Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 42(4): 719-30, 2015 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-25902483

RESUMEN

Within secondary lymphoid tissues, stromal reticular cells support lymphocyte function, and targeting reticular cells is a potential strategy for controlling pathogenic lymphocytes in disease. However, the mechanisms that regulate reticular cell function are not well understood. Here we found that during an immune response in lymph nodes, dendritic cells (DCs) maintain reticular cell survival in multiple compartments. DC-derived lymphotoxin beta receptor (LTßR) ligands were critical mediators, and LTßR signaling on reticular cells mediated cell survival by modulating podoplanin (PDPN). PDPN modulated integrin-mediated cell adhesion, which maintained cell survival. This DC-stromal axis maintained lymphocyte survival and the ongoing immune response. Our findings provide insight into the functions of DCs, LTßR, and PDPN and delineate a DC-stromal axis that can potentially be targeted in autoimmune or lymphoproliferative diseases.


Asunto(s)
Células Dendríticas/citología , Ganglios Linfáticos/citología , Receptor beta de Linfotoxina/inmunología , Glicoproteínas de Membrana/inmunología , Células del Estroma/citología , Animales , Adhesión Celular , Supervivencia Celular/inmunología , Células Dendríticas/inmunología , Regulación de la Expresión Génica , Inmunofenotipificación , Ganglios Linfáticos/inmunología , Depleción Linfocítica , Receptor beta de Linfotoxina/genética , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Transducción de Señal , Células del Estroma/inmunología
2.
PLoS Comput Biol ; 17(6): e1009031, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34106916

RESUMEN

Treating macaques with an anti-α4ß7 antibody under the umbrella of combination antiretroviral therapy (cART) during early SIV infection can lead to viral remission, with viral loads maintained at < 50 SIV RNA copies/ml after removal of all treatment in a subset of animals. Depletion of CD8+ lymphocytes in controllers resulted in transient recrudescence of viremia, suggesting that the combination of cART and anti-α4ß7 antibody treatment led to a state where ongoing immune responses kept the virus undetectable in the absence of treatment. A previous mathematical model of HIV infection and cART incorporates immune effector cell responses and exhibits the property of two different viral load set-points. While the lower set-point could correspond to the attainment of long-term viral remission, attaining the higher set-point may be the result of viral rebound. Here we expand that model to include possible mechanisms of action of an anti-α4ß7 antibody operating in these treated animals. We show that the model can fit the longitudinal viral load data from both IgG control and anti-α4ß7 antibody treated macaques, suggesting explanations for the viral control associated with cART and an anti-α4ß7 antibody treatment. This effective perturbation to the virus-host interaction can also explain observations in other nonhuman primate experiments in which cART and immunotherapy have led to post-treatment control or resetting of the viral load set-point. Interestingly, because the viral kinetics in the various treated animals differed-some animals exhibited large fluctuations in viral load after cART cessation-the model suggests that anti-α4ß7 treatment could act by different primary mechanisms in different animals and still lead to post-treatment viral control. This outcome is nonetheless in accordance with a model with two stable viral load set-points, in which therapy can perturb the system from one set-point to a lower one through different biological mechanisms.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antivirales/uso terapéutico , Integrinas/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/terapia , Animales , Anticuerpos Monoclonales/inmunología , Antivirales/farmacología , Linfocitos T CD8-positivos/inmunología , Terapia Combinada , Depleción Linfocítica , Macaca , Síndrome de Inmunodeficiencia Adquirida del Simio/tratamiento farmacológico , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/aislamiento & purificación , Carga Viral/efectos de los fármacos , Carga Viral/inmunología
3.
Eur J Immunol ; 50(3): 418-425, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32012252

RESUMEN

To investigate the role of lymphotoxin (LT) in Sjögren's syndrome (SS) and in mucosal associated lymphoid tissue (MALT)-lymphoma, we made transgenic mice (Amy1-LTαß) that targeted LTα and LTß to the salivary and lacrimal glands. Amy1-LTαß mice developed atrophic salivary and lacrimal glands that contained tertiary lymphoid organs (TLOs) and had reduced tear production. Amy1-LTαß mice developed cervical lymphadenopathy but not MALT-lymphoma. TLO formation in the salivary and lacrimal glands of Amy1-LTαß was not sufficient to induce autoimmunity as measured by autoantibody titres.


Asunto(s)
Aparato Lagrimal/patología , Linfadenopatía/patología , Linfotoxina-alfa/metabolismo , Glándulas Salivales/patología , Estructuras Linfoides Terciarias/patología , Animales , Linfadenopatía/genética , Linfoma de Células B de la Zona Marginal/genética , Linfoma de Células B de la Zona Marginal/inmunología , Linfoma de Células B de la Zona Marginal/patología , Linfotoxina-alfa/genética , Ratones , Ratones Transgénicos , Síndrome de Sjögren/genética , Síndrome de Sjögren/inmunología , Síndrome de Sjögren/patología , Lágrimas/metabolismo , Estructuras Linfoides Terciarias/genética
4.
Curr Top Microbiol Immunol ; 426: 1-19, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32588229

RESUMEN

Tertiary lymphoid organs (TLOs), also known as inducible lymphoid organs, tertiary lymphoid structures, tertiary lymphoid tissues, or ectopic lymphoid organs are accumulations of cells in chronic inflammation that have been observed in most tissues in autoimmunity, infection, and cancer in mouse and man. They share many properties with secondary lymphoid organs (SLOs), particularly lymph nodes, with regard to cellular composition, function, and regulation. TLOs include T and B cells, dendritic cells, follicular dendritic cells, and many other stromal cells, and high endothelial venules (HEVs) and lymphatic vessels. They serve as sites of antigen presentation and tolerance induction; they are harmful in autoimmunity and can be both harmful and beneficial in cancer. SLO induction in ontogeny is mediated by interactions of several cell types, including CD4+ CD3- lymphoid tissue inducer (LTi) RORγt+ cells that express LTαß and interact with mesenchymal lymphoid tissue organizer (LTo) FAP+ cells in the presence of lymphatic and blood vessels. A variety of inducer cells initiate TLOs, including bona fide LTi cells, T cells, B cells, and NK cells. The mesenchymal organizer cells are less well characterized but can include FAP+ cells. Current challenges include identification of methods to inhibit TLOs in autoimmunity without affecting SLOs, and enhancement of TLOs for defense against tumors.


Asunto(s)
Inflamación/inmunología , Tejido Linfoide/inmunología , Animales , Humanos , Ganglios Linfáticos
5.
Kidney Int ; 89(1): 113-26, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26398497

RESUMEN

Accumulation of inflammatory cells in different renal compartments is a hallmark of progressive kidney diseases including glomerulonephritis (GN). Lymphotoxin ß receptor (LTßR) signaling is crucial for the formation of lymphoid tissue, and inhibition of LTßR signaling has ameliorated several non-renal inflammatory models. Therefore, we tested whether LTßR signaling could also have a role in renal injury. Renal biopsies from patients with GN were found to express both LTα and LTß ligands, as well as LTßR. The LTßR protein and mRNA were localized to tubular epithelial cells, parietal epithelial cells, crescents, and cells of the glomerular tuft, whereas LTß was found on lymphocytes and tubular epithelial cells. Human tubular epithelial cells, mesangial cells, and mouse parietal epithelial cells expressed both LTα and LTß mRNA upon stimulation with TNF in vitro. Several chemokine mRNAs and proteins were expressed in response to LTßR signaling. Importantly, in a murine lupus model, LTßR blockade improved renal function without the reduction of serum autoantibody titers or glomerular immune complex deposition. Thus, a preclinical mouse model and human studies strongly suggest that LTßR signaling is involved in renal injury and may be a suitable therapeutic target in renal diseases.


Asunto(s)
Glomerulonefritis por IGA/metabolismo , Nefritis Lúpica/metabolismo , Receptor beta de Linfotoxina/antagonistas & inhibidores , Receptor beta de Linfotoxina/metabolismo , ARN Mensajero/análisis , Transducción de Señal , Adulto , Animales , Línea Celular , Quimiocinas/genética , Quimiocinas/metabolismo , Modelos Animales de Enfermedad , Células Epiteliales/química , Células Epiteliales/metabolismo , Femenino , Glomerulonefritis por IGA/genética , Humanos , Inmunoglobulinas/farmacología , Glomérulos Renales/química , Glomérulos Renales/patología , Túbulos Renales/química , Túbulos Renales/metabolismo , Túbulos Renales/patología , Ligandos , Nefritis Lúpica/genética , Linfocitos/química , Receptor beta de Linfotoxina/análisis , Receptor beta de Linfotoxina/genética , Linfotoxina-alfa/análisis , Linfotoxina-alfa/genética , Linfotoxina-alfa/metabolismo , Linfotoxina beta/análisis , Linfotoxina beta/genética , Linfotoxina beta/metabolismo , Masculino , Células Mesangiales/metabolismo , Ratones , Persona de Mediana Edad , Transducción de Señal/efectos de los fármacos , Transcriptoma
6.
PLoS Biol ; 11(10): e1001672, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24130458

RESUMEN

Lymph node (LN) stromal cells provide survival signals and adhesive substrata to lymphocytes. During an immune response, B cell follicles enlarge, questioning how LN stromal cells manage these cellular demands. Herein, we used a murine fate mapping system to describe a new stromal cell type that resides in the T cell zone of resting LNs. We demonstrated that upon inflammation, B cell follicles progressively trespassed into the adjacent T cell zone and surrounded and converted these stromal cells into CXCL13 secreting cells that in return delineated the new boundaries of the growing follicle. Acute B cell ablation in inflamed LNs abolished CXCL13 secretion in these cells, while LT-ß deficiency in B cells drastically affected this conversion. Altogether, we reveal the existence of a dormant stromal cell subset that can be functionally awakened by B cells to delineate the transient boundaries of their expanding territories upon inflammation.


Asunto(s)
Linfocitos B/patología , Inflamación/inmunología , Inflamación/patología , Animales , Quimiocina CXCL13/metabolismo , Células Dendríticas/patología , Fibroblastos/patología , Ganglios Linfáticos/patología , Depleción Linfocítica , Linfocitos/patología , Ratones , Receptores CXCR5/deficiencia , Receptores CXCR5/metabolismo , Receptores de Complemento 3d/metabolismo , Células del Estroma/metabolismo , Células del Estroma/patología , Linfocitos T
7.
J Neuroinflammation ; 11: 10, 2014 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-24444311

RESUMEN

BACKGROUND: Expression of chemokine CCL2 in the normal central nervous system (CNS) is nearly undetectable, but is significantly upregulated and drives neuroinflammation during experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis which is considered a contributing factor in the human disease. As astrocytes and brain microvascular endothelial cells (BMEC) forming the blood-brain barrier (BBB) are sources of CCL2 in EAE and other neuroinflammatory conditions, it is unclear if one or both CCL2 pools are critical to disease and by what mechanism(s). METHODS: Mice with selective CCL2 gene knockout (KO) in astrocytes (Astro KO) or endothelial cells (Endo KO) were used to evaluate the respective contributions of these sources to neuroinflammation, i.e., clinical disease progression, BBB damage, and parenchymal leukocyte invasion in a myelin oligodendrocyte glycoprotein peptide (MOG35-55)-induced EAE model. High-resolution 3-dimensional (3D) immunofluorescence confocal microscopy and colloidal gold immuno-electron microscopy were employed to confirm sites of CCL2 expression, and 3D immunofluorescence confocal microscopy utilized to assess inflammatory responses along the CNS microvasculature. RESULTS: Cell-selective loss of CCL2 immunoreactivity was demonstrated in the respective KO mice. Compared to wild-type (WT) mice, Astro KO mice showed reduced EAE severity but similar onset, while Endo KO mice displayed near normal severity but significantly delayed onset. Neither of the KO mice showed deficits in T cell proliferation, or IL-17 and IFN-γ production, following MOG35-55 exposure in vitro, or altered MOG-major histocompatibility complex class II tetramer binding. 3D confocal imaging further revealed distinct actions of the two CCL2 pools in the CNS. Astro KOs lacked the CNS leukocyte penetration and disrupted immunostaining of CLN-5 at the BBB seen during early EAE in WT mice, while Endo KOs uniquely displayed leukocytes stalled in the microvascular lumen. CONCLUSIONS: These results point to astrocyte and endothelial pools of CCL2 each regulating different stages of neuroinflammation in EAE, and carry implications for drug delivery in neuroinflammatory disease.


Asunto(s)
Astrocitos/patología , Quimiocina CCL2/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Endotelio/patología , Imagenología Tridimensional , Microscopía Confocal , Animales , Sistema Nervioso Central/patología , Quimiocina CCL2/deficiencia , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inducido químicamente , Ratones , Ratones Noqueados , Microvasos/patología , Glicoproteína Mielina-Oligodendrócito , Fragmentos de Péptidos
8.
J Immunol ; 188(12): 6001-9, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22586035

RESUMEN

IFN-γ-inducible lysosomal thiol reductase (GILT) is an enzyme located in the Lamp-2-positive compartments of APC. GILT(-/-) mice are phenotypically normal, but their T cells exhibit reduced proliferation to several exogenously administered Ags that include cysteine residues and disulfide bonds. We undertook the present studies to determine if GILT(-/-) mice would process exogenously administered myelin oligodendrocyte glycoprotein (MOG), which contains disulfide bonds, to generate experimental autoimmune encephalomyelitis (EAE) to the endogenous protein. One possibility was that MOG(35-55) peptide would induce EAE, but that MOG protein would not. GILT(-/-) mice were relatively resistant to MOG(35-55)-induced EAE but slightly more susceptible to rat MOG protein-induced EAE than wild-type (WT) mice. Even though MOG(35-55) was immunogenic in GILT(-/-) mice, GILT APCs could not generate MOG(35-55) from MOG protein in vitro, suggesting that the endogenous MOG protein was not processed to the MOG(35-55) peptide in vivo. Immunization of GILT(-/-) mice with rat MOG protein resulted in a switch in pathogenic mechanism from that seen in WT mice; the CNS infiltrate included large numbers of plasma cells; and GILT(-/-) T cells proliferated to peptides other than MOG(35-55). In contrast to WT rat MOG-immunized mice, rat MOG-immunized GILT(-/-) mice generated Abs that transferred EAE to MOG(35-55)-primed GILT(-/-) mice, and these Abs bound to oligodendrocytes. These studies, demonstrating the key role of a processing enzyme in autoimmunity, indicate that subtle phenotypic changes have profound influences on pathogenic mechanisms and are directly applicable to the outbred human population.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Glicoproteínas/inmunología , Proteínas de la Mielina/inmunología , Oxidorreductasas/inmunología , Fragmentos de Péptidos/inmunología , Secuencia de Aminoácidos , Animales , Células Presentadoras de Antígenos/inmunología , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/metabolismo , Técnica del Anticuerpo Fluorescente , Humanos , Immunoblotting , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Proteínas de la Mielina/metabolismo , Glicoproteína Mielina-Oligodendrócito , Oxidorreductasas/deficiencia , Oxidorreductasas/genética , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro , Ratas , Homología de Secuencia de Aminoácido
9.
Proc Natl Acad Sci U S A ; 108(46): 18784-9, 2011 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-22065738

RESUMEN

To trigger an effective immune response, antigen and antigen-presenting cells travel to the lymph nodes via collecting lymphatic vessels. However, our understanding of the regulation of collecting lymphatic vessel function and lymph transport is limited. To dissect the molecular control of lymphatic function, we developed a unique mouse model that allows intravital imaging of autonomous lymphatic vessel contraction. Using this method, we demonstrated that endothelial nitric oxide synthase (eNOS) in lymphatic endothelial cells is required for robust lymphatic contractions under physiological conditions. By contrast, under inflammatory conditions, inducible NOS (iNOS)-expressing CD11b(+)Gr-1(+) cells attenuate lymphatic contraction. This inhibition of lymphatic contraction was associated with a reduction in the response to antigen in a model of immune-induced multiple sclerosis. These results suggest the suppression of lymphatic function by the CD11b(+)Gr-1(+) cells as a potential mechanism of self-protection from autoreactive responses during on-going inflammation. The central role for nitric oxide also suggests that other diseases such as cancer and infection may also mediate lymphatic contraction and thus immune response. Our unique method allows the study of lymphatic function and its molecular regulation during inflammation, lymphedema, and lymphatic metastasis.


Asunto(s)
Terapia de Inmunosupresión , Sistema Linfático/fisiología , Vasos Linfáticos/efectos de los fármacos , Animales , Células de la Médula Ósea/citología , Antígeno CD11b/biosíntesis , Sistema Inmunológico , Inflamación , Cinética , Metástasis Linfática , Vasos Linfáticos/patología , Ratones , Ratones Endogámicos C57BL , Microscopía/métodos , Óxido Nítrico Sintasa de Tipo III/metabolismo , Oxazolona/farmacología , Piel/efectos de los fármacos
10.
Am J Pathol ; 180(4): 1715-25, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22310467

RESUMEN

Lymphatic vessels (LVs) are important structures for antigen presentation, for lipid metabolism, and as conduits for tumor metastases, but they have been difficult to visualize in vivo. Prox1 is a transcription factor that is necessary for lymphangiogenesis in ontogeny and the maintenance of LVs. To visualize LVs in the lymph node of a living mouse in real time, we made the ProxTom transgenic mouse in a C57BL/6 background using red fluorescent LVs that are suitable for in vivo imaging. The ProxTom transgene contained all Prox1 regulatory sequences and was faithfully expressed in LVs coincident with endogenous Prox1 expression. The progenies of a ProxTom × Hec6stGFP cross were imaged using two-photon laser scanning microscopy, allowing the simultaneous visualization of LVs and high endothelial venules in a lymph node of a living mouse for the first time. We confirmed the expression of Prox1 in the adult liver, lens, and dentate gyrus. These intensely fluorescent mice revealed the expression of Prox1 in three novel sites: the neuroendocrine cells of the adrenal medulla, megakaryocytes, and platelets. The novel sites identified herein suggest previously unknown roles for Prox1. The faithful expression of the fluorescent reporter in ProxTom LVs indicates that these mice have potential utility in the study of diseases as diverse as lymphedema, filariasis, transplant rejection, obesity, and tumor metastasis.


Asunto(s)
Médula Suprarrenal/metabolismo , Plaquetas/metabolismo , Proteínas de Homeodominio/metabolismo , Vasos Linfáticos/metabolismo , Megacariocitos/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Células Cultivadas , Citoplasma/metabolismo , Células Endoteliales/metabolismo , Regulación de la Expresión Génica/fisiología , Genotipo , Glicoproteínas/metabolismo , Proteínas de Homeodominio/genética , Proteínas Luminiscentes/metabolismo , Ganglios Linfáticos/metabolismo , Proteínas de Transporte de Membrana , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Fluorescente , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor/genética , Proteína Fluorescente Roja
11.
FASEB J ; 26(1): 51-62, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21926237

RESUMEN

Tertiary lymphoid organs (TLOs) may develop within allografts, but their contribution to graft rejection remains unclear. Here, we study a mouse model of autoantibody-mediated cardiac allograft vasculopathy to clarify the alloimmune responses mediated by intragraft TLOs and whether blocking lymphotoxin-ß-receptor (LTßR) signaling, a pathway essential for lymphoid organogenesis, abrogates TLO development. TLOs (defined as discrete lymphoid aggregates associated with high endothelial venules) were detectable in 9 of 13 heart allografts studied and were predominantly B cell in composition, harboring germinal-center activity. These are most likely manifestations of the humoral autoimmunity triggered in this model after transplantation; TLOs did not develop if autoantibody production was prevented. Treatment with inhibitory LTßR-Ig fusion protein virtually abolished allograft TLO formation (mean TLOs/heart: 0.2 vs. 2.2 in control recipients; P=0.02), with marked attenuation of the autoantibody response. Recipients primed for autoantibody before transplantation rejected grafts rapidly, but this accelerated rejection was prevented by postoperative administration of LTßR-Ig (median survival time: 18 vs. >50 d, respectively, P=0.003). Our results provide the first demonstration that TLOs develop within chronically rejecting heart allografts, are predominantly B cell in origin, and can be targeted pharmacologically to inhibit effector humoral responses.


Asunto(s)
Coristoma/prevención & control , Trasplante de Corazón/inmunología , Tejido Linfoide/patología , Receptor beta de Linfotoxina/metabolismo , Linfotoxina beta/metabolismo , Transducción de Señal/inmunología , Animales , Linfocitos B/inmunología , Médula Ósea/inmunología , Médula Ósea/patología , Linfocitos T CD4-Positivos/inmunología , Coristoma/inmunología , Coristoma/patología , Enfermedad Crónica , Rechazo de Injerto/inmunología , Isoanticuerpos/inmunología , Tejido Linfoide/irrigación sanguínea , Tejido Linfoide/inmunología , Receptor beta de Linfotoxina/genética , Receptor beta de Linfotoxina/inmunología , Linfotoxina beta/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/inmunología , Miocardio/patología , Neovascularización Patológica/inmunología , Neovascularización Patológica/patología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/metabolismo , Bazo/inmunología , Bazo/patología , Trasplante Homólogo
12.
Transplantation ; 2023 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-37917987

RESUMEN

Tertiary lymphoid organs (TLOs), also known as tertiary or ectopic lymphoid structures or tissues, are accumulations of lymphoid cells in sites other than canonical lymphoid organs, that arise through lymphoid neogenesis during chronic inflammation in autoimmunity, microbial infection, cancer, aging, and transplantation, the focus of this review. Lymph nodes and TLOs are compared regarding their cellular composition, organization, vascular components, and migratory signal regulation. These characteristics of posttransplant TLOs (PT-TLOs) are described with individual examples in a wide range of organs including heart, kidney, trachea, lung, artery, skin, leg, hand, and face, in many species including human, mouse, rat, and monkey. The requirements for induction and maintenance of TLOs include sustained exposure to autoantigens, alloantigens, tumor antigens, ischemic reperfusion, nephrotoxic agents, and aging. Several staging schemes have been put forth regarding their function in organ rejection. PT-TLOs most often are associated with organ rejection, but in some cases contribute to tolerance. The role of PT-TLOs in cancer is considered in the case of immunosuppression. Furthermore, TLOs can be associated with development of lymphomas. Challenges for PT-TLO research are considered regarding staging, imaging, and opportunities for their therapeutic manipulation to inhibit rejection and encourage tolerance.

13.
Curr Opin Physiol ; 362023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38523879

RESUMEN

High endothelial venules (HEVs), high walled cuboidal blood vessels, through their expression of adhesion molecules and chemokines, allow the entrance of lymphoid cells into primary, secondary, and tertiary lymphoid structures (aka tertiary lymphoid organs). HEV heterogeneity exists between various lymphoid organs in their expression of peripheral node addressin (PNAd) and mucosal vascular addressin adhesion molecule 1(MAdCAM-1). Transcriptomic analyses reveal extensive heterogeneity, plasticity, and regulation of HEV gene expression in ontogeny, acute inflammation, and chronic inflammation within and between lymphoid organs. Rules regulating HEV development are flexible in inflammation. HEVs in tumor tertiary lymphoid structures are diagnostic of favorable clinical outcome and response to Immunotherapy, including immune check point blockade. Immunotherapy induces HEVs and provides an entrance for naïve, central memory, and effector cells and a niche for stem like precursor cells. Understanding HEV regulation will permit their exploitation as routes for drug delivery to autoimmune lesions, rejecting organs, and tumors.

14.
Blood ; 116(12): 2173-82, 2010 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-20566898

RESUMEN

Lymphotoxin-α (LTα), lymphotoxin-ß (LTß), and tumor necrosis factor-α (TNFα) are inflammatory mediators that play crucial roles in lymphoid organ development. We demonstrate here that LTα also contributes to the function of lymphatic vessels and to lymphangiogenesis during inflammation. LTα(-/-) mice exhibited reduced lymph flow velocities and increased interstitial fluid pressure. Airways of LTß(-/-) mice infected with Mycoplasma pulmonis had significantly more lymphangiogenesis than wild type (WT) or LTα(-/-) mice, as did the skin draining immunization sites of LTß(-/-) mice. Macrophages, B cells, and T cells, known sources of LT and TNFα, were apparent in the skin surrounding the immunization sites as were LTα, LTß, and TNFα mRNAs. Ectopic expression of LTα led to the development of LYVE-1 and Prox1-positive lymphatic vessels within tertiary lymphoid organs (TLOs). Quantification of pancreatic lymphatic vessel density in RIPLTαLTß(-/-) and WT mice revealed that LTα was sufficient for inducing lymphangiogenesis and that LTß was not required for this process. Kidneys of inducible LTα transgenic mice developed lymphatic vessels before the appearance of obvious TLOs. These data indicate that LTα plays a significant role in lymphatic vessel function and in inflammation-associated lymphangiogenesis.


Asunto(s)
Linfangiogénesis , Linfotoxina-alfa/fisiología , Animales , Inmunización , Inflamación , Riñón/inmunología , Vasos Linfáticos , Linfotoxina-alfa/deficiencia , Linfotoxina-alfa/genética , Linfotoxina beta , Ratones , Ratones Noqueados , Infecciones por Mycoplasma/patología , Mycoplasma pulmonis , Piel
15.
Acta Neuropathol ; 124(6): 861-73, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22842876

RESUMEN

While the role of T cells has been studied extensively in multiple sclerosis (MS), the pathogenic contribution of B cells has only recently attracted major attention, when it was shown that B cell aggregates can develop in the meninges of a subset of MS patients and were suggested to be correlates of late-stage and more aggressive disease in this patient population. However, whether these aggregates actually exist has subsequently been questioned and their functional significance has remained unclear. Here, we studied myelin basic protein (MBP)-proteolipid protein (PLP)-induced experimental autoimmune encephalomyelitis (EAE), which is one of the few animal models for MS that is dependent on B cells. We provide evidence that B cell aggregation is reflective of lymphoid neogenesis in the central nervous system (CNS) in MBP-PLP-elicited EAE. B cell aggregation was present already few days after disease onset. With disease progression CNS B cell aggregates increasingly displayed the phenotype of tertiary lymphoid organs (TLOs). Our results further imply that these TLOs were not merely epiphenomena of the disease, but functionally active, supporting intrathecal determinant spreading of the myelin-specific T cell response. Our data suggest that the CNS is not a passive "immune-privileged" target organ, but rather a compartment, in which highly active immune responses can perpetuate and amplify the autoimmune pathology and thereby autonomously contribute to disease progression.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Tejido Linfoide/inmunología , Proteína Proteolipídica de la Mielina/metabolismo , Vaina de Mielina/inmunología , Linfocitos T/inmunología , Animales , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/patología , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/patología , Femenino , Ratones , Ratones Endogámicos C57BL , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/patología , Proteína Básica de Mielina/inmunología , Proteína Básica de Mielina/metabolismo , Proteína Proteolipídica de la Mielina/inmunología , Vaina de Mielina/patología , Linfocitos T/patología
16.
J Immunol ; 183(4): 2205-12, 2009 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-19661265

RESUMEN

Secondary lymphoid organs (SLOs) include lymph nodes, spleen, Peyer's patches, and mucosal tissues such as the nasal-associated lymphoid tissue, adenoids, and tonsils. Less discretely anatomically defined cellular accumulations include the bronchus-associated lymphoid tissue, cryptopatches, and isolated lymphoid follicles. All SLOs serve to generate immune responses and tolerance. SLO development depends on the precisely regulated expression of cooperating lymphoid chemokines and cytokines such as LTalpha, LTbeta, RANKL, TNF, IL-7, and perhaps IL-17. The relative importance of these factors varies between the individual lymphoid organs. Participating in the process are lymphoid tissue initiator, lymphoid tissue inducer, and lymphoid tissue organizer cells. These cells and others that produce crucial cytokines maintain SLOs in the adult. Similar signals regulate the transition from inflammation to ectopic or tertiary lymphoid tissues.


Asunto(s)
Compartimento Celular/genética , Compartimento Celular/inmunología , Linfangiogénesis/genética , Linfangiogénesis/inmunología , Tejido Linfoide/inmunología , Tejido Linfoide/metabolismo , Animales , Humanos , Tejido Linfoide/citología , Tejido Linfoide/crecimiento & desarrollo
18.
Lancet Microbe ; 2(12): e666-e675, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34632431

RESUMEN

BACKGROUND: Among the most consequential unknowns of the devastating COVID-19 pandemic are the durability of immunity and time to likely reinfection. There are limited direct data on SARS-CoV-2 long-term immune responses and reinfection. The aim of this study is to use data on the durability of immunity among evolutionarily close coronavirus relatives of SARS-CoV-2 to estimate times to reinfection by a comparative evolutionary analysis of related viruses SARS-CoV, MERS-CoV, human coronavirus (HCoV)-229E, HCoV-OC43, and HCoV-NL63. METHODS: We conducted phylogenetic analyses of the S, M, and ORF1b genes to reconstruct a maximum-likelihood molecular phylogeny of human-infecting coronaviruses. This phylogeny enabled comparative analyses of peak-normalised nucleocapsid protein, spike protein, and whole-virus lysate IgG antibody optical density levels, in conjunction with reinfection data on endemic human-infecting coronaviruses. We performed ancestral and descendent states analyses to estimate the expected declines in antibody levels over time, the probabilities of reinfection based on antibody level, and the anticipated times to reinfection after recovery under conditions of endemic transmission for SARS-CoV-2, as well as the other human-infecting coronaviruses. FINDINGS: We obtained antibody optical density data for six human-infecting coronaviruses, extending from 128 days to 28 years after infection between 1984 and 2020. These data provided a means to estimate profiles of the typical antibody decline and probabilities of reinfection over time under endemic conditions. Reinfection by SARS-CoV-2 under endemic conditions would likely occur between 3 months and 5·1 years after peak antibody response, with a median of 16 months. This protection is less than half the duration revealed for the endemic coronaviruses circulating among humans (5-95% quantiles 15 months to 10 years for HCoV-OC43, 31 months to 12 years for HCoV-NL63, and 16 months to 12 years for HCoV-229E). For SARS-CoV, the 5-95% quantiles were 4 months to 6 years, whereas the 95% quantiles for MERS-CoV were inconsistent by dataset. INTERPRETATION: The timeframe for reinfection is fundamental to numerous aspects of public health decision making. As the COVID-19 pandemic continues, reinfection is likely to become increasingly common. Maintaining public health measures that curb transmission-including among individuals who were previously infected with SARS-CoV-2-coupled with persistent efforts to accelerate vaccination worldwide is critical to the prevention of COVID-19 morbidity and mortality. FUNDING: US National Science Foundation.


Asunto(s)
COVID-19 , Coronavirus Humano 229E , Coronavirus Humano NL63 , Coronavirus Humano OC43 , Coronavirus del Síndrome Respiratorio de Oriente Medio , Anticuerpos Antivirales/genética , COVID-19/epidemiología , Reacciones Cruzadas , Humanos , Pandemias , Filogenia , Reinfección/epidemiología , SARS-CoV-2/genética , Glicoproteína de la Espiga del Coronavirus/genética
19.
Glia ; 58(13): 1523-31, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20645414

RESUMEN

Various models of experimental autoimmune encephalomyelitis (EAE) have led to insights into the pathogenesis and novel therapies for multiple sclerosis. One generalized EAE model uses immunizing the Lewis Rat with myelin oligodendrocyte glycoprotein (MOG) and complete Freund's adjuvant that induces systemic disease and inflammatory lesions at random central nervous system (CNS) locations. These lesions result from a combination of sensitized T cells and pathogenic antibodies gaining access to the CNS to cause an immune assault on myelin-expressing oligodendrocytes. We report a focal and temporal variant of the EAE model that results in immune-mediated demyelination at a predictable time and location. Lewis rats were immunized with the extracellular domain (1-125) of recombinant rat MOG in incomplete Freund's adjuvant (IFA) to induce a clinically silent humoral response. Vascular endothelial growth factor (VEGF) was then microinjected into the spinal cord to induce a transient, focal breakdown of the blood brain barrier (BBB). Clinical signs were apparent within 72 hours and began to resolve by day 21. The histopathology at the site of injection consisted of a focal region containing OX-42(+) cells, phagocytic cells with debris, extensive demyelination, and some lymphocyte infiltration. Neither intraspinal injection of PBS into immunized animals nor VEGF into animals treated with IFA alone resulted in clinical lesions. Thus, a transient, focal opening of the BBB with VEGF in animals with subclinical MOG immunization leads to a discrete inflammatory demyelinating lesion. This model may be useful for the study of transplanted myelin-forming cells in a discrete inflammatory demyelinating lesion.


Asunto(s)
Encefalomielitis Autoinmune Experimental/etiología , Encefalomielitis Autoinmune Experimental/inmunología , Glicoproteína Asociada a Mielina/inmunología , Factor A de Crecimiento Endotelial Vascular/efectos adversos , Análisis de Varianza , Animales , Anticuerpos/metabolismo , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/fisiopatología , Complejo CD3/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/patología , Ensayo de Inmunoadsorción Enzimática/métodos , Femenino , Adyuvante de Freund/efectos adversos , Inyecciones Espinales/métodos , Lípidos/efectos adversos , Microscopía Electrónica de Transmisión/métodos , Proteínas de la Mielina , Glicoproteína Mielina-Oligodendrócito , Ratas , Ratas Endogámicas Lew , Médula Espinal/patología , Médula Espinal/ultraestructura , Factores de Tiempo
20.
J Exp Med ; 197(9): 1153-63, 2003 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-12732657

RESUMEN

Lymph node (LN) function depends on T and B cell compartmentalization, antigen presenting cells, and high endothelial venules (HEVs) expressing mucosal addressin cell adhesion molecule (MAdCAM-1) and peripheral node addressin (PNAd), ligands for naive cell entrance into LNs. Luminal PNAd expression requires a HEV-restricted sulfotransferase (HEC-6ST). To investigate LT alpha beta's activities in lymphoid organogenesis, mice simultaneously expressing LT alpha and LT beta under rat insulin promoter II (RIP) control were compared with RIPLT alpha mice in a model of lymphoid neogenesis and with LT beta-/- mice. RIPLT alpha beta pancreata exhibited massive intra-islet mononuclear infiltrates that differed from the more sparse peri-islet cell accumulations in RIPLT alpha pancreata: separation into T and B cell areas was more distinct with prominent FDC networks, expression of lymphoid chemokines (CCL21, CCL19, and CXCL13) was more intense, and L-selectin+ cells were more frequent. In contrast to the predominant abluminal PNAd pattern of HEV in LT beta-/- MLN and RIPLT alpha pancreatic infiltrates, PNAd was expressed at the luminal and abluminal aspects of HEV in wild-type LN and in RIPLT alpha beta pancreata, coincident with HEC-6ST. These data highlight distinct roles of LT alpha and LT alpha beta in lymphoid organogenesis supporting the notion that HEC-6ST-dependent luminal PNAd is under regulation by LT alpha beta.


Asunto(s)
Tejido Linfoide/embriología , Linfotoxina-alfa/fisiología , Proteínas de la Membrana/fisiología , Sulfotransferasas/metabolismo , Animales , Inducción Enzimática , Inmunohistoquímica , Linfotoxina beta , Ratones , Ratones Transgénicos , Sulfotransferasas/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA