Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros











Intervalo de año de publicación
1.
Front Immunol ; 13: 1002629, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36439150

RESUMEN

Immune mediated inflammatory diseases (IMIDs) are a heterogeneous group of debilitating, multifactorial and unrelated conditions featured by a dysregulated immune response leading to destructive chronic inflammation. The immune dysregulation can affect various organ systems: gut (e.g., inflammatory bowel disease), joints (e.g., rheumatoid arthritis), skin (e.g., psoriasis, atopic dermatitis), resulting in significant morbidity, reduced quality of life, increased risk for comorbidities, and premature death. As there are no reliable disease progression and therapy response biomarkers currently available, it is very hard to predict how the disease will develop and which treatments will be effective in a given patient. In addition, a considerable proportion of patients do not respond sufficiently to the treatment. ImmUniverse is a large collaborative consortium of 27 partners funded by the Innovative Medicine Initiative (IMI), which is sponsored by the European Union (Horizon 2020) and in-kind contributions of participating pharmaceutical companies within the European Federation of Pharmaceutical Industries and Associations (EFPIA). ImmUniverse aims to advance our understanding of the molecular mechanisms underlying two immune-mediated diseases, ulcerative colitis (UC) and atopic dermatitis (AD), by pursuing an integrative multi-omics approach. As a consequence of the heterogeneity among IMIDs patients, a comprehensive, evidence-based identification of novel biomarkers is necessary to enable appropriate patient stratification that would account for the inter-individual differences in disease severity, drug efficacy, side effects or prognosis. This would guide clinicians in the management of patients and represent a major step towards personalized medicine. ImmUniverse will combine the existing and novel advanced technologies, including multi-omics, to characterize both the tissue microenvironment and blood. This comprehensive, systems biology-oriented approach will allow for identification and validation of tissue and circulating biomarker signatures as well as mechanistic principles, which will provide information about disease severity and future disease progression. This truly makes the ImmUniverse Consortium an unparalleled approach.


Asunto(s)
Dermatitis Atópica , Medicina de Precisión , Humanos , Calidad de Vida , Biomarcadores , Progresión de la Enfermedad
2.
Nat Commun ; 8(1): 1576, 2017 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-29146903

RESUMEN

Dysregulation of autophagy and inflammasome activity contributes to the development of auto-inflammatory diseases. Emerging evidence highlights the importance of the actin cytoskeleton in modulating inflammatory responses. Here we show that deficiency of Wiskott-Aldrich syndrome protein (WASp), which signals to the actin cytoskeleton, modulates autophagy and inflammasome function. In a model of sterile inflammation utilizing TLR4 ligation followed by ATP or nigericin treatment, inflammasome activation is enhanced in monocytes from WAS patients and in WAS-knockout mouse dendritic cells. In ex vivo models of enteropathogenic Escherichia coli and Shigella flexneri infection, WASp deficiency causes defective bacterial clearance, excessive inflammasome activation and host cell death that are associated with dysregulated septin cage-like formation, impaired autophagic p62/LC3 recruitment and defective formation of canonical autophagosomes. Taken together, we propose that dysregulation of autophagy and inflammasome activities contribute to the autoinflammatory manifestations of WAS, thereby identifying potential targets for therapeutic intervention.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Autofagia/inmunología , Inflamasomas/inmunología , Proteína del Síndrome de Wiskott-Aldrich/genética , Proteína del Síndrome de Wiskott-Aldrich/metabolismo , Síndrome de Wiskott-Aldrich/inmunología , Animales , Autofagia/genética , Carga Bacteriana/inmunología , Línea Celular Tumoral , Células Dendríticas/inmunología , Escherichia coli Enteropatógena/inmunología , Humanos , Inmunidad Innata/inmunología , Interferón Tipo I/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Nigericina/farmacología , Septinas/metabolismo , Shigella flexneri/inmunología , Células THP-1 , Receptor Toll-Like 4/inmunología , Síndrome de Wiskott-Aldrich/metabolismo
3.
J Leukoc Biol ; 99(5): 699-710, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26590149

RESUMEN

The immunological synapse is a highly structured and molecularly dynamic interface between communicating immune cells. Although the immunological synapse promotes T cell activation by dendritic cells, the specific organization of the immunological synapse on the dendritic cell side in response to T cell engagement is largely unknown. In this study, confocal and electron microscopy techniques were used to investigate the role of dendritic cell actin regulation in immunological synapse formation, stabilization, and function. In the dendritic cell-restricted absence of the Wiskott-Aldrich syndrome protein, an important regulator of the actin cytoskeleton in hematopoietic cells, the immunological synapse contact with T cells occupied a significantly reduced surface area. At a molecular level, the actin network localized to the immunological synapse exhibited reduced stability, in particular, of the actin-related protein-2/3-dependent, short-filament network. This was associated with decreased polarization of dendritic cell-associated ICAM-1 and MHC class II, which was partially dependent on Wiskott-Aldrich syndrome protein phosphorylation. With the use of supported planar lipid bilayers incorporating anti-ICAM-1 and anti-MHC class II antibodies, the dendritic cell actin cytoskeleton organized into recognizable synaptic structures but interestingly, formed Wiskott-Aldrich syndrome protein-dependent podosomes within this area. These findings demonstrate that intrinsic dendritic cell cytoskeletal remodeling is a key regulatory component of normal immunological synapse formation, likely through consolidation of adhesive interaction and modulation of immunological synapse stability.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Comunicación Celular/inmunología , Células Dendríticas/inmunología , Sinapsis Inmunológicas/metabolismo , Linfocitos T/citología , Linfocitos T/inmunología , Proteína del Síndrome de Wiskott-Aldrich/metabolismo , Animales , Recuperación de Fluorescencia tras Fotoblanqueo , Molécula 1 de Adhesión Intercelular/metabolismo , Membrana Dobles de Lípidos/metabolismo , Activación de Linfocitos/inmunología , Ratones Endogámicos C57BL , Podosomas/metabolismo
4.
Blood ; 126(13): 1527-35, 2015 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-26224645

RESUMEN

Megakaryoblastic leukemia 1 (MKL1), also known as MAL or myocardin-related transcription factor A (MRTF-A), is a coactivator of serum response factor, which regulates transcription of actin and actin cytoskeleton-related genes. MKL1 is known to be important for megakaryocyte differentiation and function in mice, but its role in immune cells is unexplored. Here we report a patient with a homozygous nonsense mutation in the MKL1 gene resulting in immunodeficiency characterized predominantly by susceptibility to severe bacterial infection. We show that loss of MKL1 protein expression causes a dramatic loss of filamentous actin (F-actin) content in lymphoid and myeloid lineage immune cells and widespread cytoskeletal dysfunction. MKL1-deficient neutrophils displayed reduced phagocytosis and almost complete abrogation of migration in vitro. Similarly, primary dendritic cells were unable to spread normally or to form podosomes. Silencing of MKL1 in myeloid cell lines revealed that F-actin assembly was abrogated through reduction of globular actin (G-actin) levels and disturbed expression of multiple actin-regulating genes. Impaired migration of these cells was associated with failure of uropod retraction likely due to altered contractility and adhesion, evidenced by reduced expression of the myosin light chain 9 (MYL9) component of myosin II complex and overexpression of CD11b integrin. Together, our results show that MKL1 is a nonredundant regulator of cytoskeleton-associated functions in immune cells and fibroblasts and that its depletion underlies a novel human primary immunodeficiency.


Asunto(s)
Codón sin Sentido , Síndromes de Inmunodeficiencia/genética , Infecciones por Pseudomonas/genética , Transactivadores/genética , Actinas/metabolismo , Actinas/ultraestructura , Línea Celular , Movimiento Celular , Células Cultivadas , Citoesqueleto/metabolismo , Citoesqueleto/ultraestructura , Células Dendríticas/citología , Células Dendríticas/metabolismo , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Homocigoto , Humanos , Síndromes de Inmunodeficiencia/complicaciones , Síndromes de Inmunodeficiencia/diagnóstico , Síndromes de Inmunodeficiencia/metabolismo , Neutrófilos/citología , Neutrófilos/metabolismo , Pseudomonas/aislamiento & purificación , Infecciones por Pseudomonas/complicaciones , Infecciones por Pseudomonas/diagnóstico , Infecciones por Pseudomonas/metabolismo
5.
Eur J Immunol ; 44(9): 2692-702, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24945741

RESUMEN

Patients deficient in the cytoskeletal regulator Wiskott-Aldrich syndrome protein (WASp) are predisposed to varied autoimmunity, suggesting it has an important controlling role in participating cells. IL-10-producing regulatory B (Breg) cells are emerging as important mediators of immunosuppressive activity. In experimental, antigen-induced arthritis WASp-deficient (WASp knockout [WAS KO]) mice developed exacerbated disease associated with decreased Breg cells and regulatory T (Treg) cells, but increased Th17 cells in knee-draining LNs. Arthritic WAS KO mice showed increased serum levels of B-cell-activating factor, while their B cells were unresponsive in terms of B-cell-activating factor induced survival and IL-10 production. Adoptive transfer of WT Breg cells ameliorated arthritis in WAS KO recipients and restored a normal balance of Treg and Th17 cells. Mice with B-cell-restricted WASp deficiency, however, did not develop exacerbated arthritis, despite exhibiting reduced Breg- and Treg-cell numbers during active disease, and Th17 cells were not increased over equivalent WT levels. These findings support a contributory role for defective Breg cells in the development of WAS-related autoimmunity, but demonstrate that functional competence in other regulatory populations can be compensatory. A properly regulated cytoskeleton is therefore important for normal Breg-cell activity and complementation of defects in this lineage is likely to have important therapeutic benefits.


Asunto(s)
Artritis Experimental/inmunología , Subgrupos de Linfocitos B/inmunología , Proteína del Síndrome de Wiskott-Aldrich/inmunología , Animales , Artritis Experimental/genética , Artritis Experimental/patología , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Subgrupos de Linfocitos B/patología , Femenino , Humanos , Interleucina-10/genética , Interleucina-10/inmunología , Masculino , Ratones , Ratones Noqueados , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología , Células Th17/inmunología , Células Th17/patología , Proteína del Síndrome de Wiskott-Aldrich/genética
6.
Blood ; 121(1): 72-84, 2013 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-23160469

RESUMEN

Wiskott Aldrich syndrome (WAS), an X-linked immunodeficiency, results from loss-of-function mutations in the human hematopoietic cytoskeletal regulator gene WAS. Many missense mutations in the Ena Vasp homology1 (EVH1) domain preserve low-level WAS protein (WASp) expression and confer a milder clinical phenotype. Although disrupted binding to WASp-interacting protein (WIP) leads to enhanced WASp degradation in vivo, the intrinsic function of EVH1-mutated WASp is poorly understood. In the present study, we show that, despite mediating enhanced actin polymerization compared with wild-type WASp in vitro, EVH1 missense mutated proteins did not support full biologic function in cells, even when levels were restored by forced overexpression. Podosome assembly was aberrant and associated with dysregulated lamellipodia formation and impaired persistence of migration. At sites of residual podosome-associated actin polymerization, localization of EVH1-mutated proteins was preserved even after deletion of the entire domain, implying that WIP-WASp complex formation is not absolutely required for WASp localization. However, retention of mutant proteins in podosomes was significantly impaired and associated with reduced levels of WASp tyrosine phosphorylation. Our results indicate that the EVH1 domain is important not only for WASp stability, but also for intrinsic biologic activity in vivo.


Asunto(s)
Células Dendríticas/patología , Mutación Missense , Proteína del Síndrome de Wiskott-Aldrich/genética , Actinas/metabolismo , Animales , Biopolímeros , Proteínas Portadoras/metabolismo , Movimiento Celular , Células Cultivadas , Proteínas del Citoesqueleto , Células Dendríticas/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Fosforilación , Polimerizacion , Mapeo de Interacción de Proteínas , Procesamiento Proteico-Postraduccional , Estabilidad Proteica , Estructura Terciaria de Proteína , Seudópodos/patología , Proteínas Recombinantes de Fusión/fisiología , Eliminación de Secuencia , Organismos Libres de Patógenos Específicos , Proteína del Síndrome de Wiskott-Aldrich/química , Proteína del Síndrome de Wiskott-Aldrich/deficiencia , Proteína del Síndrome de Wiskott-Aldrich/metabolismo , Proteína del Síndrome de Wiskott-Aldrich/fisiología
7.
Blood ; 119(12): 2819-28, 2012 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-22302739

RESUMEN

Wiskott Aldrich syndrome (WAS) is caused by mutations in the WAS gene that encodes for a protein (WASp) involved in cytoskeleton organization in hematopoietic cells. Several distinctive abnormalities of T, B, and natural killer lymphocytes; dendritic cells; and phagocytes have been found in WASp-deficient patients and mice; however, the in vivo consequence of WASp deficiency within individual blood cell lineages has not been definitively evaluated. By conditional gene deletion we have generated mice with selective deficiency of WASp in the B-cell lineage (B/WcKO mice). We show that this is sufficient to cause a severe reduction of marginal zone B cells and inability to respond to type II T-independent Ags, thereby recapitulating phenotypic features of complete WASp deficiency. In addition, B/WcKO mice showed prominent signs of B-cell dysregulation, as indicated by an increase in serum IgM levels, expansion of germinal center B cells and plasma cells, and elevated autoantibody production. These findings are accompanied by hyperproliferation of WASp-deficient follicular and germinal center B cells in heterozygous B/WcKO mice in vivo and excessive differentiation of WASp-deficient B cells into class-switched plasmablasts in vitro, suggesting that WASp-dependent B cell-intrinsic mechanisms critically contribute to WAS-associated autoimmunity.


Asunto(s)
Linfocitos B/citología , Linfocitos B/inmunología , Proteína del Síndrome de Wiskott-Aldrich/inmunología , Animales , Autoanticuerpos/sangre , Autoanticuerpos/inmunología , Autoantígenos/inmunología , Recuento de Células , Modelos Animales de Enfermedad , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína del Síndrome de Wiskott-Aldrich/deficiencia , Proteína del Síndrome de Wiskott-Aldrich/genética
8.
Blood ; 118(9): 2492-501, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21690559

RESUMEN

Rearrangement of the cytoskeleton in T cells plays a critical role in the organization of a complex signaling interface referred to as immunologic synapse (IS). Surprisingly, the contribution of antigen presenting cells, in particular dendritic cells (DCs), to the structure and function of the IS has not been investigated in as much detail. We have used a natural model of cytoskeletal dysfunction caused by deficiency of the Wiskott-Aldrich syndrome protein (WASp) to explore the contribution of the DC cytoskeleton to IS formation and to T-cell priming. In an antigen-specific system, T-DC contacts were found to be less stable when DCs alone lacked WASp, and associated with multiple defects of IS structure. As a consequence, DCs were unable to support normal IL-12 secretion, and events downstream of TCR signaling were abrogated, including increased calcium flux, microtubule organizing center (MTOC) polarization, phosphorylation of ZAP-70, and T-cell proliferation. Formation of an effective signaling interface is therefore dependent on active cytoskeletal rearrangements in DCs even when T cells are functionally competent. Deficiency of DC-mediated activities may contribute significantly to the varied immunodysregulation observed in patients with WAS, and also in those with limited myeloid reconstitution after allogeneic hematopoietic stem cell transplantation.


Asunto(s)
Citoesqueleto/ultraestructura , Células Dendríticas/ultraestructura , Sinapsis Inmunológicas/ultraestructura , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T/inmunología , Proteína del Síndrome de Wiskott-Aldrich/fisiología , Animales , Proteínas Bacterianas/análisis , Proteínas Bacterianas/genética , Señalización del Calcio/inmunología , Movimiento Celular , Cruzamientos Genéticos , Genes Reporteros , Prueba de Complementación Genética , Humanos , Sinapsis Inmunológicas/inmunología , Proteínas Luminiscentes/análisis , Proteínas Luminiscentes/genética , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Ovalbúmina/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/fisiología , Linfocitos T/ultraestructura , Síndrome de Wiskott-Aldrich/genética , Síndrome de Wiskott-Aldrich/patología , Proteína del Síndrome de Wiskott-Aldrich/deficiencia , Proteína del Síndrome de Wiskott-Aldrich/genética , Proteína del Síndrome de Wiskott-Aldrich/inmunología
9.
Dis Markers ; 29(3-4): 157-75, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21178275

RESUMEN

Wiskott-Aldrich syndrome (WAS) is a rare X-linked recessive primary immunodeficiency characterised by immune dysregulation, microthrombocytopaenia, eczema and lymphoid malignancies. Mutations in the WAS gene can lead to distinct syndrome variations which largely, although not exclusively, depend upon the mutation. Premature termination and deletions abrogate Wiskott-Aldrich syndrome protein (WASp) expression and lead to severe disease (WAS). Missense mutations usually result in reduced protein expression and the phenotypically milder X-linked thrombocytopenia (XLT) or attenuated WAS [1-3]. More recently however novel activating mutations have been described that give rise to X-linked neutropenia (XLN), a third syndrome defined by neutropenia with variable myelodysplasia [4-6]. WASP is key in transducing signals from the cell surface to the actin cytoskeleton, and a lack of WASp results in cytoskeletal defects that compromise multiple aspects of normal cellular activity including proliferation, phagocytosis, immune synapse formation, adhesion and directed migration.


Asunto(s)
Proteína del Síndrome de Wiskott-Aldrich/genética , Proteína del Síndrome de Wiskott-Aldrich/inmunología , Síndrome de Wiskott-Aldrich/genética , Síndrome de Wiskott-Aldrich/inmunología , Actinas/metabolismo , Animales , Autoinmunidad , Citoesqueleto/metabolismo , Citoesqueleto/patología , Eccema/genética , Eccema/patología , Femenino , Genes Ligados a X , Humanos , Ratones , Ratones Noqueados , Mutación/inmunología , Estructura Terciaria de Proteína , Transducción de Señal/genética , Trombocitopenia/genética , Trombocitopenia/patología , Transcripción Genética , Síndrome de Wiskott-Aldrich/metabolismo , Síndrome de Wiskott-Aldrich/patología , Síndrome de Wiskott-Aldrich/terapia , Proteína del Síndrome de Wiskott-Aldrich/metabolismo , Proteína de Unión al GTP cdc42/metabolismo
10.
Proc Natl Acad Sci U S A ; 106(37): 15738-43, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19805221

RESUMEN

The Wiskott-Aldrich syndrome protein (WASp) is a key cytoskeletal regulator in hematopoietic cells. Covalent modification of a conserved tyrosine by phosphorylation has emerged as an important potential determinant of activity, although the physiological significance remains uncertain. In a murine knockin model, mutation resulting in inability to phosphorylate Y293 (Y293F) mimicked many features of complete WASp-deficiency. Although a phosphomimicking mutant Y293E conferred enhanced actin-polymerization, the cellular phenotype was similar due to functional dysregulation. Furthermore, steady-state levels of Y293E-WASp were markedly reduced compared to wild-type WASp and Y293F-WASp, although partially recoverable by treatment of cells with proteasome inhibitors. Consequently, tyrosine phosphorylation plays a critical role in normal activation of WASp in vivo, and is indispensible for multiple tasks including proliferation, phagocytosis, chemotaxis, and assembly of adhesion structures. Furthermore, it may target WASp for proteasome-mediated degradation, thereby providing a default mechanism for self-limiting stimulation of the Arp2/3 complex.


Asunto(s)
Proteína del Síndrome de Wiskott-Aldrich/metabolismo , Actinas/metabolismo , Sustitución de Aminoácidos , Animales , Sitios de Unión/genética , Células COS , Línea Celular , Movimiento Celular , Chlorocebus aethiops , Hematopoyesis , Humanos , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Mutagénesis Sitio-Dirigida , Fagocitosis , Fosforilación , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Tirosina/química , Síndrome de Wiskott-Aldrich/genética , Síndrome de Wiskott-Aldrich/metabolismo , Síndrome de Wiskott-Aldrich/patología , Proteína del Síndrome de Wiskott-Aldrich/química , Proteína del Síndrome de Wiskott-Aldrich/genética
11.
Cancer Res ; 69(16): 6598-606, 2009 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-19654308

RESUMEN

The CD11c(int) B220(+) NK1.1(+) CD49(+) subset of cells has recently been described as IFN-producing killer dendritic cells (IKDC), which share phenotypic and functional properties with both dendritic cells and natural killer cells. We have previously shown that IKDCs within murine bone marrow-derived DC preparations are essential for the antitumor activity of unpulsed DCs. Here we show that bone marrow-derived IKDCs (BM-IKDC) migrate in vivo into tumors and thence to tumor draining lymph nodes, where they highly express MHC class II and costimulatory molecules. In vitro, freshly isolated BM-IKDCs, fluorescence-activated cell sorted to homogeneity, have no intrinsic antigen presentation function unless cocultured with tumor target cells. On killing of target cells, they can cross-present antigens to stimulate antigen-primed CD8 T cells and can also present antigens to antigen-primed CD4 cells. In vivo, in mice lacking class I-restricted antigen-presenting cell function, robust proliferation of antigen-specific T cells is achieved after adoptive transfer of BM-IKDCs at an injection site distant to the tumor site. Therefore, BM-IKDCs are capable of cytotoxic killing of tumor targets and also of potent antigen presentation after encountering antigen in the context of a viable target cell.


Asunto(s)
Presentación de Antígeno/fisiología , Movimiento Celular , Células Dendríticas/inmunología , Células Dendríticas/fisiología , Interferones/metabolismo , Animales , Movimiento Celular/inmunología , Células Cultivadas , Citotoxicidad Inmunológica/fisiología , Células Dendríticas/metabolismo , Genes RAG-1 , Melanoma Experimental/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Células 3T3 NIH , Microglobulina beta-2/genética
12.
Immunobiology ; 214(9-10): 778-90, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19628299

RESUMEN

Regulation of the actin cytoskeleton is crucial for many aspects of correct and cooperative functioning of immune cells, such as migration, antigen uptake and cell activation. The Wiskott-Aldrich Syndrome protein (WASp) is an important regulator of actin cytoskeletal rearrangements and lack of this protein results in impaired immune function. This review discusses recent new insights of the role of WASp at molecular and cellular level and evaluates how WASp deficiency affects important immunological features and how defective immune cell function contributes to compromised host defence.


Asunto(s)
Proteína del Síndrome de Wiskott-Aldrich/deficiencia , Síndrome de Wiskott-Aldrich/inmunología , Actinas/metabolismo , Animales , Movimiento Celular/inmunología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Granulocitos/inmunología , Granulocitos/metabolismo , Humanos , Sinapsis Inmunológicas , Activación de Linfocitos/inmunología , Linfocitos/inmunología , Linfocitos/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Monocitos/inmunología , Monocitos/metabolismo
13.
J Virol ; 83(7): 3094-103, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19176629

RESUMEN

Lentiviral vectors (lentivectors) are effective for stimulation of cell-mediated and humoral immunity following subcutaneous and intramuscular immunization. However, lentivector genome integration carries a risk of perturbation of host gene expression. Here, we demonstrate that lentivectors with multiple mutations that prevent integration are also effective immunogens. First, systemic CD8(+) T-cell responses to the model antigen ovalbumin were detected following subcutaneous injection of nonintegrating lentivectors. Transfer of transgenic OT1 T cells demonstrated that antigen presentation persisted for at least 30 days. Furthermore, an enhanced CD8(+) T-cell response, peaking at 7 days, was stimulated by coexpression of p38 MAP kinase or an NF-kappaB activator from the same vector. Second, we demonstrated systemic CD8(+) T-cell and antibody responses to the secreted hepatitis B virus (HBV) surface antigen expressed from a nonintegrating lentivector injected intramuscularly. The induction, specificity, and kinetics of antibody production closely mimicked those of natural HBV infection. In this case, both the vector genome and the immune response were maintained for at least 2 months. Together, our data indicate that nonintegrating lentivectors can be employed to generate effective vaccines.


Asunto(s)
Anticuerpos Antineoplásicos/sangre , Anticuerpos Antivirales/sangre , Vacunas contra el Cáncer/inmunología , Vectores Genéticos , Vacunas contra Hepatitis B/inmunología , Lentivirus/genética , Linfocitos T/inmunología , Animales , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Neoplasias/patología , Neoplasias/prevención & control , Integración Viral
14.
Blood ; 112(10): 4158-69, 2008 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-18687984

RESUMEN

To more precisely identify the B-cell phenotype in Wiskott-Aldrich syndrome (WAS), we used 3 distinct murine in vivo models to define the cell intrinsic requirements for WAS protein (WASp) in central versus peripheral B-cell development. Whereas WASp is dispensable for early bone marrow B-cell development, WASp deficiency results in a marked reduction in each of the major mature peripheral B-cell subsets, exerting the greatest impact on marginal zone and B1a B cells. Using in vivo bromodeoxyuridine labeling and in vitro functional assays, we show that these deficits reflect altered peripheral homeostasis, partially resulting from an impairment in integrin function, rather than a developmental defect. Consistent with these observations, we also show that: (1) WASp expression levels increase with cell maturity, peaking in those subsets exhibiting the greatest sensitivity to WASp deficiency; (2) WASp(+) murine B cells exhibit a marked selective advantage beginning at the late transitional B-cell stage; and (3) a similar in vivo selective advantage is manifest by mature WASp(+) human B cells. Together, our data provide a better understanding of the clinical phenotype of WAS and suggest that gene therapy might be a useful approach to rescue altered B-cell homeostasis in this disease.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Células de la Médula Ósea/inmunología , Homeostasis/inmunología , Proteína del Síndrome de Wiskott-Aldrich/inmunología , Animales , Homeostasis/genética , Ratones , Ratones Noqueados , Proteína del Síndrome de Wiskott-Aldrich/genética
15.
Mol Ther ; 16(5): 836-44, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18388921

RESUMEN

Wiskott-Aldrich syndrome (WAS) is an X-linked hematological disease characterized by immunodeficiency, eczema, and thrombocytopaenia, and shows promise for treatment with hematopoietic stem cell gene therapy. The immunopathology of WAS is attributable at least in part to defects of cell migration and localization as a result of chemotactic, adhesive, and chemokinetic defects. Whereas previous studies using either gammaretroviral or lentiviral vectors have demonstrated variable correction of T-cell proliferation and dendritic cell (DC) cytoarchitecture, we have used a lentiviral vector expressing an eGFP-WASp fusion protein to test the potential for restoration of cell migratory defects. Multilineage expression of the fusion transgene was present for up to 10 months after primary engraftment, and also in secondary recipients analyzed after a further 9 months. Transduced bone marrow-derived dendritic cells (BMDCs) demonstrated recovery of podosome numbers and turnover, while B cells, BMDCs, and Langerhans cells (LCs) exhibited enhanced chemotactic responses to specific stimuli. As an indication of functionality in vivo, splenic marginal zone B cells and a cutaneous contact hypersensitivity (CHS) response to dinitrofluorobenzene (DNFB) were both partially restored. These proof of principle experiments demonstrate that WAS protein (WASp) transgene expression can be successfully maintained long term in primary and secondary recipients, and that it is associated with a significant repair of migratory defects.


Asunto(s)
Terapia Genética/métodos , Síndrome de Wiskott-Aldrich/genética , Síndrome de Wiskott-Aldrich/terapia , Animales , Movimiento Celular , Trasplante de Células , Quimiotaxis , Células Dendríticas/citología , Dinitrofluorobenceno/farmacología , Proteínas Fluorescentes Verdes/metabolismo , Células de Langerhans/citología , Lentivirus/genética , Ratones , Ratones Noqueados , Modelos Biológicos , Bazo
16.
Mol Ther ; 15(11): 1947-54, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17700544

RESUMEN

Human immunodeficiency virus (HIV)-based lentiviral vectors (LVs) hold immense promise for gene delivery applications because of their relatively large packaging capacity and their ability to infect a range of cell types. The genome of HIV non-specifically integrates into the host genome, and this promotes efficient, stable transgene expression in dividing cells. However, integration can also be problematic because of variations in gene expression among cells, possible gene silencing and, most importantly, insertional mutagenesis which can lead to undesirable effects such as malignant transformation. In order to alleviate these problems, we have developed a range of non-integrating LVs (NILVs) by introducing point mutations into the catalytic site, chromosome binding site, and viral DNA binding site of the viral integrase (IN). In addition, we have mutated the IN attachment (att) sites within the HIV long terminal repeats (LTRs). All of the vectors produced show efficient reverse transcription and transgene expression in dividing cells and prolonged expression in non-dividing myotubes. Finally, we show that NILV can be used for achieving highly effective gene transfer and expression in muscle in vivo.


Asunto(s)
Vectores Genéticos/genética , Lentivirus/genética , Músculos/metabolismo , Transducción Genética/métodos , Animales , Línea Celular , ADN Viral/genética , Expresión Génica , Genoma Viral/genética , Humanos , Integrasas/química , Integrasas/genética , Integrasas/metabolismo , Ratones , Mutación/genética , Transgenes/genética
17.
J Neurovirol ; 13(2): 173-84, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17505986

RESUMEN

Molecular diversity within brain-derived HIV-1 sequences is highly variable depending on the individual gene examined and the neurological status of the patient. Herein, we examined different brain-derived human immunodeficiency virus (HIV)-1 tat sequences in terms of their effects on LTR transactivation and host gene induction in neural cells. Astrocytic and monocytoid cells co-transfected with prototypic tat clones derived from non-demented (ND) (n = 3) and demented (HAD) (n = 3) AIDS patients and different HIV-LTR constructs revealed that LTR transactivation mediated by tat clones derived from HAD patients was decreased (p < 0.05). A Tat-derived peptide containing the amino acid 24-38 domain from a ND clone caused down-regulation of the LTR transactivation (p < 0.05) in contrast to peptides from other Tat regions derived from HAD and ND tat clones. Both brain-derived HAD and ND tat constructs were able to induce the host immune genes, MCP-1 and IL-1beta. Microarray analysis revealed several host genes were selectively upregulated by a HAD-derived tat clone including an enzyme mediating heparan sulphate synthesis, HS3ST3B1 (p < 0.05), which was also found to be increased in the brains of patients with HAD. Expression of the pro-apoptotic gene, PDCD7, was reduced in cells transfected with the HAD-derived tat clone and moreover, this gene was also suppressed in monocytoid cells infected with a neurotropic HIV-1 strain. Thus, mutations within the HIV-1 tat gene may exert pathogenic effects contributing to the development of HAD, which are independent of its effects on LTR transactivation.


Asunto(s)
Productos del Gen tat/genética , Infecciones por VIH/inmunología , Infecciones por VIH/virología , Duplicado del Terminal Largo de VIH/genética , VIH-1/fisiología , Complejo SIDA Demencia/inmunología , Complejo SIDA Demencia/metabolismo , Complejo SIDA Demencia/virología , Secuencia de Aminoácidos , Astrocitos/metabolismo , Astrocitos/virología , Encéfalo/metabolismo , Encéfalo/virología , Línea Celular Tumoral , Quimiocina CCL2/metabolismo , Regulación hacia Abajo , Regulación Viral de la Expresión Génica , Productos del Gen tat/química , Infecciones por VIH/metabolismo , Heparitina Sulfato/biosíntesis , Heparitina Sulfato/genética , Humanos , Interleucina-1beta/metabolismo , Datos de Secuencia Molecular , Monocitos/metabolismo , Monocitos/virología , Estructura Terciaria de Proteína , Alineación de Secuencia , Activación Transcripcional , Replicación Viral , Productos del Gen tat del Virus de la Inmunodeficiencia Humana
18.
Mol Ther ; 13(4): 729-37, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16360341

RESUMEN

The Wiskott Aldrich syndrome protein (WASP) is a hematopoietic-specific cytoskeletal regulator that is necessary for induction of normal immunity. In the context of effective gene therapy for WAS, cellular models of human WASP deficiency are important for definition of the threshold of protein expression required for optimal activity. Using lentiviral vector-mediated RNA interference (RNAi), we were able to down-regulate the levels of human WASP in cell lines and primary cells. In dendritic cells (DC), RNAi-induced WASP deficiency did not impair phenotypic maturation but perturbed cytoskeletal organization. As a result, podosomes, which are actin-rich structures present in immature adherent DC, were formed less efficiently and motility was disturbed. Overall, treatment of cells with RNAi recapitulated the phenotype of cells derived from patients or animals with inactivating mutations of the WAS gene. Interestingly, reduction of the levels of WASP to about 60% of normal was sufficient to inhibit the formation of podosomes in DC, implying that this cell type requires near-normal levels of WASP to sustain physiological cytoskeleton-dependent activities.


Asunto(s)
Movimiento Celular/fisiología , Células Dendríticas/metabolismo , Regulación hacia Abajo , ARN Interferente Pequeño/farmacología , Proteína del Síndrome de Wiskott-Aldrich/fisiología , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Relación Dosis-Respuesta a Droga , Expresión Génica , Terapia Genética , Vectores Genéticos/uso terapéutico , Humanos , Células Jurkat , Lentivirus/genética , Mutación , ARN Interferente Pequeño/metabolismo , Transducción Genética , Síndrome de Wiskott-Aldrich/genética , Proteína del Síndrome de Wiskott-Aldrich/deficiencia , Proteína del Síndrome de Wiskott-Aldrich/genética , Proteína del Síndrome de Wiskott-Aldrich/metabolismo
19.
J Leukoc Biol ; 78(1): 70-9, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15788439

RESUMEN

In the nonobese diabetic (NOD) mouse, a model of autoimmune diabetes, dendritic cells (DC), and macrophages (Mphi) is important for the initiation and progression of autoimmunity and the final destruction of beta-cells. Previous studies suggested that an aberrant development of DC and Mphi is related to their pathogenic function. To study this in vivo, we investigated NOD mouse monocytes, the direct precursors of DC, and Mphi. The recently described discrimination between immature (Ly-6C(high)) and mature (Ly-6C(low)) monocytes enabled us to investigate the apportioning between blood monocyte populations in the NOD mouse, which had an abnormally high number of mature monocytes in circulation, and this phenomenon appeared to be intrinsic to the NOD background, as nonobese resistant (NOR) and NOD-H2b mice also showed this altered balance. After depletion by apoptosis-inducing liposomes, the reappearance and transition of immature-to-mature monocytes had similar kinetics as control mice but led again to the presence of a larger, mature monocyte compartment in the blood. In addition, although monocytes from C57BL mice down-regulated their capability to adhere to fibronectin and intercellular adhesion molecule-1 upon maturation, the mature NOD monocytes retained their high adhesion capacity, characteristic of immature cells. Furthermore, both monocyte subpopulations of NOD mice showed enhanced differentiation into Mphi-like F4/80(high) cells in vitro. In conclusion, mice with the NOD background have raised numbers of mature monocytes in the circulation and a proinflammatory, Mphi-directed monocyte development.


Asunto(s)
Diferenciación Celular/inmunología , Quimiotaxis de Leucocito/inmunología , Diabetes Mellitus Tipo 1/inmunología , Macrófagos/inmunología , Monocitos/inmunología , Regulación hacia Arriba/inmunología , Animales , Apoptosis/inmunología , Adhesión Celular/genética , Adhesión Celular/inmunología , Moléculas de Adhesión Celular/inmunología , Recuento de Células , Proliferación Celular , Células Cultivadas , Diabetes Mellitus Tipo 1/fisiopatología , Modelos Animales de Enfermedad , Matriz Extracelular/inmunología , Predisposición Genética a la Enfermedad/genética , Liposomas/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Regulación hacia Arriba/genética
20.
Eur J Immunol ; 35(1): 225-35, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15593124

RESUMEN

The accumulation of macrophages (M Phi) and dendritic cells (DC) in the pancreas plays a crucial role in the pathogenesis of autoimmune diabetes. We studied the recruitment of monocytes, M Phi and DC to sites of inflammation, i.e. the peritoneal cavity and a subcutaneously elicited air pouch in the NOD mouse model of autoimmune diabetes. The leukocyte recruitment was studied from 1 to 7 days after injection of thioglycollate (peritoneum), C5a (peritoneum, air pouch), CCL2 and CCL3 (air pouch). C57BL/6 and BALB/c mice served as controls. Morphological and flow cytometric analysis of the recruited cells was performed, IL-1 beta, TNF-alpha, IL-6, IL-12 and IL-10 in exudates measured, and in vitro CCL2-chemotaxis of exudate M Phi (Boyden chamber) determined. NOD mice were strongly impaired in the recruitment of M Phi, DC, monocytes, and granulocytes. Chemokine-injected air pouches of NOD mice showed an increased IL-10 and a decreased IL-1 beta level, while the other cytokines were normally or very lowly expressed. In addition, NOD exudate M Phi displayed an impaired in vitro CCL2-induced migration. Our data show that NOD mice have an impaired ability to recruit leukocytes into sites of inflammation elicited in the peritoneum and the air pouch. A raised IL-10/IL-1 beta ratio at these sites and a deficient migratory capacity of NOD monocytes are important determinants in this impairment.


Asunto(s)
Diabetes Mellitus Tipo 1/inmunología , Inflamación/inmunología , Leucocitos/inmunología , Animales , Movimiento Celular , Quimiocina CCL2/farmacología , Células Dendríticas/inmunología , Células Dendríticas/patología , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/patología , Femenino , Inmunidad Innata , Técnicas In Vitro , Inflamación/patología , Interleucina-10/metabolismo , Leucocitos/efectos de los fármacos , Leucocitos/patología , Macrófagos/inmunología , Macrófagos/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Monocitos/inmunología , Monocitos/patología , Cavidad Peritoneal/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA