Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Front Immunol ; 14: 1275937, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37920467

RESUMO

Background: Human perinatal life is characterized by a period of extraordinary change during which newborns encounter abundant environmental stimuli and exposure to potential pathogens. To meet such challenges, the neonatal immune system is equipped with unique functional characteristics that adapt to changing conditions as development progresses across the early years of life, but the molecular characteristics of such adaptations remain poorly understood. The application of single cell genomics to birth cohorts provides an opportunity to investigate changes in gene expression programs elicited downstream of innate immune activation across early life at unprecedented resolution. Methods: In this study, we performed single cell RNA-sequencing of mononuclear cells collected from matched birth cord blood and 5-year peripheral blood samples following stimulation (18hrs) with two well-characterized innate stimuli; lipopolysaccharide (LPS) and Polyinosinic:polycytidylic acid (Poly(I:C)). Results: We found that the transcriptional response to LPS was constrained at birth and predominantly partitioned into classical proinflammatory gene upregulation primarily by monocytes and Interferon (IFN)-signaling gene upregulation by lymphocytes. Moreover, these responses featured substantial cell-to-cell communication which appeared markedly strengthened between birth and 5 years. In contrast, stimulation with Poly(I:C) induced a robust IFN-signalling response across all cell types identified at birth and 5 years. Analysis of gene regulatory networks revealed IRF1 and STAT1 were key drivers of the LPS-induced IFN-signaling response in lymphocytes with a potential developmental role for IRF7 regulation. Conclusion: Additionally, we observed distinct activation trajectory endpoints for monocytes derived from LPS-treated cord and 5-year blood, which was not apparent among Poly(I:C)-induced monocytes. Taken together, our findings provide new insight into the gene regulatory landscape of immune cell function between birth and 5 years and point to regulatory mechanisms relevant to future investigation of infection susceptibility in early life.


Assuntos
Lipopolissacarídeos , Transcriptoma , Recém-Nascido , Humanos , Lipopolissacarídeos/farmacologia , Lipopolissacarídeos/metabolismo , Monócitos , Transdução de Sinais , Regulação da Expressão Gênica , Poli I-C/farmacologia , Poli I-C/metabolismo
2.
Leukemia ; 37(1): 61-71, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36380143

RESUMO

Infants with KMT2A-rearranged B-cell acute lymphoblastic leukemia (ALL) have a dismal prognosis. Survival outcomes have remained static in recent decades despite treatment intensification and novel therapies are urgently required. KMT2A-rearranged infant ALL cells are characterized by an abundance of promoter hypermethylation and exhibit high BCL-2 expression, highlighting potential for therapeutic targeting. Here, we show that hypomethylating agents exhibit in vitro additivity when combined with most conventional chemotherapeutic agents. However, in a subset of samples an antagonistic effect was seen between several agents. This was most evident when hypomethylating agents were combined with methotrexate, with upregulation of ATP-binding cassette transporters identified as a potential mechanism. Single agent treatment with azacitidine and decitabine significantly prolonged in vivo survival in KMT2A-rearranged infant ALL xenografts. Treatment of KMT2A-rearranged infant ALL cell lines with azacitidine and decitabine led to differential genome-wide DNA methylation, changes in gene expression and thermal proteome profiling revealed the target protein-binding landscape of these agents. The selective BCL-2 inhibitor, venetoclax, exhibited in vitro additivity in combination with hypomethylating or conventional chemotherapeutic agents. The addition of venetoclax to azacitidine resulted in a significant in vivo survival advantage indicating the therapeutic potential of this combination to improve outcome for infants with KMT2A-rearranged ALL.


Assuntos
Leucemia Mieloide Aguda , Leucemia-Linfoma Linfoblástico de Células Precursoras , Humanos , Lactente , Azacitidina/farmacologia , Azacitidina/uso terapêutico , Decitabina/farmacologia , Decitabina/uso terapêutico , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Proteínas Proto-Oncogênicas c-bcl-2 , Leucemia Mieloide Aguda/genética
3.
Nat Commun ; 13(1): 4501, 2022 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-36042201

RESUMO

KMT2A-rearranged infant acute lymphoblastic leukemia (ALL) represents the most refractory type of childhood leukemia. To uncover the molecular heterogeneity of this disease, we perform RNA sequencing, methylation array analysis, whole exome and targeted deep sequencing on 84 infants with KMT2A-rearranged leukemia. Our multi-omics clustering followed by single-sample and single-cell inference of hematopoietic differentiation establishes five robust integrative clusters (ICs) with different master transcription factors, fusion partners and corresponding stages of B-lymphopoietic and early hemato-endothelial development: IRX-type differentiated (IC1), IRX-type undifferentiated (IC2), HOXA-type MLLT1 (IC3), HOXA-type MLLT3 (IC4), and HOXA-type AFF1 (IC5). Importantly, our deep mutational analysis reveals that the number of RAS pathway mutations predicts prognosis and that the most refractory subgroup of IC2 possesses 100% frequency and the heaviest burden of RAS pathway mutations. Our findings highlight the previously under-appreciated intra- and inter-patient heterogeneity of KMT2A-rearranged infant ALL and provide a rationale for the future development of genomics-guided risk stratification and individualized therapy.


Assuntos
Histona-Lisina N-Metiltransferase/genética , Proteína de Leucina Linfoide-Mieloide/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras , Fusão Gênica , Humanos , Lactente , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Fatores de Transcrição/genética
4.
Biomolecules ; 11(6)2021 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-34200313

RESUMO

Notch signaling forms an evolutionarily conserved juxtacrine pathway crucial for cellular development. Initially identified in Drosophila wing morphogenesis, Notch signaling has since been demonstrated to play pivotal roles in governing mammalian cellular development in a large variety of cell types. Indeed, abolishing Notch constituents in mouse models result in embryonic lethality, demonstrating that Notch signaling is critical for development and differentiation. In this review, we focus on the crucial role of Notch signaling in governing embryogenesis and differentiation of multiple progenitor cell types. Using hematopoiesis as a diverse cellular model, we highlight the role of Notch in regulating the cell fate of common lymphoid progenitors. Additionally, the influence of Notch through microenvironment interplay with lymphoid cells and how dysregulation influences disease processes is explored. Furthermore, bi-directional and lateral Notch signaling between ligand expressing source cells and target cells are investigated, indicating potentially novel therapeutic options for treatment of Notch-mediated diseases. Finally, we discuss the role of cis-inhibition in regulating Notch signaling in mammalian development.


Assuntos
Linhagem da Célula/fisiologia , Desenvolvimento Embrionário/fisiologia , Linfopoese/fisiologia , Receptores Notch/fisiologia , Animais , Humanos , Linfócitos/fisiologia , Transdução de Sinais/fisiologia
5.
Mol Immunol ; 128: 150-164, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33129017

RESUMO

During mammalian lymphoid development, Notch signaling is necessary at multiple stages of T lymphopoiesis, including lineage commitment, and later stages of T cell effector differentiation. In contrast, outside of a defined role in the development of splenic marginal zone B cells, there is conflicting evidence regarding whether Notch signaling plays functional roles in other B cell sub-populations. Complement receptor 2 (CR2) modulates BCR-signaling and is tightly regulated throughout differentiation. During B lymphopoiesis, CR2 is detected on immature and mature B cells with high surface expression on marginal zone B cells. Here, we have explored the possibility that Notch regulates human CR2 transcriptional activity using in vitro models including a co-culture system, co-transfection gene reporters and chromatin accessibility assays. We provide evidence that Notch signaling regulates CR2 promoter activity in a mature B cell line, as well as the induction of endogenous CR2 mRNA in a non-expressing pre-B cell line. The dynamics of endogenous gene activation suggests additional unidentified factors are required to mediate surface CR2 expression on immature and mature B lineage cells.


Assuntos
Complemento C3d/genética , Células Precursoras de Linfócitos B/fisiologia , Regiões Promotoras Genéticas/genética , Receptores de Complemento 3d/genética , Receptores Notch/genética , Transdução de Sinais/genética , Transcrição Gênica/genética , Linfócitos B/fisiologia , Diferenciação Celular/genética , Linhagem Celular , Linhagem Celular Tumoral , Cromatina/genética , Técnicas de Cocultura/métodos , Humanos , Células K562 , Ativação Linfocitária/genética , Linfopoese/genética
6.
Epigenetics ; 15(1-2): 134-144, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31423932

RESUMO

Activation of naïve CD8+ T cells stimulates proliferation and differentiation into cytotoxic T-lymphocytes (CTLs). Adoptive T Cell Therapy (ACT) involves multiple rounds of ex vivo activation to generate enough CTLs for reinfusion into patients, but this drives differentiation into terminal effector T cells. Less differentiated CTL populations, such as stem cell memory T cells, are more ideal candidates for ACT because of increased self-renewal and persistent properties. Ex vivo targeting of T cell differentiation with epigenetic modifiers is a potential strategy to improve cytotoxic T-lymphocyte (CTL) generation for ACT. We established a pipeline to assess the effects of epigenetic modifiers on CD8+ T cell proliferation, differentiation, and efficacy in a preclinical melanoma model. Single treatment with epigenetic modifiers inhibited T cell proliferation in vitro, producing CD44hiCD62Lhi effector-like T cells rather than a stem cell memory T cell phenotype. Most epigenetic modifying agents had no significant effect on ACT efficacy with the notable exception of the bromodomain and extraterminal (BET)-inhibitor JQ1 which was associated with a decrease in efficacy compared to unmodified T cells. These findings reveal the complexity of epigenetic targeting of T cell differentiation, highlighting the need to precisely define the epigenetic targeting strategies to improve CTL generation for ACT.


Assuntos
Proliferação de Células , Epigênese Genética , Imunoterapia Adotiva/métodos , Melanoma Experimental/terapia , Linfócitos T/efeitos dos fármacos , Animais , Azepinas/farmacologia , Benzodiazepinas/farmacologia , Diferenciação Celular , Linhagem Celular Tumoral , Células Cultivadas , Indolizinas/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Sulfonas/farmacologia , Linfócitos T/citologia , Linfócitos T/imunologia , Linfócitos T/fisiologia , Triazóis/farmacologia
7.
Oncoimmunology ; 8(8): 1609874, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31413920

RESUMO

Immunotherapies such as adoptive cell therapy (ACT) are promising treatments for solid cancers. However, relapsing disease remains a problem and the molecular mechanisms underlying resistance are poorly defined. We postulated that the deregulated epigenetic landscape in cancer cells could underpin the acquisition of resistance to immunotherapy. To address this question, two preclinical models of ACT were employed to study transcriptional and epigenetic regulatory processes within ACT-treated cancer cells. In these models ACT consistently causes robust tumor regression, but resistance develops and tumors relapse. We identified down-regulated expression of immunogenic antigens at the mRNA level correlated with escape from immune control. To determine whether this down-regulation was under epigenetic control, we treated escaped tumor cells with DNA demethylating agents, azacytidine (AZA) and decitabine (DEC). AZA or DEC treatment restored antigen expression in a proportion of the tumor population. To explore the importance of other epigenetic modifications we isolated tumor cells refractory to DNA demethylation and screened clones against a panel of 19 different epigenetic modifying agents (EMAs). The library of EMAs included inhibitors of a range of chromosomal and transcription regulatory protein complexes, however, when tested as single agents none restored further antigen expression. These findings suggest that tumor cells employ multiple epigenetic and genetic mechanisms to evade immune control, and a combinatorial approach employing several EMAs targeting transcription and genome stability may be required to overcome tumor resistance to immunotherapy.

9.
Oncogene ; 38(1): 140-150, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30076412

RESUMO

Overexpression of MYC oncogene is highly prevalent in many malignancies such as aggressive triple-negative breast cancers (TNBCs) and it is associated with very poor outcome. Despite decades of research, attempts to effectively inhibit MYC, particularly with small molecules, still remain challenging due to the featureless nature of its protein structure. Herein, we describe the engineering of the dominant-negative MYC peptide (OmoMYC) linked to a functional penetrating 'Phylomer' peptide (FPPa) as a therapeutic strategy to inhibit MYC in TNBC. We found FPPa-OmoMYC to be a potent inducer of apoptosis (with IC50 from 1-2 µM) in TNBC cells with negligible effects in non-tumorigenic cells. Transcriptome analysis of FPPa-OmoMYC-treated cells indicated that the fusion protein inhibited MYC-dependent networks, inducing dynamic changes in transcriptional, metabolic, and apoptotic processes. We demonstrated the efficacy of FPPa-OmoMYC in inhibiting breast cancer growth when injected orthotopically in TNBC allografts. Lastly, we identified strong pharmacological synergisms between FPPa-OmoMYC and chemotherapeutic agents. This study highlights a novel therapeutic approach to target highly aggressive and chemoresistant MYC-activated cancers.


Assuntos
Peptídeos Penetradores de Células/farmacologia , Terapia de Alvo Molecular/métodos , Proteínas de Neoplasias/antagonistas & inibidores , Fragmentos de Peptídeos/uso terapêutico , Proteínas Proto-Oncogênicas c-myc/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-myc/uso terapêutico , Proteínas Recombinantes de Fusão/uso terapêutico , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Sequência de Aminoácidos , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Linhagem Celular Tumoral , Peptídeos Penetradores de Células/administração & dosagem , Peptídeos Penetradores de Células/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Sinergismo Farmacológico , Feminino , Genes myc , Humanos , Concentração Inibidora 50 , Zíper de Leucina/genética , Camundongos , Modelos Moleculares , Mutação , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/farmacocinética , Biblioteca de Peptídeos , Conformação Proteica , Engenharia de Proteínas , Proteínas Proto-Oncogênicas c-myc/administração & dosagem , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/farmacocinética , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/farmacocinética
10.
Front Immunol ; 9: 2990, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30700986

RESUMO

Conventional dendritic cells (cDC) resident in the lymphoid organs of mice have been classically divided into CD8+ and CD8neg subsets. It is well-established that CD8+ dendritic cells (DCs) and their migratory counterparts in the periphery comprise the cross-presenting cDC1 subset. In contrast, CD8neg DCs are grouped together in the heterogeneous cDC2 subset. CD8neg DCs are relatively poor cross-presenters and drive more prominent CD4+ T cell responses against exogenous antigens. The discovery of the X-C motif chemokine receptor 1 (XCR1) as a specific marker of cross-presenting DCs, has led to the identification of a divergent subset of CD8+ DCs that lacks the ability to cross-present. Here, we report that these poorly characterized CD8+XCR1neg DCs have a gene expression profile that is consistent with both plasmacytoid DCs (pDCs) and cDC2. Our data demonstrate that CD8+XCR1neg DCs possess a unique pattern of endocytic receptors and a restricted toll-like receptor (TLR) profile that is particularly enriched for TLR5, giving them a unique position within the DC immunosurveillance network.


Assuntos
Apresentação Cruzada , Células Dendríticas/metabolismo , Receptor 5 Toll-Like/metabolismo , Animais , Antígenos CD8/metabolismo , Separação Celular , Células Dendríticas/imunologia , Endocitose/imunologia , Citometria de Fluxo , Perfilação da Expressão Gênica , Camundongos , Camundongos Endogâmicos C57BL , Análise de Sequência com Séries de Oligonucleotídeos , Receptores de Quimiocinas/metabolismo , Receptores de Reconhecimento de Padrão/imunologia , Receptores de Reconhecimento de Padrão/metabolismo , Receptor 5 Toll-Like/imunologia
11.
Cell Mol Immunol ; 13(1): 119-31, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25640655

RESUMO

Complement receptor 2 (CR2/CD21) is predominantly expressed on the surface of mature B cells where it forms part of a coreceptor complex that functions, in part, to modulate B-cell receptor signal strength. CR2/CD21 expression is tightly regulated throughout B-cell development such that CR2/CD21 cannot be detected on pre-B or terminally differentiated plasma cells. CR2/CD21 expression is upregulated at B-cell maturation and can be induced by IL-4 and CD40 signaling pathways. We have previously characterized elements in the proximal promoter and first intron of CR2/CD21 that are involved in regulating basal and tissue-specific expression. We now extend these analyses to the CR2/CD21 core promoter. We show that in mature B cells, CR2/CD21 transcription proceeds from a focused TSS regulated by a non-consensus TATA box, an initiator element and a downstream promoter element. Furthermore, occupancy of the general transcriptional machinery in pre-B versus mature B-cell lines correlate with CR2/CD21 expression level and indicate that promoter accessibility must switch from inactive to active during the transitional B-cell window.


Assuntos
Antígenos CD40/metabolismo , Interleucina-4/metabolismo , Células Precursoras de Linfócitos B/metabolismo , Regiões Promotoras Genéticas , Receptores de Complemento 3d/metabolismo , Sítio de Iniciação de Transcrição , Sequência de Bases , Antígenos CD40/genética , Antígenos CD40/imunologia , Diferenciação Celular , Linhagem Celular Tumoral , Éxons , Regulação da Expressão Gênica , Humanos , Interleucina-4/genética , Interleucina-4/imunologia , Íntrons , Células K562 , Dados de Sequência Molecular , Células Precursoras de Linfócitos B/citologia , Células Precursoras de Linfócitos B/imunologia , Receptores de Complemento 3d/genética , Receptores de Complemento 3d/imunologia , Transdução de Sinais , Transcrição Gênica
12.
Int J Biochem Cell Biol ; 64: 107-19, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25817480

RESUMO

Complement receptor 2 (CR2/CD21) plays an important role in the generation of normal B cell immune responses. As transcription appears to be the prime mechanism via which surface CR2/CD21 expression is controlled, understanding transcriptional regulation of this gene will have broader implications to B cell biology. Here we report opposing, cell-context specific control of CR2/CD21 promoter activity by tandem E-box elements, spaced 22 bp apart and within 70 bp of the transcription initiation site. We have identified E2A and USF transcription factors as binding to the distal and proximal E-box sites respectively in CR2-positive B-cells, at a site that is hypersensitive to restriction enzyme digestion compared to non-expressing K562 cells. However, additional unidentified proteins have also been found to bind these functionally important elements. By utilizing a proteomics approach we have identified a repressor protein, RP58, binding the distal E-box motif. Co-transfection experiments using RP58 overexpression constructs demonstrated a specific 10-fold repression of CR2/CD21 transcriptional activity mediated through the distal E-box repressor element. Taken together, our results indicate that repression of the CR2/CD21 promoter can occur through one of the E-box motifs via recruitment of RP58 and other factors to bring about a silenced chromatin context within CR2/CD21 non-expressing cells.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Receptores de Complemento 3d/genética , Proteínas Repressoras/fisiologia , Fatores Estimuladores Upstream/metabolismo , Sequência de Bases , Cromatina/fisiologia , Elementos E-Box , Epigênese Genética , Humanos , Células K562 , Dados de Sequência Molecular , Especificidade de Órgãos , Regiões Promotoras Genéticas , Receptores de Complemento 3d/metabolismo
13.
Genome Med ; 5(10): 96, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24134860

RESUMO

BACKGROUND: Preterm birth confers a high risk of adverse long term health outcomes for survivors, yet the underlying molecular mechanisms are unclear. We hypothesized that effects of preterm birth can be mediated through measurable epigenomic changes throughout development. We therefore used a longitudinal birth cohort to measure the epigenetic mark of DNA methylation at birth and 18 years comparing survivors of extremely preterm birth with infants born at term. METHODS: Using 12 extreme preterm birth cases and 12 matched, term controls, we extracted DNA from archived neonatal blood spots and blood collected in a similar way at 18 years of age. DNA methylation was measured at 347,789 autosomal locations throughout the genome using Infinium HM450 arrays. Representative methylation differences were confirmed by Sequenom MassArray EpiTYPER. RESULTS: At birth we found 1,555 sites with significant differences in methylation between term and preterm babies. At 18 years of age, these differences had largely resolved, suggesting that DNA methylation differences at birth are mainly driven by factors relating to gestational age, such as cell composition and/or maturity. Using matched longitudinal samples, we found evidence for an epigenetic legacy associated with preterm birth, identifying persistent methylation differences at ten genomic loci. Longitudinal comparisons of DNA methylation at birth and 18 years uncovered a significant overlap between sites that were differentially-methylated at birth and those that changed with age. However, we note that overlapping sites may either differ in the same (300/1,555) or opposite (431/1,555) direction during gestation and aging respectively. CONCLUSIONS: We present evidence for widespread methylation differences between extreme preterm and term infants at birth that are largely resolved by 18 years of age. These results are consistent with methylation changes associated with blood cell development, cellular composition, immune induction and age at these time points. Finally, we identified ten probes significantly associated with preterm individuals and with greater than 5% methylation discordance at birth and 18 years that may reflect a long term epigenetic legacy of preterm birth.

14.
Genome Biol ; 14(5): R42, 2013 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-23697701

RESUMO

BACKGROUND: The extent to which development- and age-associated epigenetic changes are influenced by genetic, environmental and stochastic factors remains to be discovered. Twins provide an ideal model with which to investigate these influences but previous cross-sectional twin studies provide contradictory evidence of within-pair epigenetic drift over time. Longitudinal twin studies can potentially address this discrepancy. RESULTS: In a pilot, genome-scale study of DNA from buccal epithelium, a relatively homogeneous tissue, we show that one-third of the CpGs assayed show dynamic methylation between birth and 18 months. Although all classes of annotated genomic regions assessed show an increase in DNA methylation over time, probes located in intragenic regions, enhancers and low-density CpG promoters are significantly over-represented, while CpG islands and high-CpG density promoters are depleted among the most dynamic probes. Comparison of co-twins demonstrated that within-pair drift in DNA methylation in our cohort is specific to a subset of pairs, who show more differences at 18 months. The rest of the pairs show either minimal change in methylation discordance, or more similar, converging methylation profiles at 18 months. As with age-associated regions, sites that change in their level of within-pair discordance between birth and 18 months are enriched in genes involved in development, but the average magnitude of change is smaller than for longitudinal change. CONCLUSIONS: Our findings suggest that DNA methylation in buccal epithelium is influenced by non-shared stochastic and environmental factors that could reflect a degree of epigenetic plasticity within an otherwise constrained developmental program.


Assuntos
DNA/análise , Gêmeos/genética , Fatores Etários , Ilhas de CpG , Metilação de DNA , Epigênese Genética , Epigenômica/métodos , Feminino , Genoma Humano , Humanos , Lactente , Recém-Nascido , Estudos Longitudinais , Masculino , Boca/citologia , Projetos Piloto
15.
Genome Med ; 4(10): 83, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23131117

RESUMO

Epigenome-wide association studies (EWAS) can be used to investigate links between early life environment, epigenetics and disease. However, such studies raise the question of which came first: the mark or the malady? A recent study has demonstrated that EWAS can be performed on neonatal 'Guthrie' heel-prick blood spots. As Guthrie cards are collected from all newborn infants and stored indefinitely in many countries, they represent an important timepoint to compare with later disease-associated epigenetic marks.

16.
Genome Res ; 22(8): 1395-406, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22800725

RESUMO

Comparison between groups of monozygotic (MZ) and dizygotic (DZ) twins enables an estimation of the relative contribution of genetic and shared and nonshared environmental factors to phenotypic variability. Using DNA methylation profiling of ∼20,000 CpG sites as a phenotype, we have examined discordance levels in three neonatal tissues from 22 MZ and 12 DZ twin pairs. MZ twins exhibit a wide range of within-pair differences at birth, but show discordance levels generally lower than DZ pairs. Within-pair methylation discordance was lowest in CpG islands in all twins and increased as a function of distance from islands. Variance component decomposition analysis of DNA methylation in MZ and DZ pairs revealed a low mean heritability across all tissues, although a wide range of heritabilities was detected for specific genomic CpG sites. The largest component of variation was attributed to the combined effects of nonshared intrauterine environment and stochastic factors. Regression analysis of methylation on birth weight revealed a general association between methylation of genes involved in metabolism and biosynthesis, providing further support for epigenetic change in the previously described link between low birth weight and increasing risk for cardiovascular, metabolic, and other complex diseases. Finally, comparison of our data with that of several older twins revealed little evidence for genome-wide epigenetic drift with increasing age. This is the first study to analyze DNA methylation on a genome scale in twins at birth, further highlighting the importance of the intrauterine environment on shaping the neonatal epigenome.


Assuntos
Metilação de DNA , Epigênese Genética , Desenvolvimento Fetal/genética , Genoma Humano , Gêmeos Dizigóticos/genética , Gêmeos Monozigóticos/genética , Células Cultivadas , Ilhas de CpG , Epigenômica/métodos , Feminino , Retardo do Crescimento Fetal/genética , Deriva Genética , Idade Gestacional , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Recém-Nascido de Baixo Peso , Recém-Nascido , Padrões de Herança , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Especificidade de Órgãos , Fenótipo , Placenta/citologia , Placenta/metabolismo , Gravidez , Análise de Regressão , Processos Estocásticos
17.
Mol Immunol ; 52(3-4): 165-73, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22673213

RESUMO

Systemic lupus erythematosus (SLE) is a complex autoimmune disease with a strong genetic component that determines risk. A common three single-nucleotide polymorphism (SNP) haplotype of the complement receptor 2 (CR2) gene has been associated with increased risk of SLE (Wu et al., 2007; Douglas et al., 2009), and a less common haplotype consisting of the major allele at SNP1 and minor alleles at SNP2 and 3 confers protection (Douglas et al., 2009). SNP1 (rs3813946), which is located in the 5' untranslated region (UTR) of the CR2 gene, altered transcriptional activity of a CR2 promoter-luciferase reporter gene construct transiently transfected into a B cell line (Wu et al., 2007) and had an independent effect in the protective haplotype (Douglas et al., 2009). In this study, we show that this SNP alters transcriptional activity in a transiently transfected non B-cell line as well as in stably transfected cell lines, supporting its relevance in vivo. Furthermore, the allele at this SNP affects chromatin accessibility of the surrounding sequence and transcription factor binding. These data confirm the effects of rs3813946 on CR2 transcription, identifying the 5' UTR to be a novel regulatory element for the CR2 gene in which variation may alter gene function and modify the development of lupus.


Assuntos
Regiões 5' não Traduzidas , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/imunologia , Polimorfismo de Nucleotídeo Único , Receptores de Complemento 3d/genética , Transcrição Gênica , Linfócitos B/imunologia , Linhagem Celular Tumoral , Predisposição Genética para Doença , Humanos , Lúpus Eritematoso Sistêmico/metabolismo , Regiões Promotoras Genéticas , Receptores de Complemento 3d/imunologia
18.
Amino Acids ; 39(5): 1087-105, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20204433

RESUMO

Eukaryotic genomic DNA is orderly compacted to fit into the nucleus and to inhibit accessibility of specific sequences. DNA is manipulated in many different ways by bound RNA and proteins within the composite material known as chromatin. All of the biological processes that require access to genomic DNA (such as replication, recombination and transcription) therefore are dependent on the precise characteristics of chromatin in eukaryotes. This distinction underlies a fundamental property of eukaryotic versus prokaryotic gene regulation such that chromatin structure must be regulated to precisely repress or relieve repression of particular regions of the genome in an appropriate spatio-temporal manner. As well as playing a key role in structuring genomic DNA, histones are subject to site-specific modifications that can influence the organization of chromatin structure. This review examines the molecular processes regulating site-specific histone acetylation, methylation and phosphorylation with an emphasis on how these processes underpin differentiation-regulated transcription.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/genética , Histonas/metabolismo , Processamento de Proteína Pós-Traducional , Humanos
19.
Immunol Cell Biol ; 88(2): 117-24, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19806164

RESUMO

The notch signaling pathway is evolutionarily conserved across the animal kingdom and regulates developmental 'decisions', such as cell fate commitment, differentiation, proliferation and apoptosis. In the mammalian immune system, notch signaling events have been extensively studied during T lymphopoiesis, and have a role both during early development, as well as differentiation into discreet effector cell compartments. In contrast, the impact of notch signaling in the B-cell compartment is less obvious. It is clear that notch signaling is crucial to generate the marginal zone B-cell population located within the spleen; however, the full effects of notch signaling during normal B-cell development remain unresolved. Nevertheless, there is compelling evidence that notch signaling regulates multiple stages of B-cell differentiation and in shaping the antibody repertoire; however, the molecular details have not been elucidated. This review explores the relationship between notch signaling and B-cell development with attention to how these processes contribute to a normal B-cell repertoire.


Assuntos
Linfócitos B/imunologia , Linfopoese , Receptores Notch/imunologia , Transdução de Sinais/imunologia , Animais , Linfócitos B/citologia , Humanos , Ativação Linfocitária/imunologia , Subpopulações de Linfócitos/citologia , Subpopulações de Linfócitos/imunologia
20.
Mol Immunol ; 46(13): 2613-22, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19487031

RESUMO

Stringent developmental transcription requires multiple transcription factor (TF) binding sites, cell-specific expression of signaling molecules, TFs and co-regulators and appropriate chromatin structure. During B-lymphopoiesis, human Complement receptor 2 (CR2/CD21) is detected on immature and mature B cells but not on B cell precursors and plasma cells. We examined cell- and stage-specific human CR2 gene regulation using cell lines modeling B-lymphopoiesis. Chromatin accessibility assays revealed a region between -409 and -262 with enhanced accessibility in mature B cells and pre-B cells, compared to either non-lymphoid or plasma cell-types, however, accessibility near the transcription start site (TSS) was elevated only in CR2-expressing B cells. A correlation between histone acetylation and CR2 expression was observed, while histone H3K4 dimethylation was enriched near the TSS in both CR2-expressing B cells and non-expressing pre-B cells. Candidate sites within the CR2 promoter were identified which could regulate chromatin, including a matrix attachment region associated with CDP, SATB1/BRIGHT and CEBP-beta sites as well as two CBF1 sites. ChIP assays verified that both CBF1 and C/EBP-beta bind the CR2 promoter in B cells raising the possibility that these factors facilitate or respond to alterations in chromatin structure to control the timing and/or level of CR2 transcription.


Assuntos
Linfócitos B/metabolismo , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Cromatina/metabolismo , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/metabolismo , Regiões Promotoras Genéticas/genética , Receptores de Complemento 3d/genética , Acetilação , Animais , Linfócitos B/citologia , Sítios de Ligação/genética , Linhagem Celular , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Citometria de Fluxo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Histonas/metabolismo , Humanos , Células Jurkat , Células K562 , Camundongos , Ligação Proteica , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Sítio de Iniciação de Transcrição , Células U937
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA