Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 185
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Blood ; 141(7): 713-724, 2023 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-36279417

RESUMO

Patients with hypomorphic mutations in the RAG1 or RAG2 gene present with either Omenn syndrome or atypical combined immunodeficiency with a wide phenotypic range. Hematopoietic stem cell transplantation (HSCT) is potentially curative, but data are scarce. We report on a worldwide cohort of 60 patients with hypomorphic RAG variants who underwent HSCT, 78% of whom experienced infections (29% active at HSCT), 72% had autoimmunity, and 18% had granulomas pretransplant. These complications are frequently associated with organ damage. Eight individuals (13%) were diagnosed by newborn screening or family history. HSCT was performed at a median of 3.4 years (range 0.3-42.9 years) from matched unrelated donors, matched sibling or matched family donors, or mismatched donors in 48%, 22%, and 30% of the patients, respectively. Grafts were T-cell depleted in 15 cases (25%). Overall survival at 1 and 4 years was 77.5% and 67.5% (median follow-up of 39 months). Infection was the main cause of death. In univariable analysis, active infection, organ damage pre-HSCT, T-cell depletion of the graft, and transplant from a mismatched family donor were predictive of worse outcome, whereas organ damage and T-cell depletion remained significant in multivariable analysis (hazard ratio [HR] = 6.01, HR = 8.46, respectively). All patients diagnosed by newborn screening or family history survived. Cumulative incidences of acute and chronic graft-versus-host disease were 35% and 22%, respectively. Cumulative incidences of new-onset autoimmunity was 15%. Immune reconstitution, particularly recovery of naïve CD4+ T cells, was faster and more robust in patients transplanted before 3.5 years of age, and without organ damage. These findings support the indication for early transplantation.


Assuntos
Doença Enxerto-Hospedeiro , Transplante de Células-Tronco Hematopoéticas , Recém-Nascido , Humanos , Doadores de Tecidos , Linfócitos T , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Diagnóstico Precoce , Efeitos Psicossociais da Doença , Doença Enxerto-Hospedeiro/diagnóstico , Doença Enxerto-Hospedeiro/etiologia , Estudos Retrospectivos , Doadores não Relacionados , Condicionamento Pré-Transplante
2.
Br J Haematol ; 204(3): 1061-1066, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37671902

RESUMO

Acute chest syndrome (ACS) is a frequent cause of hospitalization in sickle cell disease (SCD). Despite advances in acute care, many settings still lack knowledge about ACS best practices. After the AIEOP Guidelines were published in 2012, suggesting standardized management in Italy, a retrospective study was performed to assess the diagnostic and therapeutic pathways of ACS in children. From 2013 to 2018, 208 ACS episodes were presented by 122/583 kids in 11 centres. 73 were male, mean age 10.9 years, 85% African, 92% HbSS or Sß°. In our hub-and-spoke system, a good adherence to Guidelines was documented, but discrepancies between reference centres and general hospitals were noted. Improvement is needed for timely transfer to reference centres, use of incentive spirometry, oxygen therapy and pain management.


Assuntos
Síndrome Torácica Aguda , Anemia Falciforme , Criança , Humanos , Masculino , Feminino , Estudos Retrospectivos , Anemia Falciforme/tratamento farmacológico , Hemoglobina Falciforme , Hospitalização
3.
Blood Cells Mol Dis ; 108: 102860, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38889660

RESUMO

Acquired aplastic anemia (AA) is a rare heterogeneous disorder characterized by pancytopenia and hypoplastic bone marrow. The incidence is 2-3 per million population per year in the Western world, but 3 times higher in East Asia. Survival in severe aplastic anemia (SAA) has improved significantly due to advances in hematopoietic stem cell transplantation (HSCT), immunosuppressive therapy, biologic agents, and supportive care. In SAA, HSCT from a matched sibling donor (MSD) is the first-line treatment. If a MSD is not available, options include immunosuppressive therapy (IST), matched unrelated donor, or haploidentical HSCT. The purpose of this guideline is to provide health care professionals with clear guidance on the diagnosis and management of pediatric patients with AA. A preliminary evidence-based document prepared by a group of pediatric hematologists of the Bone Marrow Failure Study Group of the Italian Association of Pediatric Hemato-Oncology (AIEOP) was discussed, modified and approved during a series of consensus conferences that started online during COVID 19 and continued in the following years, according to procedures previously validated by the AIEOP Board of Directors.


Assuntos
Anemia Aplástica , Transplante de Células-Tronco Hematopoéticas , Anemia Aplástica/terapia , Anemia Aplástica/diagnóstico , Anemia Aplástica/etiologia , Humanos , Criança , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Itália , COVID-19/diagnóstico , Imunossupressores/uso terapêutico , SARS-CoV-2
4.
Blood ; 132(3): 281-292, 2018 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-29743177

RESUMO

Hypomorphic RAG1 mutations allowing residual T- and B-cell development have been found in patients presenting with delayed-onset combined immune deficiency with granulomas and/or autoimmunity (CID-G/AI) and abnormalities of the peripheral T- and B-cell repertoire. To examine how hypomorphic Rag1 mutations affect the earliest stages of lymphocyte development, we used CRISPR/Cas9 to generate mouse models with mutations equivalent to those found in patients with CID-G/AI. Immunological characterization showed partial development of T and B lymphocytes, with persistence of naïve cells and preserved serum immunoglobulin but impaired antibody responses and presence of autoantibodies, thereby recapitulating the phenotype seen in patients with CID-G/AI. By using high-throughput sequencing, we identified marked skewing of Igh V and Trb V gene usage in early progenitors, with a bias for productive Igh and Trb rearrangements after selection occurred and increased apoptosis of B-cell progenitors. Rearrangement at the Igk locus was impaired, and polyreactive immunoglobulin M antibodies were detected. This study provides novel insights into how hypomorphic Rag1 mutations alter the primary repertoire of T and B cells, setting the stage for immune dysregulation frequently seen in patients.


Assuntos
Diferenciação Celular/genética , Genes RAG-1 , Linfopoese/genética , Mutação , Alelos , Animais , Linfócitos B/citologia , Linfócitos B/imunologia , Linfócitos B/metabolismo , Biomarcadores , Suscetibilidade a Doenças/imunologia , Edição de Genes , Regulação da Expressão Gênica , Predisposição Genética para Doença , Imunidade Humoral , Imunofenotipagem , Camundongos , Camundongos Transgênicos , Fenótipo , Linfócitos T/citologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Recombinação V(D)J
5.
Genes Immun ; 16(2): 151-61, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25569260

RESUMO

Lymphocyte apoptosis is mainly induced by either death receptor-dependent activation of caspase-8 or mitochondria-dependent activation of caspase-9. Mutations in caspase-8 lead to autoimmunity/lymphoproliferation and immunodeficiency. This work describes a heterozygous H237P mutation in caspase-9 that can lead to similar disorders. H237P mutation was detected in two patients: Pt1 with autoimmunity/lymphoproliferation, severe hypogammaglobulinemia and Pt2 with mild hypogammaglobulinemia and Burkitt lymphoma. Their lymphocytes displayed defective caspase-9 activity and decreased apoptotic and activation responses. Transfection experiments showed that mutant caspase-9 display defective enzyme and proapoptotic activities and a dominant-negative effect on wild-type caspase-9. Ex vivo analysis of the patients' lymphocytes and in vitro transfection experiments showed that the expression of mutant caspase-9 correlated with a downregulation of BAFFR (B-cell-activating factor belonging to the TNF family (BAFF) receptor) in B cells and ICOS (inducible T-cell costimulator) in T cells. Both patients carried a second inherited heterozygous mutation missing in the relatives carrying H237P: Pt1 in the transmembrane activator and CAML interactor (TACI) gene (S144X) and Pt2 in the perforin (PRF1) gene (N252S). Both mutations have been previously associated with immunodeficiencies in homozygosis or compound heterozygosis. Taken together, these data suggest that caspase-9 mutations may predispose to immunodeficiency by cooperating with other genetic factors, possibly by downregulating the expression of BAFFR and ICOS.


Assuntos
Receptor do Fator Ativador de Células B/biossíntese , Caspase 9/genética , Síndromes de Imunodeficiência/genética , Proteína Coestimuladora de Linfócitos T Induzíveis/biossíntese , Transtornos Linfoproliferativos/genética , Mutação , Adolescente , Adulto , Apoptose/genética , Apoptose/imunologia , Receptor do Fator Ativador de Células B/genética , Receptor do Fator Ativador de Células B/imunologia , Caspase 9/imunologia , Regulação para Baixo , Células HEK293 , Humanos , Síndromes de Imunodeficiência/imunologia , Síndromes de Imunodeficiência/metabolismo , Proteína Coestimuladora de Linfócitos T Induzíveis/genética , Proteína Coestimuladora de Linfócitos T Induzíveis/imunologia , Transtornos Linfoproliferativos/imunologia , Transtornos Linfoproliferativos/metabolismo , Masculino , Linhagem
7.
Infection ; 41(4): 827-31, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23575516

RESUMO

BACKGROUND: Interferon-gamma release assays (IGRAs) have high specificity and sensitivity for the diagnosis of tuberculosis (TB) infection. However, their role as a screening tool in children with immunodeficiency disorders is still unclear. In the present study, we performed a contact investigation using serial IGRAs on children with immunodeficiency conditions exposed to a contagious TB patient. METHODS: Children who were exposed to a contagious TB case underwent serial QuantiFERON(®) TB Gold In-Tube (QFT-GIT) and T-SPOT(®).TB (T-SPOT) testing. RESULTS: Eighteen children were tested. At the first testing, only two children (11 %) were positive to T-SPOT. Indeterminate results were more frequent with QFT-GIT (35 %) than with T-SPOT (12 %). In the multivariable analysis, a statistically significant association of lymphocyte count <500 cells/mm(3) (p < 0.00005) and low age (p = 0.03) with indeterminate results for the QFT-GIT test but not for T-SPOT (p = 0.10 and p = 0.88, respectively) was found. At the end of October 2012, 15 of the 18 children were alive and none developed active TB disease. CONCLUSION: T-SPOT provided more determinate results and was less influenced by low age and lymphocytopenia than QFT-GIT in this population of immunodeficient children. These findings suggest that T-SPOT is a more accurate test for the identification of TB infection in young children with lymphocytopenia and should be preferred to QFT-GIT under such specific conditions.


Assuntos
Busca de Comunicante/métodos , Testes de Liberação de Interferon-gama/métodos , Tuberculose Pulmonar/diagnóstico , Tuberculose Pulmonar/transmissão , Adolescente , Criança , Pré-Escolar , Infecção Hospitalar/diagnóstico , Infecção Hospitalar/transmissão , Feminino , Neoplasias Hematológicas/complicações , Humanos , Lactente , Masculino , Programas de Rastreamento/métodos , Adulto Jovem
8.
Nat Genet ; 9(4): 414-7, 1995 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-7795648

RESUMO

X-linked thrombocytopenia (XLT) is a rare recessive hereditary disorder characterized by isolated thrombocytopenia with small-sized platelets. The XLT locus has been located to chromosome Xp11 by linkage analysis, which is also where the recently cloned Wiskott-Aldrich syndrome (WAS) gene, maps. The relationship between XLT and WAS has long been debated; they might be due to different mutations of the same gene or to mutations in different genes. We now show that mutations in the WAS gene, different from those found in WAS patients, are present in three unrelated male patients with isolated thrombocytopenia and small-sized platelets. Our results demonstrate that XLT and WAS are allelic forms of the same disease, but the causes of the differences need to be further investigated.


Assuntos
Alelos , Mutação , Proteínas/genética , Trombocitopenia/genética , Síndrome de Wiskott-Aldrich/genética , Cromossomo X , Sequência de Bases , Criança , DNA/genética , Primers do DNA/genética , Éxons , Mutação da Fase de Leitura , Genes , Humanos , Íntrons , Masculino , Dados de Sequência Molecular , Mutação Puntual , Reação em Cadeia da Polimerase , Proteína da Síndrome de Wiskott-Aldrich
9.
Nat Genet ; 25(3): 343-6, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10888887

RESUMO

Osteopetrosis includes a group of inherited diseases in which inadequate bone resorption is caused by osteoclast dysfunction. Although molecular defects have been described for many animal models of osteopetrosis, the gene responsible for most cases of the severe human form of the disease (infantile malignant osteopetrosis) is unknown. Infantile malignant autosomal recessive osteopetrosis (MIM 259700) is a severe bone disease with a fatal outcome, generally within the first decade of life. Osteoclasts are present in normal or elevated numbers in individuals affected by autosomal recessive osteopetrosis, suggesting that the defect is not in osteoclast differentiation, but in a gene involved in the functional capacity of mature osteoclasts. Some of the mouse mutants have a decreased number of osteoclasts, which suggests that the defect directly interferes with osteoclast differentiation. In other mutants, it is the function of the osteoclast that seems to be affected, as they show normal or elevated numbers of non-functioning osteoclasts. Here we show that TCIRG1, encoding the osteoclast-specific 116-kD subunit of the vacuolar proton pump, is mutated in five of nine patients with a diagnosis of infantile malignant osteopetrosis. Our data indicate that mutations in TCIRG1 are a frequent cause of autosomal recessive osteopetrosis in humans.


Assuntos
Osteopetrose/genética , Bombas de Próton/genética , ATPases Translocadoras de Prótons/genética , ATPases Vacuolares Próton-Translocadoras , Processamento Alternativo , Sequência de Bases , Medula Óssea/patologia , DNA Complementar , Éxons , Feminino , Mutação da Fase de Leitura , Genes Recessivos , Humanos , Lactente , Íntrons , Masculino , Dados de Sequência Molecular , Osteopetrose/patologia
10.
Clin Exp Immunol ; 169(1): 57-69, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22670779

RESUMO

Primary immunodeficiency diseases (PIDs) comprise a heterogeneous group of rare disorders. This study was devised in order to compare management of these diseases in the northern hemisphere, given the variability of practice among clinicians in North America. The members of two international societies for clinical immunologists were asked about their management protocols in relation to their PID practice. An anonymous internet questionnaire, used previously for a survey of the American Academy of Allergy, Asthma and Immunology (AAAAI), was offered to all full members of the European Society for Immunodeficiency (ESID). The replies were analysed in three groups, according to the proportion of PID patients in the practice of each respondent; this resulted in two groups from North America and one from Europe. The 123 responses from ESID members (23·7%) were, in the majority, very similar to those of AAAAI respondents, with > 10% of their practice devoted to primary immunodeficiency. There were major differences between the responses of these two groups and those of the general AAAAI respondents whose clinical practice was composed of < 10% of PID patients. These differences included the routine use of intravenous immunoglobulin therapy (IVIg) for particular types of PIDs, initial levels of IVIg doses, dosing intervals, routine use of prophylactic antibiotics, perceptions of the usefulness of subcutaneous immunoglobulin therapy (SCIg) and of the risk to patients' health of policies adopted by health-care funders. Differences in practice were identified and are discussed in terms of methods of health-care provision, which suggest future studies for ensuring continuation of appropriate levels of immunoglobulin replacement therapies.


Assuntos
Imunoglobulinas Intravenosas/uso terapêutico , Síndromes de Imunodeficiência/terapia , Padrões de Prática Médica/estatística & dados numéricos , Academias e Institutos , Antibacterianos/uso terapêutico , Europa (Continente) , Humanos , Internet , América do Norte , Guias de Prática Clínica como Assunto , Inquéritos e Questionários
11.
Blood Cancer J ; 11(9): 151, 2021 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-34521813

RESUMO

The ability of patients with hematologic malignancies (HM) to develop an effective humoral immune response after COVID-19 is unknown. A prospective study was performed to monitor the immune response to SARS-CoV-2 of patients with follicular lymphoma (FL), diffuse large B-cell lymphoma (DLBCL), chronic lymphoproliferative disorders (CLD), multiple myeloma (MM), or myelodysplastic/myeloproliferative syndromes (MDS/MPN). Antibody (Ab) levels to the SARS-CoV-2 nucleocapsid (N) and spike (S) protein were measured at +1, +3, +6 months after nasal swabs became PCR-negative. Forty-five patients (9 FL, 8 DLBCL, 8 CLD, 10 MM, 10 MDS/MPS) and 18 controls were studied. Mean anti-N and anti-S-Ab levels were similar between HM patients and controls, and shared the same behavior, with anti-N Ab levels declining at +6 months and anti-S-Ab remaining stable. Seroconversion rates were lower in HM patients than in controls. In lymphoma patients mean Ab levels and seroconversion rates were lower than in other HM patients, primarily because all nine patients who had received rituximab within 6 months before COVID-19 failed to produce anti-N and anti-S-Ab. Only one patient requiring hematological treatment after COVID-19 lost seropositivity after 6 months. No reinfections were observed. These results may inform vaccination policies and clinical management of HM patients.


Assuntos
COVID-19/imunologia , Neoplasias Hematológicas/imunologia , Imunidade Humoral/efeitos dos fármacos , Rituximab/farmacologia , SARS-CoV-2/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Anticorpos Antivirais/efeitos dos fármacos , Anticorpos Antivirais/metabolismo , Formação de Anticorpos/efeitos dos fármacos , Formação de Anticorpos/fisiologia , Especificidade de Anticorpos/efeitos dos fármacos , COVID-19/complicações , COVID-19/epidemiologia , COVID-19/terapia , Estudos de Casos e Controles , Feminino , Seguimentos , Neoplasias Hematológicas/complicações , Neoplasias Hematológicas/tratamento farmacológico , Neoplasias Hematológicas/epidemiologia , Hospitalização , Humanos , Itália/epidemiologia , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Rituximab/uso terapêutico
12.
Genes Immun ; 11(8): 665-70, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20927127

RESUMO

Cystic fibrosis (CF) is a common life-threatening autosomal recessive disorder in the Caucasian population, and the gene responsible is the CF transmembrane conductance regulator (CFTR). Patients with CF have repeated bacterial infection of the airways caused by Pseudomonas aeruginosa (PA), which is one of the predominant pathogen, and endobronchial chronic infection represents a major cause of morbidity and mortality. Pentraxin 3 (PTX3) is a gene that encodes the antimicrobial protein, PTX3, which is believed to have an important role in innate immunity of lung. To address the role of PTX3 in the risk of PA lung colonization, we investigated five single nucleotide polymorphisms of PTX3 gene in 172 Caucasian CF patients who were homozygous for the F508del mutation. We observed that PTX3 haplotype frequencies were significantly different between patients with PA colonization, as compared with noncolonized patients. Moreover, a protective effect was found in association with a specific haplotype (odds ratio 0.524). Our data suggest that variations within PTX3 affect lung colonization of Pseudomonas in patients with CF.


Assuntos
Proteína C-Reativa/genética , Fibrose Cística/genética , Fibrose Cística/microbiologia , Componente Amiloide P Sérico/genética , Proteína C-Reativa/metabolismo , Fibrose Cística/complicações , Fibrose Cística/imunologia , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Variação Genética , Genótipo , Haplótipos , Homozigoto , Humanos , Imunidade Inata , Polimorfismo de Nucleotídeo Único , Infecções por Pseudomonas/imunologia , Pseudomonas aeruginosa/metabolismo , Componente Amiloide P Sérico/metabolismo
13.
J Exp Med ; 183(6): 2687-92, 1996 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-8676091

RESUMO

Mutations affecting the expression of the Janus family kinase JAK3 were recently shown to be responsible for autosomal recessive severe combined immunodeficiency (SCID). JAK3-deficient patients present with a clinical phenotype virtually indistinguishable from boys affected by X-linked SCID, a disease caused by genetic defects of the common gamma chain (gamma c) that is a shared component of the receptors for IL-2, IL-4, IL-7, IL-9, and IL-15. The specific interaction of JAK3 and gamma c represents the biochemical basis for the similarities between these two immunodeficiencies. Both forms of SCID are characterized by recurrent, severe infections leading to death in infancy unless successfully treated by allogeneic bone marrow transplantation. Because of the potentially lethal complications associated with allogeneic bone marrow transplantation and the frequent lack of suitable marrow donors, the development of alternative forms of therapy is highly desirable. To this end, we investigated a retroviral-mediated gene correction approach for JAK3-deficiency. A vector carrying a copy of JAK3 cDNA was constructed and used to transduce B cell lines derived from patients with JAK3-deficient SCID. We demonstrate restoration of JAK3 expression and phosphorylation upon IL-2 and IL-4 stimulation. Furthermore, patients' cells transduced with JAK3 acquired the ability to proliferate normally in response to IL-2. These data indicate that the biological defects of JAK3-deficient cells can be efficiently corrected in vitro by retroviral-mediated gene transfer, thus providing the basis for future investigation of gene therapy as treatment for JAK3-deficient SCID.


Assuntos
Terapia Genética , Proteínas Tirosina Quinases/deficiência , Proteínas Tirosina Quinases/genética , Receptores de Interleucina/genética , Imunodeficiência Combinada Severa/genética , Imunodeficiência Combinada Severa/terapia , Linfócitos B/imunologia , Western Blotting , Células Cultivadas , Criança , Clonagem Molecular , Consanguinidade , Genes Recessivos , Vetores Genéticos , Humanos , Interleucina-2/farmacologia , Janus Quinase 3 , Ativação Linfocitária/efeitos dos fármacos , Substâncias Macromoleculares , Masculino , Proteínas Tirosina Quinases/biossíntese , Proteínas Recombinantes/biossíntese , Retroviridae , Imunodeficiência Combinada Severa/enzimologia , Transfecção , Cromossomo X
14.
J Exp Med ; 194(3): 235-46, 2001 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-11489943

RESUMO

In humans, natural killer (NK) cell function is regulated by a series of receptors and coreceptors with either triggering or inhibitory activity. Here we describe a novel 60-kD glycoprotein, termed NTB-A, that is expressed by all human NK, T, and B lymphocytes. Monoclonal antibody (mAb)-mediated cross-linking of NTB-A results in the induction of NK-mediated cytotoxicity. Similar to 2B4 (CD244) functioning as a coreceptor in the NK cell activation, NTB-A also triggers cytolytic activity only in NK cells expressing high surface densities of natural cytotoxicity receptors. This suggests that also NTB-A may function as a coreceptor in the process of NK cell activation. Molecular cloning of the cDNA coding for NTB-A molecule revealed a novel member of the immunoglobulin superfamily belonging to the CD2 subfamily. NTB-A is characterized, in its extracellular portion, by a distal V-type and a proximal C2-type domain and by a cytoplasmic portion containing three tyrosine-based motifs. NTB-A undergoes tyrosine phosphorylation and associates with the Src homology 2 domain-containing protein (SH2D1A) as well as with SH2 domain-containing phosphatases (SHPs). Importantly, analysis of NK cells derived from patients with X-linked lymphoproliferative disease (XLP) showed that the lack of SH2D1A protein profoundly affects the function not only of 2B4 but also of NTB-A. Thus, in XLP-NK cells, NTB-A mediates inhibitory rather than activating signals. These inhibitory signals are induced by the interaction of NTB-A with still undefined ligands expressed on Epstein-Barr virus (EBV)-infected target cells. Moreover, mAb-mediated masking of NTB-A can partially revert this inhibitory effect while a maximal recovery of target cell lysis can be obtained when both 2B4 and NTB-A are simultaneously masked. Thus, the altered function of NTB-A appears to play an important role in the inability of XLP-NK cells to kill EBV-infected target cells.


Assuntos
Linfócitos B/imunologia , Linfócitos B/virologia , Herpesvirus Humano 4/imunologia , Células Matadoras Naturais/imunologia , Transtornos Linfoproliferativos/imunologia , Glicoproteínas de Membrana/imunologia , Animais , Anticorpos Monoclonais , Sequência de Bases , Citotoxicidade Imunológica , Primers do DNA/genética , Humanos , Técnicas In Vitro , Transtornos Linfoproliferativos/genética , Masculino , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Camundongos , Mutação , Receptores Imunológicos/química , Receptores Imunológicos/genética , Domínios de Homologia de src
15.
J Exp Med ; 192(3): 337-46, 2000 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-10934222

RESUMO

2B4 is a surface molecule involved in activation of the natural killer (NK) cell-mediated cytotoxicity. It binds a protein termed Src homology 2 domain-containing protein (SH2D1A) or signaling lymphocyte activation molecule (SLAM)-associated protein (SAP), which in turn has been proposed to function as a regulator of the 2B4-associated signal transduction pathway. In this study, we analyzed patients with X-linked lymphoproliferative disease (XLP), a severe inherited immunodeficiency characterized by critical mutations in the SH2D1A gene and by the inability to control Epstein-Barr virus (EBV) infections. We show that, in these patients, 2B4 not only fails to transduce triggering signals, but also mediates a sharp inhibition of the NK-mediated cytolysis. Other receptors involved in NK cell triggering, including CD16, NKp46, NKp44, and NKp30, displayed a normal functional capability. However, their activating function was inhibited upon engagement of 2B4 molecules. CD48, the natural ligand of 2B4, is highly expressed on the surface of EBV(+) B cell lines. Remarkably, NK cells from XLP patients could not kill EBV(+) B cell lines. This failure was found to be the consequence of inhibitory signals generated by the interaction between 2B4 and CD48, as the antibody-mediated disruption of the 2B4-CD48 interaction restored lysis of EBV(+) target cells lacking human histocompatibility leukocyte antigen (HLA) class I molecules. In the case of autologous or allogeneic (HLA class I(+)) EBV(+) lymphoblastoid cell lines, restoration of lysis was achieved only by the simultaneous disruption of 2B4-CD48 and NK receptor-HLA class I interactions. Molecular analysis revealed that 2B4 molecules isolated from either XLP or normal NK cells were identical. As expected, in XLP-NK cells, 2B4 did not associate with SH2D1A, whereas similar to 2B4 molecules isolated from normal NK cells, it did associate with Src homology 2 domain-containing phosphatase 1.


Assuntos
Antígenos CD , Herpesvirus Humano 4/imunologia , Peptídeos e Proteínas de Sinalização Intracelular , Células Matadoras Naturais/imunologia , Transtornos Linfoproliferativos/imunologia , Glicoproteínas de Membrana/imunologia , Receptores Imunológicos/imunologia , Transdução de Sinais , Cromossomo X , Sequência de Bases , Proteínas de Transporte/genética , Linhagem Celular , Membrana Celular/metabolismo , Pré-Escolar , DNA Complementar , Ligação Genética , Humanos , Células Matadoras Naturais/virologia , Ativação Linfocitária/imunologia , Transtornos Linfoproliferativos/sangue , Transtornos Linfoproliferativos/genética , Masculino , Glicoproteínas de Membrana/biossíntese , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/fisiologia , Pessoa de Meia-Idade , Dados de Sequência Molecular , Mutação , Receptor 1 Desencadeador da Citotoxicidade Natural , Receptores Imunológicos/metabolismo , Proteína Associada à Molécula de Sinalização da Ativação Linfocitária , Família de Moléculas de Sinalização da Ativação Linfocitária , Células Tumorais Cultivadas
16.
Clin Exp Immunol ; 158 Suppl 1: 14-22, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19883420

RESUMO

Primary immunodeficiencies (PIDs) are uncommon, chronic and severe disorders of the immune system in which patients cannot mount a sufficiently protective immune response, leading to an increased susceptibility to infections. The treatment of choice for PID patients with predominant antibody deficiency is intravenous immunoglobulin (Ig) replacement therapy. Despite major advances over the last 20 years in the molecular characterization of PIDs, many patients remain undiagnosed or are diagnosed too late, with severe consequences. Various strategies to ensure timely diagnosis of PIDs are in place, and novel approaches are being developed. In recent years, several patient registries have been established. Such registries shed light on the pathology and natural history of these varied disorders. Analyses of the registry data may also reveal which patients are likely to respond well to higher Ig infusion rates and may help to determine the optimal dosing of Ig products. Faster infusion rates may lead to improved convenience for patients and thus increase patient compliance, and may reduce nursing time and the need for hospital resources. Data from two recent studies suggest that Gamunex and Privigen are well tolerated at high infusion rates. Nevertheless, careful selection of patients for high infusion rates, based on co-morbid conditions and tolerance of the current infusion rate, is advisable. Based on the available data, intravenous Ig offers broad protection against encapsulated organisms. As vaccine trends change, careful monitoring of specific antibody levels in the general population, such as those against pneumococcal and meningococcal bacteria, should be implemented.


Assuntos
Síndromes de Imunodeficiência/diagnóstico , Anticorpos Antibacterianos/sangue , Infecções Bacterianas/imunologia , Infecções Bacterianas/prevenção & controle , Bases de Dados Factuais , Humanos , Imunoglobulinas Intravenosas/imunologia , Imunoglobulinas Intravenosas/uso terapêutico , Síndromes de Imunodeficiência/complicações , Síndromes de Imunodeficiência/epidemiologia , Síndromes de Imunodeficiência/terapia , Cooperação Internacional , Infecções Oportunistas/imunologia , Infecções Oportunistas/prevenção & controle , Sistema de Registros
17.
Science ; 270(5235): 470-5, 1995 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-7570000

RESUMO

Adenosine deaminase (ADA) deficiency results in severe combined immunodeficiency, the first genetic disorder treated by gene therapy. Two different retroviral vectors were used to transfer ex vivo the human ADA minigene into bone marrow cells and peripheral blood lymphocytes from two patients undergoing exogenous enzyme replacement therapy. After 2 years of treatment, long-term survival of T and B lymphocytes, marrow cells, and granulocytes expressing the transferred ADA gene was demonstrated and resulted in normalization of the immune repertoire and restoration of cellular and humoral immunity. After discontinuation of treatment, T lymphocytes, derived from transduced peripheral blood lymphocytes, were progressively replaced by marrow-derived T cells in both patients. These results indicate successful gene transfer into long-lasting progenitor cells, producing a functional multilineage progeny.


Assuntos
Adenosina Desaminase/deficiência , Adenosina Desaminase/genética , Técnicas de Transferência de Genes , Terapia Genética , Células-Tronco Hematopoéticas , Linfócitos , Imunodeficiência Combinada Severa/terapia , Adenosina Desaminase/administração & dosagem , Adenosina Desaminase/sangue , Adenosina Desaminase/uso terapêutico , Formação de Anticorpos , Sequência de Bases , Células da Medula Óssea , Células Cultivadas , Pré-Escolar , Vetores Genéticos , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/enzimologia , Humanos , Imunidade Celular , Transfusão de Linfócitos , Linfócitos/enzimologia , Linfócitos/imunologia , Dados de Sequência Molecular , Imunodeficiência Combinada Severa/enzimologia , Imunodeficiência Combinada Severa/genética , Imunodeficiência Combinada Severa/imunologia , Linfócitos T/enzimologia , Linfócitos T/imunologia
18.
Bone Marrow Transplant ; 41 Suppl 2: S83-6, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18545252

RESUMO

BMT is curative in almost 75% of children affected by severe primary immunodeficiencies (PIDs). Recently, the chance of cure has increased thanks to the availability of matched unrelated donors (MUDs). Nevertheless, besides the conventional indications to BMT (profound or absent T-cell function, profound or absent natural killer function, known syndromes with T-cell deficiencies), indications to BMT for PIDs affecting the quality of life or having an expectation of life that does not exceed the third-fourth decade remain unclear. Infact, if it is evident that the survival rate in an infant grafted for a PID with a MUD is expected to be more than 80%, alternative treatments such as gene therapy are now available.


Assuntos
Transplante de Medula Óssea , Síndromes de Imunodeficiência/terapia , Sistema de Registros , Transplante de Células-Tronco , Pré-Escolar , Intervalo Livre de Doença , Humanos , Lactente , Itália , Transplante Homólogo
19.
Reprod Biomed Online ; 17(2): 265-7, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18682002

RESUMO

Anti-neoplastic treatments have significantly increased the survival of cancer patients, but female patients risk premature menopause. Oocyte cryopreservation has been proposed as a fertility-saving option. This report describes the first live birth achieved with autologous cryopreserved oocytes in an ovariectomized borderline cancer patient. A patient with a borderline ovarian tumour asked for oocyte cryopreservation after a right adnexectomy. Ovulation induction resulted in the retrieval and cryopreservation of seven mature oocytes. Thirty-nine months after a left ovariectomy, the patient asked for oocyte thawing and embryo transfer. Endometrial growth was induced using hormone replacement treatment. Three of the seven cryopreserved oocytes were thawed; they survived and, after insemination, normal fertilization took place. Three embryos were transferred into the patient's uterus. A twin pregnancy was achieved with the birth of two healthy females. Oocyte cryopreservation may be a reliable option for preserving fertility in young cancer patients who risk premature menopause due to surgery, chemotherapy or radiotherapy.


Assuntos
Carcinoma/cirurgia , Criopreservação , Nascido Vivo , Oócitos , Neoplasias Ovarianas/cirurgia , Ovariectomia , Gravidez Múltipla , Adulto , Carcinoma/reabilitação , Feminino , Fertilização in vitro/métodos , Humanos , Recém-Nascido , Recuperação de Oócitos/métodos , Neoplasias Ovarianas/reabilitação , Ovariectomia/reabilitação , Gravidez , Resultado do Tratamento , Gêmeos
20.
Mucosal Immunol ; 11(1): 50-60, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28513593

RESUMO

Loss-of-function mutations in the tyrosine kinase JAK3 cause autosomal recessive severe combined immunodeficiency (SCID). Defects in this form of SCID are restricted to the immune system, which led to the development of immunosuppressive JAK inhibitors. We find that the B6.Cg-Nr1d1tm1Ven/LazJ mouse line purchased from Jackson Laboratories harbors a spontaneous mutation in Jak3, generating a SCID phenotype and an inability to generate antigen-independent professional cytokine-producing innate lymphoid cells (ILCs). Mechanistically, Jak3 deficiency blocks ILC differentiation in the bone marrow at the ILC precursor and the pre-NK cell progenitor. We further demonstrate that the pan-JAK inhibitor tofacitinib and the specific JAK3 inhibitor PF-06651600 impair the ability of human intraepithelial ILC1 (iILC1) to produce IFN-γ, without affecting ILC3 production of IL-22. Both inhibitors impaired the proliferation of iILC1 and ILC3 and differentiation of human ILC in vitro. Tofacitinib is currently approved for the treatment of moderate-to-severely active rheumatoid arthritis. Both tofacitinib and PF-06651600 are currently in clinical trials for several other immune-mediated conditions. Our data suggest that therapeutic inhibition of JAK may also impact ILCs and, to some extent, underlie clinical efficacy.


Assuntos
Artrite Reumatoide/tratamento farmacológico , Células da Medula Óssea/fisiologia , Janus Quinase 3/genética , Células Matadoras Naturais/fisiologia , Mutação/genética , Piperidinas/uso terapêutico , Pirimidinas/uso terapêutico , Pirróis/uso terapêutico , Imunodeficiência Combinada Severa/genética , Animais , Diferenciação Celular/genética , Proliferação de Células/genética , Células Cultivadas , Humanos , Imunidade Inata , Interferon gama/metabolismo , Janus Quinase 3/antagonistas & inibidores , Camundongos , Camundongos Mutantes , Fenótipo , Piperidinas/farmacologia , Pirimidinas/farmacologia , Pirróis/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA