Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
1.
Front Immunol ; 11: 830, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32431714

RESUMO

22q11.2 deletion syndrome (DiGeorge), CHARGE syndrome, Nude/SCID and otofaciocervical syndrome type 2 (OTFCS2) are distinct clinical conditions in humans that can result in hypoplasia and occasionally, aplasia of the thymus. Thymic hypoplasia/aplasia is first suggested by absence or significantly reduced numbers of recent thymic emigrants, revealed in standard-of-care newborn screens for T cell receptor excision circles (TRECs). Subsequent clinical assessments will often indicate whether genetic mutations are causal to the low T cell output from the thymus. However, the molecular mechanisms leading to the thymic hypoplasia/aplasia in diverse human syndromes are not fully understood, partly because the problems of the thymus originate during embryogenesis. Rodent and Zebrafish models of these clinical syndromes have been used to better define the underlying basis of the clinical presentations. Results from these animal models are uncovering contributions of different cell types in the specification, differentiation, and expansion of the thymus. Cell populations such as epithelial cells, mesenchymal cells, endothelial cells, and thymocytes are variably affected depending on the human syndrome responsible for the thymic hypoplasia. In the current review, findings from the diverse animal models will be described in relation to the clinical phenotypes. Importantly, these results are suggesting new strategies for regenerating thymic tissue in patients with distinct congenital disorders.


Assuntos
Síndrome Brânquio-Otorrenal/complicações , Síndrome CHARGE/complicações , Síndrome de DiGeorge/complicações , Síndromes de Imunodeficiência/etiologia , Imunodeficiência Combinada Severa/complicações , Timo/anormalidades , Animais , Síndrome Brânquio-Otorrenal/genética , Síndrome Brânquio-Otorrenal/imunologia , Síndrome CHARGE/genética , Síndrome CHARGE/imunologia , Síndrome de DiGeorge/genética , Síndrome de DiGeorge/imunologia , Modelos Animais de Doenças , Humanos , Síndromes de Imunodeficiência/imunologia , Camundongos , Mutação , Ratos , Imunodeficiência Combinada Severa/genética , Imunodeficiência Combinada Severa/imunologia , Timo/embriologia , Timo/imunologia , Peixe-Zebra
2.
Pediatr Transplant ; 23(4): e13424, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31033123

RESUMO

CHARGE syndrome is a rare congenital malformation syndrome which may share symptoms with DiGeorge syndrome. Complete DiGeorge syndrome (cDGS) is a severe form of DiGeorge syndrome, characterized by a CD3+ T-cell count of <50/mm3 due to athymia, and is fatal without immunologic intervention. We performed peripheral blood lymphocyte transfusion (PBLT) from an HLA-identical sibling without pretransplant conditioning in a CHARGE/cDGS patient with a novel CHD7 splice site mutation. Cyclosporine and short-term methotrexate were used for graft versus host disease (GVHD) prophylaxis, and neither acute nor chronic GVHD was observed. After PBLT, T-cell proliferative response to phytohemagglutinin and concanavalin A recovered, and intractable diarrhea improved. EBV infection, evidenced by a gradual increase in the viral genome copy number to a maximum of 2861 copies/µgDNA on day 42 after PBLT, resolved spontaneously. HLA A2402 restricted, EBV-specific CTLs were detected from peripheral blood on day 148, and EBV seroconversion was observed on day 181. Thus, EBV-specific immunity was successfully established by PBLT. Our results indicate that PBLT is a simple and effective therapy to reconstitute immune systems in CHARGE/DiGeorge syndrome.


Assuntos
Síndrome CHARGE/terapia , Síndrome de DiGeorge/complicações , Síndrome de DiGeorge/imunologia , Infecções por Vírus Epstein-Barr/prevenção & controle , Transfusão de Linfócitos , Complexo CD3/metabolismo , Proliferação de Células , Concanavalina A/farmacologia , Ciclosporina/administração & dosagem , Diarreia/terapia , Infecções por Vírus Epstein-Barr/imunologia , Evolução Fatal , Doença Enxerto-Hospedeiro , Antígenos HLA/química , Herpesvirus Humano 4/genética , Humanos , Recém-Nascido , Masculino , Metotrexato/administração & dosagem , Mutação , Fenótipo , Fito-Hemaglutininas/química , Irmãos , Linfócitos T/citologia
3.
J Clin Immunol ; 39(1): 65-74, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30569262

RESUMO

PURPOSE: Immunodeficiency is one of the key features of 22q11.2 deletion syndrome (del), and it is seen in approximately 75% of the patients. The degree of immunodeficiency varies widely, from no circulating T cells to normal T cell counts. It has been hypothesized that the low number of T cells may at least in part be due to increased apoptosis of T cells. Increased spontaneous T cell apoptosis has been reported in one patient with 22q11.2del, but this has not been further investigated. METHODS: A national cohort of patients with a proven heterozygous deletion of chromosome 22q11.2 diagnosed by FISH or MLPA and a group of age and sex matched controls were studied. Spontaneous and activation-induced apoptosis, in addition to FAS expression on lymphocytes, were measured using flow cytometry. Serum levels of FASL were analyzed using ELISA. RESULTS: There was no increased spontaneous apoptosis in patients with 22q11.2del. Upon activation, anti-FAS-induced apoptosis was significantly increased in patients compared to those in controls, while there was no difference in activation induced cell death or activated cell autonomous death. We also found a significant increase in expression of FAS on freshly isolated lymphocytes from patients, while there was no difference in serum levels of FASL. Patients with congenital heart defects (CHD) had significantly higher serum levels of FASL compared to non-CHD patients. CONCLUSION: We have shown increased FAS expression on lymphocytes from patients with 22q11.2del as well as increased levels of FASL in patients with CHD. Those changes may contribute to the pathophysiology of the 22q11.2del.


Assuntos
Apoptose/imunologia , Cromossomos Humanos Par 22/genética , Síndrome de DiGeorge/genética , Síndrome de DiGeorge/imunologia , Linfócitos T/imunologia , Receptor fas/genética , Adolescente , Criança , Pré-Escolar , Cromossomos Humanos Par 22/imunologia , Feminino , Humanos , Masculino
4.
Curr Allergy Asthma Rep ; 18(12): 75, 2018 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-30377837

RESUMO

PURPOSE OF REVIEW: This review provides an update on the progress in identifying the range of immunological dysfunction seen in DiGeorge syndrome and on more recent diagnostic and treatment approaches. RECENT FINDINGS: Clinically, the associated thymic hypoplasia/aplasia is well known and can have profound effects on T cell function. Further, the humoral arm of the immune system can be affected, with hypogammaglobulinemia and poor vaccine-specific antibody response. Additionally, genetic testing utilizing chromosomal microarray demonstrates a small but significant number of 22q11 deletions that are not detectable by standard FISH testing. The recent addition of a TREC assay to newborn screening can identify a subset of infants whose severe immune defects may result from 22q11 deletion. This initial presentation now also places the immunologist in the role of "first responder" with regard to diagnosis and management of these patients. DiGeorge syndrome reflects a clinical phenotype now recognized by its underlying genetic diagnosis, chromosome 22q11.2 deletion syndrome, which is associated with multisystem involvement and variable immune defects among patients. Updated genetic and molecular techniques now allow for earlier identification of immune defects and confirmatory diagnoses, in this disorder with life-long clinical issues.


Assuntos
Síndrome de DiGeorge/genética , Síndrome de DiGeorge/imunologia , Deleção Cromossômica , Cromossomos Humanos Par 22/genética , Aconselhamento Genético , Transplante de Células-Tronco Hematopoéticas , Humanos , Timo/transplante
5.
Am J Med Genet A ; 173(9): 2366-2372, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28627729

RESUMO

The syndrome originally described by Dr. Angelo DiGeorge had immunodeficiency as a central component. When a 22q11.2 deletion was identified as the cause in the majority of patients with DiGeorge syndrome, the clinical features of 22q11.2 deletion syndrome became so expansive that the immunodeficiency became less prominent in our thinking about the syndrome. This review will focus on the immune system and the changes in our understanding over the past 50 years. Initially characterized as a pure defect in T cell development, we now appreciate that many of the clinical features related to the immunodeficiency are well downstream of the limitation imposed by a small thymus. Dysfunctional B cells presumed to be secondary to compromised T cell help, issues related to T cell exhaustion, and high rates of atopy and autoimmunity are aspects of management that require consideration for optimal clinical care and for designing a cogent monitoring approach. New data on atopy are presented to further demonstrate the association.


Assuntos
Cromossomos Humanos Par 22/genética , Síndrome de DiGeorge/genética , Síndromes de Imunodeficiência/genética , Linfócitos T/imunologia , Deleção Cromossômica , Síndrome de DiGeorge/imunologia , Síndrome de DiGeorge/patologia , Humanos , Síndromes de Imunodeficiência/imunologia , Síndromes de Imunodeficiência/patologia , Linfócitos T/patologia
6.
J Allergy Clin Immunol ; 140(6): 1660-1670.e16, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28400115

RESUMO

BACKGROUND: Thymus transplantation is a promising strategy for the treatment of athymic complete DiGeorge syndrome (cDGS). METHODS: Twelve patients with cDGS underwent transplantation with allogeneic cultured thymus. OBJECTIVE: We sought to confirm and extend the results previously obtained in a single center. RESULTS: Two patients died of pre-existing viral infections without having thymopoiesis, and 1 late death occurred from autoimmune thrombocytopenia. One infant had septic shock shortly after transplantation, resulting in graft loss and the need for a second transplant. Evidence of thymopoiesis developed from 5 to 6 months after transplantation in 10 patients. Median circulating naive CD4 counts were 44 × 106/L (range, 11-440 × 106/L) and 200 × 106/L (range, 5-310 × 106/L) at 12 and 24 months after transplantation and T-cell receptor excision circles were 2,238/106 T cells (range, 320-8,807/106 T cells) and 4,184/106 T cells (range, 1,582-24,596/106 T cells). Counts did not usually reach normal levels for age, but patients were able to clear pre-existing infections and those acquired later. At a median of 49 months (range, 22-80 months), 8 have ceased prophylactic antimicrobials, and 5 have ceased immunoglobulin replacement. Histologic confirmation of thymopoiesis was seen in 7 of 11 patients undergoing biopsy of transplanted tissue, including 5 showing full maturation through to the terminal stage of Hassall body formation. Autoimmune regulator expression was also demonstrated. Autoimmune complications were seen in 7 of 12 patients. In 2 patients early transient autoimmune hemolysis settled after treatment and did not recur. The other 5 experienced ongoing autoimmune problems, including thyroiditis (3), hemolysis (1), thrombocytopenia (4), and neutropenia (1). CONCLUSIONS: This study confirms the previous reports that thymus transplantation can reconstitute T cells in patients with cDGS but with frequent autoimmune complications in survivors.


Assuntos
Doenças Autoimunes/imunologia , Síndrome de DiGeorge/terapia , Transplante de Órgãos , Complicações Pós-Operatórias/imunologia , Linfócitos T/imunologia , Timo/transplante , Doenças Autoimunes/etiologia , Células Cultivadas , Criança , Pré-Escolar , Síndrome de DiGeorge/imunologia , Europa (Continente) , Feminino , Humanos , Reconstituição Imune , Lactente , Masculino , Técnicas de Cultura de Órgãos , Transplante Homólogo , Resultado do Tratamento
8.
Int Rev Immunol ; 35(1): 25-38, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-25774666

RESUMO

Primary immunodeficiencies (PIDs) include a heterogeneous group of mostly monogenic diseases characterized by functional/developmental alterations of the immune system. Skin and skin annexa abnormalities may be a warning sign of immunodeficiency, since both epidermal and thymic epithelium have ectodermal origin. In this review, we will focus on the most common immune disorders associated with ectodermal alterations. Elevated IgE levels represent the immunological hallmark of hyper-IgE syndrome, characterized by severe eczema and susceptibility to infections. Ectodermal dysplasia (ED) is a group of rare disorders that affect tissues of ectodermal origin. Hypoidrotic ED (HED), the most common form, is inherited as autosomal dominant, autosomal recessive or X-linked trait (XLHED). HED and XLHED are caused by mutations in NEMO and EDA-1 genes, respectively, and show similarities in the cutaneous involvement but differences in the susceptibility to infections and immunological phenotype. Alterations in the transcription factor FOXN1 gene, expressed in the mature thymic and skin epithelia, are responsible for human and murine athymia and prevent the development of the T-cell compartment associated to ectodermal abnormalities such as alopecia and nail dystrophy. The association between developmental abnormalities of the skin and immunodeficiencies suggest a role of the skin as a primary lymphoid organ. Recently, it has been demonstrated that a co-culture of human skin-derived keratinocytes and fibroblasts, in the absence of thymic components, can support the survival of human haematopoietic stem cells and their differentiation into T-lineage committed cells.


Assuntos
Síndrome de DiGeorge/imunologia , Displasia Ectodérmica/imunologia , Síndrome de Job/imunologia , Imunodeficiência Combinada Severa/imunologia , Pele/imunologia , Linfócitos T/imunologia , Animais , Síndrome de DiGeorge/genética , Displasia Ectodérmica/genética , Eczema/imunologia , Epiderme/crescimento & desenvolvimento , Epiderme/imunologia , Humanos , Síndrome de Job/genética , Camundongos , Mutação , Fenótipo , Imunodeficiência Combinada Severa/genética , Linfócitos T/metabolismo , Timo/crescimento & desenvolvimento , Timo/imunologia
9.
Clin Immunol ; 161(2): 144-9, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26341314

RESUMO

PURPOSE: Patients with DiGeorge syndrome suffer from T-lymphopenia. T-cells are important for the maturation and regulation of B-cell function. Our aim was to characterize the B-cell compartment in DiGeorge syndrome patients. METHODS: B-cell subset phenotypization using flow cytometry. Serum BAFF (B-cell activating factor) and serum anti-alpha-galactosyl IgM measurement using ELISA. Serum IgG measurement using nephelometry. RESULTS: We observed a significantly increased number of naïve B-cells and decreased number of switched memory B-cells in DiGeorge patients. Furthermore, we observed increased BAFF levels and a trend toward hypergammaglobulinemia later in life. Surprisingly, we detected a decrease in marginal zone-like (MZ-like) B-cells and natural antibodies in DiGeorge patients. CONCLUSION: The maturation of B-cells is impaired in DiGeorge patients, with high naïve and low switched memory B-cell numbers being observed. There is a clear trend toward hypergammaglobulinemia later in life, coupled with increased serum BAFF levels. Surprisingly, the T-independent humoral response is also impaired, with low numbers of MZ-like B-cell and low levels of anti-alpha-galactosyl IgM natural antibodies being detected.


Assuntos
Anticorpos/imunologia , Subpopulações de Linfócitos B/imunologia , Síndrome de DiGeorge/imunologia , Adolescente , Adulto , Fator Ativador de Células B/imunologia , Estudos de Casos e Controles , Criança , Pré-Escolar , Feminino , Humanos , Hipergamaglobulinemia/imunologia , Imunidade Humoral/imunologia , Imunoglobulina G/imunologia , Imunoglobulina M/imunologia , Memória Imunológica/imunologia , Lactente , Recém-Nascido , Masculino , Adulto Jovem
10.
J Clin Immunol ; 35(5): 435-8, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26048260

RESUMO

PURPOSE: Complete DiGeorge syndrome (cDGS) describes a subset of patients with DiGeorge syndrome that have thymic aplasia, and thus are at risk for severe opportunistic infections. Patients with cDGS and mycobacterial infection have not previously been described. We present this case to illustrate that patients with cDGS are at risk for nontuberculous mycobacterial infections and to discuss further antimicrobial prophylaxis prior to thymic transplantation. METHODS: A 13-month old male was identified as T cell deficient by the T cell receptor excision circle (TREC) assay on newborn screening, and was subsequently confirmed to have cDGS. He presented with fever and cough, and was treated for chronic aspiration pneumonia as well as Pneumocystis jirovecii infection without significant improvement. It was only after biopsy of mediastinal lymph nodes seen on CT that the diagnosis of disseminated Mycobacterium kansasii was made. We reviewed the literature regarding atypical mycobacterial infections and prophylaxis used in other immunocompromised patients, as well as the current data regarding cDGS detection through TREC newborn screening. RESULTS: Multiple cases of cDGS have been diagnosed via TREC newborn screening, however this is the first patient with cDGS and disseminated mycobacterial infection to be reported in literature. Thymic transplantation is the definitive treatment of choice for cDGS. Prophylaxis with either clarithromycin or azithromycin has been shown to reduce mycobacterial infections in children with advanced human immunodeficiency virus infection. CONCLUSIONS: Children with cDGS should receive thymic transplantion as soon as possible, but prior to this are at risk for nontuberculous mycobacterial infections. Severe, opportunistic infections may require invasive testing for diagnosis in patients with cDGS. Antimicrobial prophylaxis should be considered to prevent disseminated mycobacterial infection in these patients.


Assuntos
Antibioticoprofilaxia , Síndrome de DiGeorge/diagnóstico , Infecções por Mycobacterium não Tuberculosas/diagnóstico , Mycobacterium kansasii , Linfócitos T/imunologia , Timo/transplante , Azitromicina/uso terapêutico , Biópsia , Claritromicina/uso terapêutico , Síndrome de DiGeorge/complicações , Síndrome de DiGeorge/imunologia , Humanos , Lactente , Masculino , Infecções por Mycobacterium não Tuberculosas/imunologia , Infecções por Mycobacterium não Tuberculosas/prevenção & controle , Transplante de Órgãos , Timo/patologia , Tomografia Computadorizada por Raios X
11.
J Allergy Clin Immunol ; 135(5): 1293-302, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25748067

RESUMO

BACKGROUND: DiGeorge syndrome affects more than 3.5 million persons worldwide. Partial DiGeorge syndrome (pDGS), which is characterized by a number of gene deletions in chromosome 22, including the chicken tumor virus number 10 regulator of kinase (Crk)-like (CrkL) gene, is one of the most common genetic disorders in human subjects. To date, the role of natural killer (NK) cells in patients with pDGS remains unclear. OBJECTIVE: We sought to define the effect of pDGS-related Crk haploinsufficiency on NK cell activation and cytotoxic immunological synapse (IS) structure and function. METHODS: Inducible CrkL-silenced NK cells were used to recapitulate the pDGS, CrkL-haploinsufficient phenotype. Findings were validated by using NK cells from patients with actual pDGS. Ultimately, deficits in the function of NK cells from patients with pDGS were restored by lentiviral transduction of CrkL. RESULTS: Silencing of CrkL expression inhibits NK cell function. Specifically, pDGS haploinsufficiency of CrkL inhibits accumulation of activating receptors, polarization of cytolytic machinery and key signaling molecules, and activation of ß2-integrin at the IS. Reintroduction of CrkL protein restores NK cell cytotoxicity. CONCLUSION: CrkL haploinsufficiency causes functional NK deficits in patients with pDGS by disrupting both ß2-integrin activation and activating receptor accumulation at the IS. Our results suggest that NK cell IS quality can directly affect immune status, providing a potential target for diagnosis and therapeutic manipulation in patients with pDGS and in other patients with functional NK cell deficiencies.


Assuntos
Síndrome de DiGeorge/genética , Síndrome de DiGeorge/imunologia , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Citotoxicidade Imunológica/genética , Inativação Gênica , Humanos , Sinapses Imunológicas/genética , Sinapses Imunológicas/metabolismo , Integrinas/metabolismo , Modelos Biológicos , Proteínas Nucleares/genética , Proteínas Proto-Oncogênicas c-vav/metabolismo
12.
Pediatr Res ; 77(4): 579-85, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25580739

RESUMO

BACKGROUND: The 22q11.2 deletion syndrome (22q11.2DS) is a congenital multisystem anomaly characterized by typical facial features, palatal anomalies, congenital heart defects, hypocalcemia, immunodeficiency, and cognitive and neuropsychiatric symptoms. The aim of our study was to investigate T- and B-lymphocyte characteristics associated with 22q11.2DS. METHODS: Seventy-five individuals with 22q11.2DS were tested for T and B lymphocytes by examination of T-cell receptor rearrangement excision circles (TRECs) and B-cell κ-deleting recombination excision circles (KRECs), respectively. RESULTS: The 22q11.2DS individuals displayed low levels of TRECs, while exhibiting normal levels of KRECs. There was a significant positive correlation between TREC and KREC in the 22q11.2DS group, but not in controls. Both TREC and KREC levels showed a significant decrease with age and only TREC was low in 22q11.2DS individuals with recurrent infections. No difference in TREC levels was found between 22q11.2DS individuals who underwent heart surgery (with or without thymectomy) and those who did not. CONCLUSION: T-cell immunodeficiency in 22q11.2DS includes low TREC levels, which may contribute to recurrent infections in individuals with this syndrome. A correlation between T- and B-cell abnormalities in 22q11.2DS was identified. The B-cell abnormalities could account for part of the immunological deficiency seen in 22q11.2DS.


Assuntos
Medula Óssea/patologia , Síndrome de DiGeorge/genética , Síndrome de DiGeorge/patologia , Timo/patologia , Adolescente , Adulto , Linfócitos B/citologia , Estudos de Casos e Controles , Criança , Síndrome de DiGeorge/imunologia , Feminino , Rearranjo Gênico do Linfócito B , Rearranjo Gênico do Linfócito T , Humanos , Masculino , Recombinação Genética , Linfócitos T/citologia , Adulto Jovem
13.
J Clin Immunol ; 34(7): 864-70, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25008482

RESUMO

PURPOSE: Syndrome diGeorge is associated amongst other clinical signs with various degrees of thymic dysplasia, related immunodeficiency and autoimmune disorders. Helios, a transcription factor from Ikaros family, has been proposed as a marker for thymus derived Tregs. We therefore examined Helios + Tregs in a cohort of patients with genetically proven diGeorge syndrome with typical T cell lymphopenia due to the thymic pathology. METHODS: T cells, FoxP3+ Tregs and Helios + FoxP3+ Tregs were examined in 52 samples from 37 patients. One patient with diGeorge/CHARGE syndrome with total thymic aplasia was also included. Statistical analysis was performed using a linear regression comparison. RESULTS: Total absolute Tregs were significantly lower in diGeorge patients as compared to controls in all age groups (0-20 years) (p = 0.0016). The difference was more expressed in the first four years of age. Relative Treg numbers expressed as the percentage of Tregs in CD4+ T-cells, however, were not different in patients and controls in all age groups (p = 0.661), neither could we find any significant difference in the percentage of Helios + Tregs between patients and controls (p = 0.238). Helios + Tregs were still present in a patient with diGeorge/CHARGE syndrome with complete athymia 7 years after partially matched unrelated repeated T lymphocytes infusions. CONCLUSION: Our findings show that while there was a significant decrease in absolute numbers of Tregs in patients with diGeorge syndrome, the relative percentage of this population did not differ between patients and controls. Low absolute Tregs thus reflected typical T cells lymphopenia in patients. Helios expression was not affected in diGeorge syndrome.


Assuntos
Biomarcadores/metabolismo , Síndrome de DiGeorge/imunologia , Fator de Transcrição Ikaros/metabolismo , Linfócitos T Reguladores/imunologia , Timo/imunologia , Adolescente , Adulto , Contagem de Células , Criança , Pré-Escolar , Estudos de Coortes , Feminino , Fatores de Transcrição Forkhead/metabolismo , Humanos , Fator de Transcrição Ikaros/genética , Lactente , Recém-Nascido , Masculino , Timo/patologia , Adulto Jovem
14.
J Pediatr ; 163(3): 908-10, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23660376

RESUMO

Adenovirus causes significant morbidity and mortality in immunocompromised children. We report how an infusion of HLA-matched sibling donor T lymphocytes rapidly eradicated life-threatening, high-level adenoviremia in a child with complete DiGeorge syndrome (22q11.2 deletion) who went on to reconstitute a diverse, donor-derived, postthymic T-cell repertoire.


Assuntos
Infecções por Adenovirus Humanos/cirurgia , Transplante de Medula Óssea , Síndrome de DiGeorge/complicações , Hospedeiro Imunocomprometido , Linfócitos T/transplante , Infecções por Adenovirus Humanos/diagnóstico , Infecções por Adenovirus Humanos/imunologia , Síndrome de DiGeorge/imunologia , Feminino , Humanos , Lactente
15.
Curr Opin Otolaryngol Head Neck Surg ; 20(6): 502-6, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23000736

RESUMO

PURPOSE OF REVIEW: Improved recognition of velocardiofacial syndrome (VCFS) has led to increasing awareness of VCFS by otolaryngologists. Understanding the developmental biologic processes affected in VCFS patients will help improve treatment and outcomes. Advanced application of molecular labeling techniques has better outlined the role of T-Box transcription factor 1 (TBX1) as the primary genetic anomaly leading to VCFS. TBX1 plays multiple roles during branchial, cardiac, and craniofacial development and increased understanding of how these systems are affected by TBX1 mutations will improve patient outcomes. Furthermore, additional modifiers of TBX1 expression have been identified that may explain the variability of VCFS phenotypes. The phenotypic spectrum of VCFS may include cardiac anomalies, velopharyngeal insufficiency, aberrant calcium metabolism, and immune dysfunction. Recent interest has focused on the cognitive and neuropsychiatric manifestations of VCFS. Improved understanding of the biology of VCFS associated mutations has the potential to improve therapeutic outcomes. RECENT FINDINGS: This article will discuss recent developmental biologic understanding of the role of TBX1 and genetic modifiers generating the phenotypic variability seen in VCFS patients. Special attention is given to advances in the realms of immunodeficiency, hypocalcemia, cardiac and arterial patterning anomalies, velopharyngeal insufficiency, as well as cognitive and psychiatric problems. SUMMARY: Enhanced understanding of the multiple systems affected by TBX1 mutations will result in improved patient outcomes and improved family education. Future research will lead to improved detection of potential targets for gene therapy and change the way physicians counsel families and treat patients.


Assuntos
Síndrome de DiGeorge/genética , Animais , Região Branquial/embriologia , Cálcio/metabolismo , Síndrome de DiGeorge/complicações , Síndrome de DiGeorge/imunologia , Síndrome de DiGeorge/terapia , Terapia Genética , Homeostase , Humanos , Glândulas Paratireoides/transplante , Fenótipo , Proteínas com Domínio T/metabolismo , Linfócitos T/imunologia , Timo/anormalidades , Timo/transplante , Tretinoína/metabolismo
16.
Clin Dev Immunol ; 2012: 467101, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22474479

RESUMO

Primary immunodeficiencies (PIDs) are disorders of the immune system, which lead to increased susceptibility to infections. T-cell defects, which may affect T-cell development/function, are approximately 11% of reported PIDs. The pathogenic mechanisms are related to molecular alterations not only of genes selectively expressed in hematopoietic cells but also of the stromal component of the thymus that represents the primary lymphoid organ for T-cell differentiation. With this regard, the prototype of athymic disorders due to abnormal stroma is the Nude/SCID syndrome, first described in mice in 1966. In man, the DiGeorge Syndrome (DGS) has long been considered the human prototype of a severe T-cell differentiation defect. More recently, the human equivalent of the murine Nude/SCID has been described, contributing to unravel important issues of the T-cell ontogeny in humans. Both mice and human diseases are due to alterations of the FOXN1, a developmentally regulated transcription factor selectively expressed in skin and thymic epithelia.


Assuntos
Síndrome de DiGeorge/imunologia , Fatores de Transcrição Forkhead/imunologia , Imunodeficiência Combinada Severa/imunologia , Linfócitos T/imunologia , Animais , Diferenciação Celular/imunologia , Síndrome de DiGeorge/genética , Síndrome de DiGeorge/patologia , Modelos Animais de Doenças , Células Epiteliais/imunologia , Células Epiteliais/patologia , Fatores de Transcrição Forkhead/genética , Humanos , Camundongos , Camundongos Nus , Camundongos SCID , Imunodeficiência Combinada Severa/genética , Imunodeficiência Combinada Severa/patologia , Pele/imunologia , Pele/patologia , Linfócitos T/patologia , Timo/imunologia , Timo/patologia
17.
Zhonghua Bing Li Xue Za Zhi ; 41(11): 742-6, 2012 Nov.
Artigo em Chinês | MEDLINE | ID: mdl-23302334

RESUMO

OBJECTIVE: To investigate clinicopathological features of DiGeorge syndrome (DGS). METHOD: The clinical features, histological and immunohistochemical findings were analyzed in 5 cases of DGS by autopsy. RESULTS: Five cases of DGS in male infants aged 4 days, 1 month, 7 months, 10 months, and 13 months respectively. Gross and microscopic observations revealed that thymic cortex was depleted of lymphocytes or showed few, dispersed lymphocytes. The thymic medulla showed predominantly epithelial cells with calcified Hassall bodies as well as lymphocyte depletion. T lymphocytes were also scarce in the tonsils, lymph nodes, spleen, and mucosa-associated lymphatic tissue of ileum. In addition, 3 of the 5 patients also showed parathyroid aplasia or dysplasia, and congenital hypertrophy of the ventricular septum. CONCLUSIONS: The pathological changes indicate that clinicians should be aware of defects of immune system if the infants suffer from severe infections. Pathologists should recognize the importance of abnormalities of lymphohematopoietic tissues in the diagnosis of primary immunodeficiency diseases such as DGS.


Assuntos
Síndrome de DiGeorge/patologia , Glândulas Paratireoides/patologia , Linfócitos T/patologia , Timo/patologia , Autopsia , Síndrome de DiGeorge/imunologia , Síndrome de DiGeorge/virologia , Hepatite Viral Humana/patologia , Humanos , Hipertrofia Ventricular Esquerda/patologia , Lactente , Recém-Nascido , Contagem de Linfócitos , Masculino , Pneumonia Viral/patologia , Linfócitos T/imunologia
18.
J Pediatr Hematol Oncol ; 33(6): 465-6, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21792043

RESUMO

The absence of an appropriate central tolerance in primary immunodeficiencies favors proliferation of autoreactive lymphocyte clones, causing a greater incidence of autoimmunity. Del 22q11.2 syndrome presents an increased incidence of allergic and autoimmune diseases. One of the most relevant and frequent immune manifestations is autoimmune thrombocytopenia. We present the case of a pediatric patient with autoimmune thrombocytopenia due to the immunological dysregulation observed in partial DiGeorge syndrome.


Assuntos
Síndrome de DiGeorge/complicações , Síndrome de DiGeorge/imunologia , Púrpura Trombocitopênica Idiopática/etiologia , Adolescente , Deleção Cromossômica , Feminino , Humanos
19.
Hum Pathol ; 42(12): 2037-41, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21683977

RESUMO

Reported herein was the first autopsy case of Epstein-Barr virus-associated T-cell lymphoma in a 25-year-old man with DiGeorge syndrome. Systemic lymph nodes demonstrated diffuse encasement by large lymphoma cells positive for CD45, CD2, CD3, CD5, CD7, CD8, TIA1, and granzyme B, accompanied with marked hemophagocytosis. Almost 100% of lymphoma cells were both EBER- and LMP-1-positive, and EBNA2-negative. The rearrangement of T-cell receptor ß gene was proved by polymerase chain reaction. Clinical and pathologic features coincided with Epstein-Barr virus-associated T/NK-cell lymphoproliferative disorder preceded by chronic active Epstein-Barr virus infection. A fluorescence in situ hybridization using paraffin-embedded tissues demonstrated a mosaic chromosome 22q11.2 deletion with both host cardiac myocytes and lymphoma cells, suggesting that Epstein-Barr virus-associated T-cell lymphoma was associated with and derived from the cells carrying the chromosomal abnormality. Furthermore, the lymphomagenesis of our case correlated with defect of cellular immunity in DiGeorge syndrome.


Assuntos
Deleção Cromossômica , Cromossomos Humanos Par 22/genética , Síndrome de DiGeorge/complicações , Infecções por Vírus Epstein-Barr/complicações , Linfoma de Células T/genética , Adulto , Autopsia , Síndrome de DiGeorge/genética , Síndrome de DiGeorge/imunologia , Infecções por Vírus Epstein-Barr/patologia , Evolução Fatal , Rearranjo Gênico da Cadeia beta dos Receptores de Antígenos dos Linfócitos T/genética , Herpesvirus Humano 4/fisiologia , Humanos , Imunidade Celular , Hibridização in Situ Fluorescente , Células Matadoras Naturais/virologia , Linfonodos/imunologia , Linfonodos/patologia , Linfoma de Células T/complicações , Linfoma de Células T/patologia , Linfoma de Células T/virologia , Masculino , Miócitos Cardíacos/virologia , Inclusão em Parafina , Deleção de Sequência , Linfócitos T/virologia
20.
Clin Immunol ; 140(3): 244-59, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21565561

RESUMO

A functional thymus develops after cultured thymus tissue is transplanted into subjects with complete DiGeorge anomaly. To gain insight into how the process occurs, 7 post-transplantation thymus biopsy tissues were evaluated. In 5 of 7 biopsies, the thymus appeared to be predominantly cortex with thymocytes expressing cortical markers. Unexpectedly, the epithelium expressed both cortical [cortical dendritic reticulum antigen 2 (CDR2)] and medullary [cytokeratin (CK) 14] markers. Early medullary development was suggested by epithelial cell adhesion molecule (EpCAM) reactivity in small areas of biopsies. Two other biopsies had distinct mature cortex and medulla with normal restriction of CK14 to the medulla and subcapsular cortex, and of CDR2 to cortex. These data are consistent with a model in which thymic epithelium contains CK14+ "progenitor epithelial cells". After transplantation these cells proliferate as CK14+CDR2+ thymic epithelial cells that are associated with cortical thymocytes. Later these cells differentiate into distinct cortical and medullary epithelia.


Assuntos
Síndrome de DiGeorge/cirurgia , Timo/imunologia , Timo/transplante , Biópsia , Diferenciação Celular/imunologia , Síndrome de DiGeorge/imunologia , Humanos , Lactente , Recém-Nascido , Queratina-14/imunologia , Ativação Linfocitária/imunologia , Proteínas do Tecido Nervoso/imunologia , Células-Tronco/imunologia , Linfócitos T/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA