Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 152
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancer Immunol Res ; 10(4): 371, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-35362048

RESUMO

Infiltration of lymphocytes into solid tumors represents a significant bottleneck to successful control of tumor growth. The nature of tumor blood vessels is an important factor governing both quantitative and qualitative features of the immune infiltrate. In this issue, Sawada and colleagues identify a genetic signature for blood vessels, most notably high endothelial venules, which are associated with tertiary lymphoid structures and improved clinical outcome. See related article by Sawada et al., p. 468 (4).


Assuntos
Neoplasias , Estruturas Linfoides Terciárias , Humanos , Linfonodos , Linfócitos , Neoplasias/genética , Vênulas/imunologia
2.
J Immunother Cancer ; 9(10)2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34670828

RESUMO

BACKGROUND: High endothelial venule (HEV) is a specialized vasculature for lymphocyte trafficking. While HEVs are frequently observed within gastric cancer (GC), the vascular-immune interaction between HEV and tumor-infiltrating lymphocytes (TILs) has not been well elucidated. In this study, we aimed to unveil the potential value of HEVs as a surrogate marker for T-cell inflamed immune microenvironment in GC using a large number of prospectively collected surgical specimens of GC. METHODS: We included 460 patients with GC who underwent surgical resection. Nanostring PanCancer immune profiling was performed to evaluate the immunological phenotype of GCs. HEV density and three distinct patterns of TILs (Crohn-like lymphoid reaction, peritumoral lymphoid reaction, and intratumoral lymphoid reaction) were analyzed for their relationship and evaluated as prognostic factors for relapse-free survival (RFS) and overall survival (OS). RESULTS: HEV-high GC revealed increased infiltration by immune cell subsets, including dendritic cells, CD8+ cytotoxic T cells, and CD4+ helper T cells. In addition, HEV-high GC demonstrated increased immune-modulating chemokines, type I or II interferon pathway, and immune checkpoints, all of which indicate the inflamed tumor microenvironment (TME). All three distinct patterns of TILs were associated with HEV density. In survival analysis, patients with HEV-high GC displayed significantly longer RFS and OS than those with HEV-low GC (p<0.001 for RFS, p<0.001 for OS). Multivariate analysis demonstrated that HEV was the most significant immunological prognostic factor for RFS (patients with high HEV compared with those with low HEV; HR 0.412, 95% CI 0.241 to 0.705, p=0.001) and OS (HR 0.547, 95% CI 0.329 to 0.909, p=0.02) after adjustment for age, stage, and TIL. CONCLUSION: HEV is the most significant immunological prognosticator for RFS and OS in resected GC, indicating inflamed TME.


Assuntos
Biomarcadores Tumorais/metabolismo , Biomarcadores/metabolismo , Neoplasias Gástricas/genética , Vênulas/imunologia , Adulto , Idoso , Humanos , Pessoa de Meia-Idade , Prognóstico , Microambiente Tumoral , Adulto Jovem
3.
Front Immunol ; 12: 736670, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34484246

RESUMO

High endothelial venules (HEVs) are specialized postcapillary venules composed of cuboidal blood endothelial cells that express high levels of sulfated sialomucins to bind L-Selectin/CD62L on lymphocytes, thereby facilitating their transmigration from the blood into the lymph nodes (LN) and other secondary lymphoid organs (SLO). HEVs have also been identified in human and murine tumors in predominantly CD3+T cell-enriched areas with fewer CD20+B-cell aggregates that are reminiscent of tertiary lymphoid-like structures (TLS). While HEV/TLS areas in human tumors are predominantly associated with increased survival, tumoral HEVs (TU-HEV) in mice have shown to foster lymphocyte-enriched immune centers and boost an immune response combined with different immunotherapies. Here, we discuss the current insight into TU-HEV formation, function, and regulation in tumors and elaborate on the functional implication, opportunities, and challenges of TU-HEV formation for cancer immunotherapy.


Assuntos
Células Endoteliais/imunologia , Linfócitos/imunologia , Neoplasias/irrigação sanguínea , Neoplasias/imunologia , Estruturas Linfoides Terciárias/imunologia , Vênulas/imunologia , Animais , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Humanos , Imunoterapia , Selectina L/metabolismo , Linfócitos/metabolismo , Neoplasias/patologia , Neoplasias/terapia , Sialomucinas/metabolismo , Transdução de Sinais , Estruturas Linfoides Terciárias/metabolismo , Estruturas Linfoides Terciárias/patologia , Migração Transendotelial e Transepitelial , Microambiente Tumoral , Vênulas/metabolismo , Vênulas/patologia
4.
Life Sci Alliance ; 4(8)2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34187874

RESUMO

High endothelial venules (HEVs) effectively recruit circulating lymphocytes from the blood to lymph nodes. HEVs have endothelial cells (ECs) and perivascular sheaths consisting of fibroblastic reticular cells (FRCs). Yet, post-luminal lymphocyte migration steps are not well elucidated. Herein, we performed intravital imaging to investigate post-luminal T- and B-cell migration in popliteal lymph node, consisting of trans-EC migration, crawling in the perivascular channel (a narrow space between ECs and FRCs) and trans-FRC migration. The post-luminal migration of T cells occurred in a PNAd-dependent manner. Remarkably, we found hot spots for the trans-EC and trans-FRC migration of T- and B cells. Interestingly, T- and B cells preferentially shared trans-FRC migration hot spots but not trans-EC migration hot spots. Furthermore, the trans-FRC T-cell migration was confined to fewer sites than trans-EC T-cell migration, and trans-FRC migration of T- and B cells preferentially occurred at FRCs covered by CD11c+ dendritic cells in HEVs. These results suggest that HEV ECs and FRCs with perivascular DCs delicately regulate T- and B-cell entry into peripheral lymph nodes.


Assuntos
Linfócitos B/metabolismo , Linfócitos T/metabolismo , Vênulas/imunologia , Animais , Microscopia Intravital , Linfonodos/imunologia , Camundongos , Migração Transendotelial e Transepitelial
5.
Immunity ; 54(7): 1494-1510.e7, 2021 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-34033752

RESUMO

Aging is associated with dysregulated immune functions. Here, we investigated the impact of age on neutrophil diapedesis. Using confocal intravital microscopy, we found that in aged mice, neutrophils adhered to vascular endothelium in inflamed tissues but exhibited a high frequency of reverse transendothelial migration (rTEM). This retrograde breaching of the endothelium by neutrophils was governed by enhanced production of the chemokine CXCL1 from mast cells that localized at endothelial cell (EC) junctions. Increased EC expression of the atypical chemokine receptor 1 (ACKR1) supported this pro-inflammatory milieu in aged venules. Accumulation of CXCL1 caused desensitization of the chemokine receptor CXCR2 on neutrophils and loss of neutrophil directional motility within EC junctions. Fluorescent tracking revealed that in aged mice, neutrophils undergoing rTEM re-entered the circulation and disseminated to the lungs where they caused vascular leakage. Thus, neutrophils stemming from a local inflammatory site contribute to remote organ damage, with implication to the dysregulated systemic inflammation associated with aging.


Assuntos
Envelhecimento/imunologia , Transporte Biológico/imunologia , Inflamação/imunologia , Neutrófilos/imunologia , Animais , Quimiocina CXCL1/imunologia , Células Endoteliais/imunologia , Endotélio Vascular/imunologia , Feminino , Junções Intercelulares/imunologia , Pulmão/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Interleucina-8B/imunologia , Vênulas/imunologia
6.
Pathol Res Pract ; 220: 153392, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33647862

RESUMO

BACKGROUND: Tumors lymphocytic infiltration has prognostic and predictive value. However, the mechanisms involved in lymphocyte recruitment remain poorly characterized. High endothelial venules (HEV) are blood vessels specialized in lymphocyte recruitment, recently showing prognostic significance in some types of cancer. Their implications in laryngeal or pharyngeal cancer is largely unknown. AIM OF THE STUDY: To investigate the possible presence of HEVs in head and neck cancer. MATERIAL AND METHODS: Oropharyngeal (n = 61), hypopharyngeal (n = 53) and laryngeal (n = 21) squamous cell carcinomas were immunohistochemically studied with the MECA-79 antibody, which specifically recognizes HEVs. Histological and clinical factors were correlated with HEVs' presence. RESULTS: HEVs were present in 34% of tumors, showing significant correlations with oropharyngeal localization, higher lymphocytic response, lower tumor budding, lower T status, absence of distant metastases and better overall and progression-free survival. CONCLUSION: HEVs represent an important prognostic factor in head and neck cancer.


Assuntos
Células Endoteliais/patologia , Neoplasias Laríngeas/patologia , Neoplasias Faríngeas/patologia , Carcinoma de Células Escamosas de Cabeça e Pescoço/secundário , Vênulas/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Células Endoteliais/imunologia , Feminino , Humanos , Neoplasias Laríngeas/imunologia , Neoplasias Laríngeas/mortalidade , Neoplasias Laríngeas/terapia , Linfócitos do Interstício Tumoral/imunologia , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Neoplasias Faríngeas/imunologia , Neoplasias Faríngeas/mortalidade , Neoplasias Faríngeas/terapia , Intervalo Livre de Progressão , Estudos Retrospectivos , Carcinoma de Células Escamosas de Cabeça e Pescoço/imunologia , Carcinoma de Células Escamosas de Cabeça e Pescoço/mortalidade , Carcinoma de Células Escamosas de Cabeça e Pescoço/terapia , Linfócitos T/imunologia , Microambiente Tumoral , Vênulas/imunologia
7.
Am J Pathol ; 191(2): 396-414, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33159887

RESUMO

Recruitment of naive T cells to lymph nodes is essential for the development of adaptive immunity. Upon pathogen infection, lymph nodes promptly increase the influx of naive T cells from the circulation in order to screen and prime the T cells. The precise contribution of the lymph node vasculature to the regulation of this process remains unclear. Here we show a role for the Ras GTPase, R-Ras, in the functional adaptation of high endothelial venules to increase naive T cell trafficking to the lymph nodes. R-Ras is transiently up-regulated in the endothelium of high endothelial venules by the inflammatory cytokine tumor necrosis factor (TNF) within 24 hours of pathogen inoculation. TNF induces R-Ras upregulation in endothelial cells via JNK and p38 mitogen-activated protein kinase but not NF-κB. Studies of T cell trafficking found that the loss of function of endothelial R-Ras impairs the rapid acceleration of naive T cell recruitment to the lymph nodes upon inflammation. This defect diminished the ability of naive OT-1 T cells to develop antitumor activity against ovalbumin-expressing melanoma. Proteomic analyses suggest that endothelial R-Ras facilitates TNF-dependent transendothelial migration (diapedesis) of naive T cells by modulating molecular assembly the at T cell-endothelial cell interface. These findings give new mechanistic insights into the functional adaptation of high endothelial venules to accelerate naive T cell recruitment to the lymph nodes.


Assuntos
Quimiotaxia de Leucócito/fisiologia , Linfócitos T/imunologia , Migração Transendotelial e Transepitelial/fisiologia , Fator de Necrose Tumoral alfa/metabolismo , Proteínas ras/metabolismo , Animais , Células Endoteliais/metabolismo , Humanos , Linfonodos/irrigação sanguínea , Linfonodos/imunologia , Linfonodos/metabolismo , Camundongos , Linfócitos T/metabolismo , Regulação para Cima , Vênulas/imunologia , Vênulas/metabolismo
8.
Front Immunol ; 11: 1028, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32536926

RESUMO

B cell adaptor molecule of 32 kDa (Bam32), known as dual adapter for phosphotyrosine and 3-phosphoinositides 1 (DAPP1), has been implicated in regulating lymphocyte proliferation and recruitment during inflammation. However, its role in neutrophils during inflammation remains unknown. Using intravital microscopy, we examined the role of Bam32 in formyl peptide receptor agonist WKYMVm-induced permeability changes in post-capillary venules and assessed simultaneously neutrophil adhesion and emigration in cremaster muscles of Bam32-deficient (Bam32-/-) and wild-type (WT) control mice. We observed significantly reduced WKYMVm-induced microvascular hyperpermeability accompanied by markedly decreased neutrophil emigration in Bam32-/- mice. The Bam32-specific decrease in WKYMVm-induced hyperpermeability was neutrophil-dependent as this was verified in bone marrow transplanted chimeric mice. We discovered that Bam32 was critically required for WKYMVm-induced intracellular and extracellular production of reactive oxygen species (ROS) in neutrophils. Pharmacological scavenging of ROS eliminated the differences in WKYMVm-induced hyperpermeability between Bam32-/- and WT mice. Deficiency of Bam32 decreased WKYMVm-induced ERK1/2 but not p38 or JNK phosphorylation in neutrophils. Inhibition of ERK1/2 signaling cascade suppressed WKYMVm-induced ROS generation in WT neutrophils and microvascular hyperpermeability in WT mice. In conclusion, our study reveals that Bam32-dependent, ERK1/2-involving ROS generation in neutrophils is critical in WKYMVm-induced microvascular hyperpermeability during neutrophil recruitment.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Permeabilidade Capilar/efeitos dos fármacos , Permeabilidade Capilar/fisiologia , Lipoproteínas/metabolismo , Neutrófilos/metabolismo , Oligopeptídeos/farmacologia , Proteínas Adaptadoras de Transdução de Sinal/deficiência , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Transplante de Medula Óssea , Permeabilidade Capilar/imunologia , Adesão Celular/efeitos dos fármacos , Adesão Celular/imunologia , Adesão Celular/fisiologia , Lipoproteínas/deficiência , Lipoproteínas/genética , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infiltração de Neutrófilos/efeitos dos fármacos , Infiltração de Neutrófilos/imunologia , Infiltração de Neutrófilos/fisiologia , Neutrófilos/efeitos dos fármacos , Neutrófilos/imunologia , Espécies Reativas de Oxigênio/metabolismo , Receptores de Formil Peptídeo/agonistas , Quimeras de Transplante/imunologia , Quimeras de Transplante/fisiologia , Vênulas/efeitos dos fármacos , Vênulas/imunologia , Vênulas/fisiologia
9.
J Immunol ; 204(9): 2552-2561, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32205425

RESUMO

The adaptive immune function of lymph nodes is dependent on constant recirculation of lymphocytes. In this article, we identify neutrophils present in the lymph node at steady state, exhibiting the same capacity for recirculation. In germ-free mice, neutrophils still recirculate through lymph nodes, and in mice cohoused with wild microbiome mice, the level of neutrophils in lymph nodes increases significantly. We found that at steady state, neutrophils enter the lymph node entirely via L-selectin and actively exit via efferent lymphatics via an S1P dependent mechanism. The small population of neutrophils in the lymph node can act as reconnaissance cells to recruit additional neutrophils in the event of bacterial dissemination to the lymph node. Without these reconnaissance cells, there is a delay in neutrophil recruitment to the lymph node and a reduction in swarm formation following Staphylococcus aureus infection. This ability to recruit additional neutrophils by lymph node neutrophils is initiated by LTB4. This study establishes the capacity of neutrophils to recirculate, much like lymphocytes via L-selectin and high endothelial venules in lymph nodes and demonstrates how the presence of neutrophils at steady state fortifies the lymph node in case of an infection disseminating through lymphatics.


Assuntos
Linfonodos/imunologia , Infiltração de Neutrófilos/imunologia , Neutrófilos/imunologia , Infecções Estafilocócicas/imunologia , Animais , Endotélio/imunologia , Endotélio/microbiologia , Feminino , Selectina L/imunologia , Linfonodos/microbiologia , Vasos Linfáticos/imunologia , Vasos Linfáticos/microbiologia , Linfócitos/imunologia , Linfócitos/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microbiota/imunologia , Receptores de Esfingosina-1-Fosfato/imunologia , Infecções Estafilocócicas/microbiologia , Vênulas/imunologia , Vênulas/microbiologia
10.
Am J Pathol ; 188(7): 1653-1665, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29929915

RESUMO

The innate immune system is the primary defense against cryptococcal infection, but paradoxically it promotes infection of the central nervous system. We performed a detailed longitudinal study of neurocryptococcosis in normal, chimeric, green fluorescent protein phagocyte-positive mice and phagocyte-depleted mice and interrogated the central nervous system innate immune response to Cryptococcus neoformans H99 using confocal microscopy, histology, flow cytometry, and quantification of brain cytokine/chemokines and fungal burdens. C. neoformans was present in the perivascular space (PVS) of post-capillary venules. This was associated with a massive influx of blood-derived monocytes, neutrophils, and T lymphocytes into the PVS and a predominantly proinflammatory cytokine/chemokine response. Phagocytes containing cryptococci were present only in the lumen and corresponding PVS of post-capillary venules. Free cryptococci were observed breaching the glia limitans, the protective barrier between the PVS and the cerebral parenchyma. Parenchymal cryptococcomas were typically in direct contact with post-capillary venules and lacked surrounding immune cell infiltrates. Phagocyte depletion abrogated cryptococcoma formation and PVS infiltrates. Together, these observations suggest that cryptococcomas can originate via phagocyte-dependent transport across post-capillary venular endothelium into the PVS and thence via passage of free cryptococci into the brain. In conclusion, we demonstrate for the first time that the PVS of cortical post-capillary venules is the major site of the early innate immune response to, and phagocyte-dependent entry of, C. neoformans.


Assuntos
Encéfalo/imunologia , Cryptococcus neoformans/imunologia , Imunidade Inata/imunologia , Meningite Criptocócica/imunologia , Fagócitos/imunologia , Linfócitos T/imunologia , Vênulas/imunologia , Animais , Encéfalo/microbiologia , Encéfalo/patologia , Modelos Animais de Doenças , Feminino , Meningite Criptocócica/microbiologia , Meningite Criptocócica/patologia , Camundongos , Camundongos Endogâmicos C57BL , Monócitos , Fagócitos/microbiologia , Fagócitos/patologia , Linfócitos T/microbiologia , Linfócitos T/patologia , Vênulas/microbiologia , Vênulas/patologia
11.
Mod Pathol ; 31(6): 910-922, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29416107

RESUMO

Oral squamous cell carcinomas are associated with a poor prognosis, which may be partly due to functional impairment of the immune response. Lymphocyte recruitment to the tumor site is facilitated by high-endothelial venules, whereas expression of programmed-death ligand 1 (PD-L1) can impair T-cell function. Thus, we hypothesize that these factors are important in shaping the immune response in oral squamous cell carcinoma. In the present study, we characterized the immune infiltrate in formalin-fixed, paraffin-embedded tumor samples from 75 oral squamous cell carcinoma patients. We used immunohistochemistry to determine the distribution of immune cell subsets, high-endothelial venules, and PD-L1, as well as quantitative real-time polymerase chain reaction to assess the expression of inflammatory cytokines and chemokines associated with lymphocyte trafficking. Finally, we calculated correlations between the presence of immune cell subsets, the gene expression patterns, high-endothelial venules, PD-L1, and the clinicopathological parameters, including patient survival. The presence of high-endothelial venules correlated with increased number of CD3+ T cells and CD20+ B cells, higher levels of the chemokines CXCL12 and CCL21, and lower levels of CCL20, irrespective of the tumors' T stage. In univariate analysis, high levels of CD20+ B cells and CD68+ macrophages, positive high-endothelial venule status, and low T and N stages predicted longer patient survival. However, only the presence of high-endothelial venules and a low T stage were independent positive prognosticators. This indicates that high-endothelial venules are important mediators and a convenient marker of an antitumor immune response in oral squamous cell carcinoma. Our findings suggest that these vessels are a potential immunomodulatory target in this type of cancer. PD-L1 staining in tumor cells correlated with lower T stage, increased infiltration of CD4+ cells, and higher expression of several inflammation-related cytokines. Thus, oral squamous cell carcinomas rich in CD4+ cells may preferentially respond to PD-1/PD-L1 blockade therapy.


Assuntos
Carcinoma de Células Escamosas/patologia , Neoplasias Bucais/patologia , Microambiente Tumoral/imunologia , Vênulas/patologia , Biomarcadores Tumorais , Carcinoma de Células Escamosas/imunologia , Células Endoteliais/imunologia , Células Endoteliais/patologia , Humanos , Imuno-Histoquímica , Neoplasias Bucais/imunologia , Estadiamento de Neoplasias , Prognóstico , Estudos Retrospectivos , Vênulas/imunologia
12.
PLoS One ; 12(4): e0176110, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28419144

RESUMO

Identification of the structure-function relationship of heparin, particularly between 2-O-, 6-O-, and N-sulfation and its anticoagulant or anti-inflammatory activities, is critical in order to evaluate the biological effects of heparin, especially in conjunction with modifications for oral formulation. In this study, we demonstrated that removal of 2-O, 6-O, or N-desulfation and their hydrophobic modifications have differential effects on the blocking of interactions between sLeX and P-and L-selectins, with highest inhibition by 6-O desulfation, which was consistent with their in vivo therapeutic efficacies on CIA mice. The 6-O desulfation of lower molecular weight heparin (LMWH) retained the ability of LMWH to interfere with T cell adhesion via selectin-sLeX interactions. Furthermore, 6DSHbD coated on the apical surface of inflamed endothelium directly blocked the adhesive interactions of circulating T cells, which was confirmed in vivo by suppressing T cell adhesion at post-capillary venular endothelium. Thus, in series with our previous study demonstrating inhibition of transendothelial migration, oral delivery of low anticoagulant LMWH to venular endothelium of inflamed joint tissues ameliorated arthritis by the stepwise inhibition of T cell recruitment and provides a rationale for the development of modified oral heparins as innovative agents for the treatment of chronic inflammatory arthritis.


Assuntos
Anti-Inflamatórios/química , Anti-Inflamatórios/uso terapêutico , Anticoagulantes/química , Anticoagulantes/uso terapêutico , Artrite/tratamento farmacológico , Heparina/química , Heparina/uso terapêutico , Linfócitos T/efeitos dos fármacos , Animais , Anti-Inflamatórios/farmacologia , Anticoagulantes/farmacologia , Artrite/imunologia , Artrite/patologia , Adesão Celular/efeitos dos fármacos , Heparina/farmacologia , Heparina de Baixo Peso Molecular/química , Heparina de Baixo Peso Molecular/farmacologia , Heparina de Baixo Peso Molecular/uso terapêutico , Humanos , Masculino , Camundongos , Camundongos Endogâmicos DBA , Sulfatos/química , Linfócitos T/imunologia , Linfócitos T/patologia , Migração Transendotelial e Transepitelial/efeitos dos fármacos , Vênulas/efeitos dos fármacos , Vênulas/imunologia , Vênulas/patologia
13.
J Clin Invest ; 126(11): 4125-4139, 2016 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-27701149

RESUMO

Neutrophils need to penetrate the perivascular basement membrane for successful extravasation into inflamed tissue, but this process is incompletely understood. Recent findings have associated mammalian sterile 20-like kinase 1 (MST1) loss of function with a human primary immunodeficiency disorder, suggesting that MST1 may be involved in immune cell migration. Here, we have shown that MST1 is a critical regulator of neutrophil extravasation during inflammation. Mst1-deficient (Mst1-/-) neutrophils were unable to migrate into inflamed murine cremaster muscle venules, instead persisting between the endothelium and the basement membrane. Mst1-/- neutrophils also failed to extravasate from gastric submucosal vessels in a murine model of Helicobacter pylori infection. Mechanistically, we observed defective translocation of VLA-3, VLA-6, and neutrophil elastase from intracellular vesicles to the surface of Mst1-/- neutrophils, indicating that MST1 is required for this crucial step in neutrophil transmigration. Furthermore, we found that MST1 associates with the Rab27 effector protein synaptotagmin-like protein 1 (JFC1, encoded by Sytl1 in mice), but not Munc13-4, thereby regulating the trafficking of Rab27-positive vesicles to the cellular membrane. Together, these findings highlight a role for MST1 in vesicle trafficking and extravasation in neutrophils, providing an additional mechanistic explanation for the severe immune defect observed in patients with MST1 deficiency.


Assuntos
Fator de Crescimento de Hepatócito/imunologia , Neutrófilos/imunologia , Proteínas Proto-Oncogênicas/imunologia , Vesículas Secretórias/imunologia , Migração Transendotelial e Transepitelial/imunologia , Músculos Abdominais/irrigação sanguínea , Músculos Abdominais/imunologia , Animais , Membrana Basal/imunologia , Transporte Biológico Ativo/genética , Transporte Biológico Ativo/imunologia , Mucosa Gástrica/química , Mucosa Gástrica/imunologia , Fator de Crescimento de Hepatócito/genética , Humanos , Síndromes de Imunodeficiência/genética , Síndromes de Imunodeficiência/imunologia , Integrina alfa3beta1/genética , Integrina alfa3beta1/imunologia , Integrina alfa6beta1/genética , Integrina alfa6beta1/imunologia , Elastase de Leucócito/genética , Elastase de Leucócito/imunologia , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Camundongos , Camundongos Knockout , Proteínas Proto-Oncogênicas/genética , Vesículas Secretórias/genética , Migração Transendotelial e Transepitelial/genética , Vênulas/imunologia , Proteínas de Transporte Vesicular
14.
J Immunol ; 197(6): 2400-8, 2016 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-27534549

RESUMO

A classical hallmark of acute inflammation is neutrophil infiltration of tissues, a multistep process that involves sequential cell-cell interactions of circulating leukocytes with IL-1- or TNF-activated microvascular endothelial cells (ECs) and pericytes (PCs) that form the wall of the postcapillary venules. The initial infiltrating cells accumulate perivascularly in close proximity to PCs. IL-17, a proinflammatory cytokine that acts on target cells via a heterodimeric receptor formed by IL-17RA and IL-17RC subunits, also promotes neutrophilic inflammation but its effects on vascular cells are less clear. We report that both cultured human ECs and PCs strongly express IL-17RC and, although neither cell type expresses much IL-17RA, PCs express significantly more than ECs. IL-17, alone or synergistically with TNF, significantly alters inflammatory gene expression in cultured human PCs but not ECs. RNA sequencing analysis identifies many IL-17-induced transcripts in PCs encoding proteins known to stimulate neutrophil-mediated immunity. Conditioned media from IL-17-activated PCs, but not ECs, induce pertussis toxin-sensitive neutrophil polarization, likely mediated by PC-secreted chemokines, and they also stimulate neutrophil production of proinflammatory molecules, including TNF, IL-1α, IL-1ß, and IL-8. Furthermore, IL-17-activated PCs, but not ECs, can prolong neutrophil survival by producing G-CSF and GM-CSF, delaying the mitochondrial outer membrane permeabilization and caspase-9 activation. Importantly, neutrophils exhibit enhanced phagocytic capacity after activation by conditioned media from IL-17-treated PCs. We conclude that PCs, not ECs, are the major target of IL-17 within the microvessel wall and that IL-17-activated PCs can modulate neutrophil functions within the perivascular tissue space.


Assuntos
Endotélio Vascular/fisiologia , Interleucina-17/imunologia , Neutrófilos/imunologia , Pericitos/fisiologia , Receptores de Interleucina-17/imunologia , Caspase 9/metabolismo , Células Cultivadas , Meios de Cultura , Citocinas/biossíntese , Citocinas/imunologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/imunologia , Fator Estimulador de Colônias de Granulócitos/biossíntese , Fator Estimulador de Colônias de Granulócitos/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/biossíntese , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Humanos , Interleucina-17/genética , Interleucina-17/farmacologia , Infiltração de Neutrófilos , Neutrófilos/fisiologia , Pericitos/efeitos dos fármacos , Pericitos/imunologia , Receptores de Interleucina-17/fisiologia , Análise de Sequência de RNA , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/farmacologia , Vênulas/citologia , Vênulas/imunologia
15.
Cell Immunol ; 304-305: 59-62, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27131737

RESUMO

During inflammation, the selectin-induced slow rolling of neutrophils on venules cooperates with chemokine signaling to mediate neutrophil recruitment into tissues. Previous studies identified P-selectin glycoprotein ligand-1 (PSGL-1) and CD44 as E-selectin ligands that activate integrins to induce slow rolling. We show here that in TNF-α-treated cremaster muscle venules, slow leukocyte rolling was impaired in mice deficient in moesin, a member of the ezrin-radixin-moesin (ERM) family. Accordingly, neutrophil recruitment in a peritonitis model was decreased in moesin-deficient mice when chemokine signaling was blocked with pertussis toxin. These results suggest that moesin contributes to the slow rolling and subsequent recruitment of neutrophils during inflammation.


Assuntos
Movimento Celular , Proteínas dos Microfilamentos/metabolismo , Neutrófilos/imunologia , Peritonite/imunologia , Vênulas/imunologia , Músculos Abdominais/irrigação sanguínea , Animais , Movimento Celular/genética , Proteínas do Citoesqueleto/metabolismo , Modelos Animais de Doenças , Humanos , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas dos Microfilamentos/genética , Transdução de Sinais
16.
J Biol Chem ; 291(3): 1441-7, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26631722

RESUMO

In humans and mice, megakaryocytes/platelets and endothelial cells constitutively synthesize P-selectin and mobilize it to the plasma membrane to mediate leukocyte rolling during inflammation. TNF-α, interleukin 1ß, and LPS markedly increase P-selectin mRNA in mice but decrease P-selectin mRNA in humans. Transgenic mice bearing the entire human SELP gene recapitulate basal and inducible expression of human P-selectin and reveal human-specific differences in P-selectin function. Differences in the human SELP and murine Selp promoters account for divergent expression in vitro, but their significance in vivo is not known. Here we generated knockin mice that replace the 1.4-kb proximal Selp promoter with the corresponding SELP sequence (Selp(KI)). Selp(KI) (/) (KI) mice constitutively expressed more P-selectin on platelets and more P-selectin mRNA in tissues but only slightly increased P-selectin mRNA after injection of TNF-α or LPS. Consistent with higher basal expression, leukocytes rolled more slowly on P-selectin in trauma-stimulated venules of Selp(KI) (/) (KI) mice. However, TNF-α did not further reduce P-selectin-dependent rolling velocities. Blunted up-regulation of P-selectin mRNA during contact hypersensitivity reduced P-selectin-dependent inflammation in Selp(KI) (/-) mice. Higher basal P-selectin in Selp(KI) (/) (KI) mice compensated for this defect. Therefore, divergent sequences in a short promoter mediate most of the functionally significant differences in expression of human and murine P-selectin in vivo.


Assuntos
Regulação da Expressão Gênica , Selectina-P/metabolismo , Regiões Promotoras Genéticas , Animais , Sequência de Bases , Cruzamentos Genéticos , Dermatite de Contato/imunologia , Dermatite de Contato/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Migração e Rolagem de Leucócitos/efeitos dos fármacos , Migração e Rolagem de Leucócitos/imunologia , Lipopolissacarídeos/toxicidade , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Especificidade de Órgãos , Selectina-P/química , Selectina-P/genética , Regiões Promotoras Genéticas/efeitos dos fármacos , RNA Mensageiro/metabolismo , Especificidade da Espécie , Organismos Livres de Patógenos Específicos , Fator de Necrose Tumoral alfa/metabolismo , Vênulas/efeitos dos fármacos , Vênulas/imunologia
17.
Nat Commun ; 6: 7965, 2015 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-26242575

RESUMO

Inflammatory cell recruitment to local sites of tissue injury and/or infection is controlled by a plethora of signalling processes influencing cell-to-cell interactions between the vascular endothelial cells (ECs) in post-capillary venules and circulating leukocytes. Recently, ATP-sensitive P2Y purinergic receptors have emerged as downstream regulators of EC activation in vascular inflammation. However, the mechanism(s) regulating cellular ATP release in this response remains elusive. Here we report that the ATP-release channel Pannexin1 (Panx1) opens downstream of EC activation by TNF-α. This process involves activation of type-1 TNF receptors, recruitment of Src family kinases (SFK) and SFK-dependent phosphorylation of Panx1. Using an inducible, EC-specific Panx1 knockout mouse line, we report a previously unidentified role for Panx1 channels in promoting leukocyte adhesion and emigration through the venous wall during acute systemic inflammation, placing Panx1 channels at the centre of cytokine crosstalk with purinergic signalling in the endothelium.


Assuntos
Conexinas/metabolismo , Células Endoteliais/metabolismo , Inflamação/metabolismo , Leucócitos/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Migração Transendotelial e Transepitelial , Trifosfato de Adenosina/metabolismo , Animais , Adesão Celular , Células Cultivadas , Endotélio Vascular/imunologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Masculino , Camundongos Endogâmicos C57BL , Fosforilação , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima , Vênulas/imunologia , Quinases da Família src/metabolismo
19.
Science ; 346(6214): 1234-8, 2014 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-25477463

RESUMO

Immune and inflammatory responses require leukocytes to migrate within and through the vasculature, a process that is facilitated by their capacity to switch to a polarized morphology with an asymmetric distribution of receptors. We report that neutrophil polarization within activated venules served to organize a protruding domain that engaged activated platelets present in the bloodstream. The selectin ligand PSGL-1 transduced signals emanating from these interactions, resulting in the redistribution of receptors that drive neutrophil migration. Consequently, neutrophils unable to polarize or to transduce signals through PSGL-1 displayed aberrant crawling, and blockade of this domain protected mice against thromboinflammatory injury. These results reveal that recruited neutrophils scan for activated platelets, and they suggest that the neutrophils' bipolarity allows the integration of signals present at both the endothelium and the circulation before inflammation proceeds.


Assuntos
Plaquetas/imunologia , Inflamação/imunologia , Neutrófilos/imunologia , Ativação Plaquetária , Trombose/imunologia , Animais , Circulação Sanguínea , Movimento Celular , Polaridade Celular , Endotélio Vascular/imunologia , Inflamação/sangue , Masculino , Glicoproteínas de Membrana , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais , Vênulas/imunologia
20.
J Immunol ; 193(10): 5284-93, 2014 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-25320278

RESUMO

Although the homing of lymphocytes to GALT has been extensively studied, little is known about how high endothelial venules (HEVs) within Peyer's patches (PPs) are patterned to display dominantly mucosal addressin cell adhesion molecule 1 (MAdCAM-1). In this study, we report that Nkx2-3-deficient mice show gradual loss of MAdCAM-1 in PPs postnatally and increased levels of mRNA for peripheral lymph node addressin (PNAd) backbone proteins as well as enhanced expression of MECA79 sulfated glycoepitope at the luminal aspect of HEVs, thus replacing MAdCAM-1 with PNAd. Induction of PNAd in mutant PPs requires lymphotoxin ß receptor activity, and its upregulation needs the presence of mature T and B cells. Furthermore, treatment with MECA-79 anti-PNAd mAb in vivo effectively blocks lymphocyte homing to mutant PPs. Despite the replacement of MAdCAM-1 by PNAd in HEV endothelia, lymphocytes could efficiently home to PPs in mutant mice. We conclude that although Nkx2-3 activity controls the addressin balance of HEVs in GALT, the general HEV functionality is preserved independently from Nkx2-3, indicating a substantial plasticity in the specification of GALT HEV endothelium.


Assuntos
Linfócitos B/metabolismo , Proteínas de Homeodomínio/imunologia , Nódulos Linfáticos Agregados/metabolismo , Linfócitos T/metabolismo , Fatores de Transcrição/imunologia , Animais , Animais Recém-Nascidos , Anticorpos Monoclonais/farmacologia , Antígenos de Superfície/genética , Antígenos de Superfície/imunologia , Linfócitos B/citologia , Linfócitos B/imunologia , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/imunologia , Regulação da Expressão Gênica , Proteínas de Homeodomínio/genética , Mucosa Intestinal/metabolismo , Intestinos/citologia , Intestinos/imunologia , Linfonodos/citologia , Linfonodos/imunologia , Linfonodos/metabolismo , Receptor beta de Linfotoxina/genética , Receptor beta de Linfotoxina/imunologia , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Mucoproteínas , Nódulos Linfáticos Agregados/citologia , Nódulos Linfáticos Agregados/imunologia , Transdução de Sinais , Linfócitos T/citologia , Linfócitos T/imunologia , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Vênulas/citologia , Vênulas/imunologia , Vênulas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...