Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Int J Mol Sci ; 22(3)2021 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-33572538

RESUMO

Inborn errors of immunity (IEI) are a group of disorders that are mostly caused by genetic mutations affecting immune host defense and immune regulation. Although IEI present with a wide spectrum of clinical features, in about one third of them various degrees of gastrointestinal (GI) involvement have been described and for some IEI the GI manifestations represent the main and peculiar clinical feature. The microbiome plays critical roles in the education and function of the host's innate and adaptive immune system, and imbalances in microbiota-immunity interactions can contribute to intestinal pathogenesis. Microbial dysbiosis combined to the impairment of immunosurveillance and immune dysfunction in IEI, may favor mucosal permeability and lead to inflammation. Here we review how immune homeostasis between commensals and the host is established in the gut, and how these mechanisms can be disrupted in the context of primary immunodeficiencies. Additionally, we highlight key aspects of the first studies on gut microbiome in patients affected by IEI and discuss how gut microbiome could be harnessed as a therapeutic approach in these diseases.


Assuntos
Disbiose/microbiologia , Microbioma Gastrointestinal/fisiologia , Interações entre Hospedeiro e Microrganismos , Doenças da Imunodeficiência Primária/microbiologia , Imunidade Adaptativa , Disbiose/imunologia , Microbioma Gastrointestinal/imunologia , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/microbiologia , Trato Gastrointestinal/patologia , Homeostase , Humanos , Doenças da Imunodeficiência Primária/imunologia , Doenças da Imunodeficiência Primária/patologia
2.
Pediatr Allergy Immunol ; 31 Suppl 26: 57-59, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33236415

RESUMO

Inborn errors of immunity (IEI), also referred to as primary immunodeficiencies (PID), are disorders that, for the most part, result from mutations in genes involved in immune host defense and immune regulation. Thanks to the increased availability of high-throughput DNA sequencing and the improvement in genomic data interpretation, the number of newly identified genes associated with IEI has exponentially increased over the last decade. We reviewed four recently described monogenic IEI and discussed the clinical and immunologic features of these new conditions.


Assuntos
Síndromes de Imunodeficiência , Doenças da Imunodeficiência Primária , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Síndromes de Imunodeficiência/genética , Mutação
3.
Med Princ Pract ; 29(2): 101-112, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31597133

RESUMO

Primary immunodeficiencies are disorders resulting from mutations in genes involved in immune defense and immune regulation. These conditions are characterized by various combinations of recurrent infections, autoimmunity, lymphoproliferation, inflammatory manifestations, and malignancy. In the last 20 years, newborn screening programs and next generation sequencing techniques have increased the ability to diagnose primary immunodeficiencies. Furthermore, an advanced understanding of the molecular basis of these inherited disorders has led to the implementation of targeted therapies that utilize small molecules and biologics to modulate the activity of impaired intracellular pathways. This article will discuss selected primary immunodeficiencies, the genetic defects of which have been recently studied and are amenable to targeted therapy as a reflection of the potential of precision medicine in the future.


Assuntos
Produtos Biológicos/uso terapêutico , Doenças da Imunodeficiência Primária/tratamento farmacológico , Humanos , Medicina de Precisão/métodos , Doenças da Imunodeficiência Primária/genética
4.
Clin Immunol ; 173: 121-123, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27713031

RESUMO

Recombination-activating gene (RAG) 1 and 2 mutations in humans cause T- B- NK+ SCID and Omenn syndrome, but milder phenotypes associated with residual protein activity have been recently described. We report a male patient with a diagnosis of common variable immunodeficiency (CVID) born from non-consanguineous parents, whose immunological phenotype was characterized by severe reduction of B cells and agammaglobulinemia for which several candidate genes were excluded by targeted Sanger sequencing. Next Generation Sequencing revealed two compound heterozygous mutations in the RAG1 gene: the previously described p.R624H, and the novel p.Y728H mutation, as well as the known polymorphism p.H249R. This case reinforces the notion of large phenotypic spectrum in RAG deficiency and opens questions on the management and follow-up of these patients.


Assuntos
Agamaglobulinemia/genética , Imunodeficiência de Variável Comum/genética , Proteínas de Homeodomínio/genética , Pólipos Nasais/genética , Agamaglobulinemia/imunologia , Linfócitos B/imunologia , Criança , Imunodeficiência de Variável Comum/imunologia , Humanos , Masculino , Mutação , Pólipos Nasais/imunologia , Fenótipo
6.
J Allergy Clin Immunol ; 131(2): 314-23, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23374262

RESUMO

The characterization of primary immunodeficiencies (PIDs) in human subjects is crucial for a better understanding of the biology of the immune response. New achievements in this field have been possible in light of collaborative studies; attention paid to new phenotypes, infectious and otherwise; improved immunologic techniques; and use of exome sequencing technology. The International Union of Immunological Societies Expert Committee on PIDs recently reported on the updated classification of PIDs. However, new PIDs are being discovered at an ever-increasing rate. A series of 19 novel primary defects of immunity that have been discovered after release of the International Union of Immunological Societies report are discussed here. These new findings highlight the molecular pathways that are associated with clinical phenotypes and suggest potential therapies for affected patients.


Assuntos
Síndromes de Imunodeficiência/classificação , Síndromes de Imunodeficiência/imunologia , Animais , Humanos , Imunidade/genética , Imunidade/imunologia , Síndromes de Imunodeficiência/terapia , Fenótipo
7.
Blood ; 116(26): 5867-74, 2010 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-20702779

RESUMO

CD40/CD40 ligand (CD40L) cross-talk plays a key role in B-cell terminal maturation in the germinal centers. Genetic defects affecting CD40 cause a rare form of hyper-immunoglobulin M (IgM) syndrome, a disorder characterized by low or absent serum IgG and IgA, associated with recurrent infections. We previously reported on a few patients with homozygous CD40 mutations resulting in lack or severe reduction of CD40 cell surface expression. Here we characterize the 3 CD40 mutants due to missense mutations or small in-frame deletions, and show that the mutated proteins are synthesized but retained in the endoplasmic reticulum (ER), likely due to protein misfolding. Interestingly, the intracellular behavior and fate differ significantly among the mutants: progressive accumulation of the P2 mutant causes endoplasmic reticulum stress and the activation of an unfolded protein response; the mutant P4 is rather efficiently disposed by the ER-associated degradation pathway, while the P5 mutant partially negotiates transport to the plasma membrane, and is competent for CD40L binding. Interestingly, this latter mutant activates downstream signaling elements when overexpressed in transfected cells. These results give new important insights into the molecular pathogenesis of HIGM disease, and suggest that CD40 deficiency can also be regarded as an ER-storage disease.


Assuntos
Antígenos CD40/genética , Antígenos CD40/metabolismo , Mutação da Fase de Leitura/genética , Síndrome de Imunodeficiência com Hiper-IgM/genética , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutação de Sentido Incorreto/genética , Sequência de Aminoácidos , Linfócitos B/metabolismo , Western Blotting , Antígenos CD40/química , Membrana Celular/metabolismo , Células Cultivadas , Criança , Pré-Escolar , Retículo Endoplasmático/metabolismo , Feminino , Citometria de Fluxo , Glicosilação , Humanos , Síndrome de Imunodeficiência com Hiper-IgM/imunologia , Lactente , Rim/citologia , Rim/metabolismo , Masculino , Dados de Sequência Molecular , Proteínas Mutantes/química , Linhagem , Conformação Proteica , Dobramento de Proteína , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência de Aminoácidos
8.
Front Immunol ; 13: 845496, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35371100

RESUMO

Background: Severe skeletal muscle damage has been recently reported in patients with SARS-CoV-2 infection and as a rare vaccination complication. Case summary: On Apr 28, 2021 a 68-year-old man who was previously healthy presented with an extremely severe rhabdomyolysis that occurred nine days following the first dose of SARS-CoV-2 ChAdOx1 nCov-19 vaccination. He had no risk factors, and denied any further assumption of drugs except for fermented red rice, and berberine supplement. The clinical scenario was complicated by a multi organ failure involving bone marrow, liver, lung, and kidney. For the rapid increase of the inflammatory markers, a cytokine storm was suspected and multi-target biologic immunosuppressive therapy was started, consisting of steroids, anakinra, and eculizumab, which was initially successful resulting in close to normal values of creatine phosphokinase after 17 days of treatment. Unfortunately, 48 days after the vaccination an accelerated phase of deterioration, characterized by severe multi-lineage cytopenia, untreatable hypotensive shock, hypoglycemia, and dramatic increase of procalcitonin (PCT), led to patient death. Conclusion: Physicians should be aware that severe and fatal rhabdomyolysis may occur after SARS-CoV2 vaccine administration.


Assuntos
COVID-19 , Rabdomiólise , Trombocitopenia , Idoso , Vacinas contra COVID-19/efeitos adversos , ChAdOx1 nCoV-19 , Humanos , Masculino , Insuficiência de Múltiplos Órgãos/etiologia , RNA Viral , Rabdomiólise/etiologia , SARS-CoV-2 , Vacinação/efeitos adversos
9.
Am J Pathol ; 176(3): 1104-12, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20093495

RESUMO

Autoimmune regulator (AIRE) modulates the expression of tissue-restricted antigens (TSAs) and promotes central tolerance in the thymus. However, few autoreactive T cells escape negative selection and reach the periphery, where peripheral tolerance is required to avoid autoimmunity. Murine lymph nodes (LNs) have been shown to contain "stromal" cells expressing AIRE and TSAs. Here we report the occurrence of AIRE-expressing cells in human peripheral lymphoid tissues, including LNs, tonsils, and gut-associated lymphoid tissue, with the exception of the spleen. Notably, AIRE+ cells are absent in fetal LNs and, in postnatal life, they are more numerous in abdominal than in superficial LNs, thus suggesting that their development in periphery may depend on instructive signals from microenvironment and antigen challenge. Extrathymic AIRE+ cells show a dendritic morphology, consistently express human leukocyte antigen-DR (HLADR) and fascin, and are largely positive for CD11c and S100 and for the dendritic cell-activation markers CD40, CD83, DC-LAMP/CD208, and CCR7. Lymphoid, myelomonocytic, mesenchymal, and epithelial cell lineage markers are negative. The HLADRhigh/AIRE+ cell fraction isolated from mesenteric LNs expressed TSAs (insulin, CYP17A1, and CYP21A2), as well as molecules associated with tolerogenic functions, such as interleukin-10 and indoleamine 2,3-dioxygenase. Data indicate that AIRE+ cells in human peripheral lymphoid tissues correspond to a subset of activated interdigitating dendritic cells expressing TSAs and the tolerogenic molecules indoleamine 2,3-dioxygenase and interleukin-10, suggestive of a potential tolerogenic function.


Assuntos
Células Dendríticas/metabolismo , Tecido Linfoide/citologia , Fatores de Transcrição/metabolismo , Adolescente , Adulto , Antígenos/imunologia , Células Dendríticas/citologia , Feminino , Feto/metabolismo , Humanos , Tolerância Imunológica/imunologia , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Especificidade de Órgãos , Fenótipo , Proteína AIRE
10.
Am J Hematol ; 84(8): 473-9, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19507210

RESUMO

Infantile malignant osteopetrosis (IMO) includes various genetic disorders that affect osteoclast development and/or function. Genotype-phenotype correlation studies in IMO have been hampered by the rarity and heterogeneity of the disease and by the severity of the clinical course, which often leads to death early in life. We report on the clinical and molecular findings and treatment in 20 consecutive patients (11 males, nine females) with IMO, diagnosed at a single center in the period 1991-2008. Mean age at diagnosis was 3.9 months, and mean follow-up was 66.75 months. Mutations in TCIRG1, OSTM1, ClCN7, and TNFRSF11A genes were detected in nine, three, one, and one patients, respectively. Six patients remain genetically undefined. OSTM1 and ClCN7 mutations were associated with poor neurologic outcome. Among nine patients with TCIRG1 defects, six presented with hypogammaglobulinemia, and one showed primary pulmonary hypertension. Fourteen patients received hematopoietic cell transplantation; of these, nine are alive and eight of them have evidence of osteoclast function. These data may provide a basis for informed decisions regarding the care of patients with IMO.


Assuntos
Doenças Genéticas Inatas/terapia , Transplante de Células-Tronco Hematopoéticas , Osteopetrose/terapia , Agamaglobulinemia/genética , Agamaglobulinemia/mortalidade , Agamaglobulinemia/terapia , Canais de Cloreto/genética , Intervalo Livre de Doença , Feminino , Seguimentos , Doenças Genéticas Inatas/genética , Doenças Genéticas Inatas/mortalidade , Humanos , Hipertensão Pulmonar/genética , Hipertensão Pulmonar/mortalidade , Hipertensão Pulmonar/terapia , Lactente , Recém-Nascido , Masculino , Proteínas de Membrana/genética , Osteopetrose/genética , Osteopetrose/mortalidade , Receptor Ativador de Fator Nuclear kappa-B/genética , Estudos Retrospectivos , Taxa de Sobrevida , Transplante Homólogo , Ubiquitina-Proteína Ligases/genética , ATPases Vacuolares Próton-Translocadoras/genética
11.
Front Pediatr ; 7: 353, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31508401

RESUMO

Inborn errors of immunity are genetic disorders with broad clinical manifestations, ranging from increased susceptibility to infections to significant immune dysregulation, often leading to multiple autoimmune phenomena, lymphoproliferation, and malignancy. The treatment is challenging as it requires careful balancing of immunosuppression in subjects at increased risk of infections. Recently, the improved ability to define inborn errors of immunity pathophysiology at the molecular level has set the basis for the development of targeted therapeutic interventions. Such a "precision medicine" approach is mainly bases on the use of available small molecules and biologics to target a specific cell function. In this article, we summarize the clinical and laboratory features of various recently described inborn errors of immunity associated with immune dysregulation and hyperinflammation in which mechanism-based therapeutic approaches have been implemented.

12.
Front Pediatr ; 7: 295, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31440487

RESUMO

Primary immunodeficiencies (PID) are disorders that for the most part result from mutations in genes involved in immune host defense and immunoregulation. These conditions are characterized by various combinations of recurrent infections, autoimmunity, lymphoproliferation, inflammatory manifestations, atopy, and malignancy. Most PID are due to genetic defects that are intrinsic to hematopoietic cells. Therefore, replacement of mutant cells by healthy donor hematopoietic stem cells (HSC) represents a rational therapeutic approach. Full or partial ablation of the recipient's marrow with chemotherapy is often used to allow stable engraftment of donor-derived HSCs, and serotherapy may be added to the conditioning regimen to reduce the risks of graft rejection and graft versus host disease (GVHD). Initially, hematopoietic stem cell transplantation (HSCT) was attempted in patients with severe combined immunodeficiency (SCID) as the only available curative treatment. It was a challenging procedure, associated with elevated rates of morbidity and mortality. Overtime, outcome of HSCT for PID has significantly improved due to availability of high-resolution HLA typing, increased use of alternative donors and new stem cell sources, development of less toxic, reduced-intensity conditioning (RIC) regimens, and cellular engineering techniques for graft manipulation. Early identification of infants affected by SCID, prior to infectious complication, through newborn screening (NBS) programs and prompt genetic diagnosis with Next Generation Sequencing (NGS) techniques, have also ameliorated the outcome of HSCT. In addition, HSCT has been applied to treat a broader range of PID, including disorders of immune dysregulation. Yet, the broad spectrum of clinical and immunological phenotypes associated with PID makes it difficult to define a universal transplant regimen. As such, integration of knowledge between immunologists and transplant specialists is necessary for the development of innovative transplant protocols and to monitor their results during follow-up. Despite the improved outcome observed after HSCT, patients with severe forms of PID still face significant challenges of short and long-term transplant-related complications. To address this issue, novel HSCT strategies are being implemented aiming to improve both survival and long-term quality of life. This article will discuss the current status and latest developments in HSCT for PID, and present data regarding approach and outcome of HSCT in recently described PID, including disorders associated with immune dysregulation.

13.
Haematologica ; 88(10): 1169-75, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14555314

RESUMO

BACKGROUND AND OBJECTIVES: Standard or investigative immunosuppressive therapies for cutaneous chronic graft-versus-host disease (GVHD) may prove not only ineffective but also cause serious adverse effects. Repeated exposure of the skin to ultraviolet radiation in the wavelength range 340-400 nm (so-called ultraviolet A1) was recently reported to have a strong local (intracutaneous) immunomodulatory activity. This study was undertaken to evaluate efficacy and safety of this phototherapy. DESIGN AND METHODS: Nine patients with cutaneous (4 lichenoid and 5 sclerodermoid) GVHD and mild or no other organ involvement were enrolled. All patients had developed serious drug toxicity and/or opportunistic infections. Phototherapy was administered three times a week. RESULTS: Complete remission was seen in 5 (2 lichenoid and 3 sclerodermoid) cases and a partial improvement in 4 (2 lichenoid and 2 sclerodermoid) after having received 15.8+/-3.8 (lichenoid GVHD) or 21.6+/-8.0 (sclerodermoid GVHD) sessions of phototherapy. Adverse effects were not registered. At follow-up (range: 6-25 months), two patients with sclerodermoid lesions relapsed after 5 months but responded to another treatment cycle. Patients with lichenoid GVHD showed relapses within one month and prolonged maintenance phototherapy was needed. Problems of drug toxicity and opportunistic infections improved as phototherapy allowed the reduction or interruption of systemic drug therapies. INTERPRETATION AND CONCLUSIONS: Ultraviolet A1 phototherapy may be considered as an appropriate therapeutic approach for sclerodermoid GVHD with no or mild involvement of internal organs. Patients with lichenoid GVHD should be treated only if they develop serious adverse effects to immunosuppressive therapies and opportunistic infections because of the carcinogenic hazard of high cumulative doses of ultraviolet A1 radiation.


Assuntos
Doença Enxerto-Hospedeiro/complicações , Erupções Liquenoides/etiologia , Erupções Liquenoides/terapia , Terapia PUVA/métodos , Esclerodermia Localizada/etiologia , Esclerodermia Localizada/terapia , Adulto , Transplante de Medula Óssea/métodos , Criança , Pré-Escolar , Feminino , Humanos , Imunossupressores/efeitos adversos , Imunossupressores/uso terapêutico , Leucemia/cirurgia , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Transplante Homólogo , Resultado do Tratamento
14.
Haematologica ; 89(4): 450-61, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15075079

RESUMO

BACKGROUND AND OBJECTIVES: In utero transplantation of hematopoietic stem cells allows immune reconstitution of fetuses with severe combined immunodeficiency. The objective of this work was to study the quality of T-cell reconstitution following this procedure. DESIGN AND METHODS: We evaluated the kinetics and extent of T-cell reconstitution in five infants with severe combined immune deficiency (SCID), three with a B+ and two with a B- phenotype, who received haploidentical stem cell transplantation before birth. To this end, we measured the frequency of T-cell receptor excision circles (TREC) and the diversity of the T-cell repertoire. RESULTS: In utero transplantation led to engraftment of donor-derived T lymphocytes which attained normal numbers in four infants, who are in good health. In the three patients with a B+ phenotype, generation of a heterogeneous T-cell repertoire was associated with development of TREC levels comparable to those of SCID patients treated by post-natal transplantation and of healthy babies. Of the two patients with a B- phenotype, one developed mixed T-cell chimerism and a substantial number of circulating T cells, associated with a variable heterogeneity of the T-cell repertoire; TREC levels were normal soon after birth, but declined thereafter. The remaining B- patient remained lymphopenic with a skewed T-cell repertoire and very low TREC levels. This patient eventually required transplantation from a matched unrelated donor at 5 years of age, but died of EBV-related lymphoproliferative disease. INTERPRETATION AND CONCLUSIONS: These data indicate that in utero transplantation of fetuses with B+ SCID allows generation of newly diversified T lymphocytes and ensures long-term reconstitution of cell-mediated immunity.


Assuntos
Terapias Fetais , Transplante de Células-Tronco Hematopoéticas/métodos , Linfócitos T/fisiologia , Adulto , Feminino , Humanos , Sistema Imunitário/fisiologia , Lactente , Gravidez , Receptores de Antígenos de Linfócitos T/genética , Regeneração , Imunodeficiência Combinada Severa/terapia , Timo/fisiologia
15.
Front Immunol ; 5: 259, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25076946

RESUMO

Thymocytes and thymic epithelial cells (TECs) cross-talk is essential to support T cell development and preserve thymic architecture and maturation of TECs and Foxp3(+) natural regulatory T cells. Accordingly, disruption of thymic lymphostromal cross-talk may have major implications on the thymic mechanisms that govern T cell tolerance. Several genetic defects have been described in humans that affect early stages of T cell development [leading to severe combined immune deficiency (SCID)] or late stages in thymocyte maturation (resulting in combined immunodeficiency). Hypomorphic mutations in SCID-causing genes may allow for generation of a limited pool of T lymphocytes with a restricted repertoire. These conditions are often associated with infiltration of peripheral tissues by activated T cells and immune dysregulation, as best exemplified by Omenn syndrome (OS). In this review, we will discuss our recent findings on abnormalities of thymic microenvironment in OS with a special focus of defective maturation of TECs, altered distribution of thymic dendritic cells and impairment of deletional and non-deletional mechanisms of central tolerance. Here, taking advantage of mouse models of OS and atypical SCID, we will discuss how modifications in stromal compartment impact and shape lymphocyte differentiation, and vice versa how inefficient T cell signaling results in defective stromal maturation. These findings are instrumental to understand the extent to which novel therapeutic strategies should act on thymic stroma to achieve full immune reconstitution.

16.
J Exp Med ; 210(2): 355-74, 2013 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-23337808

RESUMO

Mutations in Wiskott-Aldrich syndrome (WAS) protein (WASp), a regulator of actin dynamics in hematopoietic cells, cause WAS, an X-linked primary immunodeficiency characterized by recurrent infections and a marked predisposition to develop autoimmune disorders. The mechanisms that link actin alterations to the autoimmune phenotype are still poorly understood. We show that chronic activation of plasmacytoid dendritic cells (pDCs) and elevated type-I interferon (IFN) levels play a role in WAS autoimmunity. WAS patients display increased expression of type-I IFN genes and their inducible targets, alteration in pDCs numbers, and hyperresponsiveness to TLR9. Importantly, ablating IFN-I signaling in WASp null mice rescued chronic activation of conventional DCs, splenomegaly, and colitis. Using WASp-deficient mice, we demonstrated that WASp null pDCs are intrinsically more responsive to multimeric agonist of TLR9 and constitutively secrete type-I IFN but become progressively tolerant to further stimulation. By acute silencing of WASp and actin inhibitors, we show that WASp-mediated actin polymerization controls intracellular trafficking and compartmentalization of TLR9 ligands in pDCs restraining exaggerated activation of the TLR9-IFN-α pathway. Together, these data highlight the role of actin dynamics in pDC innate functions and imply the pDC-IFN-α axis as a player in the onset of autoimmune phenomena in WAS disease.


Assuntos
Actinas/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Interferon Tipo I/biossíntese , Proteína da Síndrome de Wiskott-Aldrich/metabolismo , Actinas/antagonistas & inibidores , Animais , Autoimunidade , Sequência de Bases , Células Dendríticas/patologia , Modelos Animais de Doenças , Endocitose , Feminino , Humanos , Imunidade Inata , Interferon Tipo I/genética , Interferon-alfa/biossíntese , Interferon-alfa/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor de Interferon alfa e beta/deficiência , Receptor de Interferon alfa e beta/genética , Transdução de Sinais , Receptor Toll-Like 9/metabolismo , Síndrome de Wiskott-Aldrich/genética , Síndrome de Wiskott-Aldrich/imunologia , Síndrome de Wiskott-Aldrich/metabolismo , Síndrome de Wiskott-Aldrich/patologia , Proteína da Síndrome de Wiskott-Aldrich/deficiência , Proteína da Síndrome de Wiskott-Aldrich/genética
17.
Curr Pharm Des ; 18(35): 5729-34, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22726115

RESUMO

Inflammatory and autoimmune diseases are classically considered as disorders arising from hyper-activation of immunity and hence are treated with drugs that suppress the lymphocyte activation and inflammation. Although this strategy has proven useful to cure symptoms, it rarely can heal the disease and long-term treatments are usually needed. Inflammatory and autoimmune diseases frequently occur also in patients with primary immune deficiency disease, proving that immune hyper-activation may paradoxically arise from defective function of immune genes. In these cases, the phenotype of hyper-activation is believed to reflect the attempts of the immune system to compensate for immune defects. Recent data suggest that similar mechanisms could be involved also in the pathogenesis of some multifactorial disorders, such as Crohn's disease and systemic lupus erythematosus. Based on these considerations, novel therapies could be developed to cure severe autoimmune and inflammatory disorders, not only by aiming to hyper-activation but as well by focusing on the possible underlying immune defects.


Assuntos
Doenças Autoimunes/tratamento farmacológico , Desenho de Fármacos , Inflamação/tratamento farmacológico , Doenças Autoimunes/genética , Doenças Autoimunes/imunologia , Criança , Humanos , Síndromes de Imunodeficiência/tratamento farmacológico , Síndromes de Imunodeficiência/genética , Síndromes de Imunodeficiência/imunologia , Inflamação/genética , Inflamação/imunologia , Ativação Linfocitária/efeitos dos fármacos , Índice de Gravidade de Doença
18.
Front Immunol ; 2(15)2011 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-21822418

RESUMO

Lymphostromal cross-talk in the thymus is essential to allow generation of a diversified repertoire of T lymphocytes and to prevent autoimmunity by self-reactive T cells. Hypomorphic mutations in genes that control T cell development have been associated with immunodeficiency and immune dysregulation both in humans and in mice. We have studied T cell development and thymic stroma architecture and maturation in two mouse models of leaky severe combined immune deficiency, carrying hypomorphic mutations in rag1 and lig4 genes. Defective T cell development was associated with abnormalities of thymic architecture that predominantly affect the thymic medulla, with reduction of the pool of mature medullary thymic epithelial cells (mTECs). While the ability of mTECs to express autoimmune regulator (Aire) is preserved in mutant mice, the frequency of mature mTECs expressing Aire and tissue-specific antigens is severely reduced. Similarly, the ability of CD4(+) T cells to differentiate into Foxp3(+) natural regulatory T cells is preserved in rag1 and lig4 mutant mice, but their number is greatly reduced. These data indicate that hypomorphic defects in T cell development may cause defective lymphostromal cross-talk and impinge on thymic stromal cells maturation, and thus favor immune dysregulation.

19.
Front Immunol ; 2: 54, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22566844

RESUMO

We report the updated classification of primary immunodeficiency diseases, compiled by the ad hoc Expert Committee of the International Union of Immunological Societies. As compared to the previous edition, more than 15 novel disease entities have been added in the updated version. For each disorders, the key clinical and laboratory features are provided. This updated classification is meant to help in the diagnostic approach to patients with these diseases.

20.
J Comput Assist Tomogr ; 31(4): 620-8, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17882044

RESUMO

OBJECTIVE: The aims of the study were 1) to identify and quantify pulmonary changes in subjects affected by agammaglobulinemia (AG), and common variable immunodeficiency (CVID) and 2) to assess the incidence, type, and degree of chronic sinusitis and their relation to pulmonary changes. METHODS: Forty-five patients affected by AG (18) and CVID (27) underwent computed tomography of lungs and paranasal sinuses. RESULTS: Of 45 patients, 26 (57.7%) had pulmonary changes, more frequent among CVID than AG patients (P = 0.37). Bronchiectases were detected in 7 of 12 AG and in 9 of 14 CVID; the difference is not statistically significant (P = 0.53). Computed tomographic findings of chronic sinusitis were detected in 41 of 45 patients. There was no statistically significant difference between AG and CVID patients. Bronchial and sinusal abnormalities did not correlate in 11 patients followed longitudinally. CONCLUSIONS: On computed tomography, the type and severity of lung lesions do not correlate either with the type of immunodeficiency or with the severity of the sinusal involvement.


Assuntos
Agamaglobulinemia/diagnóstico por imagem , Imunodeficiência de Variável Comum/diagnóstico por imagem , Pulmão/diagnóstico por imagem , Seios Paranasais/diagnóstico por imagem , Tomografia Computadorizada por Raios X , Adolescente , Adulto , Bronquiectasia/diagnóstico por imagem , Bronquiectasia/etiologia , Criança , Doença Crônica , Feminino , Humanos , Estudos Longitudinais , Masculino , Sinusite/diagnóstico por imagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA