Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros











Intervalo de año de publicación
1.
Science ; 376(6599): eabm6380, 2022 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-35587511

RESUMEN

The molecular basis of interindividual clinical variability upon infection with Staphylococcus aureus is unclear. We describe patients with haploinsufficiency for the linear deubiquitinase OTULIN, encoded by a gene on chromosome 5p. Patients suffer from episodes of life-threatening necrosis, typically triggered by S. aureus infection. The disorder is phenocopied in patients with the 5p- (Cri-du-Chat) chromosomal deletion syndrome. OTULIN haploinsufficiency causes an accumulation of linear ubiquitin in dermal fibroblasts, but tumor necrosis factor receptor-mediated nuclear factor κB signaling remains intact. Blood leukocyte subsets are unaffected. The OTULIN-dependent accumulation of caveolin-1 in dermal fibroblasts, but not leukocytes, facilitates the cytotoxic damage inflicted by the staphylococcal virulence factor α-toxin. Naturally elicited antibodies against α-toxin contribute to incomplete clinical penetrance. Human OTULIN haploinsufficiency underlies life-threatening staphylococcal disease by disrupting cell-intrinsic immunity to α-toxin in nonleukocytic cells.


Asunto(s)
Toxinas Bacterianas , Síndrome del Maullido del Gato , Endopeptidasas , Haploinsuficiencia , Proteínas Hemolisinas , Infecciones Estafilocócicas , Staphylococcus aureus , Toxinas Bacterianas/inmunología , Síndrome del Maullido del Gato/genética , Síndrome del Maullido del Gato/inmunología , Endopeptidasas/genética , Haploinsuficiencia/genética , Haploinsuficiencia/inmunología , Proteínas Hemolisinas/inmunología , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Celular/genética , Necrosis , Infecciones Estafilocócicas/genética , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/patología
2.
Nat Med ; 27(9): 1646-1654, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34183838

RESUMEN

The pathophysiology of adverse events following programmed cell death protein 1 (PD-1) blockade, including tuberculosis (TB) and autoimmunity, remains poorly characterized. We studied a patient with inherited PD-1 deficiency and TB who died of pulmonary autoimmunity. The patient's leukocytes did not express PD-1 or respond to PD-1-mediated suppression. The patient's lymphocytes produced only small amounts of interferon (IFN)-γ upon mycobacterial stimuli, similarly to patients with inborn errors of IFN-γ production who are vulnerable to TB. This phenotype resulted from a combined depletion of Vδ2+ γδ T, mucosal-associated invariant T and CD56bright natural killer lymphocytes and dysfunction of other T lymphocyte subsets. Moreover, the patient displayed hepatosplenomegaly and an expansion of total, activated and RORγT+ CD4-CD8- double-negative αß T cells, similar to patients with STAT3 gain-of-function mutations who display lymphoproliferative autoimmunity. This phenotype resulted from excessive amounts of STAT3-activating cytokines interleukin (IL)-6 and IL-23 produced by activated T lymphocytes and monocytes, and the STAT3-dependent expression of RORγT by activated T lymphocytes. Our work highlights the indispensable role of human PD-1 in governing both antimycobacterial immunity and self-tolerance, while identifying potentially actionable molecular targets for the diagnostic and therapeutic management of TB and autoimmunity in patients on PD-1 blockade.


Asunto(s)
Autoinmunidad/genética , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Receptor de Muerte Celular Programada 1/genética , Factor de Transcripción STAT3/genética , Tuberculosis/inmunología , Autoinmunidad/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Antígeno CD56/genética , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Niño , Humanos , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunoterapia/efectos adversos , Interleucina-23/genética , Interleucina-6/genética , Linfocitos Intraepiteliales/inmunología , Linfocitos Intraepiteliales/patología , Masculino , Mycobacterium tuberculosis/patogenicidad , Neoplasias/complicaciones , Neoplasias/tratamiento farmacológico , Neoplasias/mortalidad , Receptor de Muerte Celular Programada 1/deficiencia , Tuberculosis/genética , Tuberculosis/mortalidad
3.
Proc Natl Acad Sci U S A ; 118(15)2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33876776

RESUMEN

Human inborn errors of IFN-γ underlie mycobacterial disease, due to insufficient IFN-γ production by lymphoid cells, impaired myeloid cell responses to this cytokine, or both. We report four patients from two unrelated kindreds with intermittent monocytosis and mycobacterial disease, including bacillus Calmette-Guérin-osis and disseminated tuberculosis, and without any known inborn error of IFN-γ. The patients are homozygous for ZNFX1 variants (p.S959* and p.E1606Rfs*10) predicted to be loss of function (pLOF). There are no subjects homozygous for pLOF variants in public databases. ZNFX1 is a conserved and broadly expressed helicase, but its biology remains largely unknown. It is thought to act as a viral double-stranded RNA sensor in mice, but these patients do not suffer from severe viral illnesses. We analyze its subcellular localization upon overexpression in A549 and HeLa cell lines and upon stimulation of THP1 and fibroblastic cell lines. We find that this cytoplasmic protein can be recruited to or even induce stress granules. The endogenous ZNFX1 protein in cell lines of the patient homozygous for the p.E1606Rfs*10 variant is truncated, whereas ZNFX1 expression is abolished in cell lines from the patients with the p.S959* variant. Lymphocyte subsets are present at normal frequencies in these patients and produce IFN-γ normally. The hematopoietic and nonhematopoietic cells of the patients tested respond normally to IFN-γ. Our results indicate that human ZNFX1 is associated with stress granules and essential for both monocyte homeostasis and protective immunity to mycobacteria.


Asunto(s)
Antígenos de Neoplasias/genética , Leucocitosis/genética , Infecciones por Mycobacterium no Tuberculosas/genética , Células A549 , Adolescente , Antígenos de Neoplasias/metabolismo , Células Cultivadas , Niño , Gránulos Citoplasmáticos/metabolismo , Femenino , Células HEK293 , Células HeLa , Homocigoto , Humanos , Lactante , Interferón gamma/metabolismo , Leucocitosis/patología , Masculino , Mutación , Infecciones por Mycobacterium no Tuberculosas/patología , Linaje , Células THP-1 , Adulto Joven
4.
J Clin Invest ; 129(2): 583-597, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30422821

RESUMEN

X-linked dominant incontinentia pigmenti (IP) and X-linked recessive anhidrotic ectodermal dysplasia with immunodeficiency (EDA-ID) are caused by loss-of-function and hypomorphic IKBKG (also known as NEMO) mutations, respectively. We describe a European mother with mild IP and a Japanese mother without IP, whose 3 boys with EDA-ID died from ID. We identify the same private variant in an intron of IKBKG, IVS4+866 C>T, which was inherited from and occurred de novo in the European mother and Japanese mother, respectively. This mutation creates a new splicing donor site, giving rise to a 44-nucleotide pseudoexon (PE) generating a frameshift. Its leakiness accounts for NF-κB activation being impaired but not abolished in the boys' cells. However, aberrant splicing rates differ between cell types, with WT NEMO mRNA and protein levels ranging from barely detectable in leukocytes to residual amounts in induced pluripotent stem cell-derived (iPSC-derived) macrophages, and higher levels in fibroblasts and iPSC-derived neuronal precursor cells. Finally, SRSF6 binds to the PE, facilitating its inclusion. Moreover, SRSF6 knockdown or CLK inhibition restores WT NEMO expression and function in mutant cells. A recurrent deep intronic splicing mutation in IKBKG underlies a purely quantitative NEMO defect in males that is most severe in leukocytes and can be rescued by the inhibition of SRSF6 or CLK.


Asunto(s)
Displasia Ectodérmica , Mutación del Sistema de Lectura , Quinasa I-kappa B , Incontinencia Pigmentaria , Intrones , Displasia Ectodérmica/genética , Displasia Ectodérmica/metabolismo , Displasia Ectodérmica/patología , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Quinasa I-kappa B/deficiencia , Quinasa I-kappa B/metabolismo , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/metabolismo , Síndromes de Inmunodeficiencia/patología , Incontinencia Pigmentaria/genética , Incontinencia Pigmentaria/metabolismo , Incontinencia Pigmentaria/patología , Macrófagos/metabolismo , Macrófagos/patología , Masculino
5.
Cell ; 168(5): 789-800.e10, 2017 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-28235196

RESUMEN

The molecular basis of the incomplete penetrance of monogenic disorders is unclear. We describe here eight related individuals with autosomal recessive TIRAP deficiency. Life-threatening staphylococcal disease occurred during childhood in the proband, but not in the other seven homozygotes. Responses to all Toll-like receptor 1/2 (TLR1/2), TLR2/6, and TLR4 agonists were impaired in the fibroblasts and leukocytes of all TIRAP-deficient individuals. However, the whole-blood response to the TLR2/6 agonist staphylococcal lipoteichoic acid (LTA) was abolished only in the index case individual, the only family member lacking LTA-specific antibodies (Abs). This defective response was reversed in the patient, but not in interleukin-1 receptor-associated kinase 4 (IRAK-4)-deficient individuals, by anti-LTA monoclonal antibody (mAb). Anti-LTA mAb also rescued the macrophage response in mice lacking TIRAP, but not TLR2 or MyD88. Thus, acquired anti-LTA Abs rescue TLR2-dependent immunity to staphylococcal LTA in individuals with inherited TIRAP deficiency, accounting for incomplete penetrance. Combined TIRAP and anti-LTA Ab deficiencies underlie staphylococcal disease in this patient.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Lipopolisacáridos/metabolismo , Glicoproteínas de Membrana/deficiencia , Receptores de Interleucina-1/deficiencia , Infecciones Estafilocócicas/genética , Infecciones Estafilocócicas/inmunología , Ácidos Teicoicos/metabolismo , Inmunidad Adaptativa , Niño , Femenino , Fibroblastos/metabolismo , Humanos , Inmunidad Innata , Lipopolisacáridos/inmunología , Macrófagos/inmunología , Masculino , Glicoproteínas de Membrana/análisis , Glicoproteínas de Membrana/genética , Monocitos/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Linaje , Fagocitos/metabolismo , Mutación Puntual , Isoformas de Proteínas/análisis , Isoformas de Proteínas/genética , Receptores de Interleucina-1/análisis , Receptores de Interleucina-1/genética , Infecciones Estafilocócicas/tratamiento farmacológico , Ácidos Teicoicos/inmunología , Receptor Toll-Like 2/metabolismo , Receptores Toll-Like/agonistas , Receptores Toll-Like/metabolismo
6.
Am J Hum Genet ; 99(2): 407-13, 2016 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-27426734

RESUMEN

Cardiospondylocarpofacial (CSCF) syndrome is characterized by growth retardation, dysmorphic facial features, brachydactyly with carpal-tarsal fusion and extensive posterior cervical vertebral synostosis, cardiac septal defects with valve dysplasia, and deafness with inner ear malformations. Whole-exome sequencing identified heterozygous MAP3K7 mutations in six distinct CSCF-affected individuals from four families and ranging in age from 5 to 37 years. MAP3K7 encodes transforming growth factor ß (TGF-ß)-activated kinase 1 (TAK1), which is involved in the mitogen-activated protein kinase (MAPK)-p38 signaling pathway. MAPK-p38 signaling was markedly altered when expression of non-canonical TGF-ß-driven target genes was impaired. These findings support the loss of transcriptional control of the TGF-ß-MAPK-p38 pathway in fibroblasts obtained from affected individuals. Surprisingly, although TAK1 is located at the crossroad of inflammation, immunity, and cancer, this study reports MAP3K7 mutations in a developmental disorder affecting mainly cartilage, bone, and heart.


Asunto(s)
Huesos del Carpo/anomalías , Vértebras Cervicales/anomalías , Pérdida Auditiva Conductiva/genética , Heterocigoto , Quinasas Quinasa Quinasa PAM/genética , Insuficiencia de la Válvula Mitral/genética , Mutación/genética , Huesos Tarsianos/anomalías , Anomalías Múltiples , Adolescente , Adulto , Niño , Preescolar , Femenino , Fibroblastos , Regulación de la Expresión Génica , Pérdida Auditiva Bilateral , Humanos , Interleucina-1beta/metabolismo , Sistema de Señalización de MAP Quinasas , Masculino , Osteosclerosis , Síndrome , Factor de Crecimiento Transformador beta/metabolismo , Adulto Joven , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
8.
J Exp Med ; 212(6): 939-51, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-26008899

RESUMEN

Inherited, complete deficiency of human HOIL-1, a component of the linear ubiquitination chain assembly complex (LUBAC), underlies autoinflammation, infections, and amylopectinosis. We report the clinical description and molecular analysis of a novel inherited disorder of the human LUBAC complex. A patient with multiorgan autoinflammation, combined immunodeficiency, subclinical amylopectinosis, and systemic lymphangiectasia, is homozygous for a mutation in HOIP, the gene encoding the catalytic component of LUBAC. The missense allele (L72P, in the PUB domain) is at least severely hypomorphic, as it impairs HOIP expression and destabilizes the whole LUBAC complex. Linear ubiquitination and NF-κB activation are impaired in the patient's fibroblasts stimulated by IL-1ß or TNF. In contrast, the patient's monocytes respond to IL-1ß more vigorously than control monocytes. However, the activation and differentiation of the patient's B cells are impaired in response to CD40 engagement. These cellular and clinical phenotypes largely overlap those of HOIL-1-deficient patients. Clinical differences between HOIL-1- and HOIP-mutated patients may result from differences between the mutations, the loci, or other factors. Our findings show that human HOIP is essential for the assembly and function of LUBAC and for various processes governing inflammation and immunity in both hematopoietic and nonhematopoietic cells.


Asunto(s)
Regulación de la Expresión Génica , Ubiquitina-Proteína Ligasas/deficiencia , Alelos , Secuencia de Aminoácidos , Ligando de CD40/metabolismo , Catálisis , Femenino , Fibroblastos/metabolismo , Prueba de Complementación Genética , Mutación de Línea Germinal , Enfermedad del Almacenamiento de Glucógeno Tipo IV/patología , Homocigoto , Humanos , Síndromes de Inmunodeficiencia/patología , Inflamación , Linfangiectasia/patología , Monocitos/metabolismo , Mutación Missense , FN-kappa B/metabolismo , Homología de Secuencia de Aminoácido , Factores de Transcripción , Ubiquitina/química , Ubiquitina-Proteína Ligasas/genética , Adulto Joven
9.
Blood ; 120(25): 4992-5001, 2012 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-23002119

RESUMEN

We studied the distribution of peripheral B-cell subsets in patients deficient for key factors of the TLR-signaling pathways (MyD88, TIRAP/MAL, IL-1 receptor-associated kinase 4 [IRAK-4], TLR3, UNC-93B, TRIF). All TLRs, except TLR3, which signals through the TRIF adaptor, require MyD88 and IRAK-4 to mediate their function. TLR4 and the TLR2 heterodimers (with TLR1, TLR6, and possibly TLR10) require in addition the adaptor TIRAP, whereas UNC-93B is needed for the proper localization of intracellular TLR3, TLR7, TLR8, and TLR9. We found that IgM(+)IgD(+)CD27(+) but not switched B cells were strongly reduced in MyD88-, IRAK-4-, and TIRAP-deficient patients. This defect did not appear to be compensated with age. However, somatic hypermutation of Ig genes and heavy-chain CDR3 size distribution of IgM(+)IgD(+)CD27(+) B cells were not affected in these patients. In contrast, the numbers of IgM(+)IgD(+)CD27(+) B cells were normal in the absence of TLR3, TRIF, and UNC-93B, suggesting that UNC-93B-dependent TLRs, and notably TLR9, are dispensable for the presence of this subset in peripheral blood. Interestingly, TLR10 was found to be expressed at greater levels in IgM(+)IgD(+)CD27(+) compared with switched B cells in healthy patients. Hence, we propose a role for TIRAP-dependent TLRs, possibly TLR10 in particular, in the development and/or maintenance of IgM(+)IgD(+)CD27(+) B cells in humans.


Asunto(s)
Linfocitos B/inmunología , Inmunoglobulina D/inmunología , Inmunoglobulina M/inmunología , Quinasas Asociadas a Receptores de Interleucina-1/genética , Glicoproteínas de Membrana/genética , Proteínas de Transporte de Membrana/genética , Factor 88 de Diferenciación Mieloide/genética , Receptores de Interleucina-1/genética , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología , Adolescente , Adulto , Linfocitos B/patología , Niño , Preescolar , Citocinas/inmunología , Humanos , Inmunoglobulina D/análisis , Inmunoglobulina M/análisis , Mutación , Receptor Toll-Like 10/genética , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/análisis , Adulto Joven
10.
Blood ; 118(4): 926-35, 2011 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-21622647

RESUMEN

Nuclear factor-κB essential modulator (NEMO), the regulatory subunit of the IκB kinase complex, is a critical component of the NF-κB pathway. Hypomorphic mutations in the X-linked human NEMO gene cause various forms of anhidrotic ectodermal dysplasia with immunodeficiency (EDA-ID). All known X-linked EDA-ID-causing mutations impair NEMO protein expression, folding, or both. We describe here 2 EDA-ID-causing missense mutations that affect the same residue in the CC2-LZ domain (D311N and D311G) that do not impair NEMO production or folding. Structural studies based on pull-down experiments showed a defect in noncovalent interaction with K63-linked and linear polyubiquitin chains for these mutant proteins. Functional studies on the patients' cells showed an impairment of the classic NF-κB signaling pathways after activation of 2 NEMO ubiquitin-binding-dependent receptors, the TNF and IL-1ß receptors, and in the CD40-dependent NF-κB pathway. We report the first human NEMO mutations responsible for X-linked EDA-ID found to affect the polyubiquitin binding of NEMO rather than its expression and folding. These experiments demonstrate that the binding of human NEMO to polyubiquitin is essential for NF-κB activation. They also demonstrate that the normal expression and folding of NEMO do not exclude a pathogenic role for NEMO mutations in patients with EDA-ID.


Asunto(s)
Displasia Ectodermal Anhidrótica Tipo 1/genética , Quinasa I-kappa B/genética , Síndromes de Inmunodeficiencia/genética , Ubiquitina/metabolismo , Western Blotting , Displasia Ectodermal Anhidrótica Tipo 1/metabolismo , Activación Enzimática/genética , Femenino , Humanos , Quinasa I-kappa B/metabolismo , Síndromes de Inmunodeficiencia/metabolismo , Masculino , Mutación Missense , FN-kappa B/metabolismo , Linaje , Unión Proteica , Pliegue de Proteína , Transducción de Señal/genética , Adulto Joven
11.
J Exp Med ; 207(2): 291-7, 2010 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-20123958

RESUMEN

Most patients with autoimmune polyendocrine syndrome type I (APS-I) display chronic mucocutaneous candidiasis (CMC). We hypothesized that this CMC might result from autoimmunity to interleukin (IL)-17 cytokines. We found high titers of autoantibodies (auto-Abs) against IL-17A, IL-17F, and/or IL-22 in the sera of all 33 patients tested, as detected by multiplex particle-based flow cytometry. The auto-Abs against IL-17A, IL-17F, and IL-22 were specific in the five patients tested, as shown by Western blotting. The auto-Abs against IL-17A were neutralizing in the only patient tested, as shown by bioassays of IL-17A activity. None of the 37 healthy controls and none of the 103 patients with other autoimmune disorders tested had such auto-Abs. None of the patients with APS-I had auto-Abs against cytokines previously shown to cause other well-defined clinical syndromes in other patients (IL-6, interferon [IFN]-gamma, or granulocyte/macrophage colony-stimulating factor) or against other cytokines (IL-1beta, IL-10, IL-12, IL-18, IL-21, IL-23, IL-26, IFN-beta, tumor necrosis factor [alpha], or transforming growth factor beta). These findings suggest that auto-Abs against IL-17A, IL-17F, and IL-22 may cause CMC in patients with APS-I.


Asunto(s)
Candidiasis Mucocutánea Crónica/inmunología , Interleucina-17/inmunología , Interleucinas/inmunología , Poliendocrinopatías Autoinmunes/inmunología , Adolescente , Adulto , Autoanticuerpos/sangre , Autoanticuerpos/inmunología , Autoinmunidad , Western Blotting , Candidiasis Mucocutánea Crónica/sangre , Candidiasis Mucocutánea Crónica/etiología , Niño , Preescolar , Femenino , Citometría de Flujo , Humanos , Interferón gamma/inmunología , Masculino , Persona de Mediana Edad , Poliendocrinopatías Autoinmunes/sangre , Poliendocrinopatías Autoinmunes/complicaciones , Adulto Joven , Interleucina-22
12.
J Immunol ; 180(1): 647-54, 2008 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-18097067

RESUMEN

We investigated an otherwise healthy patient presenting two episodes of staphylococcal cellulitis and abscesses, accompanied by high fever and biological signs of inflammation but, paradoxically, with no detectable increase in serum levels of C-reactive protein (CRP), an IL-6-responsive protein synthesized in the liver. Following in vitro activation of whole blood cells from the patient with multiple cytokines, TLR agonists, heat-killed bacteria, and mitogens, we observed a profound and specific impairment of IL-6 secretion. However, the patient's PBMCs, activated in the same conditions but in the absence of the patient's plasma, secreted IL-6 normally. The patient's serum contained high titers of IgG1 autoantibodies against IL-6, which specifically neutralized IL-6 production by control PBMCs as well as IL-6 responses in the human hepatocellular carcinoma cell line Hep3B. These anti-IL-6 autoantibodies were detected over a period of 4 years, in the absence of any other autoantibodies. Our results indicate that these Abs probably prevented an increase in CRP concentration during infection and that impaired IL-6-mediated immunity may have contributed to staphylococcal disease. Patients with severe bacterial infections and low serum CRP concentrations should be tested for anti-IL-6 autoantibodies, especially in the presence of other clinical and biological signs of inflammation.


Asunto(s)
Absceso/diagnóstico , Autoanticuerpos/sangre , Celulitis (Flemón)/diagnóstico , Interleucina-6/inmunología , Piel/inmunología , Infecciones Estafilocócicas/diagnóstico , Absceso/inmunología , Absceso/microbiología , Autoanticuerpos/farmacología , Proteína C-Reactiva/análisis , Línea Celular Tumoral , Celulitis (Flemón)/inmunología , Celulitis (Flemón)/microbiología , Preescolar , Humanos , Inmunoglobulina G/sangre , Interleucina-6/análisis , Interleucina-6/antagonistas & inhibidores , Linfocitos/inmunología , Masculino , Recurrencia , Piel/microbiología , Infecciones Estafilocócicas/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA