Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Cell Transplant ; 32: 9636897231178460, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37278405

RESUMEN

Previous studies transplanted human-induced pluripotent stem cells (hiPSCs)-derived mesenchymal stem cells (iMSCs) into thyroid cartilage defect of X-liked severe combined immunodeficiency (X-SCID) rats and confirmed transplanted cell survival and cartilage regeneration. Thus, this study aimed to investigate the contribution of iMSC transplantation to thyroid cartilage regeneration of nude rats. iMSCs were induced from hiPSCs via a neural crest cell lineage. Then, clumps formed from an iMSC/extracellular matrix complex were transplanted into thyroid cartilage defects in nude rats. The larynx was removed and histological and immunohistochemical analyses were performed 4 or 8 weeks after the transplantation. Human nuclear antigen (HNA)-positive cells were observed in 11 of 12 (91.7%) rats, which indicated that transplanted iMSCs survived in thyroid cartilage defects in nude rats. HNA-positive cells co-expressed SOX9, and type II collagen was identified around HNA-positive cells in 8 of 12 rats (66.7%), which indicated cartilage-like regeneration. Cartilage-like regeneration in nude rats in this study was comparable to the previous report on X-SCID rats (HNA-positive cells were observed in all 14 rats and cartilage-like regeneration was observed in 10 of 14 rats). This result suggests that nude rats could be an alternative to X-SCID rats in thyroid cartilage regeneration experiments using iMSCs, and this nude rat cartilage transplantation model may develop cartilage regeneration research concerning fewer problems such as infection due to immunosuppression.


Asunto(s)
Células Madre Pluripotentes Inducidas , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X , Humanos , Ratas , Animales , Células Madre Pluripotentes Inducidas/metabolismo , Ratas Desnudas , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/metabolismo , Diferenciación Celular , Cartílagos Laríngeos , Células Madre Mesenquimatosas/metabolismo
2.
Nutrients ; 13(1)2021 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-33435521

RESUMEN

Reduced magnesium (Mg) intake is a frequent cause of deficiency with age together with reduced absorption, renal wasting, and polypharmacotherapy. Chronic Mg deficiency may result in increased oxidative stress and low-grade inflammation, which may be linked to several age-related diseases, including higher predisposition to infectious diseases. Mg might play a role in the immune response being a cofactor for immunoglobulin synthesis and other processes strictly associated with the function of T and B cells. Mg is necessary for the biosynthesis, transport, and activation of vitamin D, another key factor in the pathogenesis of infectious diseases. The regulation of cytosolic free Mg in immune cells involves Mg transport systems, such as the melastatin-like transient receptor potential 7 channel, the solute carrier family, and the magnesium transporter 1 (MAGT1). The functional importance of Mg transport in immunity was unknown until the description of the primary immunodeficiency XMEN (X-linked immunodeficiency with Mg defect, Epstein-Barr virus infection, and neoplasia) due to a genetic deficiency of MAGT1 characterized by chronic Epstein-Barr virus infection. This and other research reporting associations of Mg deficit with viral and bacterial infections indicate a possible role of Mg deficit in the recent coronavirus disease 2019 (COVID-19) and its complications. In this review, we will discuss the importance of Mg for the immune system and for infectious diseases, including the recent pandemic of COVID-19.


Asunto(s)
Envejecimiento/fisiología , COVID-19/metabolismo , Enfermedades Transmisibles/metabolismo , Deficiencia de Magnesio/complicaciones , Magnesio/metabolismo , Anciano , COVID-19/etiología , COVID-19/inmunología , COVID-19/virología , Proteínas de Transporte de Catión/metabolismo , Enfermedades Transmisibles/inmunología , Enfermedades Transmisibles/microbiología , Enfermedades Transmisibles/virología , Infecciones por Virus de Epstein-Barr/metabolismo , Femenino , Humanos , Magnesio/inmunología , Deficiencia de Magnesio/inmunología , Deficiencia de Magnesio/metabolismo , Masculino , SARS-CoV-2/inmunología , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/metabolismo
3.
Front Immunol ; 11: 581758, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33162995

RESUMEN

We previously reported the Bruton's tyrosine kinase (BTK) inhibitors ibrutinib and acalabrutinib improve outcomes in a mouse model of polymicrobial sepsis. Now we show that genetic deficiency of the BTK gene alone in Xid mice confers protection against cardiac, renal, and liver injury in polymicrobial sepsis and reduces hyperimmune stimulation ("cytokine storm") induced by an overwhelming bacterial infection. Protection is due in part to enhanced bacterial phagocytosis in vivo, changes in lipid metabolism and decreased activation of NF-κB and the NLRP3 inflammasome. The inactivation of BTK leads to reduced innate immune cell recruitment and a phenotypic switch from M1 to M2 macrophages, aiding in the resolution of sepsis. We have also found that BTK expression in humans is increased in the blood of septic non-survivors, while lower expression is associated with survival from sepsis. Importantly no further reduction in organ damage, cytokine production, or changes in plasma metabolites is seen in Xid mice treated with the BTK inhibitor ibrutinib, demonstrating that the protective effects of BTK inhibitors in polymicrobial sepsis are mediated solely by inhibition of BTK and not by off-target effects of this class of drugs.


Asunto(s)
Agammaglobulinemia Tirosina Quinasa/metabolismo , Insuficiencia Multiorgánica/metabolismo , Sepsis/metabolismo , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/metabolismo , Adenina/análogos & derivados , Adenina/farmacología , Animales , Benzamidas/farmacología , Modelos Animales de Enfermedad , Inflamasomas/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos CBA , Insuficiencia Multiorgánica/tratamiento farmacológico , Fagocitosis/efectos de los fármacos , Piperidinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Pirazinas/farmacología , Sepsis/tratamiento farmacológico , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/tratamiento farmacológico
4.
Dis Model Mech ; 13(8)2020 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-32651192

RESUMEN

Model animals are indispensable for the study of human diseases, and in general, of complex biological processes. The Syrian hamster is an important model animal for infectious diseases, behavioral science and metabolic science, for which more experimental tools are becoming available. Here, we describe the generation and characterization of an interleukin-2 receptor subunit gamma (Il2rg) knockout (KO) Syrian hamster strain. In humans, mutations in IL2RG can result in a total failure of T and natural killer (NK) lymphocyte development and nonfunctional B lymphocytes (X-linked severe combined immunodeficiency; XSCID). Therefore, we sought to develop a non-murine model to study XSCID and the infectious diseases associated with IL2RG deficiency. We demonstrated that the Il2rg KO hamsters have a lymphoid compartment that is greatly reduced in size and diversity, and is impaired in function. As a result of the defective adaptive immune response, Il2rg KO hamsters developed a more severe human adenovirus infection and cleared virus less efficiently than immune competent wild-type hamsters. Because of this enhanced virus replication, Il2rg KO hamsters developed more severe adenovirus-induced liver pathology than wild-type hamsters. This novel hamster strain will provide researchers with a new tool to investigate human XSCID and its related infections.


Asunto(s)
Inmunidad Adaptativa , Infecciones por Adenovirus Humanos/virología , Adenovirus Humanos/patogenicidad , Huésped Inmunocomprometido , Subunidad gamma Común de Receptores de Interleucina/genética , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/genética , Células A549 , Infecciones por Adenovirus Humanos/genética , Infecciones por Adenovirus Humanos/inmunología , Infecciones por Adenovirus Humanos/metabolismo , Adenovirus Humanos/crecimiento & desarrollo , Animales , Animales Modificados Genéticamente , Modelos Animales de Enfermedad , Femenino , Técnicas de Inactivación de Genes , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Subunidad gamma Común de Receptores de Interleucina/deficiencia , Hígado/inmunología , Hígado/metabolismo , Hígado/virología , Linfocitos/inmunología , Linfocitos/metabolismo , Linfocitos/virología , Masculino , Mesocricetus/genética , Carga Viral , Replicación Viral , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/inmunología , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/metabolismo
5.
J Leukoc Biol ; 108(2): 739-748, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32392633

RESUMEN

X-linked severe combined immunodeficiency (X-SCID) caused by full mutation of the IL2RG gene leads to T- B+ NK- phenotype and is usually associated with severe opportunistic infections, diarrhea, and failure to thrive. When IL2RG hypomorphic mutation occurs, diagnosis could be delayed and challenging since only moderate reduction of T and NK cells may be present. Here, we explored phenotypic insights and the impact of the p.R222C hypomorphic mutation (IL2RGR222C ) in distinct cell subsets in an 8-month-old patient with atypical X-SCID. We found reduced CD4+ T cell counts, a decreased frequency of naïve CD4+ and CD8+ T cells, and an expansion of B cells. Ex vivo STAT5 phosphorylation was impaired in CD4+ CD45RO+ T cells, yet compensated by supraphysiological doses of IL-2. Sanger sequencing on purified cell subsets showed a partial reversion of the mutation in total CD3+ cells, specifically in recent thymic emigrants (RTE), effector memory (EM), and CD45RA+ terminally differentiated EM (EMRA) CD4+ T cells. Of note, patient's NK cells had a normal frequency compared to age-matched healthy subjects, but displayed an expansion of CD56bright cells with higher perforin content and cytotoxic potential, associated with accumulation of NK-cell stimulatory cytokines (IL-2, IL-7, IL-15). Overall, this report highlights an alteration in the NK-cell compartment that, together with the high disease-phenotype variability, should be considered in the suspicion of X-SCID with hypomorphic IL2RG mutation.


Asunto(s)
Subunidad gamma Común de Receptores de Interleucina/genética , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Mutación , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/etiología , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/metabolismo , Linfocitos B/inmunología , Linfocitos B/metabolismo , Biomarcadores , Susceptibilidad a Enfermedades , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Inmunofenotipificación , Lactante , Masculino
6.
Mol Genet Genomic Med ; 6(5): 713-721, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30032486

RESUMEN

BACKGROUND: Mutations in the Janus Kinase 3 (JAK3) gene cause an autosomal recessive form of severe combined immunodeficiency (SCID) usually characterized by the absence of both T and NK cells, but preserved numbers of B lymphocytes (T-B+NK-SCID). The detection of larger (>100 bp) genomic duplications or deletions can be more difficult to be detected by PCR-based methods or standard NGS protocols, and a broad range of mutation detection techniques are necessary. METHODS: We report four unrelated Italian patients (two females and two males) with SCID phenotype. Protein expression, functional studies, molecular analysis by standard methods and NGS, and transcripts studies were performed to obtain a definitive diagnosis. RESULTS: Here, we describe four JAK3-deficient patients from four unrelated families. The first patient is homozygous for the known c.1951 C>T mutation causing the amino acidic change p.R651W. The other two patients, originating from the same small Italian town, resulted compound heterozygotes for the same g.15410_16542del deletion and two different novel mutations, g.13319_13321delTTC and c.933T>G (p.F292V), respectively. The fourth patient was compound heterozygous for the novel mutations p.V599G and p.W709R. Defective STAT5 phosphorylation after IL2 or IL15 stimulation corroborated the mutation pathogenicity. Concerning g.15410_16542del mutation, probably due to an unequal homologous recombination between Alu elements of JAK3 gene, microsatellites analysis revealed that both unrelated Pt2 and Pt3 and their carrier family members shared the same haplotype. These data support the hypothesis of a founder effect for the g.15410_16542del mutation that might have inherited in both unrelated families from the same ancient progenitor. CONCLUSION: Different molecular techniques are still required to obtain a definitive diagnosis of AR-SCID particularly in all cases in which a monoallelic mutation is found by standard mutation scanning methods.


Asunto(s)
Secuencia de Bases , Janus Quinasa 3/genética , Mutación Missense , Eliminación de Secuencia , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/genética , Sustitución de Aminoácidos , Femenino , Humanos , Lactante , Recién Nacido , Italia , Janus Quinasa 3/metabolismo , Masculino , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/metabolismo , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/patología
7.
Magnes Res ; 28(2): 46-55, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26422833

RESUMEN

Disturbances in magnesium homeostasis, often linked to altered expression and/or function of magnesium channels, have been implicated in a plethora of diseases. This review focuses on magnesium transporter 1 (MAGT1), as recently described changes in this gene have further extended our understanding of the role of magnesium in human health and disease. The identification of genetic changes and their functional consequences in patients with immunodeficiency revealed that magnesium and MAGT1 are key molecular players for T cell-mediated immune responses. This led to the description of XMEN (X-linked immunodeficiency with magnesium defect, Epstein Barr Virus infection, and neoplasia) syndrome, for which Mg2+ supplementation has been shown to be beneficial. Similarly, the identification of a copy-number variation (CNV) leading to dysfunctional MAGT1 in a family with atypical ATRX syndrome and skin abnormalities, suggested that the MAGT1 defect could be responsible for the cutaneous problems. On the other hand, recent genetic investigations question the previously proposed role for MAGT1 in intellectual disability. Understanding the molecular basis of the involvement of magnesium and its channels in human pathogenesis will improve opportunities for Mg2+ therapies in the clinic.


Asunto(s)
Proteínas de Transporte de Catión/fisiología , Síndromes de Inmunodeficiencia/genética , Discapacidad Intelectual/genética , Animales , Humanos , Síndromes de Inmunodeficiencia/diagnóstico , Síndromes de Inmunodeficiencia/metabolismo , Discapacidad Intelectual/diagnóstico , Discapacidad Intelectual/metabolismo , Magnesio/metabolismo , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/diagnóstico , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/genética , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/metabolismo
8.
J Immunol ; 192(8): 3582-95, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24646744

RESUMEN

Immune response to T cell independent type 2 (TI-2) Ags, such as bacterial polysaccharides, is severely impaired in X-linked immunodeficient (XID) mice. In this study, we investigated the involvement of a proliferation-inducing ligand (APRIL) or BAFF and their receptors in the unresponsiveness of XID mouse to TI-2 Ags. We discovered that whereas serum BAFF levels were increased, the expression of the APRIL and BAFF receptor transmembrane activator and calcium-modulator and cyclophilin ligand interactor (TACI) was severely reduced in XID B cells. Moreover, B cells from XID mouse were unable to secrete Igs in response to APRIL or BAFF. In correlation with reduced TACI expression and impaired TACI function, APRIL or BAFF did not activate the classical NF-κB pathway in XID cells. Also correlating with the unaltered expression of BAFF receptor, BAFF stimulation induced the activation of the alternative NF-κB pathway in XID cells. Moreover, activation of MAPK pathway was ablated in APRIL-stimulated XID cells. Prestimulation of XID B cells with the TLR9 agonist, CpG led to a significant increase in TACI expression and restored TACI-mediated functions. CpG prestimulation also restored TACI-mediated signaling in APRIL- or BAFF-stimulated XID B cells. Finally, immunization of XID mouse with the prototype TI-2 Ag NP-Ficoll induced IgG and IgM Abs when CpG was given with NP-Ficoll. Collectively, these results suggest that reduced TACI expression is responsible for the unresponsiveness of XID mouse to TI-2 Ags and BCR activation controls TACI expression.


Asunto(s)
Receptores de Antígenos de Linfocitos B/metabolismo , Transducción de Señal , Proteína Activadora Transmembrana y Interactiva del CAML/metabolismo , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/inmunología , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/metabolismo , Agammaglobulinemia Tirosina Quinasa , Animales , Factor Activador de Células B/sangre , Factor Activador de Células B/metabolismo , Subgrupos de Linfocitos B/inmunología , Subgrupos de Linfocitos B/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Sistema de Señalización de MAP Quinasas , Ratones , FN-kappa B/metabolismo , Proteínas Tirosina Quinasas/deficiencia , Proteína Activadora Transmembrana y Interactiva del CAML/genética , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/genética
9.
Blood ; 118(26): 6824-35, 2011 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-22039266

RESUMEN

SCID resulting from mutations in IL2RG or JAK3 is characterized by lack of T and natural killer cells; B cells are present in normal number, but antibody responses are defective. Hematopoietic cell transplantation (HCT) is curative for SCID. However, B-cell dysfunction persists in a substantial proportion of patients. We hypothesized that impaired B-cell responses after HCT in IL2RG/JAK3 deficiency results from poor donor B-cell engraftment and defective γc-dependent cytokine signaling in host B cells. To test this, and to identify which γc cytokine(s) is critical for humoral immunity, we studied 28 transplanted patients with IL2RG/JAK3 deficiency. Lack of donor B-cell engraftment associated with persistent humoral dysfunction and significantly reduced memory B cells. B-cell proliferation induced by CD40L alone or together with CpG, anti-Ig, IL-4, IL-10, or IL-13 was comparable in healthy controls and in post-HCT SCID patients, irrespective of their chimerism status. However, in vitro stimulation with CD40L/IL-21 induced B-cell proliferation, plasmablast differentiation, and antibody secretion in patients with donor B cells, but not in patients with autologous B cells. These data imply that IL-21-mediated signaling is critical for long-lived humoral immunity and to restore antibody responses in IL2RG/JAK3-deficient patients after HCT. Furthermore, in vitro stimulation with CD40L/IL-21 can predict in vivo B-cell immunity in IL2RG/JAK3 SCID after transplantation.


Asunto(s)
Linfocitos B/inmunología , Diferenciación Celular/inmunología , Subunidad gamma Común de Receptores de Interleucina/inmunología , Interleucinas/inmunología , Linfocitos B/metabolismo , Ligando de CD40/inmunología , Ligando de CD40/metabolismo , Células Cultivadas , Citometría de Flujo , Trasplante de Células Madre Hematopoyéticas , Humanos , Inmunoglobulina A/sangre , Inmunoglobulina A/inmunología , Inmunoglobulina M/sangre , Inmunoglobulina M/inmunología , Memoria Inmunológica/inmunología , Subunidad gamma Común de Receptores de Interleucina/genética , Subunidad gamma Común de Receptores de Interleucina/metabolismo , Interleucinas/metabolismo , Janus Quinasa 3/genética , Janus Quinasa 3/inmunología , Janus Quinasa 3/metabolismo , Mutación , Unión Proteica , Inmunodeficiencia Combinada Grave/genética , Inmunodeficiencia Combinada Grave/inmunología , Inmunodeficiencia Combinada Grave/metabolismo , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/genética , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/inmunología , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/metabolismo
10.
Sci Transl Med ; 3(97): 97ra79, 2011 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-21865537

RESUMEN

X-linked severe combined immunodeficiency (SCID-X1) is caused by mutations in the common cytokine receptor γ chain. These mutations classically lead to complete absence of functional T and natural killer cell lineages as well as to intrinsically compromised B cell function. Although human leukocyte antigen (HLA)-matched hematopoietic stem cell transplantation (HSCT) is highly successful in SCID-X1 patients, HLA-mismatched procedures can be associated with prolonged immunodeficiency, graft-versus-host disease, and increased overall mortality. Here, 10 children were treated with autologous CD34(+) hematopoietic stem and progenitor cells transduced with a conventional gammaretroviral vector. The patients did not receive myelosuppressive conditioning and were monitored for immunological recovery after cell infusion. All patients were alive after a median follow-up of 80 months (range, 54 to 107 months), and a functional polyclonal T cell repertoire was restored in all patients. Humoral immunity only partially recovered but was sufficient in some patients to allow for withdrawal of immunoglobulin replacement; however, three patients developed antibiotic-responsive acute pulmonary infection after discontinuation of antibiotic prophylaxis and/or immunoglobulin replacement. One patient developed acute T cell acute lymphoblastic leukemia because of up-regulated expression of the proto-oncogene LMO-2 from insertional mutagenesis, but maintained a polyclonal T cell repertoire through chemotherapy and entered remission. Therefore, gene therapy for SCID-X1 without myelosuppressive conditioning effectively restored T cell immunity and was associated with high survival rates for up to 9 years. Further studies using vectors designed to limit mutagenesis and strategies to enhance B cell reconstitution are warranted to define the role of this treatment modality alongside conventional HSCT for SCID-X1.


Asunto(s)
Terapia Genética/métodos , Linfocitos T/inmunología , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/inmunología , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/terapia , Antígenos CD34/genética , Antígenos CD34/metabolismo , Preescolar , Femenino , Gammaretrovirus/genética , Vectores Genéticos/genética , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Lactante , Masculino , Proto-Oncogenes Mas , Trasplante de Células Madre/métodos , Trasplante Autólogo/métodos , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/metabolismo
11.
Cell ; 136(6): 1098-109, 2009 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-19303852

RESUMEN

Activation of nuclear factor-kappaB (NF-kappaB), a key mediator of inducible transcription in immunity, requires binding of NF-kappaB essential modulator (NEMO) to ubiquitinated substrates. Here, we report that the UBAN (ubiquitin binding in ABIN and NEMO) motif of NEMO selectively binds linear (head-to-tail) ubiquitin chains. Crystal structures of the UBAN motif revealed a parallel coiled-coil dimer that formed a heterotetrameric complex with two linear diubiquitin molecules. The UBAN dimer contacted all four ubiquitin moieties, and the integrity of each binding site was required for efficient NF-kappaB activation. Binding occurred via a surface on the proximal ubiquitin moiety and the canonical Ile44 surface on the distal one, thereby providing specificity for linear chain recognition. Residues of NEMO involved in binding linear ubiquitin chains are required for NF-kappaB activation by TNF-alpha and other agonists, providing an explanation for the detrimental effect of NEMO mutations in patients suffering from X-linked ectodermal dysplasia and immunodeficiency.


Asunto(s)
Quinasa I-kappa B/metabolismo , Subunidad p50 de NF-kappa B/metabolismo , Ubiquitina/metabolismo , Secuencias de Aminoácidos , Displasia Ectodérmica/metabolismo , Humanos , Quinasa I-kappa B/química , Modelos Moleculares , Unión Proteica , Ubiquitina/química , Ubiquitinas/química , Ubiquitinas/metabolismo , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/metabolismo
12.
J Immunol ; 182(5): 3304-9, 2009 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-19234229

RESUMEN

Mutations of the IL2RG encoding the common gamma-chain (gamma(c)) lead to the X-linked SCID disease. Gene correction through ex vivo retroviral transduction restored the immunological impairment in the most of treated patients, although lymphoproliferative events occurred in five of them. Even though in two cases it was clearly documented an insertional mutagenesis in LMO2, it is conceivable that gamma(c) could have a role per se in malignant lymphoproliferation. The gamma(c) is a shared cytokine receptor subunit, involved also in growth hormone (GH) receptor signaling. Through short interfering RNA or using X-linked SCID B lymphoblastoid cell lines lacking gamma(c), we demonstrate that self-sufficient growth was strongly dependent on gamma(c) expression. Furthermore, a correlation between gamma(c) amount and the extent of constitutive activation of JAK3 was found. The reduction of gamma(c) protein expression also reduced GH-induced proliferation and STAT5 nuclear translocation in B lymphoblastoid cell lines. Hence, our data demonstrate that gamma(c) plays a remarkable role in either spontaneous or GH-induced cell cycle progression depending on the amount of protein expression, suggesting a potential role as enhancing cofactor in lymphoproliferation.


Asunto(s)
Linfocitos B/inmunología , Linfocitos B/metabolismo , Proliferación Celular , Subunidad gamma Común de Receptores de Interleucina/biosíntesis , Activación de Linfocitos/inmunología , Linfocitos B/enzimología , Linfocitos B/patología , Ciclo Celular/genética , Ciclo Celular/inmunología , Línea Celular Transformada , Células Cultivadas , Relación Dosis-Respuesta Inmunológica , Técnicas de Silenciamiento del Gen , Sustancias de Crecimiento/genética , Sustancias de Crecimiento/fisiología , Humanos , Subunidad gamma Común de Receptores de Interleucina/antagonistas & inhibidores , Subunidad gamma Común de Receptores de Interleucina/deficiencia , Subunidad gamma Común de Receptores de Interleucina/genética , Janus Quinasa 3/antagonistas & inhibidores , Janus Quinasa 3/genética , Janus Quinasa 3/metabolismo , Activación de Linfocitos/genética , Recuento de Linfocitos , ARN Interferente Pequeño/genética , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/inmunología , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/metabolismo , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/patología
13.
J Biol Chem ; 284(13): 8548-56, 2009 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-19155218

RESUMEN

Macrophages act to protect the body against inflammation and infection by engaging in chemotaxis and phagocytosis. In chemotaxis, macrophages use an actin-based membrane structure, the podosome, to migrate to inflamed tissues. In phagocytosis, macrophages form another type of actin-based membrane structure, the phagocytic cup, to ingest foreign materials such as bacteria. The formation of these membrane structures is severely affected in macrophages from patients with Wiskott-Aldrich syndrome (WAS), an X chromosome-linked immunodeficiency disorder. WAS patients lack WAS protein (WASP), suggesting that WASP is required for the formation of podosomes and phagocytic cups. Here we have demonstrated that formin-binding protein 17 (FBP17) recruits WASP, WASP-interacting protein (WIP), and dynamin-2 to the plasma membrane and that this recruitment is necessary for the formation of podosomes and phagocytic cups. The N-terminal EFC (extended FER-CIP4 homology)/F-BAR (FER-CIP4 homology and Bin-amphiphysin-Rvs) domain of FBP17 was previously shown to have membrane binding and deformation activities. Our results suggest that FBP17 facilitates membrane deformation and actin polymerization to occur simultaneously at the same membrane sites, which mediates a common molecular step in the formation of podosomes and phagocytic cups. These results provide a potential mechanism underlying the recurrent infections in WAS patients.


Asunto(s)
Proteínas Portadoras/metabolismo , Macrófagos/metabolismo , Síndrome de Wiskott-Aldrich/metabolismo , Actinas/inmunología , Actinas/metabolismo , Proteínas Portadoras/inmunología , Línea Celular , Estructuras de la Membrana Celular , Proteínas del Citoesqueleto/inmunología , Proteínas del Citoesqueleto/metabolismo , Dinamina II/inmunología , Dinamina II/metabolismo , Proteínas de Unión a Ácidos Grasos , Humanos , Péptidos y Proteínas de Señalización Intracelular/inmunología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Macrófagos/inmunología , Macrófagos/patología , Estructura Terciaria de Proteína , Síndrome de Wiskott-Aldrich/inmunología , Síndrome de Wiskott-Aldrich/patología , Proteína del Síndrome de Wiskott-Aldrich/inmunología , Proteína del Síndrome de Wiskott-Aldrich/metabolismo , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/inmunología , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/metabolismo , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/patología
14.
Gastroenterology ; 132(5): 1705-17, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17484868

RESUMEN

BACKGROUND & AIMS: Immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX; OMIM 304930) syndrome is a congenital syndrome characterized by autoimmune enteropathy, endocrinopathy, dermatitis, and other autoimmune phenomena. In the present work, we aimed to uncover the molecular basis of a distinct form of IPEX syndrome presenting at the edge of autoimmunity and severe allergy. METHODS: The FOXP3 gene was sequenced, FOXP3 messenger RNA (mRNA) was quantified by real-time polymerase chain reaction (PCR), and protein expression in peripheral blood lymphocytes was analyzed by flow cytometry after intracellular staining. In coculture experiments (CD4(+)CD25(-) and CD4(+)CD25(+) cells), the functions of regulatory T cells were analyzed. Expression of interferon gamma and interleukin 2 and 4 mRNA within the inflamed intestinal mucosa was quantified by real-time PCR. RESULTS: Here, we describe a distinct familial form of IPEX syndrome that combines autoimmune and allergic manifestations including severe enteropathy, food allergies, atopic dermatitis, hyper-IgE, and eosinophilia. We have identified a 1388-base pair deletion (g.del-6247_-4859) of the FOXP3 gene encompassing a portion of an upstream noncoding exon (exon -1) and the adjacent intron (intron -1). This deletion impairs mRNA splicing, resulting in accumulation of unspliced pre-mRNA and alternatively spliced mRNA. This causes low FOXP3 mRNA levels and markedly decreased protein expression in peripheral blood lymphocytes of affected patients. Numbers of CD4(+)CD25(+)FOXP3(+) regulatory T cells are extremely low, and the CD4(+)CD25(+) T cells that are present exhibit little regulatory function. CONCLUSIONS: A new mutation within an upstream noncoding region of FOXP3 results in a variant of IPEX syndrome associating autoimmune and severe immunoallergic symptoms.


Asunto(s)
Eccema/genética , Hipersensibilidad a los Alimentos/genética , Factores de Transcripción Forkhead/genética , Eliminación de Gen , Enfermedades Intestinales/genética , Poliendocrinopatías Autoinmunes/genética , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/genética , Secuencia de Bases , Antígenos CD4/genética , Antígenos CD4/metabolismo , Niño , Preescolar , Eccema/metabolismo , Eccema/patología , Femenino , Factores de Transcripción Forkhead/metabolismo , Humanos , Subunidad alfa del Receptor de Interleucina-2/genética , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Mucosa Intestinal/patología , Masculino , Datos de Secuencia Molecular , Linaje , Poliendocrinopatías Autoinmunes/metabolismo , Poliendocrinopatías Autoinmunes/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/patología , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/metabolismo , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...