Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
1.
Nucleic Acids Res ; 50(D1): D1358-D1367, 2022 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-34751399

RESUMO

We describe a comprehensive and unique database 'Priority index' (Pi; http://pi.well.ox.ac.uk) of prioritized genes encoding potential therapeutic targets that encompasses all major immune-mediated diseases. We provide targets at the gene level, each receiving a 5-star rating supported by: genomic evidence arising from disease genome-wide associations and functional immunogenomics, annotation evidence using ontologies restricted to genes with genomic evidence, and network evidence from protein interactions. Target genes often act together in related molecular pathways. The underlying Pi approach is unique in identifying a network of highly rated genes that mediate pathway crosstalk. In the Pi website, disease-centric pages are specially designed to enable the users to browse a complete list of prioritized genes and also a manageable list of nodal genes at the pathway crosstalk level; both switchable by clicks. Moreover, target genes are cross-referenced and supported using additional information, particularly regarding tractability, including druggable pockets viewed in 3D within protein structures. Target genes highly rated across diseases suggest drug repurposing opportunity, while genes in a particular disease reveal disease-specific targeting potential. To facilitate the ease of such utility, cross-disease comparisons involving multiple diseases are also supported. This facility, together with the faceted search, enhances integrative mining of the Pi resource to accelerate early-stage therapeutic target identification and validation leveraging human genetics.


Assuntos
Bases de Dados Genéticas , Genoma Humano/imunologia , Doenças do Sistema Imunitário/genética , Software , Biologia Computacional , Genoma Humano/genética , Genômica , Humanos , Doenças do Sistema Imunitário/imunologia , Imunogenética/normas
2.
PLoS Comput Biol ; 17(8): e1009254, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34343164

RESUMO

Driven by the necessity to survive environmental pathogens, the human immune system has evolved exceptional diversity and plasticity, to which several factors contribute including inheritable structural polymorphism of the underlying genes. Characterizing this variation is challenging due to the complexity of these loci, which contain extensive regions of paralogy, segmental duplication and high copy-number repeats, but recent progress in long-read sequencing and optical mapping techniques suggests this problem may now be tractable. Here we assess this by using long-read sequencing platforms from PacBio and Oxford Nanopore, supplemented with short-read sequencing and Bionano optical mapping, to sequence DNA extracted from CD14+ monocytes and peripheral blood mononuclear cells from a single European individual identified as HV31. We use this data to build a de novo assembly of eight genomic regions encoding four key components of the immune system, namely the human leukocyte antigen, immunoglobulins, T cell receptors, and killer-cell immunoglobulin-like receptors. Validation of our assembly using k-mer based and alignment approaches suggests that it has high accuracy, with estimated base-level error rates below 1 in 10 kb, although we identify a small number of remaining structural errors. We use the assembly to identify heterozygous and homozygous structural variation in comparison to GRCh38. Despite analyzing only a single individual, we find multiple large structural variants affecting core genes at all three immunoglobulin regions and at two of the three T cell receptor regions. Several of these variants are not accurately callable using current algorithms, implying that further methodological improvements are needed. Our results demonstrate that assessing haplotype variation in these regions is possible given sufficiently accurate long-read and associated data. Continued reductions in the cost of these technologies will enable application of these methods to larger samples and provide a broader catalogue of germline structural variation at these loci, an important step toward making these regions accessible to large-scale genetic association studies.


Assuntos
Variação Genética , Genoma Humano/imunologia , Sistema Imunitário , Algoritmos , Biologia Computacional , Variações do Número de Cópias de DNA , Genômica/métodos , Genômica/estatística & dados numéricos , Antígenos HLA/genética , Haplótipos , Sequenciamento de Nucleotídeos em Larga Escala/estatística & dados numéricos , Humanos , Fenômenos Imunogenéticos , Imunoglobulinas/genética , Receptores de Antígenos de Linfócitos T/genética , Receptores KIR/genética , Análise de Sequência de DNA/estatística & dados numéricos
3.
Signal Transduct Target Ther ; 6(1): 312, 2021 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-34417437

RESUMO

Immunotherapies play critical roles in cancer treatment. However, given that only a few patients respond to immune checkpoint blockades and other immunotherapeutic strategies, more novel technologies are needed to decipher the complicated interplay between tumor cells and the components of the tumor immune microenvironment (TIME). Tumor immunomics refers to the integrated study of the TIME using immunogenomics, immunoproteomics, immune-bioinformatics, and other multi-omics data reflecting the immune states of tumors, which has relied on the rapid development of next-generation sequencing. High-throughput genomic and transcriptomic data may be utilized for calculating the abundance of immune cells and predicting tumor antigens, referring to immunogenomics. However, as bulk sequencing represents the average characteristics of a heterogeneous cell population, it fails to distinguish distinct cell subtypes. Single-cell-based technologies enable better dissection of the TIME through precise immune cell subpopulation and spatial architecture investigations. In addition, radiomics and digital pathology-based deep learning models largely contribute to research on cancer immunity. These artificial intelligence technologies have performed well in predicting response to immunotherapy, with profound significance in cancer therapy. In this review, we briefly summarize conventional and state-of-the-art technologies in the field of immunogenomics, single-cell and artificial intelligence, and present prospects for future research.


Assuntos
Antígenos de Neoplasias/genética , Genoma Humano/imunologia , Neoplasias/imunologia , Microambiente Tumoral/imunologia , Inteligência Artificial , Biomarcadores Tumorais/imunologia , Linhagem da Célula , Humanos , Imunidade Inata/genética , Imunogenética/tendências , Imunoterapia , Neoplasias/genética , Neoplasias/terapia , Proteoma/imunologia , Análise de Célula Única , Transcriptoma/imunologia
4.
Int J Biochem Cell Biol ; 131: 105882, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33189874

RESUMO

The Major Histocompatibility Complex (MHC) on the short arm of chromosome 6 is associated with more diseases than any other region of the genome; it encodes the antigen-presenting Human Leukocyte Antigen (HLA) proteins and is one of the key immunogenetic regions of the genome. Accurate genome inference and interpretation of MHC association signals have traditionally been hampered by the region's uniquely complex features, such as high levels of polymorphism; inter-gene sequence homologies; structural variation; and long-range haplotype structures. Recent algorithmic and technological advances have, however, significantly increased the accessibility of genetic variation in the MHC; these developments include (i) accurate SNP-based HLA type imputation; (ii) genome graph approaches for variation-aware genome inference from next-generation sequencing data; (iii) long-read-based diploid de novo assembly of the MHC; (iv) cost-effective targeted MHC sequencing methods. Applied to hundreds of thousands of samples over the last years, these technologies have already enabled significant biological discoveries, for example in the field of autoimmune disease genetics. Remaining challenges concern the development of integrated methods that leverage haplotype-resolved de novo assembly of the MHC for the development of improved MHC genotyping methods for short reads and the construction of improved reference panels for SNP-based imputation. Improved genome inference in the MHC can crucially contribute to an improved genetic and functional understanding of many immune-related phenotypes and diseases.


Assuntos
Algoritmos , Mapeamento Cromossômico/métodos , Genoma Humano/imunologia , Antígenos HLA/genética , Teste de Histocompatibilidade/métodos , Complexo Principal de Histocompatibilidade/genética , Alelos , Sequência de Bases , Biologia Computacional/métodos , Heterogeneidade Genética , Antígenos HLA/classificação , Antígenos HLA/imunologia , Haplótipos , Sequenciamento de Nucleotídeos em Larga Escala/estatística & dados numéricos , Humanos , Desequilíbrio de Ligação , Polimorfismo de Nucleotídeo Único
5.
Antioxid Redox Signal ; 34(14): 1128-1150, 2021 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-33143450

RESUMO

Significance: Genomic instability, a hallmark of cancer, renders cancer cells susceptible to genomic stress from both endogenous and exogenous origins, resulting in the increased tendency to accrue DNA damage, chromosomal instability, or aberrant DNA localization. Apart from the cell autonomous tumor-promoting effects, genomic stress in cancer cells could have a profound impact on the tumor microenvironment. Recent Advances: Recently, it is increasingly appreciated that harnessing genomic stress could provide a promising strategy to revive antitumor immunity, and thereby offer new therapeutic opportunities in cancer treatment. Critical Issues: Genomic stress is closely intertwined with antitumor immunity via mechanisms involving the direct crosstalk with DNA damage response components, upregulation of immune-stimulatory/inhibitory ligands, release of damage-associated molecular patterns, increase of neoantigen repertoire, and activation of DNA sensing pathways. A better understanding of these mechanisms will provide molecular basis for exploiting the genomic stress to boost antitumor immunity. Future Directions: Future research should pay attention to the heterogeneity between individual cancers in the genomic instability and the associated immune response, and how to balance the toxicity and benefit by specifying the types, potency, and treatment sequence of genomic stress inducer in therapeutic practice. Antioxid. Redox Signal. 34, 1128-1150.


Assuntos
Instabilidade Genômica , Genômica , Imunidade Inata/genética , Neoplasias/genética , Dano ao DNA/genética , Dano ao DNA/imunologia , Reparo do DNA/genética , Reparo do DNA/imunologia , Genoma Humano/genética , Genoma Humano/imunologia , Humanos , Neoplasias/imunologia , Neoplasias/metabolismo , Neoplasias/patologia , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia
6.
DNA Res ; 27(5)2020 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-33084858

RESUMO

Accumulating evidence suggests that human genome can fold into non-B DNA structures, when appropriate sequence and favourable conditions are present. Among these, G-quadruplexes (G4-DNA) are associated with gene regulation, chromosome fragility and telomere maintenance. Although several techniques are used in detecting such structures in vitro, understanding their intracellular existence has been challenging. Recently, an antibody, BG4, was described to study G4 structures within cells. Here, we characterize BG4 for its affinity towards G4-DNA, using several biochemical and biophysical tools. BG4 bound to G-rich DNA derived from multiple genes that form G-quadruplexes, unlike complementary C-rich or random sequences. BLI studies revealed robust binding affinity (Kd = 17.4 nM). Gel shift assays show BG4 binds to inter- and intramolecular G4-DNA, when it is in parallel orientation. Mere presence of G4-motif in duplex DNA is insufficient for antibody recognition. Importantly, BG4 can bind to G4-DNA within telomere sequence in a supercoiled plasmid. Finally, we show that BG4 binds to form efficient foci in four cell lines, irrespective of their lineage, demonstrating presence of G4-DNA in genome. Importantly, number of BG4 foci within the cells can be modulated, upon knockdown of G4-resolvase, WRN. Thus, we establish specificity of BG4 towards G4-DNA and discuss its potential applications.


Assuntos
Anticorpos , DNA/imunologia , Quadruplex G , Genoma Humano/imunologia , Linhagem Celular , Células HEK293 , Células HeLa , Humanos
7.
Ann Clin Transl Neurol ; 7(10): 2041-2046, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32951359

RESUMO

The precise genetic diagnosis of dystrophinopathies can be challenging, largely due to rare deep intronic variants and more complex structural variants (SVs). We report on the genetic characterization of a dystrophinopathy patient. He remained without a genetic diagnosis after routine genetic testing, dystrophin protein and mRNA analysis, and short- and long-read whole DMD gene sequencing. We finally identified a novel complex SV in DMD via long-read whole-genome sequencing. The variant consists of a large-scale (~1Mb) inversion/deletion-insertion rearrangement mediated by LINE-1s. Our study shows that long-read whole-genome sequencing can serve as a clinical diagnostic tool for genetically unsolved dystrophinopathies.


Assuntos
Distrofina/metabolismo , Genoma Humano/imunologia , Mutação/efeitos dos fármacos , RNA Mensageiro/metabolismo , Distrofina/genética , Humanos , Masculino , Distrofia Muscular de Duchenne/diagnóstico , Sequenciamento Completo do Genoma/métodos
8.
Nat Rev Immunol ; 20(12): 771-780, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32612208

RESUMO

The human genome encodes more than 300 potential immune inhibitory receptors. The reason for this large number of receptors remains unclear. We suggest that inhibitory receptors operate as two distinct functional categories: receptors that control the signalling threshold for immune cell activation and receptors involved in the negative feedback of immune cell activation. These two categories have characteristic receptor expression patterns: 'threshold' receptors are expressed at steady state and their expression remains high or is downregulated upon activation, whereas 'negative feedback' receptors are induced upon immune cell activation. We use mathematical models to illustrate their possible modes of operation in different scenarios for different purposes. We discuss how this categorization may impact the choice of therapeutic targets for immunotherapy of malignant, infectious and autoimmune diseases.


Assuntos
Receptores Imunológicos/imunologia , Animais , Doenças Autoimunes/imunologia , Regulação para Baixo/imunologia , Genoma Humano/imunologia , Humanos , Transdução de Sinais/imunologia
9.
J Cell Physiol ; 235(12): 9834-9850, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32452055

RESUMO

Renal clear cell carcinoma (ccRCC) is the most common type of renal cell carcinoma, which has strong immunogenicity. A comprehensive study of the role of immune-related genes (IRGs) in ccRCC is of great significance in finding ccRCC treatment targets and improving patient prognosis. In this study, we comprehensively analyzed the expression of IRGs in ccRCC based on The Cancer Genome Atlas datasets. The mechanism of differentially expressed IRGs in ccRCC was analyzed by bioinformatics. In addition, Cox regression analysis was used to screen prognostic related IRGs from differentially expressed IRGs. We also identified a four IRGs signature consisting of four IRGs (CXCL2, SEMA3G, PDGFD, and UCN) through lasso regression and multivariate Cox regression analysis. Further analysis results showed that the four IRGs signature could effectively predict the prognosis of patients with ccRCC, and its predictive power is independent of other clinical factors. In addition, the correlation analysis of immune cell infiltration showed that this four IRGs signature could effectively reflect the level of immune cell infiltration of ccRCC. We also found that the expression of immune checkpoint genes CTLA-4, LAG3, and PD-1 in the high-risk group was higher than that in the low-risk group. Our research revealed the role of IRGs in ccRCC, and developed a four IRGs signature that could be used to evaluate the prognosis of patients with ccRCC, which will help to develop personalized treatment strategies for patients with ccRCC and improve their prognosis. In addition, these four IRGs may be effective therapeutic targets for ccRCC.


Assuntos
Carcinoma de Células Renais/imunologia , Quimiocina CXCL2/genética , Linfocinas/genética , Fator de Crescimento Derivado de Plaquetas/genética , Semaforinas/genética , Urocortinas/genética , Adulto , Idoso , Biomarcadores Tumorais/imunologia , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/patologia , Quimiocina CXCL2/imunologia , Intervalo Livre de Doença , Feminino , Regulação Neoplásica da Expressão Gênica/imunologia , Genoma Humano/imunologia , Humanos , Proteínas de Checkpoint Imunológico/genética , Proteínas de Checkpoint Imunológico/imunologia , Imunogenética , Linfocinas/imunologia , Masculino , Pessoa de Meia-Idade , Fator de Crescimento Derivado de Plaquetas/imunologia , Prognóstico , Modelos de Riscos Proporcionais , Semaforinas/imunologia , Transcriptoma , Urocortinas/imunologia
10.
Proc Natl Acad Sci U S A ; 117(14): 7905-7916, 2020 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-32193341

RESUMO

Transposable elements (TEs) compose nearly half of mammalian genomes and provide building blocks for cis-regulatory elements. Using high-throughput sequencing, we show that 84 TE subfamilies are overrepresented, and distributed in a lineage-specific fashion in core and boundary domains of CD8+ T cell enhancers. Endogenous retroviruses are most significantly enriched in core domains with accessible chromatin, and bear recognition motifs for immune-related transcription factors. In contrast, short interspersed elements (SINEs) are preferentially overrepresented in nucleosome-containing boundaries. A substantial proportion of these SINEs harbor a high density of the enhancer-specific histone mark H3K4me1 and carry sequences that match enhancer boundary nucleotide composition. Motifs with regulatory features are better preserved within enhancer-enriched TE copies compared to their subfamily equivalents located in gene deserts. TE-rich and TE-poor enhancers associate with both shared and unique gene groups and are enriched in overlapping functions related to lymphocyte and leukocyte biology. The majority of T cell enhancers are shared with other immune lineages and are accessible in common hematopoietic progenitors. A higher proportion of immune tissue-specific enhancers are TE-rich compared to enhancers specific to other tissues, correlating with higher TE occurrence in immune gene-associated genomic regions. Our results suggest that during evolution, TEs abundant in these regions and carrying motifs potentially beneficial for enhancer architecture and immune functions were particularly frequently incorporated by evolving enhancers. Their putative selection and regulatory cooption may have accelerated the evolution of immune regulatory networks.


Assuntos
Elementos de DNA Transponíveis/genética , Elementos Facilitadores Genéticos/genética , Evolução Molecular , Linfócitos T/imunologia , Animais , Cromatina/genética , Cromatina/imunologia , Elementos de DNA Transponíveis/imunologia , Retrovirus Endógenos/genética , Retrovirus Endógenos/imunologia , Elementos Facilitadores Genéticos/imunologia , Redes Reguladoras de Genes/genética , Genoma Humano/genética , Genoma Humano/imunologia , Genômica/métodos , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Elementos Nucleotídeos Curtos e Dispersos/genética , Elementos Nucleotídeos Curtos e Dispersos/imunologia
11.
J Genet Genomics ; 47(1): 27-35, 2020 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-32111437

RESUMO

B cells express B-cell receptors (BCRs) which recognize antigen to trigger signaling cascades for B-cell activation and subsequent antibody production. BCR activation has a crucial influence on B-cell fate. How BCR is activated upon encountering antigen remains to be solved, although tremendous progresses have been achieved in the past few years. Here, we summarize the models that have been proposed to explain BCR activation, including the cross-linking model, the conformation-induced oligomerization model, the dissociation activation model, and the conformational change model. Especially, we elucidate the partially resolved structures of antibodies and/or BCRs by far and discusse how these current structural and further immunogenomic messages and more importantly the future studies may shed light on the explanation of BCR activation and the relevant diseases in the case of dysregulation.


Assuntos
Linfócitos B/imunologia , Ativação Linfocitária/imunologia , Conformação Proteica , Receptores de Antígenos de Linfócitos B/genética , Genoma Humano/imunologia , Humanos , Ativação Linfocitária/genética , Receptores de Antígenos de Linfócitos B/imunologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia
12.
Biomed Res Int ; 2019: 2183510, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31781598

RESUMO

Immunotherapy directed against cancer-specific neoantigens derived from non-silent mutants is a promising individualized strategy for cancer treatment. Neoantigens shared across patients could be used as a public resource for developing T cell-based therapy. To identify potential public neoantigens for therapy in gastric cancer (GC), 74 GC patients were enrolled in this study. Combined with the TCGA cohort and other published studies, whole exome sequencing data from 942 GC patients were used to detect somatic mutations and predict neoantigens shared by GC patients. The mutations pattern between our study and the TCGA cohort is comparable, and C > T is the most common substitution. The number of neoantigens was significantly higher in older patients (age ≥60) compared to younger patients (age <60), both in this study and the TCGA cohort. Recurrent neoantigens were found in eight genes (TP53, PIK3CA, PGM5, ERBB3, C6, TRIM49C, OR4C16, and KRAS) in this study. The neoantigen-associated mutations PIK3CA (p.H1047R) and TP53 (p.R175H) are common across several cancer types, indicating their potential usage. Overall, our study illustrates a comprehensive genomic landscape of GC and provides the recurrent neoantigens to facilitate further immunotherapy.


Assuntos
Antígenos de Neoplasias/genética , Classe I de Fosfatidilinositol 3-Quinases/genética , Neoplasias Gástricas/imunologia , Proteína Supressora de Tumor p53/genética , Idoso , Antígenos de Neoplasias/imunologia , Terapia Baseada em Transplante de Células e Tecidos/métodos , Classe I de Fosfatidilinositol 3-Quinases/imunologia , Bases de Dados Genéticas , Exoma/genética , Exoma/imunologia , Feminino , Genoma Humano/genética , Genoma Humano/imunologia , Genômica , Humanos , Imunoterapia/métodos , Masculino , Mutação/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/terapia , Linfócitos T/imunologia , Proteína Supressora de Tumor p53/imunologia
13.
Front Immunol ; 10: 1572, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31333681

RESUMO

V(D)J recombination generates antigen receptor diversity by mixing and matching individual variable (V), diversity (D), and joining (J) gene segments. An obligate by-product of many of these reactions is the excised signal circle (ESC), generated by excision of the DNA from between the gene segments. Initially, the ESC was believed to be inert and formed to protect the genome from reactive broken DNA ends but more recent work suggests that the ESC poses a substantial threat to genome stability. Crucially, the recombinase re-binds to the ESC, which can result in it being re-integrated back into the genome, to cause potentially oncogenic insertion events. In addition, very recently, the ESC/recombinase complex was found to catalyze breaks at recombination signal sequences (RSSs) throughout the genome, via a "cut-and-run" mechanism. Remarkably, the ESC/recombinase complex triggers these breaks at key leukemia driver genes, implying that this reaction could be a significant cause of lymphocyte genome instability. Here, we explore these alternate pathways and discuss their relative dangers to lymphocyte genome stability.


Assuntos
Genoma Humano/imunologia , Instabilidade Genômica/imunologia , Leucemia/imunologia , Recombinação V(D)J/imunologia , Animais , Humanos , Leucemia/genética , Leucemia/patologia
14.
Nutrients ; 11(3)2019 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-30901909

RESUMO

Nutrigenomics studies how environmental factors, such as food intake and lifestyle, influence the expression of the genome. Vitamin D3 represents a master example of nutrigenomics, since via its metabolite 1α,25-dihydroxyvitamin D3, which binds with high-affinity to the vitamin D receptor, the secosteroid directly affects the epigenome and transcriptome at thousands of loci within the human genome. Vitamin D is important for both cellular metabolism and immunity, as it controls calcium homeostasis and modulates the response of the innate and adaptive immune system. At sufficient UV-B exposure, humans can synthesize vitamin D3 endogenously in their skin, but today's lifestyle often makes the molecule a true vitamin and micronutrient that needs to be taken up by diet or supplementation with pills. The individual's molecular response to vitamin D requires personalized supplementation with vitamin D3, in order to obtain optimized clinical benefits in the prevention of osteoporosis, sarcopenia, autoimmune diseases, and possibly different types of cancer. The importance of endogenous synthesis of vitamin D3 created an evolutionary pressure for reduced skin pigmentation, when, during the past 50,000 years, modern humans migrated from Africa towards Asia and Europe. This review will discuss different aspects of how vitamin D interacts with the human genome, focusing on nutritional epigenomics in context of immune responses. This should lead to a better understanding of the clinical benefits of vitamin D.


Assuntos
Colecalciferol/genética , Genoma Humano/imunologia , Sistema Imunitário/metabolismo , Nutrigenômica , Vitamina D/genética , Epigenômica , Humanos , Transcriptoma
15.
Annu Rev Genomics Hum Genet ; 20: 73-97, 2019 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-30848957

RESUMO

Pregnancy presents a singular physiological scenario during which the maternal immune system must accommodate the semiallogeneic fetus. Fluctuations between pro- and anti-inflammatory states are required throughout gestation to facilitate uterine tissue remodeling, fetal growth and development, and finally birth. Tolerance for the fetus must be established and maintained without fundamentally compromising the maternal immune system function, so that both the mother and fetus are protected from foreign insults. Here, we review our current understanding of how genetic variation at both maternal and fetal loci affects implantation and placenta formation, thereby determining the likelihood of a successful pregnancy outcome or the development of pregnancy-related complications. We also consider the impact of pregnancy on both the maternal and fetal systemic immune systems and the related implications for modulating ongoing autoimmune diseases and triggering their development.


Assuntos
Doenças Autoimunes/genética , Feto/imunologia , Genoma Humano/imunologia , Sistema Imunitário/metabolismo , Placenta/imunologia , Complicações na Gravidez/genética , Doenças Autoimunes/imunologia , Doenças Autoimunes/patologia , Feminino , Feto/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Loci Gênicos , Variação Genética , Antígenos HLA/genética , Antígenos HLA/imunologia , Humanos , Sistema Imunitário/crescimento & desenvolvimento , Tolerância Imunológica , Imunogenética/métodos , Placenta/metabolismo , Gravidez , Complicações na Gravidez/imunologia , Complicações na Gravidez/patologia , Receptores KIR/genética , Receptores KIR/imunologia
16.
EMBO Rep ; 19(10)2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30061100

RESUMO

Endogenous retroviruses (ERVs) have accumulated in vertebrate genomes and contribute to the complexity of gene regulation. KAP1 represses ERVs during development by its recruitment to their repetitive sequences through KRAB zinc-finger proteins (KZNFs), but little is known about the regulation of ERVs in adult tissues. We observed that KAP1 repression of HERVK14C was conserved in differentiated human cells and performed KAP1 knockout to obtain an overview of KAP1 function. Our results show that KAP1 represses ERVs (including HERV-T and HERV-S) and ZNF genes, both of which overlap with KAP1 binding sites and H3K9me3 in multiple cell types. Furthermore, this pathway is functionally conserved in adult human peripheral blood mononuclear cells. Cytosine methylation that acts on KAP1 regulated loci is necessary to prevent an interferon response, and KAP1-depletion leads to activation of some interferon-stimulated genes. Finally, loss of KAP1 leads to a decrease in H3K9me3 enrichment at ERVs and ZNF genes and an RNA-sensing response mediated through MAVS signaling. These data indicate that the KAP1-KZNF pathway contributes to genome stability and innate immune control in adult human cells.


Assuntos
Retrovirus Endógenos/genética , Imunidade Inata/genética , Proteínas Repressoras/genética , Proteína 28 com Motivo Tripartido/genética , Sítios de Ligação/genética , Metilação de DNA/genética , Retrovirus Endógenos/imunologia , Retrovirus Endógenos/patogenicidade , Regulação da Expressão Gênica/imunologia , Técnicas de Inativação de Genes , Genoma Humano/imunologia , Histonas/genética , Histonas/imunologia , Humanos , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/virologia , Regiões Promotoras Genéticas
17.
Clin Cancer Res ; 24(15): 3560-3571, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29661778

RESUMO

Purpose: Ovarian cancer is one of the first human cancers for which in situ immune response was reported to be important for the clinical outcome. To elucidate the mechanistic relationship between immune repertoire and cancer genotype in ovarian cancer, the development of a well-defined immune score for ovarian cancer is required.Experimental Design: From a collection of 2,203 patient samples of advanced ovarian cancer from public available resources, we evaluated the prognostic values for a compendium of immune marker genes and proposed an immune score. The relationships between immune score, tumor-infiltrating immune cells, cancer genotypes, and their impact on patient outcome were characterized.Results: Loss of chemokine and IFNγ pathway genes is frequent in ovarian cancer and is significantly associated with low immune score and poor outcome. Chemotherapy can increase the immune score of tumors by inducing the expression of IFNγ inducible chemokines. High immune score is significantly associated with BRCA1/2 mutation status and the response to chemotherapy. Multivariate analysis revealed that immune score is a strong predictor of patient survival and the response to immunotherapy.Conclusions: Our results reveal the drivers of the immune repertoire of advanced ovarian cancer and demonstrate the importance of immune score as an independent prognostic signature and a potent indicator of intratumoral immune status. Clin Cancer Res; 24(15); 3560-71. ©2018 AACR.


Assuntos
Cistadenocarcinoma Seroso/imunologia , Genoma Humano/imunologia , Imunidade Inata/genética , Neoplasias Ovarianas/imunologia , Cistadenocarcinoma Seroso/genética , Cistadenocarcinoma Seroso/patologia , Variações do Número de Cópias de DNA/genética , Variações do Número de Cópias de DNA/imunologia , Feminino , Regulação Neoplásica da Expressão Gênica/imunologia , Genótipo , Humanos , Estimativa de Kaplan-Meier , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Prognóstico
19.
Hum Biol ; 89(1): 47-65, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-29285966

RESUMO

The human immune system is under great pathogen-mediated selective pressure. Divergent infectious disease pathogenesis across human populations combined with the overrepresentation of "immune genes" in genomic regions with signatures of positive selection suggests that pathogens have significantly altered the human genome. However, important features of the human immune system can confound searches for and interpretations of signatures of pathogen-mediated evolution. Immune system redundancy, immune gene pleiotropy, host ability to acquire immunity and alter the immune repertoire of offspring through "priming," and host microbiome complicate evolutionary interpretations of host-pathogen interactions. The overall promiscuity and sensitivity of the immune system to local environments can also muddy assumptions about the origins of a selective pressure on a given set of genes. This review addresses (a) how features of the immune system, the primary buffer between a pathogen and the human genome, affect evolutionary signal and (b) the considerations that must be made when assessing how pathogens have contributed to human diversification.


Assuntos
Doenças Transmissíveis/genética , Doenças Transmissíveis/imunologia , Variação Genética/genética , Genoma Humano/genética , Genoma Humano/imunologia , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Seleção Genética/imunologia , Evolução Molecular , Humanos , Imunidade/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...