Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Curr Opin Hematol ; 29(4): 218-224, 2022 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-35787550

RESUMO

PURPOSE OF REVIEW: This review highlights findings describing the role of interleukin (IL)-10-producing Type 1 regulatory T (Tr1) cells in controlling autoimmune diseases and possible approaches to restore their function and number. RECENT FINDINGS: Reduced frequency and/or function of cell subsets playing a role in Tr1 cell induction (e.g., DC-10 and Bregs), was found in patients with autoimmunity and may impact on Tr1 cell frequency. SUMMARY: IL-10 is a pleiotropic cytokine with fundamental anti-inflammatory functions acting as negative regulator of immune responses. IL-10 is critically involved in the induction and functions of Tr1 cells, a subset of memory CD4+ T cells induced in the periphery to suppress immune responses to a variety of antigens (Ags), including self-, allogeneic, and dietary Ags. Alterations in IL-10-related pathways and/or in the frequency and activities of Tr1 cells have been associated to several autoimmune diseases. We will give an overview of the alterations of IL-10 and IL-10-producing Tr1 cells in Multiple Sclerosis, Type 1 Diabetes, and Celiac Disease, in which similarities in the role of these tolerogenic mechanisms are present. Current and future approaches to overcome Tr1 cell defects and restore tolerance in these diseases will also be discussed.


Assuntos
Doenças Autoimunes , Interleucina-10/imunologia , Linfócitos T Reguladores/imunologia , Animais , Doenças Autoimunes/imunologia , Doenças Autoimunes/patologia , Autoimunidade , Humanos
2.
J Allergy Clin Immunol ; 142(6): 1909-1921.e9, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29705245

RESUMO

BACKGROUND: Forkhead box P3 (FOXP3) is a key transcription factor in regulatory T (Treg) cell function. FOXP3 gene mutations cause immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome, a fatal autoimmune syndrome. FOXP3 has also been proposed to act in effector T (Teff) cells, but to date, this role has not been confirmed. OBJECTIVE: We sought to evaluate the effect of reduced FOXP3 expression on human Treg and Teff cell development and correlate it with IPEX syndrome immune pathology. METHODS: We developed a model of humanized mice (huMice) in which the human hematopoietic system is stably knocked down or knocked out for the FOXP3 gene (knockdown [KD]/knockout [KO] huMice). RESULTS: Because FOXP3-KD/KO was not 100% effective, residual FOXP3 expression in hematopoietic stem progenitor cells was sufficient to give rise to Treg cells with normal expression of FOXP3. However, numerous defects appeared in the Teff cell compartment. Compared with control mice, FOXP3-KD/KO huMice showed altered thymocyte differentiation, with KD/KO thymocytes displaying significantly reduced T-cell receptor (TCR) signaling strength and increased TCR repertoire diversity. Peripheral KD/KO Teff cells were expanded and showed signs of homeostatic proliferation, such as a significantly contracted TCR repertoire, a severely reduced naive compartment, decreased telomeric repeat-binding factor 2 expression, and a skew toward a TH2 profile, resembling an aged immune system. Consistent with results in FOXP3-KD/KO huMice, analysis of patients with IPEX syndrome provided evidence of defects in the Teff cell compartment at both the thymic and peripheral levels. CONCLUSIONS: These findings support an intrinsic role for human FOXP3 in controlling thymocyte maturation and peripheral expansion of Teff cells and reveal a previously undescribed pathogenic mechanism through an altered Teff cell compartment in patients with IPEX syndrome.


Assuntos
Diabetes Mellitus Tipo 1/congênito , Diarreia/imunologia , Fatores de Transcrição Forkhead/imunologia , Doenças Genéticas Ligadas ao Cromossomo X/imunologia , Doenças do Sistema Imunitário/congênito , Linfócitos T/imunologia , Timo/imunologia , Adolescente , Adulto , Animais , Diferenciação Celular , Criança , Pré-Escolar , Diabetes Mellitus Tipo 1/imunologia , Homeostase , Humanos , Doenças do Sistema Imunitário/imunologia , Lactente , Recém-Nascido , Masculino , Camundongos Transgênicos , Adulto Jovem
3.
Blood ; 119(20): 4675-85, 2012 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-22452978

RESUMO

Chromatin remodeling is fundamental for B-cell differentiation. In the present study, we explored the role of KAP1, the cofactor of KRAB-ZFP transcriptional repressors, in this process. B-lymphoid-specific Kap1-KO mice displayed reduced numbers of mature B cells, lower steady-state levels of Abs, and accelerated rates of decay of neutralizing Abs after viral immunization. Transcriptome analyses of Kap1-deleted B splenocytes revealed an up-regulation of PTEN, the enzymatic counteractor of PIK3 signaling, and of genes encoding DNA-damage response factors, cell-cycle regulators, and chemokine receptors. ChIP/seq studies established that KAP1 bound at or close to several of these genes and controlled chromatin status at their promoters. Genome wide, KAP1 binding sites lacked active B cell-specific enhancers and were enriched in repressive histone marks, further supporting a role for this molecule in gene silencing in vivo. Likely responsible for tethering KAP1 to at least some of these targets, a discrete subset of KRAB-ZFPs is enriched in B lymphocytes. Our results therefore reveal the role of KRAB/KAP1-mediated epigenetic regulation in B-cell development and homeostasis.


Assuntos
Linfócitos B/fisiologia , Diferenciação Celular/genética , Linfócitos/fisiologia , Proteínas Nucleares/fisiologia , Proteínas Repressoras/fisiologia , Animais , Formação de Anticorpos/genética , Formação de Anticorpos/imunologia , Linfócitos B/imunologia , Linfócitos B/metabolismo , Proteínas de Bactérias/genética , Diferenciação Celular/imunologia , Diferenciação Celular/fisiologia , Cromatina/metabolismo , Epigênese Genética/genética , Epigênese Genética/imunologia , Epigênese Genética/fisiologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Redes Reguladoras de Genes/genética , Redes Reguladoras de Genes/fisiologia , Proteínas Luminescentes/genética , Contagem de Linfócitos , Linfócitos/imunologia , Linfócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Análise em Microsséries , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Proteína 28 com Motivo Tripartido
4.
Hepatology ; 56(4): 1279-90, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22684873

RESUMO

UNLABELLED: The liver is characterized by sexually dimorphic gene expression translating into sex-specific differences in lipid, drug, steroid hormone, and xenobiotic metabolism, with distinct responses of males and females to environmental challenges. Here, we investigated the role of the Krüppel-associated box (KRAB)-associated protein 1 (KAP1) epigenetic regulator in this process. Liver-specific KAP1 knockout (KO) led to strikingly sexually dimorphic phenotypic disturbances, including male-predominant steatosis and hepatic tumors with up-regulation of protein kinase B and extracellular signal-related kinases 1/2 mitogen-activated protein kinase signaling. This correlated with the sex-specific transcriptional dysregulation of a wide range of metabolic genes, notably those involved in retinol and sex hormone processing as well as in detoxification. Furthermore, chromatin immunoprecipitation followed by deep sequencing indicated that a number of dysregulated genes are direct targets of the KRAB/KAP1 repression system. Those genes include sexually dimorphic cytochrome P 450 Cyp2d9, glutathione S-transferase π, Cyp2a, Cyp2b, and Cyp3a gene clusters. Additionally, we identified a male-restricted KAP1-binding site in the fat-specific protein 27 gene, correlating with its male-predominant up-regulation upon Kap1 deletion, suggesting that the latter might be an important trigger in the development of male-specific hepatosteatosis and secondary tumorigenesis. CONCLUSION: This work reveals KRAB/KAP1-mediated transcriptional regulation as a central event in metabolic control hormones, drugs, and xenobiotics in the liver and further links disturbances in these processes with hepatic carcinogenesis.


Assuntos
Adenoma/genética , Transformação Celular Neoplásica/genética , Fígado Gorduroso/genética , Predisposição Genética para Doença , Neoplasias Hepáticas/genética , Proteínas Nucleares/metabolismo , Proteínas Repressoras/metabolismo , Adenoma/patologia , Animais , Biópsia por Agulha , Transformação Celular Neoplásica/patologia , Proteínas de Ligação a DNA/genética , Modelos Animais de Doenças , Fígado Gorduroso/patologia , Feminino , Regulação da Expressão Gênica , Imuno-Histoquímica , Neoplasias Hepáticas/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Distribuição Aleatória , Sensibilidade e Especificidade , Fatores Sexuais , Proteína 28 com Motivo Tripartido , Dedos de Zinco/genética
5.
FASEB J ; 26(11): 4561-75, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22872677

RESUMO

Chromatin remodeling at specific genomic loci controls lymphoid differentiation. Here, we investigated the role played in this process by Kruppel-associated box (KRAB)-associated protein 1 (KAP1), the universal cofactor of KRAB-zinc finger proteins (ZFPs), a tetrapod-restricted family of transcriptional repressors. T-cell-specific Kap1-deleted mice displayed a significant expansion of immature thymocytes, imbalances in CD4(+)/CD8(+) cell ratios, and altered responses to TCR and TGFß stimulation when compared to littermate KAP1 control mice. Transcriptome and chromatin studies revealed that KAP1 binds T-cell-specific cis-acting regulatory elements marked by the H3K9me3 repressive mark and enriched in Ikaros/NuRD complexes. Also, KAP1 directly controls the expression of several genes involved in TCR and cytokine signaling. Among these, regulation of FoxO1 seems to play a major role in this system. Likely responsible for tethering KAP1 to at least part of its genomic targets, a small number of KRAB-ZFPs are selectively expressed in T-lymphoid cells. These results reveal the so far unsuspected yet important role of KAP1-mediated epigenetic regulation in T-lymphocyte differentiation and activation.


Assuntos
Regulação da Expressão Gênica/fisiologia , Proteínas Nucleares/metabolismo , Proteínas Repressoras/metabolismo , Linfócitos T/fisiologia , Animais , Sítios de Ligação , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/fisiologia , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/fisiologia , DNA/genética , DNA/metabolismo , Epigênese Genética , Camundongos , Camundongos Knockout , Proteínas Nucleares/genética , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Filogenia , Ligação Proteica , RNA/genética , RNA/metabolismo , Proteínas Repressoras/genética , Linfócitos T/citologia , Transcriptoma , Proteína 28 com Motivo Tripartido
6.
Cell Rep ; 42(3): 112193, 2023 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-36870061

RESUMO

Interleukin (IL)-10 is a main player in peripheral immune tolerance, the physiological mechanism preventing immune reactions to self/harmless antigens. Here, we investigate IL-10-induced molecular mechanisms generating tolerogenic dendritic cells (tolDC) from monocytes. Using genomic studies, we show that IL-10 induces a pattern of accessible enhancers exploited by aryl hydrocarbon receptor (AHR) to promote expression of a set of core genes. We demonstrate that AHR activity occurs downstream of IL-10 signaling in myeloid cells and is required for the induction of tolerogenic activities in DC. Analyses of circulating DCs show that IL-10/AHR genomic signature is active in vivo in health. In multiple sclerosis patients, we instead observe significantly altered signature correlating with functional defects and reduced frequencies of IL-10-induced-tolDC in vitro and in vivo. Our studies identify molecular mechanisms controlling tolerogenic activities in human myeloid cells and may help in designing therapies to re-establish immune tolerance.


Assuntos
Interleucina-10 , Receptores de Hidrocarboneto Arílico , Humanos , Células Dendríticas/metabolismo , Tolerância Imunológica , Interleucina-10/metabolismo , Monócitos/metabolismo , Receptores de Hidrocarboneto Arílico/genética , Receptores de Hidrocarboneto Arílico/metabolismo
8.
Front Immunol ; 11: 1260, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32695103

RESUMO

The prominent role of dendritic cells (DC) in promoting tolerance and the development of methods to generate clinical grade products allowed the clinical application of tolerogenic DC (tolDC)-based therapies for controlling unwanted immune responses. We established an efficient method to generate tolerogenic human DC, producing supra-physiological levels of IL-10, by genetically engineering monocyte-derived DC with a bidirectional Lentiviral Vector (bdLV) encoding for IL-10 and a marker gene. DCIL-10 are mature DC, modulate T cell responses, promote T regulatory cells, and are phenotypically and functionally stable upon stimulation. Adoptive transfer of human DCIL-10 in a humanized mouse model dampens allogeneic T cell recall responses, while murine DCIL-10 delays acute graft-vs.-host disease in mice. Our report outlines an efficient method to transduce human myeloid cells with large-size LV and shows that stable over-expression of IL-10 generates an effective cell product for future clinical applications in the contest of allogeneic transplantation.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Vetores Genéticos/genética , Tolerância Imunológica , Interleucina-10/genética , Lentivirus/genética , Transdução Genética , Animais , Feminino , Expressão Gênica , Humanos , Tolerância Imunológica/genética , Imunofenotipagem , Camundongos , Monócitos/imunologia , Monócitos/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo
9.
Cell Mol Immunol ; 17(1): 95-107, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-30842629

RESUMO

Tolerogenic dendritic cells (DCs) are key players in maintaining immunological homeostasis, dampening immune responses, and promoting tolerance. DC-10, a tolerogenic population of human IL-10-producing DCs characterized by the expression of HLA-G and ILT4, play a pivotal role in promoting tolerance via T regulatory type 1 (Tr1) cells. Thus far, the absence of markers that uniquely identify DC-10 has limited in vivo studies. By in vitro gene expression profiling of differentiated human DCs, we identified CD141 and CD163 as surface markers for DC-10. The coexpression of CD141 and CD163 in combination with CD14 and CD16 enables the ex vivo isolation of DC-10 from the peripheral blood. CD14+CD16+CD141+CD163+ cells isolated from the peripheral blood of healthy subjects (ex vivo DC-10) produced spontaneously and upon activation of IL-10 and limited levels of IL-12. Moreover, in vitro stimulation of allogeneic naive CD4+ T cells with ex vivo DC-10 induced the differentiation of alloantigen-specific CD49b+LAG-3+ Tr1 cells. Finally, ex vivo DC-10 and in vitro generated DC-10 exhibited a similar transcriptional profile, which are characterized by an anti-inflammatory and pro-tolerogenic signature. These results provide new insights into the phenotype and molecular signature of DC-10 and highlight the tolerogenic properties of circulating DC-10. These findings open the opportunity to track DC-10 in vivo and to define their role in physiological and pathological settings.


Assuntos
Antígenos CD/imunologia , Antígenos de Diferenciação Mielomonocítica/imunologia , Células Dendríticas/imunologia , Regulação da Expressão Gênica/imunologia , Interleucina-10/imunologia , Receptores de Superfície Celular/imunologia , Trombomodulina/imunologia , Células Dendríticas/citologia , Humanos , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/imunologia
10.
Stem Cells ; 26(8): 2142-52, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18483423

RESUMO

The isolation of human embryonic and somatic stem cells of different types has made it possible to design novel gene and cell replacement therapies. Vectors derived from retro/lentiviruses are used to stably introduce genes into stem cells and their progeny. However, the permissivity to retroviral infection varies among cell types. We previously showed that hematopoietic stem cells are poorly permissive to human immunodeficiency virus (HIV)-derived vectors and that pharmacological inhibition of the proteasome strongly enhances gene transfer. Here we report that the proteasome limits lentiviral gene transfer in all stem cell types tested, including embryonic, mesenchymal, and neural, of both human and mouse origin. Remarkably, this inhibitory activity was sharply reduced upon differentiation of the stem cells, suggesting that it represents a novel feature of the stem cell/immature progenitor phenotype. Proteasome-mediated inhibition was specific for lentiviral vectors and occurred at a postentry infection step. It was not mediated by activation of nuclear factor-kappaB, a major signaling pathway modulated by the proteasome, and did not correlate with high proteasome activity. Interaction of the virion core with cyclophilin A was required to maximize the effect of proteasome inhibitor on the infection pathway. These findings are relevant to uncover new mediators of HIV gene transfer and help in designing more effective protocols for the genetic modification of stem cells. Disclosure of potential conflicts of interest is found at the end of this article.


Assuntos
Técnicas de Transferência de Genes , Lentivirus/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Células-Tronco/citologia , Animais , Células Cultivadas , Vetores Genéticos , HIV/genética , Humanos , Interferons/metabolismo , Camundongos , NF-kappa B/metabolismo , Fenótipo , Transdução de Sinais
11.
Methods Mol Biol ; 506: 59-70, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19110619

RESUMO

Haematopoietic Stem Cells (HSCs) are attractive targets for the gene therapy. Upon ex vivo gene transfer and transplant, they may generate a progeny of gene-corrected cells potentially for a lifespan. The viral vectors most often used for HSC gene transfer are gamma-retroviral vectors (RVs) and HIV-derived lentiviral vectors (LVs). LVs have been proposed as improved tools for this task because they are able to transduce non-proliferating cells, while RVs are not. This implies that HSCs, which are mainly quiescent cells, need to be induced to proliferate in order to be transduced by RVs, whereas a prolonged stimulation is not needed for transduction by LVs. A short in vitro manipulation should reduce the risk of altering the characteristic biological properties of HSCs.We describe here methods for short-term ex vivo culture and gene transfer into human HSCs. Cord blood-derived HSC gene transfer can be tuned to limit the average level of vector integration or instead to maximize the frequency of transduction and extent of transgene expression, according to the absence or presence of cytokines. Mobilized peripheral blood-derived HSCs need cytokine stimulation to maintain viability and obtain adequate levels of gene transfer. Although HSCs can be transduced by LVs in short ex vivo culture, they display low permissiveness to the vector. Recently, we have demonstrated that this is because proteasome activity restricts LV gene transfer in HSCs. We developed and describe here strategies that effectively overcome this restriction. Finally, we also provide methods for the assessment of the transduction efficiency.


Assuntos
Antígenos CD34/imunologia , Técnicas de Transferência de Genes , Vetores Genéticos , Células-Tronco Hematopoéticas/citologia , Lentivirus/genética , Sequência de Bases , Primers do DNA , Células-Tronco Hematopoéticas/imunologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Reação em Cadeia da Polimerase
12.
Nat Commun ; 8(1): 498, 2017 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-28887441

RESUMO

HIV-1 insertions targeting BACH2 or MLK2 are enriched and persist for decades in hematopoietic cells from patients under combination antiretroviral therapy. However, it is unclear how these insertions provide such selective advantage to infected cell clones. Here, we show that in 30/87 (34%) patients under combination antiretroviral therapy, BACH2, and STAT5B are activated by insertions triggering the formation of mRNAs that contain viral sequences fused by splicing to their first protein-coding exon. These chimeric mRNAs, predicted to express full-length proteins, are enriched in T regulatory and T central memory cells, but not in other T lymphocyte subsets or monocytes. Overexpression of BACH2 or STAT5B in primary T regulatory cells increases their proliferation and survival without compromising their function. Hence, we provide evidence that HIV-1-mediated insertional activation of BACH2 and STAT5B favor the persistence of a viral reservoir in T regulatory cells in patients under combination antiretroviral therapy.HIV insertions in hematopoietic cells are enriched in BACH2 or MLK2 genes, but the selective advantages conferred are unknown. Here, the authors show that BACH2 and additionally STAT5B are activated by viral insertions, generating chimeric mRNAs specifically enriched in T regulatory cells favoring their persistence.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica/genética , Infecções por HIV/genética , HIV-1/genética , Fator de Transcrição STAT5/genética , Linfócitos T Reguladores/metabolismo , Fármacos Anti-HIV/uso terapêutico , Células Cultivadas , Reservatórios de Doenças/virologia , Regulação da Expressão Gênica , Células HEK293 , Infecções por HIV/tratamento farmacológico , Infecções por HIV/virologia , HIV-1/efeitos dos fármacos , HIV-1/fisiologia , Humanos , Mutagênese Insercional , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/virologia , Integração Viral
13.
Nucleus ; 5(2): 138-48, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24699165

RESUMO

The ability of adaptive immune system to protect higher vertebrates from pathogens resides in the ability of B and T cells to express different antigen specific receptors and to respond to different threats by activating distinct differentiation and/or activation pathways. In the past 10 years, the major role of epigenetics in controlling molecular mechanisms responsible for these peculiar features and, more in general, for lymphocyte development has become evident. KRAB-ZFPs is the widest family of mammalian transcriptional repressors, which function through the recruitment of the co-factor KRAB-Associated Protein 1 (KAP1) that in turn engages histone modifiers inducing heterochromatin formation. Although most of the studies on KRAB proteins have been performed in embryonic cells, more recent reports highlighted a relevant role for these proteins also in adult tissues. This article will review the role of KRAB-ZFP and KAP1 in the epigenetic control of mouse and human adaptive immune cells.


Assuntos
Imunidade Adaptativa , Fatores de Transcrição Kruppel-Like/metabolismo , Animais , Humanos , Fatores de Transcrição Kruppel-Like/química , Dedos de Zinco
14.
Curr Gene Ther ; 14(6): 422-8, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25274247

RESUMO

Immune dysregulation, Polyendocrinopathy, Enteropathy, X-linked (IPEX) syndrome is a rare autoimmune disease due to mutations in the gene encoding for Forkhead box P3 (FOXP3), a transcription factor fundamental for the function of thymus-derived (t) regulatory T (Treg) cells. The dysfunction of Treg cells results in the development of devastating autoimmune manifestations affecting multiple organs, eventually leading to premature death in infants, if not promptly treated by hematopoietic stem cell transplantation (HSCT). Novel gene therapy strategies can be developed for IPEX syndrome as more definitive cure than allogeneic HSCT. Here we describe the therapeutic approaches, alternative to HSCT, currently under development. We described that effector T cells can be converted in regulatory T cells by LV-mediated FOXP3-gene transfer in differentiated T lymphocytes. Despite FOXP3 mutations mainly affect a highly specific T cell subset, manipulation of stem cells could be required for long-term remission of the disease. Therefore, we believe that a more comprehensive strategy should aim at correcting FOXP3-mutated stem cells. Potentials and hurdles of both strategies will be highlighted here.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos , Fatores de Transcrição Forkhead/genética , Terapia Genética , Transplante de Células-Tronco Hematopoéticas , Linfócitos T Reguladores/imunologia , Diabetes Mellitus Tipo 1/congênito , Diarreia , Doenças Genéticas Ligadas ao Cromossomo X/genética , Doenças Genéticas Ligadas ao Cromossomo X/imunologia , Doenças Genéticas Ligadas ao Cromossomo X/terapia , Humanos , Doenças do Sistema Imunitário/congênito
15.
Int Rev Immunol ; 33(2): 129-45, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24354325

RESUMO

Ten years ago Forkhead box P3 (FOXP3) was discovered as master gene driving CD4(+)CD25(+) T cell regulatory (Treg) function. Since then, several layers of complexity have emerged in the regulation of its expression and function, which is not only exerted in Treg cells. While the mechanisms leading to the highly selective expression of FOXP3 in thymus-derived Treg cells still remain to be elucidated, we review here the current knowledge on the role of FOXP3 in the development of Treg cells and the direct and indirect consequences of FOXP3 mutations on multiple arms of the immune response. Finally, we summarize the newly acquired knowledge on the epigenetic regulation of FOXP3, still largely undefined in human cells.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Sistema Imunitário/fisiologia , Subpopulações de Linfócitos T/imunologia , Linfócitos T Reguladores/imunologia , Animais , Antígenos CD4/metabolismo , Epigênese Genética/imunologia , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/imunologia , Homeostase , Humanos , Tolerância Imunológica , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Mutação/genética
16.
PLoS One ; 8(2): e56721, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23451074

RESUMO

Highly coordinated transcription networks orchestrate the self-renewal of pluripotent stem cell and the earliest steps of mammalian development. KRAB-containing zinc finger proteins represent the largest group of transcription factors encoded by the genomes of higher vertebrates including mice and humans. Together with their putatively universal cofactor KAP1, they have been implicated in events as diverse as the silencing of endogenous retroelements, the maintenance of imprinting and the pluripotent self-renewal of embryonic stem cells, although the genomic targets and specific functions of individual members of this gene family remain largely undefined. Here, we first generated a list of Ensembl-annotated KRAB-containing genes encoding the mouse and human genomes. We then defined the transcription levels of these genes in murine early embryonic cells. We found that the majority of KRAB-ZFP genes are expressed in mouse pluripotent stem cells and other early progenitors. However, we also identified distinctively cell- or stage-specific patterns of expression, some of which are pluripotency-restricted. Finally, we determined that individual KRAB-ZFP genes exhibit highly distinctive modes of expression, even when grouped in genomic clusters, and that these cannot be correlated with the presence of prototypic repressive or activating chromatin marks. These results pave the way to delineating the role of specific KRAB-ZFPs in early embryogenesis.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas Nucleares/metabolismo , Células-Tronco Pluripotentes/metabolismo , Proteínas Repressoras/metabolismo , Animais , Proteínas de Transporte/genética , Células Cultivadas , Imunoprecipitação da Cromatina , Humanos , Camundongos , Proteínas Nucleares/genética , Proteínas Repressoras/genética
17.
Chem Biol ; 18(6): 805-15, 2011 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-21700215

RESUMO

Protein turnover critically influences many biological functions, yet methods have been lacking to assess this parameter in vivo. Here, we demonstrate how chemical labeling of SNAP-tag fusion proteins can be exploited to measure the half-life of resident intracellular and extracellular proteins in living mice. First, we demonstrate that SNAP-tag substrates have wide bioavailability in mice and can be used for the specific in vivo labeling of SNAP-tag fusion proteins. We then apply near-infrared probes to perform noninvasive imaging of in vivo-labeled tumors. Finally, we use SNAP-mediated chemical pulse-chase labeling to perform measurement of the in vivo half-life of different extra- and intracellular proteins. These results open broad perspectives for studying protein function in living animals.


Assuntos
O(6)-Metilguanina-DNA Metiltransferase/metabolismo , Estabilidade Proteica , Animais , Antígenos CD4/química , Antígenos CD4/genética , Antígenos CD4/metabolismo , Células Cultivadas , Feminino , Fibroblastos/metabolismo , Corantes Fluorescentes/química , Meia-Vida , Camundongos , Camundongos Nus , Camundongos Transgênicos , O(6)-Metilguanina-DNA Metiltransferase/química , O(6)-Metilguanina-DNA Metiltransferase/genética , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
18.
PLoS One ; 4(8): e6571, 2009 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-19668336

RESUMO

BACKGROUND: CD4+ T lymphocytes are the primary targets of HIV1 but cannot be infected when fully quiescent, due to a post-entry block preventing the completion of reverse transcription. Chiu et al. recently proposed that this restriction reflects the action of APOBEC3G (A3G). They further suggested that T cell activation abrogates the A3G-mediated block by directing this protein to a high molecular mass complex. METHODOLOGY/PRINCIPAL FINDINGS: In the present work, we sought to explore further this model. However, we found that effective suppression of A3G by combined RNA interference and expression of HIV1 Vif does not relieve the restrictive phenotype of post-activation resting T cells. We also failed to find a correlation between HIV resistance and the presence of A3G in a low molecular complex in primary T cells. CONCLUSIONS/SIGNIFICANCE: We conclude that A3G is unlikely to play a role in the HIV restrictive phenotype of quiescent T lymphocytes.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Citosina Desaminase/imunologia , Infecções por HIV/imunologia , Desaminases APOBEC , Western Blotting , Citidina Desaminase , Citosina Desaminase/genética , Citometria de Fluxo , HIV-1 , Humanos , Ultracentrifugação
19.
Blood ; 107(11): 4257-65, 2006 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-16469870

RESUMO

The therapeutic potential of hematopoietic stem cell (HSC) gene therapy can be fully exploited only by reaching efficient gene transfer into HSCs without compromising their biologic properties. Although HSCs can be transduced by HIV-derived lentiviral vectors (LVs) in short ex vivo culture, they display low permissivity to the vector, requiring cytokine stimulation to reach high-frequency transduction. Using stringent assays of competitive xenograft repopulation, we show that early-acting cytokines synergistically enhanced human HSC gene transfer by LVs without impairing engraftment and repopulation capacity. Using S-phase suicide assays, we show that transduction enhancement by cytokines was not dependent on cell cycle progression and that LVs can transduce quiescent HSCs. Pharmacologic inhibition of the proteasome during transduction dramatically enhanced HSC gene transfer, allowing the reach of very high levels of vector integration in their progeny in vivo. Thus, LVs are effectively restricted at a postentry step by the activity of this proteolytic complex. Unexpectedly, cytokine stimulation rapidly and substantially down-regulated proteasome activity in hematopoietic progenitors, highlighting one mechanism by which cytokines may enhance permissiveness to LV gene transfer. These findings demonstrate that antiviral responses ultimately mediated by proteasomes strongly limit the efficiency of HSC transduction by LVs and establish improved conditions for HSC-based gene therapy.


Assuntos
Citocinas/farmacologia , Células-Tronco Hematopoéticas/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Transdução Genética/métodos , Animais , Antígenos CD34 , Linhagem Celular Tumoral , Regulação para Baixo/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Transplante de Células-Tronco Hematopoéticas , Humanos , Lentivirus/genética , Camundongos , Camundongos SCID , Inibidores de Proteassoma , Transdução Genética/normas
20.
Blood ; 105(6): 2307-15, 2005 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-15542582

RESUMO

Recent reports have indicated that human immunodeficiency virus (HIV) and murine leukemia virus (MLV) vectors preferentially integrate into active genes. Here, we used a novel approach based on genetic trapping to rapidly score several thousand integration sites and found that MLV vectors trapped cellular promoters more efficiently than HIV vectors. Remarkably, 1 in 5 MLV integrations trapped an active promoter in different cell lines and primary hematopoietic cells. Such frequency was even higher in growth-stimulated lymphocytes. We show that the different behavior of MLV and HIV vectors was dependent on a different integration pattern within transcribed genes. Whereas MLV-based traps showed a strong bias for promoter-proximal integration leading to efficient reporter expression, HIV-based traps integrated throughout transcriptional units and were limited for expression by the distance from the promoter and the reading frame of the targeted gene. Our results indicate a strong propensity of MLV to establish transcriptional interactions with cellular promoters, a behavior that may have evolved to enhance proviral expression and may increase the insertional mutagenesis risk. Promoter trapping efficiency provides a convenient readout to assess transcriptional interactions between the vector and its flanking genes at the integration site and to compare integration site selection among different cell types and in different growth conditions.


Assuntos
HIV/genética , Células-Tronco Hematopoéticas/fisiologia , Vírus da Leucemia Murina/genética , Linfócitos/fisiologia , Regiões Promotoras Genéticas/genética , Integração Viral/genética , Linhagem Celular , Clonagem Molecular , Vetores Genéticos , Células-Tronco Hematopoéticas/citologia , Humanos , Linfócitos/citologia , Mutagênese Insercional , Sequências Repetidas Terminais/genética , Transcrição Gênica/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA