Camundongos , NF-kappa B/metabolismo , Neoplasias/genética , Receptores CCR/genética , Transdução de Sinais , Linfócitos T/metabolismo , Receptor 4 Toll-Like/imunologia , Receptor 4 Toll-Like/metabolismo , Macrófagos Associados a Tumor/imunologia , Macrófagos Associados a Tumor/metabolismo
13.
Sheng Li Xue Bao ; 73(2): 151-159, 2021 Apr 25.
Artigo em Chinês | MEDLINE | ID: mdl-33903877

RESUMO

Integrins are a large family of heterodimeric cell adhesion molecules composed of α and ß subunits. Through interaction with their specific ligands, integrins mediate cell-cell and cell-extracellular matrix interactions. Via outside-in signaling, integrins can recruit cytoplasmic proteins to their intracellular domains and then cluster into supramolecular structures and trigger downstream signaling. Integrin activation is associated with a global conformation rearrangement from bent to extended in ectodomains and the separation of α and ß subunit cytoplasmic domains. During cell migration, integrins regulate the focal adhesion dynamics and transmit forces between the extracellular matrix and the cell cytoskeleton. In tumor microenvironment, integrins on multiple kinds of cells could be activated, which modulates cell migration into tumor and contributes to angiogenesis and tumor metastasis. Here, we review the mechanism of integrin activation, dynamics of focal adhesions during cell migration and tumor metastasis.


Assuntos
Adesões Focais , Integrinas , Adesão Celular , Moléculas de Adesão Celular , Transdução de Sinais
14.
Matrix Biol ; 95: 32-51, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33068727

RESUMO

Disturbed flow leads to increased inflammatory responses of endothelial cells (ECs) prone to atherogenic state. Currently, little is known about the physiological mechanisms protecting vasculature against disturbed flow-activated ECs leading to atherosclerosis. Understanding the protective mediators involved in EC activation could provide novel therapeutic strategies for atherosclerosis. The extracellular matrix microenvironment profoundly regulates cellular homeostasis. A non-EC resident ECM protein, cartilage oligomeric matrix protein (COMP), has diverse protective roles in the cardiovascular system. To determine whether COMP could protect against disturbed flow-activated EC and atherosclerosis, we compared oscillatory shear stress (OSS) induced EC activation coated with various ECM proteins. Purified COMP inhibited EC activation caused by OSS. EC activation was upregulated in the aortic arch where the flow is disturbed in COMP-/- mice as compared with wild-type mice under physiological conditions or pathologically in partially ligated mouse carotid arteries. Mechanistically, co-immunoprecipitation, mammalian two-hybrid and FRET assay results suggest that COMP bound directly to integrin α5 via its C-terminus. We next synthesized a COMP-derived peptidomimetics (CCPep24) mimicking a specific COMP-integrin α5 interaction and found that CCPep24 protected against EC activation and atherogenesis in vivo. This study extends our current understanding of how ECM and flow coordinately fine-tune EC homeostasis and reveals the potential therapeutic effect of COMP or COMP-derived peptidomimetics on blocking aberrant integrin α5 activation, inflammatory EC activation and atherosclerosis pathogenesis.


Assuntos
Aterosclerose/genética , Proteína de Matriz Oligomérica de Cartilagem/genética , Integrina alfa5/genética , Animais , Aterosclerose/patologia , Sistema Cardiovascular/metabolismo , Sistema Cardiovascular/patologia , Microambiente Celular/genética , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Matriz Extracelular/genética , Células Endoteliais da Veia Umbilical Humana , Humanos , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Camundongos , Camundongos Knockout , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Peptidomiméticos/farmacologia , Mapas de Interação de Proteínas/genética
15.
Front Cell Dev Biol ; 8: 603148, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33381505

RESUMO

The homing of lymphocytes from blood to gut-associated lymphoid tissue is regulated by interaction between integrin α4ß7 with mucosal vascular addressin cell adhesion molecule 1 (MAdCAM-1) expressed on the endothelium of high endothelial venules (HEVs). However, the molecular basis of mucin-like domain, a specific structure of MAdCAM-1 regulating integrin α4ß7-mediated cell adhesion remains obscure. In this study, we used heparan sulfate (HS), which is a highly acidic linear polysaccharide with a highly variable structure, to mimic the negative charges of the extracellular microenvironment and detected the adhesive behaviors of integrin α4ß7 expressing 293T cells to immobilized MAdCAM-1 in vitro. The results showed that HS on the surface significantly promoted integrin α4ß7-mediated cell adhesion, decreased the percentage of cells firmly bound and increased the rolling velocities at high wall shear stresses, which was dependent on the mucin-like domain of MAdCAM-1. Moreover, breaking the negative charges of the extracellular microenvironment of CHO-K1 cells expressing MAdCAM-1 with sialidase inhibited cell adhesion and rolling velocity of 293T cells. Mechanistically, electrostatic repulsion between mucin-like domain and negative charges of the extracellular microenvironment led to a more upright conformation of MAdCAM-1, which facilitates integrin α4ß7-mediated cell adhesion. Our findings elucidated the important role of the mucin-like domain in regulating integrin α4ß7-mediated cell adhesion, which could be applied to modulate lymphocyte homing to lymphoid tissues or inflammatory sites.

16.
BMC Biol ; 18(1): 64, 2020 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-32522281

RESUMO

BACKGROUND: ß7 integrins are responsible for the efficient recruitment of lymphocytes from the blood and their retention in gut-associated lymphoid tissues. Integrin α4ß7 binds MAdCAM-1, mediating rolling adhesion of lymphocytes on blood vessel walls when inactive and firm adhesion when activated, thereby controlling two critical steps of lymphocyte homing to the gut. By contrast, integrin αEß7 mediates the adhesion of lymphocytes to gut epithelial cells by interacting with E-cadherin. Integrin ß7 blocking antibodies have shown efficacy in clinical management of inflammatory bowel disease (IBD); however, fully blocking ß7 function leads to the depletion of colonic regulatory T (Treg) cells and exacerbates dextran sulfate sodium (DSS)-induced colitis by evoking aberrant innate immunity, implying its potential adverse effect for IBD management. Thus, a better therapeutic strategy targeting integrin ß7 is required to avoid this adverse effect. RESULTS: Herein, we inhibited integrin α4ß7 activation in vivo by creating mice that carry in their integrin ß7 gene a mutation (F185A) which from structural studies is known to lock α4ß7 in its resting state. Lymphocytes from ß7-F185A knock-in (KI) mice expressed α4ß7 integrins that could not be activated by chemokines and showed significantly impaired homing to the gut. The ß7-F185A mutation did not inhibit αEß7 activation, but led to the depletion of αEß7+ lymphocytes in the spleen and a significantly reduced population of αEß7+ lymphocytes in the gut of KI mice. ß7-F185A KI mice were resistant to T cell transfer-induced chronic colitis, but did not show an increased susceptibility to DSS-induced innate colitis, the adverse effect of fully blocking ß7 function. CONCLUSIONS: Our findings demonstrate that specific inhibition of integrin α4ß7 activation is a potentially better strategy than fully blocking α4ß7 function for IBD treatment.


Assuntos
Imunidade Adaptativa , Colite/genética , Integrinas/genética , Mutação , Animais , Colite/imunologia , Feminino , Integrinas/metabolismo , Masculino , Camundongos , Camundongos Transgênicos
17.
Gut Microbes ; 11(4): 1030-1042, 2020 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-32240032

RESUMO

Alterations of gut microbiota have been implicated in multiple diseases including cancer. However, the gut microbiota spectrum in lung cancer remains largely unknown. Here we profiled the gut microbiota composition in a discovery cohort containing 42 early-stage lung cancer patients and 65 healthy individuals through the 16S ribosomal RNA (rRNA) gene sequencing analysis. We found that lung cancer patients displayed a significant shift of microbiota composition in contrast to the healthy populations. To identify the optimal microbiota signature for noninvasive diagnosis purpose, we took advantage of Support-Vector Machine (SVM) and found that the predictive model with 13 operational taxonomic unit (OTU)-based biomarkers achieved a high accuracy in lung cancer prediction (area under curve, AUC = 97.6%). This signature performed reasonably well in the validation cohort (AUC = 76.4%), which contained 34 lung cancer patients and 40 healthy individuals. To facilitate potential clinical practice, we further constructed a 'patient discrimination index' (PDI), which largely retained the prediction efficiency in both the discovery cohort (AUC = 92.4%) and the validation cohort (AUC = 67.7%). Together, our study uncovered the microbiota spectrum of lung cancer patients and established the specific gut microbial signature for the potential prediction of the early-stage lung cancer.


Assuntos
Bactérias/classificação , Microbioma Gastrointestinal , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/microbiologia , Bactérias/genética , Biomarcadores , Estudos de Coortes , Detecção Precoce de Câncer , Fezes/microbiologia , Feminino , Genes de RNAr , Humanos , Neoplasias Pulmonares/patologia , Masculino , Redes e Vias Metabólicas , Pessoa de Meia-Idade , Estadiamento de Neoplasias , RNA Ribossômico 16S/genética , Máquina de Vetores de Suporte
18.
Int J Hyperthermia ; 36(sup1): 17-21, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31795834

RESUMO

Fever is a complex physiological response to pathogen infection and injury. One of the beneficial effects of febrile temperatures is stimulation of immune cell trafficking to the lymphoid organs and inflamed tissues, thereby enhancing immune surveillance during infection and inflammation. This trafficking process consists of a highly ordered adhesion cascade that includes tethering and rolling of immune cells along the vessel walls, chemokine-induced activation, firm arrest and diapedesis. In this review, we summarize the current findings of how febrile temperatures regulate the immune cell trafficking process. Febrile temperatures play multiple roles in the functional regulation of critical biomolecules involved in each step of the ordered adhesion cascade that includes L-selectin, chemokines, and α4 and ß2 integrins. A better understanding of febrile temperature-induced regulation of immune cell trafficking will shed light on modulating the immunity to fight against infection and inflammation.


Assuntos
Complexo Antígeno-Anticorpo/metabolismo , Quimiocinas/metabolismo , Febre/imunologia , Integrinas/metabolismo , Humanos
19.
Cell Res ; 29(10): 832-845, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31481760

RESUMO

Breast cancer is a heterogeneous disease. In particular, triple-negative breast cancer (TNBC) comprises various molecular subgroups with unclear identities and currently has few targeted treatment options. Our previous study identified protein C receptor (Procr) as a surface marker on mammary stem cells (MaSCs) located in the basal layer of the normal mammary gland. Given the possible connection of TNBC with basal layer stem cells, we conducted comparative analyses of Procr in breast cancers of mouse and human origin. In mouse mammary tumors, we showed that Procr+ cells are enriched for cancer stem cells (CSCs) in Wnt1 basal-like tumors, but not in Brca1 basal-like tumors or PyVT luminal tumors. In human cancers, PROCR was robustly expressed in half of TNBC cases. Experiments with patient-derived xenografts (PDXs) revealed that PROCR marks CSCs in this discrete subgroup (referred to as PROCR+ TNBC). Interfering with the function of PROCR using an inhibitory nanobody reduced the CSC numbers, arrested tumor growth and prevented rapid tumor recurrence. Our data suggest a key role of MaSC in breast tumorigenesis. Moreover, our work indicates that PROCR can be used as a biomarker to stratify TNBC into clinically relevant subgroups and may provide a novel targeted treatment strategy for this clinically important tumor subtype.


Assuntos
Receptor de Proteína C Endotelial/metabolismo , Células-Tronco Neoplásicas/metabolismo , Neoplasias de Mama Triplo Negativas/patologia , Animais , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Receptor de Proteína C Endotelial/antagonistas & inibidores , Receptor de Proteína C Endotelial/genética , Feminino , Humanos , Estimativa de Kaplan-Meier , Camundongos , Camundongos Nus , Camundongos SCID , Mutação , Células-Tronco Neoplásicas/imunologia , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Anticorpos de Domínio Único/imunologia , Anticorpos de Domínio Único/farmacologia , Neoplasias de Mama Triplo Negativas/metabolismo , Neoplasias de Mama Triplo Negativas/mortalidade
20.
Immunity ; 50(1): 137-151.e6, 2019 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-30650373

RESUMO

Fever is an evolutionarily conserved response that confers survival benefits during infection. However, the underlying mechanism remains obscure. Here, we report that fever promoted T lymphocyte trafficking through heat shock protein 90 (Hsp90)-induced α4 integrin activation and signaling in T cells. By inducing selective binding of Hsp90 to α4 integrins, but not ß2 integrins, fever increased α4-integrin-mediated T cell adhesion and transmigration. Mechanistically, Hsp90 bound to the α4 tail and activated α4 integrins via inside-out signaling. Moreover, the N and C termini of one Hsp90 molecule simultaneously bound to two α4 tails, leading to dimerization and clustering of α4 integrins on the cell membrane and subsequent activation of the FAK-RhoA pathway. Abolishment of Hsp90-α4 interaction inhibited fever-induced T cell trafficking to draining lymph nodes and impaired the clearance of bacterial infection. Our findings identify the Hsp90-α4-integrin axis as a thermal sensory pathway that promotes T lymphocyte trafficking and enhances immune surveillance during infection.


Assuntos
Febre/imunologia , Proteínas de Choque Térmico HSP90/metabolismo , Integrina alfa4/metabolismo , Infecções por Salmonella/imunologia , Salmonella typhimurium/imunologia , Linfócitos T/imunologia , Animais , Carga Bacteriana , Adesão Celular , Movimento Celular , Dimerização , Quinase 1 de Adesão Focal/metabolismo , Vigilância Imunológica , Integrina alfa4/genética , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ligação Proteica , Transdução de Sinais , Proteína rhoA de Ligação ao GTP/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...