Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
N Biotechnol ; 82: 43-53, 2024 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-38734368

RESUMEN

Extracellular Vesicles (EVs) have been proposed as a promising tool for drug delivery because of their natural ability to cross biological barriers, protect their cargo, and target specific cells. Moreover, EVs are not recognized by the immune system as foreign, reducing the risk of an immune response and enhancing biocompatibility. Herein, we proposed an alternative therapeutic strategy to restore STAT3 signaling exploiting STAT3 loaded EVs. This approach could be useful in the treatment of Autosomal Dominant Hyper-IgE Syndrome (AD-HIES), a rare primary immunodeficiency and multisystem disorder due to the presence of mutations in STAT3 gene. These mutations alter the signal transduction of STAT3, thereby impeding Th17 CD4+ cell differentiation that leads to the failure of immune response. We set up a simple and versatile method in which EVs were loaded with fully functional STAT3 protein. Moreover, our method allows to follow the uptake of STAT3 loaded vesicles inside cells due to the presence of EGFP in the EGFP-STAT3 fusion protein construct. Taken together, the data presented in this study could provide the scientific background for the development of new therapeutic strategy aimed to restore STAT3 signaling in STAT3 misfunction associated diseases like AD-HIES. In the future, the administration of fully functional wild type STAT3 to CD4+ T cells of AD-HIES patients might compensate its loss of function and would be beneficial for these patients, lowering the risk of infections, the use of medications, and hospitalizations.


Asunto(s)
Vesículas Extracelulares , Factor de Transcripción STAT3 , Transducción de Señal , Factor de Transcripción STAT3/metabolismo , Vesículas Extracelulares/metabolismo , Humanos , Síndrome de Job/patología , Síndrome de Job/terapia , Síndrome de Job/metabolismo , Sistemas de Liberación de Medicamentos
2.
Front Immunol ; 12: 680334, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34421895

RESUMEN

Background: Inborn errors of immunity (IEI) present with a large phenotypic spectrum of disease, which can pose diagnostic and therapeutic challenges. Suppressor of cytokine signaling 1 (SOCS1) is a key negative regulator of cytokine signaling, and has recently been associated with a novel IEI. Of patients described to date, it is apparent that SOCS1 haploinsufficiency has a pleiotropic effect in humans. Objective: We sought to investigate whether dysregulation of immune pathways, in addition to STAT1, play a role in the broad clinical manifestations of SOCS1 haploinsufficiency. Methods: We assessed impacts of reduced SOCS1 expression across multiple immune cell pathways utilizing patient cells and CRISPR/Cas9 edited primary human T cells. Results: SOCS1 haploinsufficiency phenotypes straddled across the International Union of Immunological Societies classifications of IEI. We found that reduced SOCS1 expression led to dysregulation of multiple intracellular pathways in immune cells. STAT1 phosphorylation is enhanced, comparably with STAT1 gain-of-function mutations, and STAT3 phosphorylation is similarly reduced with concurrent reduction of Th17 cells. Furthermore, reduced SOCS1 E3 ligase function was associated with increased FAK1 in immune cells, and increased AKT and p70 ribosomal protein S6 kinase phosphorylation. We also found Toll-like receptor responses are increased in SOCS1 haploinsufficiency patients. Conclusions: SOCS1 haploinsufficiency is a pleiotropic monogenic IEI. Dysregulation of multiple immune cell pathways may explain the variable clinical phenotype associated with this new condition. Knowledge of these additional dysregulated immune pathways is important when considering the optimum management for SOCS1 haploinsufficient patients.


Asunto(s)
Haploinsuficiencia , Sistema Inmunológico/metabolismo , Transducción de Señal , Proteína 1 Supresora de la Señalización de Citocinas/genética , Proteína 1 Supresora de la Señalización de Citocinas/metabolismo , Alelos , Autoinmunidad , Biomarcadores , Estudios de Casos y Controles , Niño , Preescolar , Citocinas , Femenino , Enfermedades Genéticas Congénitas/diagnóstico , Enfermedades Genéticas Congénitas/genética , Enfermedades Genéticas Congénitas/metabolismo , Humanos , Síndrome de Job/diagnóstico , Síndrome de Job/etiología , Síndrome de Job/metabolismo , Masculino , Modelos Biológicos , Linaje , Linfocitos T/inmunología , Linfocitos T/metabolismo
3.
Proc Natl Acad Sci U S A ; 118(26)2021 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-34172578

RESUMEN

Job syndrome is a rare genetic disorder caused by STAT3 mutations and primarily characterized by immune dysfunction along with comorbid skeleton developmental abnormalities including osteopenia, recurrent fracture of long bones, and scoliosis. So far, there is no definitive cure for the skeletal defects in Job syndrome, and treatments are limited to management of clinical symptoms only. Here, we have investigated the molecular mechanism whereby Stat3 regulates skeletal development and osteoblast differentiation. We showed that removing Stat3 function in the developing limb mesenchyme or osteoprogenitor cells in mice resulted in shortened and bow limbs with multiple fractures in long bones that resembled the skeleton symptoms in the Job Syndrome. However, Stat3 loss did not alter chondrocyte differentiation and hypertrophy in embryonic development, while osteoblast differentiation was severely reduced. Genome-wide transcriptome analyses as well as biochemical and histological studies showed that Stat3 loss resulted in down-regulation of Wnt/ß-catenin signaling. Restoration of Wnt/ß-catenin signaling by injecting BIO, a small molecule inhibitor of GSK3, or crossing with a Lrp5 gain of function (GOF) allele, rescued the bone reduction phenotypes due to Stat3 loss to a great extent. These studies uncover the essential functions of Stat3 in maintaining Wnt/ß-catenin signaling in early mesenchymal or osteoprogenitor cells and provide evidence that bone defects in the Job Syndrome are likely caused by Wnt/ß-catenin signaling reduction due to reduced STAT3 activities in bone development. Enhancing Wnt/ß-catenin signaling could be a therapeutic approach to reduce bone symptoms of Job syndrome patients.


Asunto(s)
Huesos/patología , Síndrome de Job/metabolismo , Síndrome de Job/patología , Células Madre Mesenquimatosas/metabolismo , Factor de Transcripción STAT3/deficiencia , Vía de Señalización Wnt , Alelos , Animales , Cartílago/patología , Diferenciación Celular , Embrión de Mamíferos/patología , Extremidades/patología , Eliminación de Gen , Humanos , Integrasas/metabolismo , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Células Madre Mesenquimatosas/patología , Mesodermo/embriología , Ratones Transgénicos , Osteoblastos/patología , Osteogénesis
4.
Int J Mol Sci ; 21(23)2020 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-33271763

RESUMEN

Hyper-immunoglobulin E syndrome (HIES) is a primary immunodeficiency disease characterized by recurrent Staphylococcus aureus (S. aureus) infections, eczema, skeletal abnormalities and high titers of serum immunoglobulin E. Although the genetic basis of HIES was not known for almost a half century, HIES most frequently exhibits autosomal dominant trait that is transmitted with variable expressivity. Careful genetic studies in recent years identified dominant-negative mutations in human signal transducer and activator of transcription 3 (STAT3) gene as the cause of sporadic and dominant forms of HIES. The STAT3 mutations were localized to DNA-binding, SRC homology 2 (SH2) and transactivating domains and disrupted T helper 17 (TH17) cell differentiation and downstream expression of TH17 cytokines IL-17 and IL-22. Deficiency of IL-17 and IL-22 in turn is responsible for suboptimal expression of anti-staphylococcal host factors, such as neutrophil-recruiting chemokines and antimicrobial peptides, by human keratinocytes and bronchial epithelial cells. TH17 cytokines deficiency thereby explains the recurrent staphylococcal lung and skin infections of HIES patients.


Asunto(s)
Susceptibilidad a Enfermedades , Síndrome de Job/complicaciones , Infecciones Estafilocócicas/etiología , Staphylococcus aureus/inmunología , Animales , Biomarcadores , Citocinas/metabolismo , Dermatitis/diagnóstico , Dermatitis/etiología , Susceptibilidad a Enfermedades/inmunología , Interacciones Huésped-Patógeno , Humanos , Síndrome de Job/diagnóstico , Síndrome de Job/etiología , Síndrome de Job/metabolismo , Proteínas Citotóxicas Formadoras de Poros/biosíntesis , Factor de Transcripción STAT3 , Infecciones Estafilocócicas/diagnóstico , Células Th17/inmunología , Células Th17/metabolismo
5.
Biol Open ; 9(9)2020 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-32580970

RESUMEN

Human induced pluripotent stem cell (iPSC) technology has opened exciting opportunities for stem-cell-based therapy. However, its wide adoption is precluded by several challenges including low reprogramming efficiency and potential for malignant transformation. Better understanding of the molecular mechanisms of the changes that cells undergo during reprograming is needed to improve iPSCs generation efficiency and to increase confidence for their clinical use safety. Here, we find that dominant negative mutations in STAT3 in patients with autosomal-dominant hyper IgE (Job's) syndrome (AD-HIES) result in greatly reduced reprograming efficiency of primary skin fibroblasts derived from skin biopsies. Analysis of normal skin fibroblasts revealed upregulation and phosphorylation of endogenous signal transducer and activator of transcription 3 (STAT3) and its binding to the NANOG promoter following transduction with OKSM factors. This coincided with upregulation of NANOG and appearance of cells expressing pluripotency markers. Upregulation of NANOG and number of pluripotent cells were greatly reduced throughout the reprograming process of AD-HIES fibroblasts that was restored by over-expression of functional STAT3. NANOGP8, the human-specific NANOG retrogene that is often expressed in human cancers, was also induced during reprogramming, to very low but detectable levels, in a STAT3-dependent manner. Our study revealed the critical role of endogenous STAT3 in facilitating reprogramming of human somatic cells.


Asunto(s)
Susceptibilidad a Enfermedades , Inmunoglobulina E/sangre , Síndrome de Job/etiología , Síndrome de Job/metabolismo , Mutación , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Adolescente , Adulto , Anciano , Alelos , Sustitución de Aminoácidos , Células Cultivadas , Niño , Fibroblastos/metabolismo , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Inmunoglobulina E/inmunología , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Mutación con Pérdida de Función , Persona de Mediana Edad , Adulto Joven
6.
J Clin Invest ; 130(8): 4167-4181, 2020 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-32369445

RESUMEN

There are more than 7000 described rare diseases, most lacking specific treatment. Autosomal-dominant hyper-IgE syndrome (AD-HIES, also known as Job's syndrome) is caused by mutations in STAT3. These patients present with immunodeficiency accompanied by severe nonimmunological features, including skeletal, connective tissue, and vascular abnormalities, poor postinfection lung healing, and subsequent pulmonary failure. No specific therapies are available for these abnormalities. Here, we investigated underlying mechanisms in order to identify therapeutic targets. Histological analysis of skin wounds demonstrated delayed granulation tissue formation and vascularization during skin-wound healing in AD-HIES patients. Global gene expression analysis in AD-HIES patient skin fibroblasts identified deficiencies in a STAT3-controlled transcriptional network regulating extracellular matrix (ECM) remodeling and angiogenesis, with hypoxia-inducible factor 1α (HIF-1α) being a major contributor. Consistent with this, histological analysis of skin wounds and coronary arteries from AD-HIES patients showed decreased HIF-1α expression and revealed abnormal organization of the ECM and altered formation of the coronary vasa vasorum. Disease modeling using cell culture and mouse models of angiogenesis and wound healing confirmed these predicted deficiencies and demonstrated therapeutic benefit of HIF-1α-stabilizing drugs. The study provides mechanistic insights into AD-HIES pathophysiology and suggests potential treatment options for this rare disease.


Asunto(s)
Matriz Extracelular/metabolismo , Síndrome de Job/metabolismo , Neovascularización Fisiológica , Piel/metabolismo , Cicatrización de Heridas , Heridas y Lesiones/metabolismo , Animales , Matriz Extracelular/genética , Matriz Extracelular/patología , Femenino , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Síndrome de Job/genética , Síndrome de Job/patología , Masculino , Ratones , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Piel/irrigación sanguínea , Piel/patología , Heridas y Lesiones/genética , Heridas y Lesiones/patología
7.
Allergy ; 74(12): 2394-2405, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31269238

RESUMEN

BACKGROUND: Signal transducer and activator of transcription 3 hyper-IgE syndrome (STAT3-HIES) is caused by heterozygous mutations in the STAT3 gene and is associated with eczema, elevated serum IgE, and recurrent infections resembling severe atopic dermatitis, while clinically relevant specific IgE is almost absent. METHODS: To investigate the impact of STAT3 signaling on B-cell responses, we assessed lymph node and bone marrow, blood B and plasma cell subsets, somatic hypermutations in Ig genes, and in vitro proliferation and antibody production in STAT3-HIES patients and healthy controls. RESULTS: Lymph nodes of STAT3-HIES patients showed normal germinal center architecture and CD138+ plasma cells residing in the paracortex, which expressed IgE, IgG, and IgM but not IgA. IgE+ plasma cells were abundantly present in STAT3-HIES bone marrow. Proliferation of naive B cells upon stimulation with CD40L and IL-4 was similar in patients and controls, while patient cells showed reduced responses to IL-21. IgE, IgG1, IgG3 and IgA1 transcripts showed reduced somatic hypermutations. Peripheral blood IgE+ memory B-cell frequencies were increased in STAT3-HIES, while other memory B-cell frequencies except for IgG4+ cells were decreased. CONCLUSIONS: Despite impaired STAT3 signaling, STAT3-HIES patients can mount in vivo T-cell-dependent B-cell responses, while circulating memory B cells, except for those expressing IgG4 and IgE, were reduced. Reduced molecular maturation demonstrated the critical need of STAT3 signaling for optimal affinity maturation and B-cell differentiation, supporting the need for immunoglobulin substitution therapy and explaining the high IgE serum level in the majority with absent allergic symptoms.


Asunto(s)
Formación de Anticuerpos/inmunología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Inmunoglobulina E/inmunología , Síndrome de Job/etiología , Síndrome de Job/metabolismo , Activación de Linfocitos/inmunología , Factor de Transcripción STAT3/metabolismo , Adolescente , Adulto , Biomarcadores , Niño , Preescolar , Susceptibilidad a Enfermedades , Femenino , Genotipo , Humanos , Inmunoglobulina E/genética , Inmunoglobulina G/genética , Inmunoglobulina G/inmunología , Cadenas Pesadas de Inmunoglobulina/genética , Región Variable de Inmunoglobulina/genética , Memoria Inmunológica , Interleucinas/biosíntesis , Síndrome de Job/diagnóstico , Activación de Linfocitos/genética , Tejido Linfoide/inmunología , Tejido Linfoide/metabolismo , Masculino , Persona de Mediana Edad , Mutación , Células Plasmáticas/inmunología , Células Plasmáticas/metabolismo , Factor de Transcripción STAT3/genética , Transducción de Señal , Adulto Joven
8.
Allergy ; 74(9): 1691-1702, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30793327

RESUMEN

BACKGROUND: Pulmonary complications are responsible for high morbidity and mortality rates in patients with the rare immunodeficiency disorder STAT3 hyper-IgE syndrome (STAT3-HIES). The aim of this study was to expand knowledge about lung disease in STAT3-HIES. METHODS: The course of pulmonary disease, radiological and histopathological interrelations, therapeutic management, and the outcome of 14 STAT3-HIES patients were assessed. RESULTS: The patients' quality of life was compromised most by pulmonary disease. All 14 patients showed first signs of lung disease at a median onset of 1.5 years of age. Lung function revealed a mixed obstructive-restrictive impairment with reduced FEV1 and FVC in 75% of the patients. The severity of lung function impairment was associated with Aspergillus fumigatus infection and prior lung surgery. Severe lung tissue damage, with reduced numbers of ATP-binding cassette sub-family A member 3 (ABCA3) positive type II pneumocytes, was observed in the histological assessment of two deceased patients. Imaging studies of all patients above 6 years of age showed severe airway and parenchyma destruction. Lung surgeries frequently led to complications, including fistula formation. Long-term antifungal and antibacterial treatment proved to be beneficial, as were inhalation therapy, chest physiotherapy, and exercise. Regular immunoglobulin replacement therapy tended to stabilize lung function. CONCLUSIONS: Due to its severity, pulmonary disease in STAT3-HIES patients requires strict monitoring and intensive therapy.


Asunto(s)
Susceptibilidad a Enfermedades , Síndrome de Job/complicaciones , Síndrome de Job/metabolismo , Enfermedades Pulmonares/etiología , Enfermedades Pulmonares/terapia , Factor de Transcripción STAT3/metabolismo , Adolescente , Adulto , Antiinfecciosos/uso terapéutico , Biopsia , Niño , Terapia Combinada , Manejo de la Enfermedad , Femenino , Humanos , Inmunohistoquímica , Síndrome de Job/genética , Síndrome de Job/mortalidad , Enfermedades Pulmonares/diagnóstico , Masculino , Persona de Mediana Edad , Pronóstico , Radiografía Torácica , Pruebas de Función Respiratoria , Factor de Transcripción STAT3/genética , Evaluación de Síntomas , Tomografía Computarizada por Rayos X , Resultado del Tratamiento , Adulto Joven
9.
Immunol Allergy Clin North Am ; 39(1): 49-61, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30466772

RESUMEN

Improvement in genetic testing has allowed specific delineation of several distinct clinical causes characterized by the hyperimmunoglobulin E (IgE) phenotype of eczema, recurrent infections, and elevated serum IgE. Mutations in STAT3, DOCK8, PGM3, ERBIN, IL6ST, and CARD11 cause clinical phenotypes that can present in this manner. This article focuses on loss of function STAT3 mutations causing autosomal-dominant hyper-IgE syndrome and dedicator of cytokinesis 8 deficiency, with discussion of other more recently described diseases.


Asunto(s)
Síndrome de Job/diagnóstico , Síndrome de Job/etiología , Fenotipo , Animales , Biomarcadores , Terapia Combinada , Diagnóstico por Imagen , Susceptibilidad a Enfermedades , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Inmunoglobulina E/sangre , Inmunoglobulina E/inmunología , Síndrome de Job/metabolismo , Síndrome de Job/terapia , Resultado del Tratamiento
10.
Haematologica ; 104(3): 609-621, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30309848

RESUMEN

Hyper-IgE syndromes comprise a group of inborn errors of immunity. STAT3-deficient hyper-IgE syndrome is characterized by elevated serum IgE levels, recurrent infections and eczema, and characteristic skeletal anomalies. A loss-of-function biallelic mutation in IL6ST encoding the GP130 receptor subunit (p.N404Y) has very recently been identified in a singleton patient (herein referred to as PN404Y) as a novel etiology of hyper-IgE syndrome. Here, we studied a patient with hyper-IgE syndrome caused by a novel homozygous mutation in IL6ST (p.P498L; patient herein referred to as PP498L) leading to abrogated GP130 signaling after stimulation with IL-6 and IL-27 in peripheral blood mononuclear cells as well as IL-6 and IL-11 in fibroblasts. Extending the initial identification of selective GP130 deficiency, we aimed to dissect the effects of aberrant cytokine signaling on T-helper cell differentiation in both patients. Our results reveal the importance of IL-6 signaling for the development of CCR6-expressing memory CD4+ T cells (including T-helper 17-enriched subsets) and non-conventional CD8+T cells which were reduced in both patients. Downstream functional analysis of the GP130 mutants (p.N404Y and p.P498L) have shown differences in response to IL-27, with the p.P498L mutation having a more severe effect that is reflected by reduced T-helper 1 cells in this patient (PP498L) only. Collectively, our data suggest that characteristic features of GP130-deficient hyper-IgE syndrome phenotype are IL-6 and IL-11 dominated, and indicate selective roles of aberrant IL-6 and IL-27 signaling on the differentiation of T-cell subsets.


Asunto(s)
Receptor gp130 de Citocinas/genética , Síndrome de Job/diagnóstico , Síndrome de Job/etiología , Mutación con Pérdida de Función , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Biomarcadores , Diferenciación Celular/genética , Niño , Preescolar , Receptor gp130 de Citocinas/química , Análisis Mutacional de ADN , Susceptibilidad a Enfermedades , Estudios de Asociación Genética , Humanos , Inmunofenotipificación , Síndrome de Job/metabolismo , Activación de Linfocitos , Masculino , Modelos Moleculares , Linaje , Fenotipo , Conformación Proteica , Radiografía
11.
Mamm Genome ; 29(7-8): 603-617, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30094507

RESUMEN

Spectacular progress has been made in the characterization of human hyper-IgE syndrome (HIES) over the last 50 years. HIES is a primary immunodeficiency defined as an association of atopy in a context of very high serum IgE levels, characteristic bacterial and fungal diseases, low-level clinical and biological inflammation, and various non-hematopoietic developmental manifestations. Somewhat arbitrarily, three disorders were successively put forward as the underlying cause of HIES: autosomal dominant (AD) STAT3 deficiency, the only disorder corresponding to the original definition of HIES, and autosomal recessive (AR) DOCK8 and PGM3 deficiencies, in which atopy and high serum IgE levels occur in a context of manifestations not seen in patients with typical HIES. Indeed, these three disorders disrupt different molecular pathways, affect different cell types, and underlie different clinical phenotypes. Surprisingly, several other inherited inborn errors of immunity in which serum IgE levels are high, sometimes almost as high as those in HIES patients, are not considered to belong to the HIES group of diseases. Studies of HIES have been further complicated by the lack of a high serum IgE phenotype in all mouse models of the disease other than two Stat3 mutant strains. The study of infections in mutant mice has helped elucidate only some forms of HIES and infection. Mouse models of these conditions have also been used to study non-hematopoietic phenotypes for STAT3 deficiency, tissue-specific immunity for DOCK8 deficiency, and cell lineage maturation for PGM3 deficiency. We review here the history of the field of HIES since the first clinical description of this condition in 1966, together with the three disorders commonly referred to as HIES, focusing, in particular, on their mouse models. We propose the restriction of the term "HIES" to patients with an AD STAT3-deficiency phenotype, including the most recently described AR ZNF341 deficiency, thus excluding AR DOCK8 and PGM3 deficiencies from the definition of this disease.


Asunto(s)
Susceptibilidad a Enfermedades , Síndrome de Job/etiología , Síndrome de Job/metabolismo , Animales , Formación de Anticuerpos/genética , Formación de Anticuerpos/inmunología , Biomarcadores , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Factores de Intercambio de Guanina Nucleótido/deficiencia , Humanos , Inmunoglobulina E/inmunología , Síndrome de Job/diagnóstico , Fenotipo , Fosfoglucomutasa/deficiencia , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal
12.
Front Immunol ; 9: 3047, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30671054

RESUMEN

The induction and action of type I interferon (IFN) is of fundamental importance in human immune defenses toward microbial pathogens, particularly viruses. Basic discoveries within the molecular and cellular signaling pathways regulating type I IFN induction and downstream actions have shown the essential role of the IFN regulatory factor (IRF) and the signal transducer and activator of transcription (STAT) families, respectively. However, the exact biological and immunological functions of these factors have been most clearly revealed through the study of inborn errors of immunity and the resultant infectious phenotypes in humans. The spectrum of human inborn errors of immunity caused by mutations in IRFs and STATs has proven very diverse. These diseases encompass herpes simplex encephalitis (HSE) and severe influenza in IRF3- and IRF7/IRF9 deficiency, respectively. They also include Mendelian susceptibility to mycobacterial infection (MSMD) in STAT1 deficiency, through disseminated measles infection associated with STAT2 deficiency, and finally staphylococcal abscesses and chronic mucocutaneous candidiasis (CMC) classically described with Hyper-IgE syndrome (HIES) in the case of STAT3 deficiency. More recently, increasing focus has been on aspects of autoimmunity and autoinflammation playing an important part in many primary immunodeficiency diseases (PID)s, as exemplified by STAT1 gain-of-function causing CMC and autoimmune thyroiditis, as well as a recently described autoinflammatory syndrome with hypogammaglobulinemia and lymphoproliferation as a result of STAT3 gain-of-function. Here I review the infectious, inflammatory, and autoimmune disorders arising from mutations in IRF and STAT transcription factors in humans, highlightning the underlying molecular mechanisms and immunopathogenesis as well as the clinical/therapeutic perspectives of these new insights.


Asunto(s)
Candidiasis Mucocutánea Crónica/metabolismo , Encefalitis por Herpes Simple/metabolismo , Gripe Humana/metabolismo , Factores Reguladores del Interferón/metabolismo , Síndrome de Job/metabolismo , Infecciones por Mycobacterium/metabolismo , Factores de Transcripción STAT/metabolismo , Autoinmunidad , Candidiasis Mucocutánea Crónica/genética , Candidiasis Mucocutánea Crónica/inmunología , Encefalitis por Herpes Simple/genética , Encefalitis por Herpes Simple/inmunología , Humanos , Inmunidad Innata , Gripe Humana/genética , Gripe Humana/inmunología , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/inmunología , Interferón Tipo I/inmunología , Interferón Tipo I/metabolismo , Quinasas Janus/metabolismo , Síndrome de Job/genética , Síndrome de Job/inmunología , Mutación , Infecciones por Mycobacterium/genética , Infecciones por Mycobacterium/inmunología , Receptor de Interferón alfa y beta/metabolismo , Factores de Transcripción STAT/genética , Factores de Transcripción STAT/inmunología
13.
Blood ; 128(26): 3061-3072, 2016 12 29.
Artículo en Inglés | MEDLINE | ID: mdl-27799162

RESUMEN

Autosomal dominant hyper-IgE syndrome (AD-HIES) is caused by dominant-negative mutations in STAT3; however, the molecular basis for mutant STAT3 allele dysfunction is unclear and treatment remains supportive. We hypothesized that AD-HIES mutations decrease STAT3 protein stability and that mutant STAT3 activity can be improved by agents that increase chaperone protein activity. We used computer modeling to characterize the effect of STAT3 mutations on protein stability. We measured STAT3 protein half-life (t1/2) and determined levels of STAT3 phosphorylated on tyrosine (Y) 705 (pY-STAT3) and mRNA levels of STAT3 gene targets in Epstein-Barr virus-transformed B (EBV) cells, human peripheral blood mononuclear cells (PBMCs), and mouse splenocytes incubated without or with chaperone protein modulators-HSF1A, a small-molecule TRiC modulator, or geranylgeranylacetone (GGA), a drug that upregulates heat shock protein (HSP) 70 and HSP90. Computer modeling predicted that 81% of AD-HIES mutations are destabilizing. STAT3 protein t1/2 in EBV cells from AD-HIES patients with destabilizing STAT3 mutations was markedly reduced. Treatment of EBV cells containing destabilizing STAT3 mutations with either HSF1A or GGA normalized STAT3 t1/2, increased pY-STAT3 levels, and increased mRNA levels of STAT3 target genes up to 79% of control. In addition, treatment of human PBMCs or mouse splenocytes containing destabilizing STAT3 mutations with either HSF1A or GGA increased levels of cytokine-activated pY-STAT3 within human CD4+ and CD8+ T cells and numbers of IL-17-producing CD4+ mouse splenocytes, respectively. Thus, most AD-HIES STAT3 mutations are destabilizing; agents that modulate chaperone protein function improve STAT3 stability and activity in T cells and may provide a specific treatment.


Asunto(s)
Síndrome de Job/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Citocinas/farmacología , Proteínas de Unión al ADN/metabolismo , Diterpenos/farmacología , Semivida , Factores de Transcripción del Choque Térmico , Herpesvirus Humano 4/fisiología , Humanos , Interleucina-17/metabolismo , Síndrome de Job/patología , Ratones , Modelos Moleculares , Proteínas Mutantes/metabolismo , Mutación/genética , Fosfotirosina/metabolismo , Unión Proteica/efectos de los fármacos , Estabilidad Proteica/efectos de los fármacos , Factor de Transcripción STAT3/química , Factor de Transcripción STAT3/genética , Bazo/patología , Factores de Transcripción/metabolismo
14.
Clin Exp Allergy ; 46(12): 1564-1574, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27474157

RESUMEN

BACKGROUND: STAT1 mutations cause chronic mucocutaneous candidiasis (CMC), while STAT3 mutations cause hyper-IgE syndrome (HIES). CMC and HIES patients have T helper (Th) 17 defects suffering from mucosal Candida infections, but only patients with HIES show an allergic phenotype with eczema, eosinophilia and high IgE levels. OBJECTIVE: We investigated whether differential Th2 and Th9 responses may explain the clinical differences. METHODS: Peripheral blood mononuclear cells of patients with CMC (n = 4), patients with HIES (n = 4), patients with atopic dermatitis (n = 4) and healthy volunteers (n = 13) were stimulated with Candida and Staphylococcus aureus, with and without IL-4. The cytokines IL-5, IL-13, IL-9, IL-17 and TGFß and regulatory T cells were measured in cell culture supernatants by ELISA or flow cytometry, respectively. RESULTS: Peripheral blood mononuclear cells of patients with CMC showed a significantly impaired production of the Th2 cytokines IL-5 and IL-13, especially in the presence of IL-4. Moreover, IL-9 production was significantly lower in patients with CMC compared to healthy controls. In contrast, patients with HIES and patients with AD showed normal IL-5 and IL-13 production, while IL-9 production was significantly lower in patients with HIES compared to healthy controls. Although TGFß was involved in the IL-4-induced IL-9 production, TGFß levels and the frequency of regulatory T cells did not differ between patients with HIES and controls. Flow cytometry analysis demonstrated an IL-9+ IL-17+ CD4+ subset in healthy controls after stimulation with Candida which was less present in patients with HIES. CONCLUSION: Patients with CMC have a general Th defect including Th2 and Th9, while patients with HIES have normal Th2 cytokines. These differences are in line with their clinical presentation. Surprisingly, the allergic cytokine IL-9 was deficient in both HIES and CMC, suggesting a Th-17-derived origin.


Asunto(s)
Candidiasis Mucocutánea Crónica/diagnóstico , Candidiasis Mucocutánea Crónica/inmunología , Síndrome de Job/diagnóstico , Síndrome de Job/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Adulto , Anciano , Biomarcadores , Candidiasis Mucocutánea Crónica/metabolismo , Candidiasis Mucocutánea Crónica/terapia , Estudios de Casos y Controles , Citocinas/sangre , Citocinas/metabolismo , Eosinófilos/inmunología , Eosinófilos/metabolismo , Femenino , Humanos , Inmunoglobulina E/sangre , Inmunoglobulina E/inmunología , Síndrome de Job/metabolismo , Síndrome de Job/terapia , Recuento de Leucocitos , Masculino , Persona de Mediana Edad , Fenotipo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Colaboradores-Inductores/metabolismo , Células Th2/inmunología , Células Th2/metabolismo
16.
Glycoconj J ; 33(3): 447-56, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26687240

RESUMEN

Glycans serve as important regulators of antibody activities and half-lives. IgE is the most heavily glycosylated antibody, but in comparison to other antibodies little is known about its glycan structure function relationships. We therefore describe the site specific IgE glycosylation from a patient with a novel hyper IgE syndrome linked to mutations in PGM3, which is an enzyme involved in synthesizing UDP-GlcNAc, a sugar donor widely required for glycosylation. A two-step method was developed to prepare two IgE samples from less than 1 mL of serum collected from a patient with PGM3 mutation and a patient with atopic dermatitis as a control subject. Then, a glycoproteomic strategy was used to study the site-specific glycosylation. No glycosylation was found at Asn264, whilst high mannose glycans were only detected at Asn275, tri-antennary glycans were exclusively observed at Asn99 and Asn252, and non-fucosylated complex glycans were detected at Asn99. The results showed similar glycosylation profiles between the two IgE samples. These observations, together with previous knowledge of IgE glycosylation, imply that IgE glycosylation is similarly regulated among healthy control, allergy and PGM3 related hyper IgE syndrome.


Asunto(s)
Inmunoglobulina E/metabolismo , Síndrome de Job/metabolismo , Mutación , Fosfoglucomutasa/metabolismo , Procesamiento Proteico-Postraduccional , Sitios de Unión , Glicoproteínas/química , Glicoproteínas/metabolismo , Glicosilación , Humanos , Inmunoglobulina E/química , Síndrome de Job/diagnóstico , Síndrome de Job/genética , Espectrometría de Masas/métodos , Técnicas de Diagnóstico Molecular/métodos , Fosfoglucomutasa/química , Fosfoglucomutasa/genética , Proteoma/química , Proteoma/metabolismo
17.
Int Rev Immunol ; 35(1): 39-56, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-25970001

RESUMEN

In recent years a number of primary immunodeficiencies (PIDs) characterized by elevated Immunoglobulin E (IgE) levels have been uncovered and termed as Hyper-IgE syndrome (HIES). In addition to the elevated levels of IgE, patients with these PIDs display a spectrum of infections by staphylococci and fungi, and in some cases viruses, particularly affecting skin and lungs. Most of these PIDs also have a non-infectious phenotype, comprising musculoskeletal, vascular, and neurological abnormalities. The genetic basis for the majority of conditions with elevated IgE has now been established and includes mutations in STAT3, DOCK8, TYK2, and most recently PGM3 molecules. However, in some patients with the relevant phenotype, mutations in these molecules are not identified, suggesting additional genetic etiologies of HIES not yet discovered. As the immunological and molecular basis of HIES is being unraveled, important insights are emerging that may have implications for our understanding of basic principles of immunology and protective immunity as well as for the pathogenesis and clinical management of patients with these complex and challenging PIDs. In this review, are presented the current knowledge on the clinical presentation, infectious phenotype, and the genetic and immunological pathogenesis of hyper-IgE syndromes as well as some other PIDs with elevated levels of IgE.


Asunto(s)
Citocinas/metabolismo , Inmunoglobulina E/metabolismo , Síndrome de Job , Inmunodeficiencia Combinada Grave , Antibacterianos/uso terapéutico , Antifúngicos/uso terapéutico , Antivirales/uso terapéutico , Linfocitos B/inmunología , Linfocitos B/metabolismo , Trasplante de Médula Ósea , Citocinas/inmunología , Diagnóstico Diferencial , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/inmunología , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Inmunoglobulina E/sangre , Síndrome de Job/diagnóstico , Síndrome de Job/genética , Síndrome de Job/metabolismo , Síndrome de Job/terapia , Mutación , Fenotipo , Fosfoglucomutasa/genética , Fosfoglucomutasa/inmunología , Fosfoglucomutasa/metabolismo , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/inmunología , Factor de Transcripción STAT3/metabolismo , Inmunodeficiencia Combinada Grave/diagnóstico , Inmunodeficiencia Combinada Grave/genética , Inmunodeficiencia Combinada Grave/metabolismo , Inmunodeficiencia Combinada Grave/terapia , Transducción de Señal , Linfocitos T/inmunología , Linfocitos T/metabolismo , TYK2 Quinasa/genética , TYK2 Quinasa/inmunología , TYK2 Quinasa/metabolismo
18.
Blood ; 124(3): 403-11, 2014 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-24891320

RESUMEN

Signal transducer and activator of transcription 3 (STAT3) is considered a negative regulator of inflammation, as inhibition of STAT3 signaling enhances antitumor immunity. However, STAT3 activation is a key oncogenic pathway in natural killer (NK)-lineage large granular lymphomas, and we recently reported enhanced proliferation and function of human NK cells activated with IL-21, which signals primarily through STAT3. These IL-21-expanded NK cells also have increased NKG2D expression, which led us to focus our investigation on whether STAT3 regulates NKG2D. In this study, we show that modulation of STAT3 phosphorylation with cytokines and small-molecule inhibitors correlates with NKG2D expression on human NK cells, leading to altered NK-cell degranulation. Moreover, NKG2D expression on murine NK cells having conditional STAT3 ablation is lower than on NK cells from wild-type mice, and human NK cells carrying dominant-negative STAT3 mutations have decreased baseline NKG2D expression and blunted responses to IL-10 and IL-21. Lastly, we show binding of STAT3 to a predicted STAT3 binding site upstream of the NKG2D gene, which is enhanced by IL-10 and IL-21 and decreased by STAT3 inhibition. Taken together, these data show that NKG2D expression in NK cells is regulated at the transcriptional level by STAT3, resulting in a functional NK cell defect in patients with STAT3 mutations.


Asunto(s)
Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Subfamilia K de Receptores Similares a Lectina de Células NK/genética , Factor de Transcripción STAT3/metabolismo , Animales , Secuencia de Bases , Sitios de Unión/genética , ADN/genética , ADN/metabolismo , Humanos , Interleucina-10/metabolismo , Interleucina-15/metabolismo , Interleucinas/metabolismo , Síndrome de Job/genética , Síndrome de Job/inmunología , Síndrome de Job/metabolismo , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Mutación , Fosforilación , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción STAT3/deficiencia , Factor de Transcripción STAT3/genética , Transducción de Señal , Transcripción Genética , Tirosina/metabolismo
19.
J Immunol ; 191(4): 1845-55, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23836059

RESUMEN

Iron is a trace element important for the proper folding and function of various proteins. Physiological regulation of iron stores is of critical importance for RBC production and antimicrobial defense. Hepcidin is a key regulator of iron levels within the body. Under conditions of iron deficiency, hepcidin expression is reduced to promote increased iron uptake from the diet and release from cells, whereas during conditions of iron excess, induction of hepcidin restricts iron uptake and movement within the body. The cytokine IL-6 is well established as an important inducer of hepcidin. The presence of this cytokine during inflammatory states can induce hepcidin production, iron deficiency, and anemia. In this study, we show that IL-22 also influences hepcidin production in vivo. Injection of mice with exogenous mouse IgG1 Fc fused to the N terminus of mouse IL-22 (Fc-IL-22), an IL-22R agonist with prolonged and enhanced functional potency, induced hepcidin production, with a subsequent decrease in circulating serum iron and hemoglobin levels and a concomitant increase in iron accumulation within the spleen. This response was independent of IL-6 and was attenuated in the absence of the IL-22R-associated signaling kinase, Tyk2. Ab-mediated blockade of hepcidin partially reversed the effects on iron biology caused by IL-22R stimulation. Taken together, these data suggest that exogenous IL-22 regulates hepcidin production to physiologically influence iron usage.


Asunto(s)
Hepcidinas/fisiología , Interleucinas/fisiología , Hierro/metabolismo , Secuencia de Aminoácidos , Anemia Ferropénica/sangre , Anemia Ferropénica/inducido químicamente , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/uso terapéutico , Células Cultivadas , Femenino , Hepatocitos/metabolismo , Hepcidinas/antagonistas & inhibidores , Hepcidinas/biosíntesis , Hepcidinas/genética , Hepcidinas/inmunología , Humanos , Fragmentos Fc de Inmunoglobulinas/genética , Inmunoglobulina G/genética , Interleucina-6/fisiología , Interleucinas/genética , Interleucinas/farmacología , Interleucinas/toxicidad , Hierro/sangre , Deficiencias de Hierro , Síndrome de Job/metabolismo , Hígado/metabolismo , Hígado/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones SCID , Datos de Secuencia Molecular , Fosforilación , Procesamiento Proteico-Postraduccional , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores de IgG/deficiencia , Receptores de Interleucina/agonistas , Receptores de Interleucina/fisiología , Proteínas Recombinantes de Fusión/farmacología , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Bazo/metabolismo , Bazo/patología , TYK2 Quinasa/deficiencia , TYK2 Quinasa/metabolismo , Interleucina-22
20.
Scand J Immunol ; 78(3): 258-65, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23659370

RESUMEN

Hyper-IgE syndrome (HIES) is a rare primary immunodeficiency disease characterized by eczema, recurrent staphylococcal aureus skin abscesses, pneumonia with pneumatocele formation, remarkably high serum IgE levels, eosinophilia and involvement of skeleton and connective tissues. Heterozygous signal transducer and activator of transcription 3 (STAT3) mutations were shown to be the cause of autosomal dominant HIES (AD-HIES). In this study, we diagnosed nine patients with HIES from 9 unrelated families on the basis of a National Institutes of Health (NIH) score of ≥40 points, sequenced the STAT3 gene of all nine patients, and quantified Th17 cells in peripheral blood of seven patients by flow cytometry in mainland China. All nine patients had characteristic manifestation of HIES with the range of NIH scores 45-77 points. STAT3 hot mutations V637M or R382W/Q were identified in five patients. We identified two novel heterozygous missense mutations (T620S and R609G) located in Src homology 2 (SH2) domain in two patients, respectively. In two other patients, no STAT3 mutations were found. Quantified Th17 cell numbers were markedly decreased or absent (0-0.28% of CD4(+) T cells) in six patients with STAT3 mutations and almost normal (0.53% of CD4(+) T cells) in one wild-type STAT3 patient compared with healthy controls (0.40-2.25% of CD4(+) T cells). These results suggest that not all patients with HIES who had NIH scores over 40 points carry STAT3 mutations, those whose Th17 cell numbers strikingly decreased probably had AD-HIES with STAT3 mutations.


Asunto(s)
Inmunoglobulina E/sangre , Síndrome de Job/genética , Síndrome de Job/inmunología , Factor de Transcripción STAT3/genética , Células Th17/inmunología , Adolescente , Niño , Preescolar , China , Femenino , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Lactante , Síndrome de Job/metabolismo , Recuento de Linfocitos , Masculino , Infecciones Estafilocócicas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...