Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
2.
Blood ; 138(12): 1019-1033, 2021 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-33876203

RESUMO

Sterile alpha motif (SAM) and Src homology-3 (SH3) domain-containing 3 (SASH3), also called SH3-containing lymphocyte protein (SLY1), is a putative adaptor protein that is postulated to play an important role in the organization of signaling complexes and propagation of signal transduction cascades in lymphocytes. The SASH3 gene is located on the X-chromosome. Here, we identified 3 novel SASH3 deleterious variants in 4 unrelated male patients with a history of combined immunodeficiency and immune dysregulation that manifested as recurrent sinopulmonary, cutaneous, and mucosal infections and refractory autoimmune cytopenias. Patients exhibited CD4+ T-cell lymphopenia, decreased T-cell proliferation, cell cycle progression, and increased T-cell apoptosis in response to mitogens. In vitro T-cell differentiation of CD34+ cells and molecular signatures of rearrangements at the T-cell receptor α (TRA) locus were indicative of impaired thymocyte survival. These patients also manifested neutropenia and B-cell and natural killer (NK)-cell lymphopenia. Lentivirus-mediated transfer of the SASH3 complementary DNA-corrected protein expression, in vitro proliferation, and signaling in SASH3-deficient Jurkat and patient-derived T cells. These findings define a new type of X-linked combined immunodeficiency in humans that recapitulates many of the abnormalities reported in mice with Sly1-/- and Sly1Δ/Δ mutations, highlighting an important role of SASH3 in human lymphocyte function and survival.


Assuntos
Cromossomos Humanos X/genética , Mutação , Doenças por Imunodeficiência Combinada Ligada ao Cromossomo X/genética , Animais , Linfócitos B/imunologia , Linfócitos T CD4-Positivos/imunologia , Pré-Escolar , Cromossomos Humanos X/imunologia , Loci Gênicos , Humanos , Células Jurkat , Células Matadoras Naturais/imunologia , Linfopenia/genética , Linfopenia/imunologia , Masculino , Camundongos , Camundongos Knockout , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Doenças por Imunodeficiência Combinada Ligada ao Cromossomo X/imunologia
3.
Clin Dev Immunol ; 2012: 257695, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22481966

RESUMO

Cancer/Testis Antigens (CTAs) are a promising class of tumor antigens that have a limited expression in somatic tissues (testis, ovary, fetal, and placental cells). Aberrant expression of CTAs in cancer cells may lead to abnormal chromosome segregation and aneuploidy. CTAs are regulated by epigenetic mechanisms (DNA methylation and acetylation of histones) and are attractive targets for immunotherapy in cancer because the gonads are immune privileged organs and anti-CTA immune response can be tumor-specific. Multiple myeloma (MM) is an incurable hematological malignancy, and several CTAs have been detected in many MM cell lines and patients. Among CTAs expressed in MM we must highlight the MAGE-C1/CT7 located on the X chromosome and expressed specificity in the malignant plasma cells. MAGE-C1/CT7 seems to be related to disease progression and functional studies suggests that this CTA might play a role in cell cycle and mainly in survival of malignant plasma cells, protecting myeloma cells against spontaneous as well as drug-induced apoptosis.


Assuntos
Antígenos de Neoplasias/imunologia , Biomarcadores Tumorais/imunologia , Imunoterapia , Mieloma Múltiplo/terapia , Proteínas de Neoplasias/imunologia , Apoptose/imunologia , Ciclo Celular/genética , Ciclo Celular/imunologia , Cromossomos Humanos X/genética , Cromossomos Humanos X/imunologia , Progressão da Doença , Epigênese Genética/imunologia , Humanos , Mieloma Múltiplo/imunologia , Mieloma Múltiplo/patologia , Plasmócitos/imunologia , Plasmócitos/patologia
4.
Int Immunopharmacol ; 101(Pt B): 108255, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34688149

RESUMO

The coronavirus disease (COVID-19) has once again reminded us of the significance of host immune response and consequential havocs of the immune dysregulation. The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) inflicts severe complications to the infected host, including cough, dyspnoea, fever, septic shock, acute respiratory distress syndrome (ARDs), and multiple organ failure. These manifestations are the consequence of the dysregulated immune system, which gives rise to excessive and unattended production of pro-inflammatory mediators. Elevated circulatory cytokine and chemokine levels are accompanied by spontaneous haemorrhage, thrombocytopenia and systemic inflammation, which are the cardinal features of life-threatening cytokine storm syndrome in advanced COVID-19 diseases. Coronavirus hijacked NF-kappa B (NF-κB) is responsible for upregulating the expressions of inflammatory cytokine, chemokine, alarmins and inducible enzymes, which paves the pathway for cytokine storm. Given the scenario, the systemic approach of simultaneous inhibition of NF-κB offers an attractive therapeutic intervention. Targeted therapies with proteasome inhibitor (VL-01, bortezomib, carfilzomib and ixazomib), bruton tyrosine kinase inhibitor (acalabrutinib), nucleotide analogue (remdesivir), TNF-α monoclonal antibodies (infliximab and adalimumab), N-acetylcysteine and corticosteroids (dexamethasone), focusing the NF-κB inhibition have demonstrated effectiveness in terms of the significant decrease in morbidity and mortality in severe COVID-19 patients. Hence, this review highlights the activation, signal transduction and cross-talk of NF-κB with regard to cytokine storm in COVID-19. Moreover, the development of therapeutic strategies based on NF-κB inhibition are also discussed herein.


Assuntos
COVID-19/imunologia , Síndrome da Liberação de Citocina/imunologia , NF-kappa B/imunologia , SARS-CoV-2 , Animais , Cromossomos Humanos X/imunologia , Exoftalmia , Hormônios Esteroides Gonadais/imunologia , Humanos , Transdução de Sinais
5.
Biol Sex Differ ; 11(1): 3, 2020 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-31937374

RESUMO

BACKGROUND: Sex bias in immune function has been contributed in part to a preponderance of immune system-related genes (ISRG) on the X-chromosome. We verified whether ISRG are more abundant on the X chromosome as compared to autosomal chromosomes and reflected on the impact of our findings. METHODS: Consulting freely accessible databases, we performed a comparative study consisting of three complementary strategies. First, among coding X/Y-linked genes, the abundance of ISRG was compared to the abundance of genes dedicated to other systems. Genes were assigned considering three criteria: disease, tissue expression, and function (DEF approach). In addition, we carried out two genome-wide approaches to compare the contribution of sex and autosomal chromosomes to immune genes defined by an elevated expression in lymphatic tissues (LTEEG approach) or annotation to an immune system process, GO:0002376 (GO approach). RESULTS: The X chromosome had less immune genes than the median of the autosomal chromosomes. Among X-linked genes, ISRG ranked fourth after the reproductive and nervous systems and genes dedicated to development, proliferation and apoptosis. On the Y chromosome, ISRG ranked second, and at the pseudoautosomal region (PAR) first. According to studies on the expression of X-linked genes in a variety of (mostly non-lymphatic) tissues, almost two-thirds of ISRG are expressed without sex bias, and the remaining ISRG presented female and male bias with similar frequency. Various epigenetic controllers, X-linked MSL3 and Y-linked KDM5D and UTY, were preferentially expressed in leukocytes and deserve further attention for a possible role in sex biased expression or its neutralisation. CONCLUSIONS: The X chromosome is not enriched for ISRG, though particular X-linked genes may be responsible for sex differences in certain immune responses. So far, there is insufficient information on sex-biased expression of X/Y-linked ISRG in leukocytes to draw general conclusions on the impact of X/Y-linked ISRG in immune function. More research on the regulation of the expression X-linked genes is required with attention to 1) female and male mechanisms that may either augment or diminish sex biased expression and 2) tissue-specific expression studies.


Assuntos
Cromossomos Humanos X/imunologia , Cromossomos Humanos Y/imunologia , Sistema Imunitário , Caracteres Sexuais , Feminino , Perfilação da Expressão Gênica , Humanos , Masculino
6.
J Autoimmun ; 33(1): 17-24, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19361956

RESUMO

Recent studies indicate that genes located on the X-chromosome play a major and unique role in autoimmunity. The fact that most X-linked primary immune deficiencies carry significant autoimmune manifestations greatly supports this notion. Autoimmunity and immune deficiency have been considered two opposite extremes resulting from immune dysregulation and failure of immune development and/or function, respectively. Growing evidence has been accumulating to indicate that autoimmune phenomena occur in patients suffering from primary immune deficiency (PID), and the molecular and cellular mechanisms that interconnect these conditions are being unraveled. The study of rare single-gene disorders associated with significant autoimmunity may shed light on the pathophysiology of more complex multifactorial and polygenic autoimmune disorders. In this regard, primary immunodeficiencies represent unique "experiments of Nature" that illustrate the critical role played by single-gene products in the development, function and homeostasis of the immune system. In this review we will focus on the clinical features and on the cellular and molecular pathophysiology of the known X-linked PID in which autoimmune manifestations are more common, in the attempt to understand what single-gene defects can teach us on the role that key immune pathways and cellular processes may play to prevent autoimmunity.


Assuntos
Autoimunidade/genética , Cromossomos Humanos X/imunologia , Doenças por Imunodeficiência Combinada Ligada ao Cromossomo X/genética , Animais , Autoimunidade/imunologia , Cromossomos Humanos X/genética , Feminino , Homeostase/genética , Homeostase/imunologia , Humanos , Tolerância Imunológica/genética , Tolerância Imunológica/imunologia , Camundongos , Camundongos Mutantes , Mutação , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Doenças por Imunodeficiência Combinada Ligada ao Cromossomo X/imunologia , Doenças por Imunodeficiência Combinada Ligada ao Cromossomo X/patologia , Doenças por Imunodeficiência Combinada Ligada ao Cromossomo X/fisiopatologia
7.
Expert Rev Clin Immunol ; 15(6): 607-615, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31033369

RESUMO

INTRODUCTION: Chronic autoimmune diseases affect 5-10% of the population worldwide and are largely predominant in women. Sex hormone changes have been widely investigated based on changes in the clinical phenotypes observed during pregnancy and menopause. It is known that females with autoimmune diseases manifest a higher rate of circulating leukocytes with a single X chromosome, and there have been several reports on the role of X chromosome gene dosage through inactivation or duplication in autoimmunity. However, it is also important not to overlook men with autoimmune diseases, who might manifest a more frequent loss of the Y chromosome in circulating leukocytes. Areas covered: In the present review, we will discuss the current evidence supporting the mechanisms of female predominance in rheumatic diseases, by discussing the role of reproductive history, sex hormones and abnormalities related to them, clinical differences between male and female patients, and epigenetic changes that have been evaluated through twin studies on genetic and environmental changes in rheumatic patients. Expert opinion: The influence of sex hormones and chromosomes on the function of the innate and adaptive immune systems needs to be clarified, to better understand the risk of autoimmune diseases, early diagnostic tools, and therapeutic response.


Assuntos
Doenças Autoimunes/imunologia , Cromossomos Humanos X/imunologia , Hormônios Esteroides Gonadais/imunologia , Doenças Reumáticas/imunologia , Caracteres Sexuais , Doenças Autoimunes/patologia , Feminino , Humanos , Masculino , Doenças Reumáticas/patologia
8.
Semin Immunopathol ; 41(2): 153-164, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30276444

RESUMO

Women develop stronger immune responses than men, with positive effects on the resistance to viral or bacterial infections but magnifying also the susceptibility to autoimmune diseases like systemic lupus erythematosus (SLE). In SLE, the dosage of the endosomal Toll-like receptor 7 (TLR7) is crucial. Murine models have shown that TLR7 overexpression suffices to induce spontaneous lupus-like disease. Conversely, suppressing TLR7 in lupus-prone mice abolishes SLE development. TLR7 is encoded by a gene on the X chromosome gene, denoted TLR7 in humans and Tlr7 in the mouse, and expressed in plasmacytoid dendritic cells (pDC), monocytes/macrophages, and B cells. The receptor recognizes single-stranded RNA, and its engagement promotes B cell maturation and the production of pro-inflammatory cytokines and antibodies. In female mammals, each cell randomly inactivates one of its two X chromosomes to equalize gene dosage with XY males. However, 15 to 23% of X-linked human genes escape X chromosome inactivation so that both alleles can be expressed simultaneously. It has been hypothesized that biallelic expression of X-linked genes could occur in female immune cells, hence fostering harmful autoreactive and inflammatory responses. We review here the current knowledge of the role of TLR7 in SLE, and recent evidence demonstrating that TLR7 escapes from X chromosome inactivation in pDCs, monocytes, and B lymphocytes from women and Klinefelter syndrome men. Female B cells where TLR7 is thus biallelically expressed display higher TLR7-driven functional responses, connecting the presence of two X chromosomes with the enhanced immunity of women and their increased susceptibility to TLR7-dependent autoimmune syndromes.


Assuntos
Cromossomos Humanos X , Dosagem de Genes/imunologia , Predisposição Genética para Doença , Lúpus Eritematoso Sistêmico , Caracteres Sexuais , Receptor 7 Toll-Like , Inativação do Cromossomo X/imunologia , Animais , Cromossomos Humanos X/genética , Cromossomos Humanos X/imunologia , Feminino , Humanos , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/imunologia , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/imunologia , Camundongos , Receptor 7 Toll-Like/genética , Receptor 7 Toll-Like/imunologia
9.
Front Immunol ; 9: 475, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29593728

RESUMO

In humans, acid-base balance is crucial to cell homeostasis. Acidosis is observed in numerous inflammatory processes, primarily acute conditions such as sepsis, trauma, or acute respiratory distress where females tend to exhibit better prognosis compared with males. The mechanisms underlying these gender-dependent differences are multiple, probably involving hormonal and genetic factors, particularly the X chromosome. Although pH influences multiple immunological functions, gender differences in acid-base balance have been poorly investigated. In this review, we provide an update on gender differences in human susceptibility to inflammatory diseases. We additionally discuss the potential impact of acid-base balance on the gender bias of the inflammatory response in view of our recent observation that girls present higher neutrophilic inflammation and lower pH with a trend toward better prognosis in severe sepsis. We also highlight the potent role played by endothelial cells in gender differences of inflammation through activation of proton-sensing G protein-coupled receptors.


Assuntos
Equilíbrio Ácido-Base/imunologia , Cromossomos Humanos X/imunologia , Caracteres Sexuais , Equilíbrio Ácido-Base/genética , Cromossomos Humanos X/genética , Feminino , Humanos , Concentração de Íons de Hidrogênio , Inflamação/genética , Inflamação/imunologia , Masculino
10.
Genome Biol Evol ; 10(3): 857-862, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29092048

RESUMO

In mammalian females, diploid somatic cells contain two X chromosomes, one of which is transcriptionally silenced, in a process termed X chromosome inactivation (XCI). Whereas XCI is largely random in placental females, many women exhibit skewed XCI (SXCI), in which the vast majority cells have the same X chromosome inactivated. SXCI has serious health consequences, associated with conditions ranging from Alzheimer's to various autoimmune disorders. SXCI is also associated with outcomes of pregnancies, with higher rates of recurrent spontaneous abortion in women with SXCI. Here, I suggest that SXCI could be driven by selfish X-linked alleles. Consistent with the association of SXCI with autoimmunity, I first note the possibility that recurrent spontaneous abortion could reflect immune rejection of fetuses inheriting alleles from the largely silenced maternal X chromosome. Preferential abortion of fetuses carrying silenced X-linked alleles implies a transmission advantage for X-linked alleles on the largely expressed chromosome, which could drive the emergence of X-linked alleles that make the chromosome resistant to XCI. I discuss the evolutionary dynamics, fitness tradeoffs and implications of this hypothesis, and suggest future directions.


Assuntos
Cromossomos Humanos X/genética , Evolução Molecular , Tolerância Imunológica/genética , Inativação do Cromossomo X/genética , Alelos , Doença de Alzheimer/genética , Cromossomos Humanos X/imunologia , Feminino , Genes Ligados ao Cromossomo X/genética , Genes Ligados ao Cromossomo X/imunologia , Humanos , Tolerância Imunológica/imunologia , Gravidez , Inativação do Cromossomo X/imunologia
11.
Ann N Y Acad Sci ; 1110: 57-64, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17911420

RESUMO

Autoimmune diseases affect approximately 5% of the population in Western countries, with high female predominance. Family and twins studies have demonstrated that genetic factors are crucial determinants of susceptibility to autoimmune disease, but no specific genes have yet been identified. Recent studies indicate that X chromosome abnormalities, such as monosomy rates and inactivation patterns, occur in a number of female-predominant autoimmune diseases. We will review herein the most recent evidence on the role of the X chromosome in loss of tolerance and discuss its potential implications. Future studies will identify the X chromosome regions containing candidate genes for autoimmune susceptibility.


Assuntos
Doenças Autoimunes/genética , Doenças Autoimunes/imunologia , Cromossomos Humanos X/genética , Cromossomos Humanos X/imunologia , Caracteres Sexuais , Feminino , Predisposição Genética para Doença/genética , Humanos , Monossomia , Inativação do Cromossomo X/genética
12.
J Clin Invest ; 125(6): 2187-93, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25915581

RESUMO

Autoimmune diseases occur when the immune system attacks and destroys the organs and tissues of its own host. Autoimmunity is the third most common type of disease in the United States. Because there is no cure for autoimmunity, it is extremely important to study the mechanisms that trigger these diseases. Most autoimmune diseases predominantly affect females, indicating a strong sex bias. Various factors, including sex hormones, the presence or absence of a second X chromosome, and sex-specific gut microbiota can influence gene expression in a sex-specific way. These changes in gene expression may, in turn, lead to susceptibility or protection from autoimmunity, creating a sex bias for autoimmune diseases. In this Review we discuss recent findings in the field of sex-dependent regulation of gene expression and autoimmunity.


Assuntos
Doenças Autoimunes/imunologia , Caracteres Sexuais , Animais , Doenças Autoimunes/epidemiologia , Cromossomos Humanos X/imunologia , Feminino , Regulação da Expressão Gênica/imunologia , Humanos , Masculino , Estados Unidos/epidemiologia
13.
PLoS One ; 9(1): e86985, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24498006

RESUMO

BACKGROUND: Maternal microchimeric cells (MMc) transfer across the placenta during pregnancy. Increased levels of MMc have been observed in several autoimmune diseases including type 1 diabetes but their role is unknown. It has been suggested that MMc are 1) effector cells of the immune response, 2) targets of the autoimmune response or 3) play a role in tissue repair. The aim of this study was to define the cellular phenotype of MMc in control (n = 14) and type 1 diabetes pancreas (n = 8). METHODS: Using sex chromosome-based fluorescence in-situ hybridization, MMc were identified in male pancreas and their phenotype determined by concomitant immunofluorescence. RESULTS: In normal pancreas, MMc positive for endocrine, exocrine, duct and acinar markers were identified suggesting that these cells are derived from maternal progenitors. Increased frequencies of MMc were observed in type 1 diabetes pancreas (p = 0.03) with particular enrichment in the insulin positive fraction (p = 0.01). MMc did not contribute to infiltrating immune cells or Ki67+ islet cell populations in type 1 diabetes. CONCLUSION: These studies provide support for the hypothesis that MMc in human pancreas are derived from pancreatic precursors. Increased frequencies of MMc beta cells may contribute to the initiation of autoimmunity or to tissue repair but do not infiltrate islets in type 1 diabetes.


Assuntos
Diabetes Mellitus Tipo 1/imunologia , Insulina/imunologia , Troca Materno-Fetal/imunologia , Pâncreas/imunologia , Adolescente , Adulto , Autoimunidade/genética , Autoimunidade/imunologia , Criança , Pré-Escolar , Quimerismo , Cromossomos Humanos X/genética , Cromossomos Humanos X/imunologia , Cromossomos Humanos Y/genética , Cromossomos Humanos Y/imunologia , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Feminino , Fator de Transcrição GATA4/imunologia , Fator de Transcrição GATA4/metabolismo , Humanos , Hibridização in Situ Fluorescente/métodos , Lactente , Insulina/metabolismo , Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/metabolismo , Antígeno Ki-67/imunologia , Antígeno Ki-67/metabolismo , Masculino , Troca Materno-Fetal/genética , Microscopia Confocal , Microscopia de Fluorescência , Pâncreas/embriologia , Pâncreas/metabolismo , Gravidez , Adulto Jovem
14.
Bone Marrow Transplant ; 40(3): 283-5, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17529999
15.
Autoimmun Rev ; 11(6-7): A531-7, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22155196

RESUMO

The number of human conditions that are currently considered to be autoimmune diseases (AID) has been steadily growing over the past decades and it is now estimated that over 10 million people are affected in the United States. One of the major shared features among AID is the predominance in the female sex which in some cases changes with the age at disease diagnosis. Numerous hypotheses have been formulated based on intuitive scientific backgrounds to justify this sex imbalance, i.e. sex hormones and reproductive factors, fetal microchimerism, other sex-related environmental factors, a skewing of the X-chromosome inactivation patterns, and major defects in sex chromosomes. Nevertheless, none of these hypotheses has thus far gathered enough convincing evidence and in most cases data are conflicting, as well illustrated by the reports on fetal microchimerism in systemic sclerosis or primary biliary cirrhosis. The present article will critically discuss the main hypotheses (loss of mosaicism, reactivation, and haploinsufficiency) that have been proposed based on findings in female patients with specific AID along with two additional mechanisms (X-chromosome vulnerability and X-linked polyamine genes) that have been observed in AID models. Further, recent data have significantly shifted the paradigm of X chromosome inactivation by demonstrating that a large number of genes can variably escape silencing on one or both chromosomes. As a result we may hypothesize that more than one mechanism may contribute to the female susceptibility to tolerance breakdown while the possibility that unknown factors may indeed protect men from AID should not be overlooked.


Assuntos
Doenças Autoimunes/genética , Autoimunidade/genética , Cromossomos Humanos X/genética , Predisposição Genética para Doença , Fatores Sexuais , Doenças Autoimunes/imunologia , Autoimunidade/imunologia , Cromossomos Humanos X/imunologia , Feminino , Humanos , Masculino , Razão de Masculinidade , Inativação do Cromossomo X
16.
Epigenetics ; 6(9): 1085-94, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21814035

RESUMO

Environment induced epigenetic effects on gene expression in early life are likely to play important roles in mediating the risk of several immune-related diseases. In order to investigate this fully, it is essential to first document temporal changes in epigenetic profile in disease-free individuals as a prelude to defining environmentally mediated changes. Mononuclear cells (MC) were collected longitudinally from a small number of females at birth, 1 year, 2.5 years and 5 years of age and examined for changes in genome-scale DNA methylation profiles using the Illumina Infinium HumanMethylation27 BeadChip array platform. MC from two males were included for comparative purposes. Flow cytometry was used to define MC cell populations in each sample in order to exclude this as the major driver of epigenetic change. The data underwent quality control and normalization within the R programming environment. Unsupervised hierarchical clustering of samples clearly delineated neonatal MC from all other ages. A further clear distinction was observed between 1 year and 5 year samples, with 2.5 year samples showing a mixed distribution between the 1 and 5 year groups. Gene ontology of probes significantly variable over the neonatal period revealed methylation changes in genes associated with cell surface receptor and signal transduction events. In the postnatal period, methylation changes were mostly associated with the development of effector immune responses and homeostasis. Unlike all other chromosomes tested, a predominantly genetic effect was identified as controlling maintenance of X-chromosome methylation profile in females, largely refractory to change over time. This data suggests that the primary driver of neonatal epigenome is determined in utero, whilst postnatally, multiple genetic and environmental factors are implicated in the development of MC epigenetic profile, particularly between the ages of 1-5 years, when the highest level of inter individual variation is apparent. This supports a model for differential sensitivity of specific individuals to disruption in the developing epigenome during the first years of life. Further studies are now needed to examine evolving epigenetic variations in specific cell populations in relation to environmental exposures, immune phenotype and subsequent disease susceptibility.


Assuntos
Cromossomos Humanos X/metabolismo , Metilação de DNA , Epigênese Genética , Imunidade , Leucócitos Mononucleares/citologia , Linfócitos B/citologia , Linfócitos B/imunologia , Pré-Escolar , Cromossomos Humanos X/genética , Cromossomos Humanos X/imunologia , Análise por Conglomerados , Meio Ambiente , Feminino , Citometria de Fluxo , Regulação da Expressão Gênica , Genoma Humano , Homeostase , Humanos , Imunofenotipagem , Lactente , Recém-Nascido , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Estudos Longitudinais , Sistema de Sinalização das MAP Quinases , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Controle de Qualidade , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/imunologia , Linfócitos T/citologia , Linfócitos T/imunologia , Fatores de Tempo
18.
Clin Rev Allergy Immunol ; 39(1): 20-9, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19653135

RESUMO

Autoimmune diseases appear to have multiple contributing factors including genetics, epigenetics, environmental factors, and aging. The predominance of females among patients with autoimmune diseases suggests possible involvement of the X chromosome and X chromosome inactivation. X chromosome inactivation is an epigenetic event resulting in multiple levels of control for modulation of the expression of X-linked genes in normal female cells such that there remains only one active X chromosome in the cell. The extent of this control is unique among the chromosomes and has the potential for problems when regulation is disrupted. Here we discuss the X chromosome inactivation process and how the X chromosome and X chromosome inactivation may be involved in development of autoimmune disorders.


Assuntos
Doenças Autoimunes/genética , Cromossomos Humanos X , Transtornos dos Cromossomos Sexuais/genética , Inativação do Cromossomo X , Animais , Doenças Autoimunes/imunologia , Doenças Autoimunes/metabolismo , Cromossomos Humanos X/imunologia , Metilação de DNA , Suscetibilidade a Doenças , Feminino , Inativação Gênica/imunologia , Doença Granulomatosa Crônica/genética , Humanos , Hidrolases/imunologia , Hidrolases/metabolismo , Proteína-Arginina Desiminase do Tipo 4 , Transtornos dos Cromossomos Sexuais/imunologia , Transtornos dos Cromossomos Sexuais/metabolismo , Fatores Sexuais , Transdução de Sinais/imunologia , Espermina/imunologia
19.
Best Pract Res Clin Rheumatol ; 22(5): 913-22, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19028371

RESUMO

Recent estimates suggest that autoimmune diseases cumulatively affect 5-10% of the general population worldwide. Although the etiology and pathogenesis remain poorly understood in most cases, similarities between diseases outnumber differences in the initiation and perpetuation of the autoimmune injury. One major example is the predominance of affected women, and perhaps its most intriguing putative mechanism is related to sex chromosomes, based on the recent observation that women with autoimmune diseases manifest a higher rate of circulating leukocytes with a single X chromosome. In a complementary fashion, there have been several reports on a role of X chromosome gene dosage through inactivation or duplication in autoimmunity. It is important not to overlook men with autoimmune diseases, who might manifest a more frequent loss of the Y chromosome in circulating leukocytes. Taken together, sex chromosome changes might constitute the common trait of autoimmunity.


Assuntos
Doenças Autoimunes/genética , Cromossomos Humanos X/genética , Cromossomos Humanos Y/genética , Predisposição Genética para Doença , Doenças Autoimunes/imunologia , Cromossomos Humanos X/imunologia , Cromossomos Humanos Y/imunologia , Metilação de DNA , Feminino , Inativação Gênica , Humanos , Masculino , Monossomia , Fatores Sexuais , Inativação do Cromossomo X
20.
J Autoimmun ; 28(4): 201-7, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17418529

RESUMO

FOXP3 is a key gene in the development of regulatory T cells (Treg). FOXP3 expression commits naïve T cells to become Treg cells. Indeed, mutations in the FOXP3 gene cause severe systemic autoimmune diseases in humans and in mice. Therefore, we hypothesized that the FOXP3 gene may be associated with thyroid autoimmunity which is among the typical autoimmune diseases that develop in individuals with FOXP3 mutations. Moreover, the FOXP3 gene is located within an X-chromosome locus (Xp11.23) previously shown to be linked with autoimmune thyroid diseases (AITD). We tested the FOXP3 gene locus for association with AITD in two large cohorts of US Caucasians and Japanese AITD patients. We analyzed 269 Caucasian AITD patients (52 males and 217 females) and 357 Caucasian controls (159 males and 198 females), as well as 377 female Japanese AITD patients and 179 female Japanese controls. The FOXP3 gene locus was analyzed using four microsatellite polymorphisms [(GT)n; (TC)n; DXS573; DXS1208] flanking the FOXP3 gene locus. Interestingly, while no association was found between FOXP3 polymorphisms and AITD in the Japanese cohort there was a significant association in the Caucasian cohort. There was a significant association of the (TC)n polymorphism with AITD in the Caucasian male AITD patients (p=0.011; 5 degrees of freedom [df]). Similarly, there was an association between the DXS573 microsatellite and AITD in the Caucasian female AITD patients (p=0.00023; 4 df). These results suggest that polymorphisms of the FOXP3 gene may play a role in the genetic susceptibility to AITD in Caucasians, perhaps by altering FOXP3 function and/or expression.


Assuntos
Povo Asiático , Fatores de Transcrição Forkhead/genética , Predisposição Genética para Doença , Linfócitos T Reguladores , Tireoidite Autoimune/genética , População Branca , Animais , Cromossomos Humanos X/genética , Cromossomos Humanos X/imunologia , Estudos de Coortes , Feminino , Fatores de Transcrição Forkhead/imunologia , Humanos , Masculino , Camundongos , Repetições de Microssatélites , Polimorfismo Genético/imunologia , Locos de Características Quantitativas/imunologia , Fatores Sexuais , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/patologia , Tireoidite Autoimune/etnologia , Tireoidite Autoimune/imunologia , Tireoidite Autoimune/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA