Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Eur J Immunol ; 49(1): 66-78, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30365177

RESUMEN

The interferon-inducible transmembrane (Ifitm/Fragilis) genes encode homologous proteins that are induced by IFNs. Here, we show that IFITM proteins regulate murine CD4+ Th cell differentiation. Ifitm2 and Ifitm3 are expressed in wild-type (WT) CD4+ T cells. On activation, Ifitm3 was downregulated and Ifitm2 was upregulated. Resting Ifitm-family-deficient CD4+ T cells had higher expression of Th1-associated genes than WT and purified naive Ifitm-family-deficient CD4+ T cells differentiated more efficiently to Th1, whereas Th2 differentiation was inhibited. Ifitm-family-deficient mice, but not Ifitm3-deficient mice, were less susceptible than WT to induction of allergic airways disease, with a weaker Th2 response and less severe disease and lower Il4 but higher Ifng expression and IL-27 secretion. Thus, the Ifitm family is important in adaptive immunity, influencing Th1/Th2 polarization, and Th2 immunopathology.


Asunto(s)
Hipersensibilidad/inmunología , Inflamación/inmunología , Proteínas de la Membrana/metabolismo , Sistema Respiratorio/inmunología , Células TH1/inmunología , Células Th2/inmunología , Animales , Diferenciación Celular/genética , Células Cultivadas , Interferón gamma/metabolismo , Interleucina-27/metabolismo , Interleucina-4/metabolismo , Activación de Linfocitos/genética , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Balance Th1 - Th2/genética
2.
Nature ; 452(7189): 877-81, 2008 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-18354397

RESUMEN

A unique feature of the germ cell lineage is the generation of totipotency. A critical event in this context is DNA demethylation and the erasure of parental imprints in mouse primordial germ cells (PGCs) on embryonic day 11.5 (E11.5) after they enter into the developing gonads. Little is yet known about the mechanism involved, except that it is apparently an active process. We have examined the associated changes in the chromatin to gain further insights into this reprogramming event. Here we show that the chromatin changes occur in two steps. The first changes in nascent PGCs at E8.5 establish a distinctive chromatin signature that is reminiscent of pluripotency. Next, when PGCs are residing in the gonads, major changes occur in nuclear architecture accompanied by an extensive erasure of several histone modifications and exchange of histone variants. Furthermore, the histone chaperones HIRA and NAP-1 (NAP111), which are implicated in histone exchange, accumulate in PGC nuclei undergoing reprogramming. We therefore suggest that the mechanism of histone replacement is critical for these chromatin rearrangements to occur. The marked chromatin changes are intimately linked with genome-wide DNA demethylation. On the basis of the timing of the observed events, we propose that if DNA demethylation entails a DNA repair-based mechanism, the evident histone replacement would represent a repair-induced response event rather than being a prerequisite.


Asunto(s)
Ensamble y Desensamble de Cromatina , Cromatina/metabolismo , Epigénesis Genética , Células Germinativas/metabolismo , Animales , Metilación de ADN , Gónadas/citología , Gónadas/metabolismo , Histonas/metabolismo , Ratones , Células Madre/metabolismo
3.
Glia ; 61(5): 679-93, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23382131

RESUMEN

Interferon-induced transmembrane protein 3 (IFITM3) iplays a crucial role in the antiviral responses of Type I interferons (IFNs). The role of IFITM3 in the central nervous system (CNS) is, however, largely unknown, despite the fact that its expression is increased in the brains of patients with neurologic and neuropsychiatric diseases. Here, we show the role of IFITM3 in long-lasting neuronal impairments in mice following polyriboinosinic-polyribocytidylic acid (polyI:C, a synthetic double-stranded RNA)-induced immune challenge during the early stages of development. We found that the induction of IFITM3 expression in the brain of mice treated with polyI:C was observed only in astrocytes. Cultured astrocytes were activated by polyI:C treatment, leading to an increase in the mRNA levels of inflammatory cytokines as well as Ifitm3. When cultured neurons were treated with the conditioned medium of polyI:C-treated astrocytes (polyI:C-ACM), neurite development was impaired. These polyI:C-ACM-induced neurodevelopmental abnormalities were alleviated by ifitm3(-/-) astrocyte-conditioned medium. Furthermore, decreases of MAP2 expression, spine density, and dendrite complexity in the frontal cortex as well as memory impairment were evident in polyI:C-treated wild-type mice, but such neuronal impairments were not observed in ifitm3(-) (/) (-) mice. We also found that IFITM3 proteins were localized to the early endosomes of astrocytes following polyI:C treatment and reduced endocytic activity. These findings suggest that the induction of IFITM3 expression in astrocytes by the activation of the innate immune system during the early stages of development has non-cell autonomous effects that affect subsequent neurodevelopment, leading to neuropathological impairments and brain dysfunction, by impairing endocytosis in astrocytes.


Asunto(s)
Astrocitos/patología , Proteínas de la Membrana/fisiología , Neuronas/patología , Animales , Animales Recién Nacidos , Astrocitos/inmunología , Células COS , Células Cultivadas , Chlorocebus aethiops , Citocinas/biosíntesis , Citocinas/genética , Endocitosis/efectos de los fármacos , Endocitosis/inmunología , Femenino , Inmunidad Innata/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Noqueados , Neuronas/inmunología , Poli I-C/farmacología , ARN Mensajero/biosíntesis , ARN Mensajero/efectos de los fármacos
4.
Nat Cell Biol ; 8(6): 623-30, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16699504

RESUMEN

Blimp1, a transcriptional repressor, has a crucial role in the specification of primordial germ cells (PGCs) in mice at embryonic day 7.5 (E7.5). This SET-PR domain protein can form complexes with various chromatin modifiers in a context-dependent manner. Here, we show that Blimp1 has a novel interaction with Prmt5, an arginine-specific histone methyltransferase, which mediates symmetrical dimethylation of arginine 3 on histone H2A and/or H4 tails (H2A/H4R3me2s). Prmt5 has been shown to associate with Tudor, a component of germ plasm in Drosophila melanogaster. Blimp1-Prmt5 colocalization results in high levels of H2A/H4 R3 methylation in PGCs at E8.5. However, at E11.5, Blimp1-Prmt5 translocates from the nucleus to the cytoplasm, resulting in the loss of H2A/H4 R3 methylation at the time of extensive epigenetic reprogramming of germ cells. Subsequently, Dhx38, a putative target of the Blimp1-Prmt5 complex, is upregulated. Interestingly, expression of Dhx38 is also seen in pluripotent embryonic germ cells that are derived from PGCs when Blimp1 expression is lost. Our study demonstrates that Blimp1 is involved in a novel transcriptional regulatory complex in the mouse germ-cell lineage.


Asunto(s)
Arginina/metabolismo , Células Germinativas/metabolismo , Histonas/metabolismo , Proteínas Represoras/fisiología , Factores de Transcripción/fisiología , Transporte Activo de Núcleo Celular , Adenosina Trifosfatasas/genética , Factores de Edad , Animales , Embrión de Mamíferos , Regulación de la Expresión Génica , Metilación , Ratones , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Unión Proteica , Proteína Metiltransferasas/metabolismo , Proteína-Arginina N-Metiltransferasas , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética
5.
BMC Med Genomics ; 16(1): 68, 2023 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-37013607

RESUMEN

BACKGROUND: Human endogenous retroviruses (HERV) are repetitive sequence elements and a substantial part of the human genome. Their role in development has been well documented and there is now mounting evidence that dysregulated HERV expression also contributes to various human diseases. While research on HERV elements has in the past been hampered by their high sequence similarity, advanced sequencing technology and analytical tools have empowered the field. For the first time, we are now able to undertake locus-specific HERV analysis, deciphering expression patterns, regulatory networks and biological functions of these elements. To do so, we inevitable rely on omics datasets available through the public domain. However, technical parameters inevitably differ, making inter-study analysis challenging. We here address the issue of confounding factors for profiling locus-specific HERV transcriptomes using datasets from multiple sources. METHODS: We collected RNAseq datasets of CD4 and CD8 primary T cells and extracted HERV expression profiles for 3220 elements, resembling most intact, near full-length proviruses. Looking at sequencing parameters and batch effects, we compared HERV signatures across datasets and determined permissive features for HERV expression analysis from multiple-source data. RESULTS: We could demonstrate that considering sequencing parameters, sequencing-depth is most influential on HERV signature outcome. Sequencing samples deeper broadens the spectrum of expressed HERV elements. Sequencing mode and read length are secondary parameters. Nevertheless, we find that HERV signatures from smaller RNAseq datasets do reliably reveal most abundantly expressed HERV elements. Overall, HERV signatures between samples and studies overlap substantially, indicating a robust HERV transcript signature in CD4 and CD8 T cells. Moreover, we find that measures of batch effect reduction are critical to uncover genic and HERV expression differences between cell types. After doing so, differences in the HERV transcriptome between ontologically closely related CD4 and CD8 T cells became apparent. CONCLUSION: In our systematic approach to determine sequencing and analysis parameters for detection of locus-specific HERV expression, we provide evidence that analysis of RNAseq datasets from multiple studies can aid confidence of biological findings. When generating de novo HERV expression datasets we recommend increased sequence depth ( > = 100 mio reads) compared to standard genic transcriptome pipelines. Finally, batch effect reduction measures need to be implemented to allow for differential expression analysis.


Asunto(s)
Retrovirus Endógenos , Humanos , Retrovirus Endógenos/genética , Provirus , Transcriptoma , Linfocitos T
6.
Viruses ; 15(11)2023 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-38005849

RESUMEN

Despite remarkable progress, a cure for HIV-1 infection remains elusive. Rebound competent latent and transcriptionally active reservoir cells persevere despite antiretroviral therapy and rekindle infection due to inefficient proviral silencing. We propose a novel "block-lock-stop" approach, entailing long term durable silencing of viral expression towards an irreversible transcriptionally inactive latent provirus to achieve long term antiretroviral free control of the virus. A graded transformation of remnant HIV-1 in PLWH from persistent into silent to permanently defective proviruses is proposed, emulating and accelerating the natural path that human endogenous retroviruses (HERVs) take over millions of years. This hypothesis was based on research into delineating the mechanisms of HIV-1 latency, lessons from latency reversing agents and advances of Tat inhibitors, as well as expertise in the biology of HERVs. Insights from elite controllers and the availability of advanced genome engineering technologies for the direct excision of remnant virus set the stage for a rapid path to an HIV-1 cure.


Asunto(s)
Retrovirus Endógenos , Infecciones por VIH , Seropositividad para VIH , VIH-1 , Humanos , VIH-1/genética , Latencia del Virus , Provirus/genética , Seropositividad para VIH/genética , Linfocitos T CD4-Positivos
7.
Bioessays ; 32(8): 659-68, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20658704

RESUMEN

During mammalian development, maintenance of cell fate through mitotic divisions require faithful replication not only of the DNA but also of a particular epigenetic state. Germline cells have the capacity of erasing this epigenetic memory at crucial times during development, thereby resetting their epigenome. Certain marks, however, appear to escape this reprogramming, which allows their transmission to the offspring and potentially guarantees transgenerational epigenetic inheritance. Here we discuss the molecular requirements for faithful transmission of epigenetic information and our current knowledge about the transmission of epigenetic information through generations.


Asunto(s)
Epigénesis Genética/genética , Animales , Metilación de ADN/genética , Histonas/genética , Humanos , Mitosis/genética
8.
PLoS One ; 16(12): e0261269, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34928979

RESUMEN

Adeno-associated viruses (AAV) are considered non-pathogenic in humans, and thus have been developed into powerful vector platforms for in vivo gene therapy. Although the various AAV serotypes display broad tropism, frequently infecting multiple tissues and cell types, vectors for specific and efficient targeting of human CD4+ T lymphocytes are largely missing. In fact, a substantial translational bottleneck exists in the field of therapeutic gene transfer that would require in vivo delivery into peripheral disease-related lymphocytes for subsequent genome editing. To solve this issue, capsid modification for retargeting AAV tropism, and in turn improving vector potency, is considered a promising strategy. Here, we genetically modified the minor AAV2 capsid proteins, VP1 and VP2, with a set of novel nanobodies with high-affinity for the human CD4 receptor. These novel vector variants demonstrated improved targeting of human CD4+ cells, including primary human peripheral blood mononuclear cells (PBMC) and purified human CD4+ T lymphocytes. Thus, the technical approach presented here provides a promising strategy for developing specific gene therapy vectors, particularly targeting disease-related peripheral blood CD4+ leukocytes.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Proteínas de la Cápside/genética , Dependovirus/genética , Vectores Genéticos/administración & dosificación , Leucocitos Mononucleares/metabolismo , Anticuerpos de Dominio Único/química , Transducción Genética , Linfocitos T CD4-Positivos/inmunología , Técnicas de Transferencia de Gen , Células HEK293 , Células HeLa , Humanos , Leucocitos Mononucleares/inmunología
9.
Virus Res ; 293: 198260, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33316352

RESUMEN

Chronic infection with human immunodeficiency virus (HIV)-1 is characterized by accumulation of proviral sequences in the genome of target cells. Integration of viral DNA in patients on long-term antiretroviral therapy selectively persists at preferential loci, suggesting site-specific crosstalk of viral sequences and human genes. This crosstalk likely contributes to chronic HIV disease through modulation of host immune pathways and emergence of clonal infected cell populations. To systematically interrogate such effects, we undertook genome engineering to generate Jurkat cell models that replicate integration of HIV-1 long terminal repeat (LTR) sequences at the BTB and CNC Homolog 2 (BACH2) integration locus. This locus is a prominent HIV-1 integration gene in chronic infection, found in 30 % of long-term treated patients with mapped proviral integrations. Using five clonal models carrying an LTR-driven reporter at different BACH2 intergenic regions, we here show that LTR transcriptional activity is repressed in BACH2 regions associated with proviral-DNA integrations in vivo but not in a control region. Our data indicates that this repression is in part epigenetically regulated, particularly through DNA methylation. Importantly, we demonstrate that transcriptional activity of the LTR is independent of BACH2 gene transcription and vice versa in our models. This suggests no transcriptional interference of endogenous and HIV-1 promoters. Taken together, our study provides first insights into how activity of HIV-1 LTR sequences is regulated at the BACH2 locus as prominent example for a recurrently-detected integration gene in chronic infection. Given the importance of integration-site dependent virus/host crosstalk for chronic HIV disease, our findings for the BACH2 locus have potential implications for future therapeutic strategies.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , VIH-1 , VIH-1/genética , Humanos , Infección Persistente , Regiones Promotoras Genéticas , Provirus/genética , Integración Viral
10.
Semin Immunopathol ; 42(2): 187-200, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32047948

RESUMEN

Human immunodeficiency virus 1 (HIV-1) replicates through the integration of its viral DNA into the genome of human immune target cells. Chronically infected individuals thus carry a genomic burden of virus-derived sequences that persists through antiretroviral therapy. This burden consists of a small fraction of intact, but transcriptionally silenced, i.e. latent, viral genomes and a dominant fraction of defective sequences. Remarkably, all viral-derived sequences are subject to interaction with host cellular physiology at various levels. In this review, we focus on epigenetic aspects of this interaction. We provide a comprehensive overview of how epigenetic mechanisms contribute to establishment and maintenance of HIV-1 gene repression during latency. We furthermore summarize findings indicating that HIV-1 infection leads to changes in the epigenome of target and bystander immune cells. Finally, we discuss how an improved understanding of epigenetic features and mechanisms involved in HIV-1 infection could be exploited for clinical use.


Asunto(s)
Infecciones por VIH , VIH-1 , Linfocitos T CD4-Positivos , Epigénesis Genética , Infecciones por VIH/genética , VIH-1/genética , Humanos , Latencia del Virus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA