Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 18(7): 744-752, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28553952

RESUMEN

The single-nucleotide polymorphism rs1990760 in the gene encoding the cytosolic viral sensor IFIH1 results in an amino-acid change (A946T; IFIH1T946) that is associated with multiple autoimmune diseases. The effect of this polymorphism on both viral sensing and autoimmune pathogenesis remains poorly understood. Here we found that human peripheral blood mononuclear cells (PBMCs) and cell lines expressing the risk variant IFIH1T946 exhibited heightened basal and ligand-triggered production of type I interferons. Consistent with those findings, mice with a knock-in mutation encoding IFIH1T946 displayed enhanced basal expression of type I interferons, survived a lethal viral challenge and exhibited increased penetrance in autoimmune models, including a combinatorial effect with other risk variants. Furthermore, IFIH1T946 mice manifested an embryonic survival defect consistent with enhanced responsiveness to RNA self ligands. Together our data support a model wherein the production of type I interferons driven by an autoimmune risk variant and triggered by ligand functions to protect against viral challenge, which probably accounts for its selection within human populations but provides this advantage at the cost of modestly promoting the risk of autoimmunity.


Asunto(s)
Autoinmunidad/genética , Infecciones por Cardiovirus/genética , Interferón Tipo I/inmunología , Helicasa Inducida por Interferón IFIH1/genética , Adolescente , Adulto , Animales , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Autoinmunidad/inmunología , Southern Blotting , Infecciones por Cardiovirus/inmunología , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , Virus de la Encefalomiocarditis/inmunología , Femenino , Predisposición Genética a la Enfermedad , Células HEK293 , Humanos , Immunoblotting , Helicasa Inducida por Interferón IFIH1/inmunología , Masculino , Ratones , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Virosis/genética , Virosis/inmunología , Adulto Joven
2.
Nat Immunol ; 14(5): 514-22, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23563688

RESUMEN

Here we identified B cells as a major source of rapid, innate-like production of interleukin 17 (IL-17) in vivo in response to infection with Trypanosoma cruzi. IL-17(+) B cells had a plasmablast phenotype, outnumbered cells of the TH17 subset of helper T cells and were required for an optimal response to this pathogen. With both mouse and human primary B cells, we found that exposure to parasite-derived trans-sialidase in vitro was sufficient to trigger modification of the cell-surface mucin CD45, which led to signaling dependent on the kinase Btk and production of IL-17A or IL-17F via a transcriptional program independent of the transcription factors RORγt and Ahr. Our combined data suggest that the generation of IL-17(+) B cells may be a previously unappreciated feature of innate immune responses required for pathogen control or IL-17-mediated autoimmunity.


Asunto(s)
Linfocitos B/inmunología , Enfermedad de Chagas/inmunología , Glicoproteínas/metabolismo , Interleucina-17/inmunología , Neuraminidasa/metabolismo , Trypanosoma cruzi/enzimología , Trypanosoma cruzi/inmunología , Animales , Linfocitos B/parasitología , Proliferación Celular , Células Cultivadas , Enfermedad de Chagas/genética , Glicoproteínas/genética , Humanos , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Neuraminidasa/genética , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/parasitología , Células Th17/inmunología , Células Th17/parasitología , Activación Transcripcional/inmunología
3.
J Immunol ; 209(10): 1880-1891, 2022 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-36426938

RESUMEN

Multiple sclerosis (MS) is an inflammatory, demyelinating CNS disease believed to be mediated by CD4 T cells specific for CNS self-antigens. CD8 T cells are also implicated in MS but their function is not well understood. MS lesions are heterogeneous and may reflect variation in the contribution of different types of lymphocytes. Understanding how lymphocytes with different effector functions contribute to MS is essential to develop effective therapies. We investigated how T cells expressing an MHC class I-restricted transgenic TCR specific for myelin basic protein (MBP) contribute to CNS autoimmunity using the mouse model of MS, experimental autoimmune encephalomyelitis. Virus infection triggered cytotoxic TCR-transgenic CD8 T cells to initiate acute experimental autoimmune encephalomyelitis in an IFN-γ- and perforin-dependent manner. Unexpectedly, spontaneous CNS autoimmunity developed in the TCR-transgenic mice that was accelerated by IFN-γ-deficiency. Spontaneous disease was associated with CD4 T cells that develop via endogenous TCR rearrangements but retain specificity for the MHC class I-restricted MBP epitope. The CD4 T cells produced TNF-α without other inflammatory cytokines and caused lesions with striking similarity to active MS lesions. Surprisingly, B cells were the predominant cell type that cross-presented MBP, and their depletion halted disease progression. This work provides a new model of spontaneous CNS autoimmunity with unique similarities to MS that is mediated by T cells with a distinct effector phenotype.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Esclerosis Múltiple , Ratones , Animales , Autoinmunidad , Linfocitos T CD4-Positivos , Sistema Nervioso Central , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T
4.
J Immunol ; 209(6): 1033-1038, 2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-35995509

RESUMEN

Germline gain-of-function mutations in the transcriptional factor STAT3 promote early-onset multisystemic autoimmunity. To investigate how increased STAT3 promotes systemic inflammation, we generated a transgenic knock-in strain expressing a pathogenic human mutation STAT3K392R within the endogenous murine locus. As predicted, STAT3K392R mice develop progressive lymphoid hyperplasia and systemic inflammation, mirroring the human disease. However, whereas the prevailing model holds that increased STAT3 activity drives human autoimmunity by dysregulating the balance between regulatory T cells and Th17 cell differentiation, we observed increased Th17 cells in the absence of major defects in regulatory T cell differentiation or function. In addition, STAT3K392R animals exhibited a prominent accumulation of IFN-γ-producing CD4+ and CD8+ T cells. Together, these data provide new insights into this complex human genetic syndrome and highlight the diverse cellular mechanisms by which dysregulated STAT3 activity promotes breaks in immune tolerance.


Asunto(s)
Autoinmunidad , Factor de Transcripción STAT3 , Linfocitos T Reguladores , Animales , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular , Mutación con Ganancia de Función , Técnicas de Sustitución del Gen , Humanos , Inflamación/patología , Ratones , Ratones Transgénicos , Factor de Transcripción STAT3/genética , Células Th17
5.
J Immunol ; 207(11): 2710-2719, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34740959

RESUMEN

The single-nucleotide polymorphism (SNP) rs3184504 is broadly associated with increased risk for multiple autoimmune and cardiovascular diseases. Although the allele is uniquely enriched in European descent, the mechanism for the widespread selective sweep is not clear. In this study, we find the rs3184504*T allele had a strong association with reduced mortality in a human sepsis cohort. The rs3184504*T allele associates with a loss-of-function amino acid change (p.R262W) in the adaptor protein SH2B3, a likely causal variant. To better understand the role of SH2B3 in sepsis, we used mouse modeling and challenged SH2B3-deficient mice with a polymicrobial cecal-ligation puncture (CLP) procedure. We found SH2B3 deficiency improved survival and morbidity with less organ damage and earlier bacterial clearance compared with control mice. The peritoneal infiltrating cells exhibited augmented phagocytosis in Sh2b3 -/- mice with enriched recruitment of Ly6Chi inflammatory monocytes despite equivalent or reduced chemokine expression. Rapid cycling of monocytes and progenitors occurred uniquely in the Sh2b3 -/- mice following CLP, suggesting augmented myelopoiesis. To model the hypomorphic autoimmune risk allele, we created a novel knockin mouse harboring a similar point mutation in the murine pleckstrin homology domain of SH2B3. At baseline, phenotypic changes suggested a hypomorphic allele. In the CLP model, homozygous knockin mice displayed improved mortality and morbidity compared with wild-type or heterozygous mice. Collectively, these data suggest that hypomorphic SH2B3 improves the sepsis response and that balancing selection likely contributed to the relative frequency of the autoimmune risk variant.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/inmunología , Sepsis/inmunología , Proteínas Adaptadoras Transductoras de Señales/genética , Adulto , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Congénicos , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Polimorfismo de Nucleótido Simple/genética , Sepsis/genética
6.
J Immunol ; 193(2): 555-63, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24913979

RESUMEN

Multiple sclerosis (MS) is an autoimmune disease in which inflammatory lesions lead to tissue injury in the brain and/or spinal cord. The specific sites of tissue injury are strong determinants of clinical outcome in MS, but the pathways that determine whether damage occurs in the brain or spinal cord are not understood. Previous studies in mouse models of MS demonstrated that IFN-γ and IL-17 regulate lesion localization within the brain; however, the mechanisms by which these cytokines mediate their effects have not been identified. In the present study, we show that IL-17 promoted, but IFN-γ inhibited, ELR(+) chemokine-mediated neutrophil recruitment to the brain, and that neutrophil infiltration was required for parenchymal tissue damage in the brain. In contrast, IFN-γ promoted ELR(+) chemokine expression and neutrophil recruitment to the spinal cord. Surprisingly, tissue injury in the spinal cord did not exhibit the same dependence on neutrophil recruitment that was observed for the brain. Our results demonstrate that the brain and spinal cord exhibit distinct sensitivities to cellular mediators of tissue damage, and that IL-17 and IFN-γ differentially regulate recruitment of these mediators to each microenvironment. These findings suggest an approach toward tailoring therapies for patients with distinct patterns of neuroinflammation.


Asunto(s)
Encéfalo/inmunología , Citocinas/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Mielitis/inmunología , Infiltración Neutrófila/inmunología , Animales , Encéfalo/metabolismo , Encéfalo/patología , Células Cultivadas , Citocinas/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/metabolismo , Citometría de Flujo , Humanos , Ratones , Ratones Endogámicos C3H , Ratones Noqueados , Ratones Transgénicos , Esclerosis Múltiple/genética , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/metabolismo , Glicoproteína Mielina-Oligodendrócito/inmunología , Mielitis/genética , Mielitis/metabolismo , Fragmentos de Péptidos/inmunología , Ratas , Receptores de Interferón/deficiencia , Receptores de Interferón/genética , Receptores de Interferón/inmunología , Receptores de Interleucina-17/deficiencia , Receptores de Interleucina-17/genética , Receptores de Interleucina-17/inmunología , Receptores de Interleucina-8B/inmunología , Receptores de Interleucina-8B/metabolismo , Receptor de Interferón gamma
7.
J Immunol ; 192(7): 3029-42, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24591371

RESUMEN

Multiple sclerosis (MS) is an inflammatory disease of the CNS that causes the demyelination of nerve cells and destroys oligodendrocytes, neurons, and axons. Historically, MS has been thought to be a CD4 T cell-mediated autoimmune disease of CNS white matter. However, recent studies identified CD8 T cell infiltrates and gray matter lesions in MS patients. These findings suggest that CD8 T cells and CNS Ags other than myelin proteins may be involved during the MS disease process. In this article, we show that CD8 T cells reactive to glial fibrillary acidic protein (GFAP), a protein expressed in astrocytes, can avoid tolerance mechanisms and, depending upon the T cell-triggering event, drive unique aspects of inflammatory CNS autoimmunity. In GFAP-specific CD8 TCR-transgenic (BG1) mice, tissue resident memory-like CD8 T cells spontaneously infiltrate the gray matter and white matter of the CNS, resulting in a relapsing-remitting CNS autoimmunity. The frequency, severity, and remissions from spontaneous disease are controlled by the presence of polyclonal B cells. In contrast, a viral trigger induces GFAP-specific CD8 T effector cells to exclusively target the meninges and vascular/perivascular space of the gray and white matter of the brain, causing a rapid, acute CNS disease. These findings demonstrate that the type of CD8 T cell-triggering event can determine the presentation of distinct CNS autoimmune disease pathologies.


Asunto(s)
Autoinmunidad/inmunología , Linfocitos T CD8-positivos/inmunología , Enfermedades del Sistema Nervioso Central/inmunología , Sistema Nervioso Central/inmunología , Proteína Ácida Fibrilar de la Glía/inmunología , Animales , Astrocitos/inmunología , Astrocitos/metabolismo , Astrocitos/patología , Encéfalo/inmunología , Encéfalo/metabolismo , Encéfalo/patología , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/patología , Células Cultivadas , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/patología , Enfermedades del Sistema Nervioso Central/genética , Enfermedades del Sistema Nervioso Central/metabolismo , Citometría de Flujo , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Inmunohistoquímica , Interferón gamma/inmunología , Interferón gamma/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Noqueados , Ratones Transgénicos , Esclerosis Múltiple/genética , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/metabolismo , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Médula Espinal/inmunología , Médula Espinal/metabolismo , Médula Espinal/patología
8.
J Immunol ; 192(10): 4525-32, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24711620

RESUMEN

Systemic lupus erythematosus is a multisystem autoimmune disease characterized by autoantibodies targeting nucleic acid-associated Ags. The endosomal TLRs TLR7 and TLR9 are critical for generation of Abs targeting RNA- or DNA-associated Ags, respectively. In murine lupus models, deletion of TLR7 limits autoimmune inflammation, whereas deletion of TLR9 exacerbates disease. Whether B cell or myeloid TLR7/TLR9 signaling is responsible for these effects has not been fully addressed. In this study, we use a chimeric strategy to evaluate the effect of B cell-intrinsic deletion of TLR7 versus TLR9 in parallel lupus models. We demonstrate that B cell-intrinsic TLR7 deletion prevents RNA-associated Ab formation, decreases production of class-switched Abs targeting nonnuclear Ags, and limits systemic autoimmunity. In contrast, B cell-intrinsic TLR9 deletion results in decreased DNA-reactive Ab, but increased Abs targeting a broad range of systemic autoantigens. Further, we demonstrate that B cell-intrinsic TLR9 deletion results in increased systemic inflammation and immune complex glomerulonephritis, despite intact TLR signaling within the myeloid compartment. These data stress the critical importance of dysregulated B cell-intrinsic TLR signaling in the pathogenesis of systemic lupus erythematosus.


Asunto(s)
Anticuerpos Antinucleares/inmunología , Linfocitos B/inmunología , Lupus Eritematoso Sistémico/inmunología , Glicoproteínas de Membrana/inmunología , Transducción de Señal/inmunología , Receptor Toll-Like 7/inmunología , Receptor Toll-Like 9/inmunología , Animales , Anticuerpos Antinucleares/genética , Linfocitos B/patología , Modelos Animales de Enfermedad , Inflamación/genética , Inflamación/inmunología , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/patología , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , ARN/genética , ARN/inmunología , Transducción de Señal/genética , Receptor Toll-Like 7/genética , Receptor Toll-Like 9/genética
9.
Chem Soc Rev ; 44(23): 8576-607, 2015 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-26390044

RESUMEN

Iron oxide nanoparticles (IONPs) have been extensively used during the last two decades, either as effective bio-imaging contrast agents or as carriers of biomolecules such as drugs, nucleic acids and peptides for controlled delivery to specific organs and tissues. Most of these novel applications require elaborate tuning of the physiochemical and surface properties of the IONPs. As new IONPs designs are envisioned, synergistic consideration of the body's innate biological barriers against the administered nanoparticles and the short and long-term side effects of the IONPs become even more essential. There are several important criteria (e.g. size and size-distribution, charge, coating molecules, and plasma protein adsorption) that can be effectively tuned to control the in vivo pharmacokinetics and biodistribution of the IONPs. This paper reviews these crucial parameters, in light of biological barriers in the body, and the latest IONPs design strategies used to overcome them. A careful review of the long-term biodistribution and side effects of the IONPs in relation to nanoparticle design is also given. While the discussions presented in this review are specific to IONPs, some of the information can be readily applied to other nanoparticle systems, such as gold, silver, silica, calcium phosphates and various polymers.


Asunto(s)
Compuestos Férricos , Nanopartículas de Magnetita , Administración Intravenosa , Administración Oral , Animales , Línea Celular , Compuestos Férricos/administración & dosificación , Compuestos Férricos/química , Compuestos Férricos/farmacocinética , Compuestos Férricos/toxicidad , Humanos , Nanopartículas de Magnetita/administración & dosificación , Nanopartículas de Magnetita/química , Nanopartículas de Magnetita/toxicidad , Ratones , Ratas , Distribución Tisular
10.
J Immunol ; 190(6): 2536-43, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23382559

RESUMEN

TLR7 activation is implicated in the pathogenesis of systemic lupus erythematosus. Mice that overexpress TLR7 develop a lupus-like disease with autoantibodies and glomerulonephritis and early death. To determine whether degradation of the TLR7 ligand RNA would alter the course of disease, we created RNase A transgenic (Tg) mice. We then crossed the RNase Tg to TLR7 Tg mice to create TLR7 × RNase double Tg (DTg) mice. DTg mice had a significantly increased survival associated with reduced activation of T and B lymphocytes and reduced kidney deposition of IgG and C3. We observed massive hepatic inflammation and cell death in TLR7 Tg mice. In contrast, hepatic inflammation and necrosis were strikingly reduced in DTg mice. These findings indicate that high concentrations of serum RNase protect against immune activation and inflammation associated with TLR7 stimulation and that RNase may be a useful therapeutic strategy in the prevention or treatment of inflammation in systemic lupus erythematosus and, possibly, liver diseases.


Asunto(s)
Regulación hacia Abajo/genética , Regulación hacia Abajo/inmunología , Glicoproteínas de Membrana/biosíntesis , Glicoproteínas de Membrana/genética , Ribonucleasa Pancreática/genética , Receptor Toll-Like 7/biosíntesis , Receptor Toll-Like 7/genética , Regulación hacia Arriba/genética , Regulación hacia Arriba/inmunología , Animales , Bovinos , Células Cultivadas , Células Madre Embrionarias , Hepatitis/enzimología , Hepatitis/inmunología , Hepatitis/patología , Humanos , Inflamación/enzimología , Inflamación/inmunología , Inflamación/prevención & control , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/mortalidad , Lupus Eritematoso Sistémico/prevención & control , Masculino , Glicoproteínas de Membrana/fisiología , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Transgénicos , Ribonucleasa Pancreática/sangre , Ribonucleasa Pancreática/fisiología , Bazo/enzimología , Bazo/inmunología , Bazo/patología , Análisis de Supervivencia , Receptor Toll-Like 7/fisiología
11.
Proc Natl Acad Sci U S A ; 107(43): 18616-21, 2010 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-20926749

RESUMEN

Most patients who die from cancer succumb to treatment-refractory advanced metastatic progression. Although the early stages of tumor metastasis result in the formation of clinically silent micrometastatic foci, its later stages primarily reflect the progressive, organ-destructive growth of already advanced metastases. Early-stage metastasis is regulated by multiple factors within tumor cells as well as by the tumor microenvironment (TME). In contrast, the molecular determinants that control advanced metastatic progression remain essentially uncharacterized, precluding the development of therapies targeted against it. Here we show that the TME, functioning in part through platelet endothelial cell adhesion molecule 1 (PECAM-1), drives advanced metastatic progression and is essential for progression through its preterminal end stage. PECAM-1-KO and chimeric mice revealed that its metastasis-promoting effects are mediated specifically through vascular endothelial cell (VEC) PECAM-1. Anti-PECAM-1 mAb therapy suppresses both end-stage metastatic progression and tumor-induced cachexia in tumor-bearing mice. It reduces proliferation, but not angiogenesis or apoptosis, within advanced tumor metastases. Because its antimetastatic effects are mediated by binding to VEC rather than to tumor cells, anti-PECAM-1 mAb appears to act independently of tumor type. A modified 3D coculture assay showed that anti-PECAM-1 mAb inhibits the proliferation of PECAM-1-negative tumor cells by altering the concentrations of secreted factors. Our studies indicate that a complex interplay between elements of the TME and advanced tumor metastases directs end-stage metastatic progression. They also suggest that some therapeutic interventions may target late-stage metastases specifically. mAb-based targeting of PECAM-1 represents a TME-targeted therapeutic approach that suppresses the end stages of metastatic progression, until now a refractory clinical entity.


Asunto(s)
Neoplasias Experimentales/secundario , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/fisiología , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/farmacología , Apoptosis , Trasplante de Médula Ósea , Caquexia/terapia , Línea Celular Tumoral , Proliferación Celular , Progresión de la Enfermedad , Células Endoteliales/fisiología , Femenino , Humanos , Neoplasias Pulmonares/secundario , Neoplasias Pulmonares/terapia , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Desnudos , Ratones Transgénicos , Neoplasias Experimentales/irrigación sanguínea , Neoplasias Experimentales/patología , Neoplasias Experimentales/terapia , Neovascularización Patológica , Comunicación Paracrina , Fenotipo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/inmunología
12.
Sci Transl Med ; 15(703): eade7028, 2023 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-37406138

RESUMEN

Heterozygous signal transducer and activator of transcription 1 (STAT1) gain-of-function (GOF) mutations promote a clinical syndrome of immune dysregulation characterized by recurrent infections and predisposition to humoral autoimmunity. To gain insights into immune characteristics of STAT1-driven inflammation, we performed deep immunophenotyping of pediatric patients with STAT1 GOF syndrome and age-matched controls. Affected individuals exhibited dysregulated CD4+ T cell and B cell activation, including expansion of TH1-skewed CXCR3+ populations that correlated with serum autoantibody titers. To dissect underlying immune mechanisms, we generated Stat1 GOF transgenic mice (Stat1GOF mice) and confirmed the development of spontaneous humoral autoimmunity that recapitulated the human phenotype. Despite clinical resemblance to human regulatory T cell (Treg) deficiency, Stat1GOF mice and humans with STAT1 GOF syndrome exhibited normal Treg development and function. In contrast, STAT1 GOF autoimmunity was characterized by adaptive immune activation driven by dysregulated STAT1-dependent signals downstream of the type 1 and type 2 interferon (IFN) receptors. However, in contrast to the prevailing type 1 IFN-centric model for STAT1 GOF autoimmunity, Stat1GOF mice lacking the type 1 IFN receptor were only partially protected from STAT1-driven systemic inflammation, whereas loss of type 2 IFN (IFN-γ) signals abrogated autoimmunity. Last, germline STAT1 GOF alleles are thought to enhance transcriptional activity by increasing total STAT1 protein, but the underlying biochemical mechanisms have not been defined. We showed that IFN-γ receptor deletion normalized total STAT1 expression across immune lineages, highlighting IFN-γ as the critical driver of feedforward STAT1 elevation in STAT1 GOF syndrome.


Asunto(s)
Autoinmunidad , Mutación con Ganancia de Función , Humanos , Niño , Ratones , Animales , Autoinmunidad/genética , Interferón gamma/metabolismo , Síndrome , Inflamación , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo
13.
Blood ; 115(11): 2146-55, 2010 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-20093406

RESUMEN

The immunodeficiency disorder, X-linked agammaglobulinemia (XLA), results from mutations in the gene encoding Bruton tyrosine kinase (Btk). Btk is required for pre-B cell clonal expansion and B-cell antigen receptor signaling. XLA patients lack mature B cells and immunoglobulin and experience recurrent bacterial infections only partially mitigated by life-long antibody replacement therapy. In pursuit of definitive therapy for XLA, we tested ex vivo gene therapy using a lentiviral vector (LV) containing the immunoglobulin enhancer (Emu) and Igbeta (B29) minimal promoter to drive B lineage-specific human Btk expression in Btk/Tec(-/-) mice, a strain that reproduces the features of human XLA. After transplantation of EmuB29-Btk-LV-transduced stem cells, treated mice showed significant, albeit incomplete, rescue of mature B cells in the bone marrow, peripheral blood, spleen, and peritoneal cavity, and improved responses to T-independent and T-dependent antigens. LV-treated B cells exhibited enhanced B-cell antigen receptor signaling and an in vivo selective advantage in the peripheral versus central B-cell compartment. Secondary transplantation showed sustained Btk expression, viral integration, and partial functional responses, consistent with long-term stem cell marking; and serial transplantation revealed no evidence for cellular or systemic toxicity. These findings strongly support pursuit of B lineage-targeted LV gene therapy in human XLA.


Asunto(s)
Agammaglobulinemia/fisiopatología , Agammaglobulinemia/terapia , Linfocitos B/fisiología , Enfermedades Genéticas Ligadas al Cromosoma X/terapia , Terapia Genética , Lentivirus/genética , Recuperación de la Función/fisiología , Agammaglobulinemia Tirosina Quinasa , Animales , Linfocitos B/citología , Células de la Médula Ósea/citología , Trasplante de Médula Ósea , Antígenos CD79/genética , Línea Celular , Linaje de la Célula , Modelos Animales de Enfermedad , Enfermedades Genéticas Ligadas al Cromosoma X/fisiopatología , Terapia Genética/efectos adversos , Vectores Genéticos/genética , Humanos , Cadenas Pesadas de Inmunoglobulina/genética , Ratones , Ratones Endogámicos C57BL , Especificidad de Órganos/genética , Proteínas Tirosina Quinasas/deficiencia , Proteínas Tirosina Quinasas/metabolismo , Proteínas Tirosina Quinasas/uso terapéutico
14.
Cancer Cell ; 1(4): 355-68, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-12086850

RESUMEN

Expression of the cyclin-dependent kinase inhibitor p27(Kip1) (p27) is frequently reduced in human colorectal cancer, and this correlates with poor patient prognosis. To clarify the role of p27 in gastrointestinal (GI) cancer, we measured p27 expression, as well as the effect of germline deletion of p27, in 3 different mouse models of GI neoplasia. p27 expression was frequently reduced in GI tumors arising in 1,2-dimethylhydrazine (DMH) treated mice, and in Apc mutant Min/+ mice, but not in GI tumors arising in Smad3 mutant mice. Germline deletion of p27 resulted in accelerated tumor development and increased tumor cell proliferation in both DMH treated and Min/+ mice, but not in Smad3 mutant mice. p27 deficiency also led to increased adenoma to adenocarcinoma progression. These results indicate that reduction of p27 cooperates with mutations in Apc but not in Smad3 during GI tumorigenesis. Thus, tumor suppression by p27 is contingent on the specific oncogenic pathway that drives tumor development.


Asunto(s)
Adenocarcinoma/patología , Adenoma/patología , Proteína de la Poliposis Adenomatosa del Colon/genética , Proteínas de Ciclo Celular/fisiología , Proteínas de Unión al ADN/genética , Neoplasias Gastrointestinales/patología , Genes Supresores de Tumor/fisiología , Transactivadores/genética , Proteínas Supresoras de Tumor/fisiología , 1,2-Dimetilhidrazina , Adenocarcinoma/inducido químicamente , Adenocarcinoma/genética , Adenoma/inducido químicamente , Adenoma/genética , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Núcleo Celular/metabolismo , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Proteínas del Citoesqueleto/metabolismo , Neoplasias Gastrointestinales/inducido químicamente , Neoplasias Gastrointestinales/genética , Pérdida de Heterocigocidad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Quinasas Asociadas a Fase-S , Transducción de Señal , Proteínas Smad , Proteínas Supresoras de Tumor/genética , beta Catenina
15.
Sci Transl Med ; 14(665): eabn1716, 2022 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-36197963

RESUMEN

Adoptive transfer of regulatory T cells (Tregs) is therapeutic in type 1 diabetes (T1D) mouse models. Tregs that are specific for pancreatic islets are more potent than polyclonal Tregs in preventing disease. However, the frequency of antigen-specific natural Tregs is extremely low, and ex vivo expansion may destabilize Tregs, leading to an effector phenotype. Here, we generated durable, antigen-specific engineered Tregs (EngTregs) from primary human CD4+ T cells by combining FOXP3 homology-directed repair editing and lentiviral T cell receptor (TCR) delivery. Using TCRs derived from clonally expanded CD4+ T cells isolated from patients with T1D, we generated islet-specific EngTregs that suppressed effector T cell (Teff) proliferation and cytokine production. EngTregs suppressed Teffs recognizing the same islet antigen in addition to bystander Teffs recognizing other islet antigens through production of soluble mediators and both direct and indirect mechanisms. Adoptively transferred murine islet-specific EngTregs homed to the pancreas and blocked diabetes triggered by islet-specific Teffs or diabetogenic polyclonal Teffs in recipient mice. These data demonstrate the potential of antigen-specific EngTregs as a targeted therapy for preventing T1D.


Asunto(s)
Diabetes Mellitus Tipo 1 , Islotes Pancreáticos , Animales , Citocinas , Diabetes Mellitus Tipo 1/genética , Factores de Transcripción Forkhead , Humanos , Ratones , Receptores de Antígenos de Linfocitos T , Linfocitos T Reguladores
17.
Mol Ther ; 17(8): 1427-33, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19471246

RESUMEN

A growing body of research supports the development of recombinant adeno-associated viral (rAAV) vectors for delivery of gene expression cassettes to striated musculature as a method of treating severe neuromuscular conditions. However, it is unclear whether delivery protocols that achieve extensive gene transfer in mice can be adapted to produce similarly extensive gene transfer in larger mammals and ultimately patients. Consequently, we sought to investigate methodological modifications that would facilitate rAAV-mediated gene transfer to the striated musculature of canines. A simple procedure incorporating acute (i) occlusion of limb blood flow, (ii) exsanguination via compression bandage, and (iii) vector "dwell" time of <20 minutes, markedly enhanced the transduction of limb muscles, compared with a simple bolus limb infusion of vector. A complementary method whereby vector was infused into the jugular vein led to efficient transduction of cardiomyocytes and to a lesser degree the diaphragm. Together these methods can be used to achieve transgene expression in heart, diaphragm, and limb muscles of juvenile dogs using rAAV6 vectors. These results establish that rAAV-mediated gene delivery is a viable approach to achieving systemic transduction of striated musculature in mammals approaching the dimensions of newborn humans.


Asunto(s)
Vasos Sanguíneos/metabolismo , Dependovirus/genética , Técnicas de Transferencia de Gen , Vectores Genéticos/administración & dosificación , Fibras Musculares Esqueléticas/metabolismo , Fosfatasa Alcalina , Animales , Vasos Sanguíneos/efectos de los fármacos , Ciclosporina/farmacología , Diafragma/metabolismo , Perros , Proteínas Ligadas a GPI , Vectores Genéticos/genética , Miembro Posterior/metabolismo , Humanos , Terapia de Inmunosupresión , Inmunosupresores/farmacología , Isoenzimas/genética , Isoenzimas/inmunología , Venas Yugulares/metabolismo , Ratones , Ácido Micofenólico/análogos & derivados , Ácido Micofenólico/farmacología , Miocardio/metabolismo
18.
J Clin Invest ; 130(1): 203-213, 2020 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-31573979

RESUMEN

Multiple sclerosis (MS) is an inflammatory, demyelinating disease of the CNS. Although CD4+ T cells are implicated in MS pathogenesis and have been the main focus of MS research using the animal model experimental autoimmune encephalomyelitis (EAE), substantial evidence from patients with MS points to a role for CD8+ T cells in disease pathogenesis. We previously showed that an MHC class I-restricted epitope of myelin basic protein (MBP) is presented in the CNS during CD4+ T cell-initiated EAE. Here, we investigated whether naive MBP-specific CD8+ T cells recruited to the CNS during CD4+ T cell-initiated EAE engaged in determinant spreading and influenced disease. We found that the MBP-specific CD8+ T cells exacerbated brain but not spinal cord inflammation. We show that a higher frequency of monocytes and monocyte-derived cells presented the MHC class I-restricted MBP ligand in the brain compared with the spinal cord. Infiltration of MBP-specific CD8+ T cells enhanced ROS production in the brain only in these cell types and only when the MBP-specific CD8+ T cells expressed Fas ligand (FasL). These results suggest that myelin-specific CD8+ T cells may contribute to disease pathogenesis via a FasL-dependent mechanism that preferentially promotes lesion formation in the brain.


Asunto(s)
Autoinmunidad/inmunología , Linfocitos T CD8-positivos/inmunología , Sistema Nervioso Central/inmunología , Encefalomielitis Autoinmune Experimental/etiología , Vaina de Mielina/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Movimiento Celular , Encefalomielitis Autoinmune Experimental/inmunología , Proteína Ligando Fas/fisiología , Femenino , Masculino , Ratones , Ratones Endogámicos C3H , Especies Reactivas de Oxígeno/metabolismo
19.
Cancer Cell ; 35(3): 489-503.e8, 2019 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-30889382

RESUMEN

Many potential targets for CAR-T cells in solid tumors are expressed in some normal tissues, raising concern for off-tumor toxicity. Following lymphodepletion, CAR-T cells targeting the tumor-associated antigen ROR1 lysed tumors in mice but induced lethal bone marrow failure due to recognition of ROR1+ stromal cells. To improve selectivity, we engineered T cells with synthetic Notch (synNotch) receptors specific for EpCAM or B7-H3, which are expressed on ROR1+ tumor cells but not ROR1+ stromal cells. SynNotch receptors induced ROR1 CAR expression selectively within the tumor, resulting in tumor regression without toxicity when tumor cells were segregated from, but not when co-localized with, normal ROR1+ cells. This strategy, thus, permits safe targeting of tumors that are sufficiently separated from normal cells.


Asunto(s)
Inmunoterapia Adoptiva/métodos , Neoplasias/terapia , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/genética , Receptores Quiméricos de Antígenos/inmunología , Animales , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Células K562 , Ratones , Neoplasias/inmunología , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/metabolismo , Linfocitos T/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Sci Adv ; 5(11): eaax0217, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31807699

RESUMEN

Recombinant adeno-associated virus (AAV) vectors are transforming therapies for rare human monogenic deficiency diseases. However, adaptive immune responses to AAV and its limited DNA insert capacity, restrict their therapeutic potential. HEDGES (high-level extended duration gene expression system), a nonviral DNA- and liposome-based gene delivery platform, overcomes these limitations in immunocompetent mice. Specifically, one systemic HEDGES injection durably produces therapeutic levels of transgene-encoded human proteins, including FDA-approved cytokines and monoclonal antibodies, without detectable integration into genomic DNA. HEDGES also controls protein production duration from <3 weeks to >1.5 years, does not induce anti-vector immune responses, is reexpressed for prolonged periods following reinjection, and produces only transient minimal toxicity. HEDGES can produce extended therapeutic levels of multiple transgene-encoded therapeutic human proteins from DNA inserts >1.5-fold larger than AAV-based therapeutics, thus creating combinatorial interventions to effectively treat common polygenic diseases driven by multigenic abnormalities.


Asunto(s)
ADN/genética , Técnicas de Transferencia de Gen , Transgenes , Animales , Línea Celular , ADN/farmacología , Femenino , Humanos , Ratones , Ratones Endogámicos ICR
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA