Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
Eur J Immunol ; 54(6): e2350761, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38566526

RESUMEN

In multiple sclerosis and its animal model, experimental autoimmune encephalomyelitis (EAE), early pathological features include immune cell infiltration into the central nervous system (CNS) and blood-brain barrier (BBB) disruption. We investigated the role of junctional adhesion molecule-A (JAM-A), a tight junction protein, in active EAE (aEAE) pathogenesis. Our study confirms JAM-A expression at the blood-brain barrier and its luminal redistribution during aEAE. JAM-A deficient (JAM-A-/-) C57BL/6J mice exhibited milder aEAE, unrelated to myelin oligodendrocyte glycoprotein-specific CD4+ T-cell priming. While JAM-A absence influenced macrophage behavior on primary mouse brain microvascular endothelial cells (pMBMECs) under flow in vitro, it did not impact T-cell extravasation across primary mouse brain microvascular endothelial cells. At aEAE onset, we observed reduced lymphocyte and CCR2+ macrophage infiltration into the spinal cord of JAM-A-/- mice compared to control littermates. This correlated with increased CD3+ T-cell accumulation in spinal cord perivascular spaces and brain leptomeninges, suggesting JAM-A absence leads to T-cell trapping in central nervous system border compartments. In summary, JAM-A plays a role in immune cell infiltration and clinical disease progression in aEAE.


Asunto(s)
Barrera Hematoencefálica , Encefalomielitis Autoinmune Experimental , Células Endoteliales , Macrófagos , Ratones Endogámicos C57BL , Ratones Noqueados , Animales , Encefalomielitis Autoinmune Experimental/inmunología , Ratones , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/inmunología , Barrera Hematoencefálica/patología , Macrófagos/inmunología , Macrófagos/metabolismo , Células Endoteliales/metabolismo , Células Endoteliales/inmunología , Médula Espinal/patología , Médula Espinal/inmunología , Médula Espinal/metabolismo , Linfocitos T CD4-Positivos/inmunología , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/patología , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Modelos Animales de Enfermedad
2.
J Cell Sci ; 134(8)2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33912914

RESUMEN

The migration of activated T cells across the blood-brain barrier (BBB) is a critical step in central nervous system (CNS) immune surveillance and inflammation. Whereas T cell diapedesis across the intact BBB seems to occur preferentially through the BBB cellular junctions, impaired BBB integrity during neuroinflammation is accompanied by increased transcellular T cell diapedesis. The underlying mechanisms directing T cells to paracellular versus transcellular sites of diapedesis across the BBB remain to be explored. By combining in vitro live-cell imaging of T cell migration across primary mouse brain microvascular endothelial cells (pMBMECs) under physiological flow with serial block-face scanning electron microscopy (SBF-SEM), we have identified BBB tricellular junctions as novel sites for T cell diapedesis across the BBB. Downregulated expression of tricellular junctional proteins or protein-based targeting of their interactions in pMBMEC monolayers correlated with enhanced transcellular T cell diapedesis, and abluminal presence of chemokines increased T cell diapedesis through tricellular junctions. Our observations assign an entirely novel role to BBB tricellular junctions in regulating T cell entry into the CNS. This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Barrera Hematoencefálica , Migración Transendotelial y Transepitelial , Animales , Transporte Biológico , Células Endoteliales , Ratones , Linfocitos T , Uniones Estrechas
3.
Eur J Immunol ; 52(1): 161-177, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34524684

RESUMEN

The migration of CD4+ effector/memory T cells across the blood-brain barrier (BBB) is a critical step in MS or its animal model, EAE. T-cell diapedesis across the BBB can occur paracellular, via the complex BBB tight junctions or transcellular via a pore through the brain endothelial cell body. Making use of primary mouse brain microvascular endothelial cells (pMBMECs) as in vitro model of the BBB, we here directly compared the transcriptome profile of pMBMECs favoring transcellular or paracellular T-cell diapedesis by RNA sequencing (RNA-seq). We identified the atypical chemokine receptor 1 (Ackr1) as one of the main candidate genes upregulated in pMBMECs favoring transcellular T-cell diapedesis. We confirmed upregulation of ACKR1 protein in pMBMECs promoting transcellular T-cell diapedesis and in venular endothelial cells in the CNS during EAE. Lack of endothelial ACKR1 reduced transcellular T-cell diapedesis across pMBMECs under physiological flow in vitro. Combining our previous observation that endothelial ACKR1 contributes to EAE pathogenesis by shuttling chemokines across the BBB, the present data support that ACKR1 mediated chemokine shuttling enhances transcellular T-cell diapedesis across the BBB during autoimmune neuroinflammation.


Asunto(s)
Barrera Hematoencefálica , Linfocitos T CD4-Positivos , Sistema del Grupo Sanguíneo Duffy , Encefalomielitis Autoinmune Experimental , Células T de Memoria , Esclerosis Múltiple , Receptores de Superficie Celular , Migración Transendotelial y Transepitelial , Animales , Ratones , Barrera Hematoencefálica/inmunología , Linfocitos T CD4-Positivos/inmunología , Sistema del Grupo Sanguíneo Duffy/genética , Sistema del Grupo Sanguíneo Duffy/inmunología , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/inmunología , Inflamación/genética , Inflamación/inmunología , Células T de Memoria/inmunología , Ratones Noqueados , Esclerosis Múltiple/genética , Esclerosis Múltiple/inmunología , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/inmunología , Migración Transendotelial y Transepitelial/genética , Migración Transendotelial y Transepitelial/inmunología
4.
Eur J Immunol ; 49(11): 2030-2043, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31318439

RESUMEN

Plasmacytoid dendritic cells (pDCs) are found in the CNS during neuroinflammation and have been reported to exert regulatory functions in multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE). However, the mechanisms of entry of pDCs into the CNS as well as their phenotype and innate functional properties, once recruited into the CNS, have not been thoroughly examined. Herein, we show that pDCs rapidly accumulate into the brain and spinal cord during the acute phase of EAE, and maintain the expression of numerous phenotypic markers typical of peripheral pDCs. Functionally, CNS-pDCs constitutively expressed IRF7 and were able to rapidly produce type I IFNs and IL-12p40 upon ex vivo TLR-9 stimulation. Using adoptive transfer experiments, we provide evidence that CNS-pDC are recruited from the blood and accumulate into the CNS during the acute phase of EAE. Accumulation of pDCs into the CNS was strongly inhibited in the absence of CD29, but not CD18, suggesting a major role for ß1 but not ß2 integrins. Indeed, blocking the CD49d α4-integrins during acute EAE drastically diminished CNS-pDC numbers. Together, our results demonstrate that circulating pDCs are actively recruited into the CNS during acute EAE through a mechanism largely dependent on CD49d/CD29-integrins.


Asunto(s)
Encéfalo/inmunología , Células Dendríticas/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Integrina alfa4/inmunología , Integrina beta1/inmunología , Médula Espinal/inmunología , Traslado Adoptivo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/patología , Movimiento Celular/inmunología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/patología , Células Dendríticas/trasplante , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/patología , Femenino , Regulación de la Expresión Génica , Inmunidad Innata , Integrina alfa4/genética , Integrina beta1/genética , Factor 7 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/inmunología , Interferón Tipo I/genética , Interferón Tipo I/inmunología , Subunidad p40 de la Interleucina-12/genética , Subunidad p40 de la Interleucina-12/inmunología , Ratones , Ratones Endogámicos C57BL , Glicoproteína Mielina-Oligodendrócito/administración & dosificación , Fragmentos de Péptidos/administración & dosificación , Toxina del Pertussis/administración & dosificación , Transducción de Señal , Médula Espinal/efectos de los fármacos , Médula Espinal/patología , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/inmunología
5.
Int J Mol Sci ; 20(6)2019 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-30901861

RESUMEN

Multiple sclerosis (MS) is the most common inflammatory disorder of the central nervous system (CNS) in young adults leading to severe disability. Besides genetic traits, environmental factors contribute to MS pathogenesis. Cigarette smoking increases the risk of MS in an HLA-dependent fashion, but the underlying mechanisms remain unknown. Here, we explored the effect of cigarette smoke exposure on spontaneous and induced models of experimental autoimmune encephalomyelitis (EAE) by evaluating clinical disease and, when relevant, blood leukocytes and histopathology. In the relapsing-remitting (RR) transgenic model in SJL/J mice, we observed very low incidence in both smoke-exposed and control groups. In the optico-spinal encephalomyelitis (OSE) double transgenic model in C57BL/6 mice, the early onset of EAE prevented a meaningful evaluation of the effects of cigarette smoke. In EAE models induced by immunization, daily exposure to cigarette smoke caused a delayed onset of EAE followed by a protracted disease course in SJL/J mice. In contrast, cigarette smoke exposure ameliorated the EAE clinical score in C57BL/6J mice. Our exploratory studies therefore show that genetic background influences the effects of cigarette smoke on autoimmune neuroinflammation. Importantly, our findings expose the challenge of identifying an animal model for studying the influence of cigarette smoke in MS.


Asunto(s)
Encefalomielitis Autoinmune Experimental/diagnóstico , Encefalomielitis Autoinmune Experimental/etiología , Antecedentes Genéticos , Fumar/efectos adversos , Edad de Inicio , Animales , Biopsia , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Encefalomielitis Autoinmune Experimental/metabolismo , Inmunohistoquímica , Ratones , Esclerosis Múltiple/diagnóstico , Esclerosis Múltiple/etiología , Esclerosis Múltiple/metabolismo , Fenotipo , Medición de Riesgo , Factores de Riesgo , Índice de Severidad de la Enfermedad , Médula Espinal/metabolismo , Médula Espinal/patología
6.
Brain Behav Immun ; 73: 3-20, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29920328

RESUMEN

In multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE) autoaggressive CD4+ T cells cross the blood-brain barrier (BBB) and cause neuroinflammation. Therapeutic targeting of CD4+ T-cell trafficking into the CNS by blocking α4-integrins has proven beneficial for the treatment of MS but comes with associated risks, probably due to blocking CD8+ T cell mediated CNS immune surveillance. Our recent observations show that CD8+ T cells also rely on α4ß1-integrins to cross the BBB. Besides vascular cell adhesion molecule-1 (VCAM-1), we identified junctional adhesion molecule-B (JAM-B) as a novel vascular α4ß1-integrin ligand involved in CD8+ T-cell migration across the BBB. This prompted us to investigate, if JAM-B also mediates CD4+ T-cell migration across the BBB. We first ensured that encephalitogenic T cells can bind to JAM-B in vitro and next compared EAE pathogenesis in JAM-B-/- C57BL/6J mice and their wild-type littermates. Following immunization with MOGaa35-55 peptide, JAM-B-/- mice developed ameliorated EAE compared to their wild-type littermates. At the same time, we isolated higher numbers of CD45+ infiltrating immune cells from the CNS of JAM-B-/- C57BL/6J mice suffering from EAE. Immunofluorescence staining revealed that the majority of CD45+ inflammatory cells accumulated in the leptomeningeal and perivascular spaces of the CNS behind the BBB but do not gain access to the CNS parenchyma. Trapping of CNS inflammatory cells was not due to increased inflammatory cell proliferation. Neither a loss of BBB integrity or BBB polarity potentially affecting local chemokine gradients nor a lack of focal gelatinase activation required for CNS parenchymal immune cell entry across the glia limitans could be detected in JAM-B-/- mice. Lack of a role for JAM-B in the effector phase of EAE was supported by the observation that we did not detect any role for JAM-B in EAE pathogenesis, when EAE was elicited by in vitro activated MOG aa35-55-specific CD4+ effector T cells. On the other hand, we also failed to demonstrate any role of JAM-B in in vivo priming, proliferation or polarization of MOGaa35-55-specific CD4+ T cells in peripheral immune organs. Finally, our study excludes expression of and thus a role for JAM-B on peripheral and CNS infiltrating myeloid cells. Taken together, although endothelial JAM-B is not required for immune cell trafficking across the BBB in EAE, in its absence accumulation of inflammatory cells mainly in CNS leptomeningeal spaces leads to amelioration of EAE.


Asunto(s)
Encefalomielitis Autoinmune Experimental/metabolismo , Molécula B de Adhesión de Unión/metabolismo , Molécula B de Adhesión de Unión/fisiología , Animales , Barrera Hematoencefálica/metabolismo , Linfocitos T CD8-positivos/metabolismo , Movimiento Celular/fisiología , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/fisiología , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/fisiopatología , Endotelio Vascular/metabolismo , Femenino , Integrina alfa4beta1/metabolismo , Molécula B de Adhesión de Unión/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/fisiopatología , Glicoproteína Mielina-Oligodendrócito/farmacología , Células Mieloides/metabolismo , Células Mieloides/fisiología , Uniones Estrechas/metabolismo
7.
Eur J Immunol ; 45(4): 1043-58, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25545837

RESUMEN

The extravasation of CD4(+) effector/memory T cells (TEM cells) across the blood-brain barrier (BBB) is a crucial step in the pathogenesis of experimental autoimmune encephalomyelitis (EAE) or multiple sclerosis (MS). Endothelial ICAM-1 and ICAM-2 are essential for CD4(+) TEM cell crawling on the BBB prior to diapedesis. Here, we investigated the influence of cell surface levels of endothelial ICAM-1 in determining the cellular route of CD4(+) TEM -cell diapedesis across cytokine treated primary mouse BBB endothelial cells under physiological flow. Inflammatory conditions, inducing high levels of endothelial ICAM-1, promoted rapid initiation of transcellular diapedesis of CD4(+) T cells across the BBB, while intermediate levels of endothelial ICAM-1 favored paracellular CD4(+) T-cell diapedesis. Importantly, the route of T-cell diapedesis across the BBB was independent of loss of BBB barrier properties. Unexpectedly, a low number of CD4(+) TEM cells was found to cross the inflamed BBB in the absence of endothelial ICAM-1 and ICAM-2 via an obviously alternatively regulated transcellular pathway. In vivo, this translated to the development of ameliorated EAE in ICAM-1(null) //ICAM-2(-/-) C57BL/6J mice. Taken together, our study demonstrates that cell surface levels of endothelial ICAM-1 rather than the inflammatory stimulus or BBB integrity influence the pathway of T-cell diapedesis across the BBB.


Asunto(s)
Barrera Hematoencefálica/inmunología , Linfocitos T CD4-Positivos/metabolismo , Encefalomielitis Autoinmune Experimental/inmunología , Molécula 1 de Adhesión Intercelular/metabolismo , Migración Transendotelial y Transepitelial/inmunología , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Linfocitos T CD4-Positivos/inmunología , Adhesión Celular/inmunología , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/inmunología , Moléculas de Adhesión Celular/metabolismo , Células Endoteliales/inmunología , Femenino , Memoria Inmunológica/inmunología , Inflamación/inmunología , Molécula 1 de Adhesión Intercelular/genética , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Esclerosis Múltiple/inmunología
8.
Nature ; 465(7297): 483-6, 2010 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-20445537

RESUMEN

In development, tissue regeneration or certain diseases, angiogenic growth leads to the expansion of blood vessels and the lymphatic vasculature. This involves endothelial cell proliferation as well as angiogenic sprouting, in which a subset of cells, termed tip cells, acquires motile, invasive behaviour and extends filopodial protrusions. Although it is already appreciated that angiogenesis is triggered by tissue-derived signals, such as vascular endothelial growth factor (VEGF) family growth factors, the resulting signalling processes in endothelial cells are only partly understood. Here we show with genetic experiments in mouse and zebrafish that ephrin-B2, a transmembrane ligand for Eph receptor tyrosine kinases, promotes sprouting behaviour and motility in the angiogenic endothelium. We link this pro-angiogenic function to a crucial role of ephrin-B2 in the VEGF signalling pathway, which we have studied in detail for VEGFR3, the receptor for VEGF-C. In the absence of ephrin-B2, the internalization of VEGFR3 in cultured cells and mutant mice is defective, which compromises downstream signal transduction by the small GTPase Rac1, Akt and the mitogen-activated protein kinase Erk. Our results show that full VEGFR3 signalling is coupled to receptor internalization. Ephrin-B2 is a key regulator of this process and thereby controls angiogenic and lymphangiogenic growth.


Asunto(s)
Efrina-B2/metabolismo , Linfangiogénesis , Neovascularización Fisiológica , Factor C de Crecimiento Endotelial Vascular/metabolismo , Animales , Células Cultivadas , Pérdida del Embrión , Embrión de Mamíferos/irrigación sanguínea , Embrión de Mamíferos/metabolismo , Endocitosis , Células Endoteliales/citología , Células Endoteliales/metabolismo , Efrina-B2/deficiencia , Efrina-B2/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Humanos , Linfangiogénesis/genética , Vasos Linfáticos , Ratones , Ratones Transgénicos , Neovascularización Fisiológica/genética , Neuropéptidos/metabolismo , Embarazo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor EphB4/deficiencia , Receptor EphB4/genética , Receptor EphB4/metabolismo , Transducción de Señal , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo , Pez Cebra , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rac1
9.
Eur J Immunol ; 44(8): 2287-94, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24740164

RESUMEN

T-cell migration across the blood-brain barrier is a crucial step in the pathogenesis of EAE, an animal model for MS. Live cell imaging studies demonstrated that P-selectin glycoprotein ligand-1 (PSGL-1) and its endothelial ligands E- and P-selectin mediate the initial rolling of T cells in brain vessels during EAE. As functional absence of PSGL-1 or E/P-selectins does not result in ameliorated EAE, we speculated that T-cell entry into the spinal cord is independent of PSGL-1 and E/P-selectin. Performing intravital microscopy, we observed the interaction of WT or PSGL-1(-/-) proteolipid protein-specific T cells in inflamed spinal cord microvessels of WT or E/P-selectin(-/-) SJL/J mice during EAE. T-cell rolling but not T-cell capture was completely abrogated in the absence of either PSGL-1 or E- and P-selectin, resulting in a significantly reduced number of T cells able to firmly adhere in the inflamed spinal cord microvessels, but did not lead to reduced T-cell invasion into the CNS parenchyma. Thus, PSGL-1 interaction with E/P-selectin is essential for T-cell rolling in inflamed spinal cord microvessels during EAE. Taken together with previous observations, our findings show that T-cell rolling is not required for successful T-cell entry into the CNS and initiation of EAE.


Asunto(s)
Selectina E/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Glicoproteínas de Membrana/inmunología , Microvasos/inmunología , Selectina-P/inmunología , Linfocitos T/inmunología , Animales , Barrera Hematoencefálica/inmunología , Adhesión Celular/inmunología , Línea Celular , Movimiento Celular/inmunología , Ligandos , Ratones , Médula Espinal/inmunología
10.
Brain ; 137(Pt 5): 1454-69, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24625696

RESUMEN

The Duffy antigen/receptor for chemokines, DARC, belongs to the family of atypical heptahelical chemokine receptors that do not couple to G proteins and therefore fail to transmit conventional intracellular signals. Here we show that during experimental autoimmune encephalomyelitis, an animal model of multiple sclerosis, the expression of DARC is upregulated at the blood-brain barrier. These findings are corroborated by the presence of a significantly increased number of subcortical white matter microvessels staining positive for DARC in human multiple sclerosis brains as compared to control tissue. Using an in vitro blood-brain barrier model we demonstrated that endothelial DARC mediates the abluminal to luminal transport of inflammatory chemokines across the blood-brain barrier. An involvement of DARC in experimental autoimmune encephalomyelitis pathogenesis was confirmed by the observed ameliorated experimental autoimmune encephalomyelitis in Darc(-/-) C57BL/6 and SJL mice, as compared to wild-type control littermates. Experimental autoimmune encephalomyelitis studies in bone marrow chimeric Darc(-/-) and wild-type mice revealed that increased plasma levels of inflammatory chemokines in experimental autoimmune encephalomyelitis depended on the presence of erythrocyte DARC. However, fully developed experimental autoimmune encephalomyelitis required the expression of endothelial DARC. Taken together, our data show a role for erythrocyte DARC as a chemokine reservoir and that endothelial DARC contributes to the pathogenesis of experimental autoimmune encephalomyelitis by shuttling chemokines across the blood-brain barrier.


Asunto(s)
Barrera Hematoencefálica , Quimiocinas , Sistema del Grupo Sanguíneo Duffy , Encefalomielitis Autoinmune Experimental , Esclerosis Múltiple , Receptores de Superficie Celular , Regulación hacia Arriba , Adulto , Anciano , Anciano de 80 o más Años , Animales , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Antígenos CD/metabolismo , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/fisiopatología , Permeabilidad Capilar/genética , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/patología , Cerebelo/metabolismo , Quimiocinas/genética , Quimiocinas/metabolismo , Modelos Animales de Enfermedad , Sistema del Grupo Sanguíneo Duffy/metabolismo , Encefalomielitis Autoinmune Experimental/sangre , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/patología , Encefalomielitis Autoinmune Experimental/fisiopatología , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Técnicas In Vitro , Ratones Endogámicos C57BL , Ratones Noqueados , Esclerosis Múltiple/patología , Receptores de Superficie Celular/deficiencia , Receptores de Superficie Celular/metabolismo , Regulación hacia Arriba/genética
11.
Eur Spine J ; 24(10): 2173-81, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25711910

RESUMEN

INTRODUCTION: Clinical treatment of spinal metastasis is gaining in complexity while the underlying biology remains unknown. Insufficient biological understanding is due to a lack of suitable experimental animal models. Intercellular adhesion molecule-1 (ICAM1) has been implicated in metastasis formation. Its role in spinal metastasis remains unclear. It was the aim to generate a reliable spinal metastasis model in mice and to investigate metastasis formation under ICAM1 depletion. MATERIAL AND METHODS: B16 melanoma cells were infected with a lentivirus containing firefly luciferase (B16-luc). Stable cell clones (B16-luc) were injected retrogradely into the distal aortic arch. Spinal metastasis formation was monitored using in vivo bioluminescence imaging/MRI. Neurological deficits were monitored daily. In vivo selected, metastasized tumor cells were isolated (mB16-luc) and reinjected intraarterially. mB16-luc cells were injected intraarterially in ICAM1 KO mice. Metastasis distribution was analyzed using organ-specific fluorescence analysis. RESULTS: Intraarterial injection of B16-luc and metastatic mB16-luc reliably induced spinal metastasis formation with neurological deficits (B16-luc:26.5, mB16-luc:21 days, p<0.05). In vivo selection increased the metastatic aggressiveness and led to a bone specific homing phenotype. Thus, mB16-luc cells demonstrated higher number (B16-luc: 1.2±0.447, mB16-luc:3.2±1.643) and increased total metastasis volume (B16-luc:2.87±2.453 mm3, mB16-luc:11.19±3.898 mm3, p<0.05) in the spine. ICAM1 depletion leads to a significantly reduced number of spinal metastasis (mB16-luc:1.2±0.84) with improved neurological outcome (29 days). General metastatic burden was significantly reduced under ICAM1 depletion (control: 3.47×10(7)±1.66×10(7); ICAM-1-/-: 5.20×10(4)±4.44×10(4), p<0.05 vs. control) CONCLUSION: Applying a reliable animal model for spinal metastasis, ICAM1 depletion reduces spinal metastasis formation due to an organ-unspecific reduction of metastasis development.


Asunto(s)
Molécula 1 de Adhesión Intercelular/fisiología , Melanoma Experimental/secundario , Neoplasias de la Columna Vertebral/secundario , Animales , Modelos Animales de Enfermedad , Vectores Genéticos , Molécula 1 de Adhesión Intercelular/genética , Melanoma Experimental/complicaciones , Melanoma Experimental/prevención & control , Ratones , Ratones Noqueados , Compresión de la Médula Espinal/etiología , Neoplasias de la Columna Vertebral/complicaciones , Neoplasias de la Columna Vertebral/prevención & control
12.
Genesis ; 52(9): 809-16, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24817584

RESUMEN

Notch signaling is important in angiogenesis during embryonic development. However, the embryonic lethal phenotypes of knock-out and transgenic mice have precluded studies of the role of Notch post-natally. To develop a mouse model that would bypass the embryonic lethal phenotype and investigate the possible role of Notch signaling in adult vessel growth, we developed transgenic mice with Cre-conditional expression of the constitutively active intracellular domain of Notch1 (IC-Notch1). Double transgenic IC-Notch1/Tie2-Cre embryos with endothelial specific IC-Notch1 expression died at embryonic day 9.5. They displayed collapsed and leaky blood vessels and defects in angiogenesis development. A tetracycline-inducible system was used to express Cre recombinase postnatally in endothelial cells. In adult mice, IC-Notch1 expression inhibited bFGF-induced neovascularization and female mice lacked mature ovarian follicles, which may reflect the block in bFGF-induced angiogenesis required for follicle growth. Our results demonstrate that Notch signaling is important for both embryonic and adult angiogenesis and indicate that the Notch signaling pathway may be a useful target for angiogenic therapies.


Asunto(s)
Inductores de la Angiogénesis/efectos adversos , Factor 2 de Crecimiento de Fibroblastos/farmacología , Neovascularización Fisiológica , Folículo Ovárico/embriología , Receptores Notch/metabolismo , Animales , Células Endoteliales/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Integrasas/genética , Integrasas/metabolismo , Ratones Transgénicos , Folículo Ovárico/crecimiento & desarrollo , Receptores Notch/genética , Transducción de Señal , Tetraciclina/farmacología
13.
Sci Signal ; 17(830): eade4335, 2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38564492

RESUMEN

Serum ferritin concentrations increase during hepatic inflammation and correlate with the severity of chronic liver disease. Here, we report a molecular mechanism whereby the heavy subunit of ferritin (FTH) contributes to hepatic inflammation. We found that FTH induced activation of the NLRP3 inflammasome and secretion of the proinflammatory cytokine interleukin-1ß (IL-1ß) in primary rat hepatic stellate cells (HSCs) through intercellular adhesion molecule-1 (ICAM-1). FTH-ICAM-1 stimulated the expression of Il1b, NLRP3 inflammasome activation, and the processing and secretion of IL-1ß in a manner that depended on plasma membrane remodeling, clathrin-mediated endocytosis, and lysosomal destabilization. FTH-ICAM-1 signaling at early endosomes stimulated Il1b expression, implying that this endosomal signaling primed inflammasome activation in HSCs. In contrast, lysosomal destabilization was required for FTH-induced IL-1ß secretion, suggesting that lysosomal damage activated inflammasomes. FTH induced IL-1ß production in liver slices from wild-type mice but not in those from Icam1-/- or Nlrp3-/- mice. Thus, FTH signals through its receptor ICAM-1 on HSCs to activate the NLRP3 inflammasome. We speculate that this pathway contributes to hepatic inflammation, a key process that stimulates hepatic fibrogenesis associated with chronic liver disease.


Asunto(s)
Inflamasomas , Hepatopatías , Ratas , Ratones , Animales , Inflamasomas/genética , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Células Estrelladas Hepáticas/metabolismo , Ferritinas/genética , Ferritinas/metabolismo , Interleucina-1beta/metabolismo , Inflamación/genética , Inflamación/metabolismo
14.
Nat Commun ; 14(1): 3106, 2023 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-37253744

RESUMEN

Blood-brain barrier (BBB) breakdown and immune cell infiltration into the central nervous system (CNS) are early hallmarks of multiple sclerosis (MS). High numbers of CD8+ T cells are found in MS lesions, and antigen (Ag) presentation at the BBB has been proposed to promote CD8+ T cell entry into the CNS. Here, we show that brain endothelial cells process and cross-present Ag, leading to effector CD8+ T cell differentiation. Under physiological flow in vitro, endothelial Ag presentation prevented CD8+ T cell crawling and diapedesis resulting in brain endothelial cell apoptosis and BBB breakdown. Brain endothelial Ag presentation in vivo was limited due to Ag uptake by CNS-resident macrophages but still reduced motility of Ag-specific CD8+ T cells within CNS microvessels. MHC class I-restricted Ag presentation at the BBB during neuroinflammation thus prohibits CD8+ T cell entry into the CNS and triggers CD8+ T cell-mediated focal BBB breakdown.


Asunto(s)
Barrera Hematoencefálica , Esclerosis Múltiple , Humanos , Barrera Hematoencefálica/metabolismo , Linfocitos T CD8-positivos , Células Endoteliales/metabolismo , Sistema Nervioso Central/metabolismo , Antígenos de Histocompatibilidad Clase I/metabolismo
15.
Nat Commun ; 14(1): 5837, 2023 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-37730744

RESUMEN

Meninges cover the surface of the brain and spinal cord and contribute to protection and immune surveillance of the central nervous system (CNS). How the meningeal layers establish CNS compartments with different accessibility to immune cells and immune mediators is, however, not well understood. Here, using 2-photon imaging in female transgenic reporter mice, we describe VE-cadherin at intercellular junctions of arachnoid and pia mater cells that form the leptomeninges and border the subarachnoid space (SAS) filled with cerebrospinal fluid (CSF). VE-cadherin expression also marked a layer of Prox1+ cells located within the arachnoid beneath and separate from E-cadherin+ arachnoid barrier cells. In vivo imaging of the spinal cord and brain in female VE-cadherin-GFP reporter mice allowed for direct observation of accessibility of CSF derived tracers and T cells into the SAS bordered by the arachnoid and pia mater during health and neuroinflammation, and detection of volume changes of the SAS during CNS pathology. Together, the findings identified VE-cadherin as an informative landmark for in vivo imaging of the leptomeninges that can be used to visualize the borders of the SAS and thus potential barrier properties of the leptomeninges in controlling access of immune mediators and immune cells into the CNS during health and neuroinflammation.


Asunto(s)
Enfermedades Neuroinflamatorias , Piamadre , Femenino , Animales , Ratones , Sistema Nervioso Central/diagnóstico por imagen , Aracnoides/diagnóstico por imagen , Cadherinas , Inflamación , Ratones Transgénicos
16.
Cancers (Basel) ; 15(20)2023 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-37894438

RESUMEN

Melanoma frequently metastasises to the brain, and a detailed understanding of the molecular and cellular mechanisms underlying melanoma cell extravasation across the blood-brain barrier (BBB) is important for preventing brain metastasis formation. Making use of primary mouse brain microvascular endothelial cells (pMBMECs) as an in vitro BBB model, we imaged the interaction of melanoma cells into pMBMEC monolayers. We observed exclusive junctional intercalation of melanoma cells and confirmed that melanoma-induced pMBMEC barrier disruption can be rescued by protease inhibition. Interleukin (IL)-1ß stimulated pMBMECs or PECAM-1-knockout (-ko) pMBMECs were employed to model compromised BBB barrier properties in vitro and to determine increased melanoma cell intercalation compared to pMBMECs with intact junctions. The newly generated brain-homing melanoma cell line YUMM1.1-BrM4 was used to reveal increased in vivo extravasation of melanoma cells across the BBB of barrier-compromised PECAM-1-deficient mice compared to controls. Taken together, our data indicate that preserving BBB integrity is an important measure to limit the formation of melanoma-brain metastasis.

17.
Eur J Immunol ; 41(3): 813-21, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21341265

RESUMEN

Inhibiting the α4 subunit of the integrin heterodimers α4ß1 and α4ß7 with the mab natalizumab is an effective treatment of multiple sclerosis (MS). Which of the two α4 heterodimers is involved in disease pathogenesis has, however, remained controversial. Whereas the development of experimental autoimmune encephalomyelitis (EAE), an animal model of MS, is ameliorated in ß7-integrin-deficient C57BL/6 mice, neutralizing antibodies against the ß7-integrin subunit or the α4ß7-integrin heterodimer fail to interfere with EAE pathogenesis in the SJL mouse. To facilitate α4ß7-integrin-mediated immune-cell trafficking across the blood-brain barrier (BBB), we established transgenic C57BL/6 mice with endothelial cell-specific, inducible expression of the α4ß7-integrin ligand mucosal addressin cell adhesion molecule (MAdCAM)-1 using the tetracycline (TET)-OFF system. Although TET-regulated MAdCAM-1 induced α4ß7-integrin mediated interaction of α4ß7(+) /α4ß1(-) T cells with the BBB in vitro and in vivo, it failed to influence EAE pathogenesis in C57BL/6 mice. TET-regulated MAdCAM-1 on the BBB neither changed the localization of central nervous system (CNS) perivascular inflammatory cuffs nor did it enhance the percentage of α4ß7-integrin(+) inflammatory cells within the CNS during EAE. In conclusion, our study demonstrates that ectopic expression of MAdCAM-1 at the BBB does not increase α4ß7-integrin-mediated immune cell trafficking into the CNS during MOG(aa35-55)-induced EAE.


Asunto(s)
Barrera Hematoencefálica/inmunología , Moléculas de Adhesión Celular/metabolismo , Encefalomielitis Autoinmune Experimental/inmunología , Integrinas/metabolismo , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Moléculas de Adhesión Celular/genética , Encefalomielitis Autoinmune Experimental/etiología , Encefalomielitis Autoinmune Experimental/patología , Expresión Génica/efectos de los fármacos , Ligandos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mucoproteínas , Fenotipo , Tetraciclina/farmacología
18.
Blood ; 116(6): 915-25, 2010 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-20395417

RESUMEN

Although it is well established that stromal intercellular adhesion molecule-1 (ICAM-1), ICAM-2, and vascular cell adhesion molecule-1 (VCAM-1) mediate lymphocyte recruitment into peripheral lymph nodes (PLNs), their precise contributions to the individual steps of the lymphocyte homing cascade are not known. Here, we provide in vivo evidence for a selective function for ICAM-1 > ICAM-2 > VCAM-1 in lymphocyte arrest within noninflamed PLN microvessels. Blocking all 3 CAMs completely inhibited lymphocyte adhesion within PLN high endothelial venules (HEVs). Post-arrest extravasation of T cells was a 3-step process, with optional ICAM-1-dependent intraluminal crawling followed by rapid ICAM-1- or ICAM-2-independent diapedesis and perivascular trapping. Parenchymal motility of lymphocytes was modestly reduced in the absence of ICAM-1, while ICAM-2 and alpha4-integrin ligands were not required for B-cell motility within follicles. Our findings highlight nonredundant functions for stromal Ig family CAMs in shear-resistant lymphocyte adhesion in steady-state HEVs, a unique role for ICAM-1 in intraluminal lymphocyte crawling but redundant roles for ICAM-1 and ICAM-2 in lymphocyte diapedesis and interstitial motility.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Movimiento Celular/inmunología , Molécula 1 de Adhesión Intercelular/metabolismo , Ganglios Linfáticos/citología , Linfocitos/citología , Molécula 1 de Adhesión Celular Vascular/metabolismo , Animales , Antígenos CD , Adhesión Celular/inmunología , Moléculas de Adhesión Celular/genética , Femenino , Molécula 1 de Adhesión Intercelular/genética , Ganglios Linfáticos/irrigación sanguínea , Recuento de Linfocitos , Linfocitos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Microcirculación/inmunología , Estrés Mecánico
19.
J Immunol ; 185(8): 4846-55, 2010 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-20861356

RESUMEN

Endothelial ICAM-1 and ICAM-2 were shown to be essential for T cell diapedesis across the blood-brain barrier (BBB) in vitro under static conditions. Crawling of T cells prior to diapedesis was only recently revealed to occur preferentially against the direction of blood flow on the endothelial surface of inflamed brain microvessels in vivo. Using live cell-imaging techniques, we prove that Th1 memory/effector T cells predominantly crawl against the direction of flow on the surface of BBB endothelium in vitro. Analysis of T cell interaction with wild-type, ICAM-1-deficient, ICAM-2-deficient, or ICAM-1 and ICAM-2 double-deficient primary mouse brain microvascular endothelial cells under physiological flow conditions allowed us to dissect the individual contributions of endothelial ICAM-1, ICAM-2, and VCAM-1 to shear-resistant T cell arrest, polarization, and crawling. Although T cell arrest was mediated by endothelial ICAM-1 and VCAM-1, T cell polarization and crawling were mediated by endothelial ICAM-1 and ICAM-2 but not by endothelial VCAM-1. Therefore, our data delineate a sequential involvement of endothelial ICAM-1 and VCAM-1 in mediating shear-resistant T cell arrest, followed by endothelial ICAM-1 and ICAM-2 in mediating T cell crawling to sites permissive for diapedesis across BBB endothelium.


Asunto(s)
Antígenos CD/metabolismo , Barrera Hematoencefálica/metabolismo , Moléculas de Adhesión Celular/metabolismo , Células Endoteliales/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Linfocitos T/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo , Animales , Antígenos CD/inmunología , Barrera Hematoencefálica/inmunología , Adhesión Celular/inmunología , Moléculas de Adhesión Celular/inmunología , Comunicación Celular/fisiología , Quimiotaxis de Leucocito/fisiología , Células Endoteliales/inmunología , Endotelio Vascular/inmunología , Endotelio Vascular/metabolismo , Femenino , Molécula 1 de Adhesión Intercelular/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Linfocitos T/inmunología , Molécula 1 de Adhesión Celular Vascular/inmunología
20.
Acta Neuropathol ; 122(5): 601-14, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21983942

RESUMEN

In experimental autoimmune encephalomyelitis (EAE), an animal model for multiple sclerosis (MS), loss of the blood-brain barrier (BBB) tight junction (TJ) protein claudin-3 correlates with immune cell infiltration into the CNS and BBB leakiness. Here we show that sealing BBB TJs by ectopic tetracycline-regulated expression of the TJ protein claudin-1 in Tie-2 tTA//TRE-claudin-1 double transgenic C57BL/6 mice had no influence on immune cell trafficking across the BBB during EAE and furthermore did not influence the onset and severity of the first clinical disease episode. However, expression of claudin-1 did significantly reduce BBB leakiness for both blood borne tracers and endogenous plasma proteins specifically around vessels expressing claudin-1. In addition, mice expressing claudin-1 exhibited a reduced disease burden during the chronic phase of EAE as compared to control littermates. Our study identifies BBB TJs as the critical structure regulating BBB permeability but not immune cell trafficking into CNS during EAE, and indicates BBB dysfunction is a potential key event contributing to disease burden in the chronic phase of EAE. Our observations suggest that stabilizing BBB barrier function by therapeutic targeting of TJs may be beneficial in treating MS, especially when anti-inflammatory treatments have failed.


Asunto(s)
Barrera Hematoencefálica/patología , Barrera Hematoencefálica/fisiología , Encefalomielitis Autoinmune Experimental/fisiopatología , Proteínas de la Membrana/fisiología , Esclerosis Múltiple/fisiopatología , Uniones Estrechas/fisiología , Animales , Movimiento Celular/fisiología , Sistema Nervioso Central/fisiopatología , Claudina-1 , Modelos Animales de Enfermedad , Endotelio Vascular/patología , Endotelio Vascular/fisiopatología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptor TIE-2/genética , Receptor TIE-2/fisiología , Tetraciclina/farmacología
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda