Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
mSystems ; 9(4): e0104823, 2024 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-38446104

RESUMO

Secondary bacterial challenges during influenza virus infection "superinfection") cause excessive mortality and hospitalization. Here, we present a longitudinal study of bulk gene expression changes in murine lungs during superinfection, with an initial influenza A virus infection and a subsequent Streptococcus pneumoniae infection. In addition to the well-characterized impairment of the host response, we identified superinfection-specific alterations in the global transcriptional program that are linked to the host's ability to resist the pathogens. Particularly, whereas superinfected mice manifested an excessive rapid induction of the resistance-to-infection program, there was a substantial tissue-level rewiring of this program: upon superinfection, interferon-regulated genes were switched from positive to negative correlations with the host's resistance state, whereas genes of fatty acid metabolism switched from negative to positive correlations with resistance states. Thus, the transcriptional resistance state in superinfection is reprogrammed toward repressed interferon signaling and induced fatty acid metabolism. Our findings suggest new insights into a tissue-level remodeling of the host defense upon superinfection, providing promising targets for future therapeutic interventions. IMPORTANCE: Secondary bacterial infections are the most frequent complications during influenza A virus (IAV) pandemic outbreaks, contributing to excessive morbidity and mortality in the human population. Most IAV-related deaths are attributed to Streptococcus pneumoniae (SP) infections, which usually begin within the first week of IAV infection in the respiratory tracts. Here, we focused on longitudinal transcriptional responses during a superinfection model consisting of an SP infection that follows an initial IAV infection, comparing superinfection to an IAV-only infection, an SP-only infection, and control treatments. Our longitudinal data allowed a fine analysis of gene expression changes during superinfection. For instance, we found that superinfected mice exhibited rapid gene expression induction or reduction within the first 12 h after encountering the second pathogen. Cell proliferation and immune response activation processes were upregulated, while endothelial processes, vasculogenesis, and angiogenesis were downregulated, providing promising targets for future therapeutic interventions. We further analyzed the longitudinal transcriptional responses in the context of a previously defined spectrum of the host's resistance state, revealing superinfection-specific reprogramming of resistance states, such as reprogramming of fatty acid metabolism and interferon signaling. The reprogrammed functions are compelling new targets for switching the pathogenic superinfection state into a single-infection state.


Assuntos
Vírus da Influenza A , Influenza Humana , Infecções Pneumocócicas , Superinfecção , Camundongos , Humanos , Animais , Streptococcus pneumoniae , Superinfecção/complicações , Estudos Longitudinais , Influenza Humana/genética , Infecções Pneumocócicas/genética , Imunidade Inata/genética , Interferons , Ácidos Graxos
2.
Cell Syst ; 13(12): 1002-1015.e9, 2022 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-36516834

RESUMO

When challenged with an invading pathogen, the host-defense response is engaged to eliminate the pathogen (resistance) and to maintain health in the presence of the pathogen (disease tolerance). However, the identification of distinct molecular programs underpinning disease tolerance and resistance remained obscure. We exploited transcriptional and physiological monitoring across 33 mouse strains, during in vivo influenza virus infection, to identify two host-defense gene programs-one is associated with hallmarks of disease tolerance and the other with hallmarks of resistance. Both programs constitute generic responses in multiple mouse and human cell types. Our study describes the organizational principles of these programs and validates Arhgdia as a regulator of disease-tolerance states in epithelial cells. We further reveal that the baseline disease-tolerance state in peritoneal macrophages is associated with the pathophysiological response to injury and infection. Our framework provides a paradigm for the understanding of disease tolerance and resistance at the molecular level.


Assuntos
Influenza Humana , Infecções por Orthomyxoviridae , Camundongos , Humanos , Animais , Influenza Humana/genética , Interações Hospedeiro-Patógeno/genética , Infecções por Orthomyxoviridae/genética , Células Epiteliais/metabolismo
3.
Genetics ; 217(4)2021 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-33734353

RESUMO

Recent computational methods have enabled the inference of the cell-type-specificity of eQTLs based on bulk transcriptomes from highly heterogeneous tissues. However, these methods are limited in their scalability to highly heterogeneous tissues and limited in their broad applicability to any cell-type specificity of eQTLs. Here we present and demonstrate Cell Lineage Genetics (CeL-Gen), a novel computational approach that allows inference of eQTLs together with the subsets of cell types in which they have an effect, from bulk transcriptome data. To obtain improved scalability and broader applicability, CeL-Gen takes as input the known cell lineage tree and relies on the observation that dynamic changes in genetic effects occur relatively infrequently during cell differentiation. CeL-Gen can therefore be used not only to tease apart genetic effects derived from different cell types but also to infer the particular differentiation steps in which genetic effects are altered.


Assuntos
Linhagem da Célula , Variação Genética , Estudo de Associação Genômica Ampla/métodos , Animais , Diferenciação Celular , Humanos , Locos de Características Quantitativas , Transcriptoma
4.
Genetics ; 213(1): 297-311, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31352366

RESUMO

Despite the importance of complex phenotypes, an in-depth understanding of the combined molecular and genetic effects on a phenotype has yet to be achieved. Here, we introduce InPhenotype, a novel computational approach for complex phenotype prediction, where gene-expression data and genotyping data are integrated to yield quantitative predictions of complex physiological traits. Unlike existing computational methods, InPhenotype makes it possible to model potential regulatory interactions between gene expression and genomic loci without compromising the continuous nature of the molecular data. We applied InPhenotype to synthetic data, exemplifying its utility for different data parameters, as well as its superiority compared to current methods in both prediction quality and the ability to detect regulatory interactions of genes and genomic loci. Finally, we show that InPhenotype can provide biological insights into both mouse and yeast datasets.


Assuntos
Variação Biológica da População , Estudo de Associação Genômica Ampla/métodos , Fenótipo , Software , Animais , Genótipo , Camundongos , Herança Multifatorial , Leveduras
5.
Nat Methods ; 16(4): 327-332, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30886410

RESUMO

Single-cell RNA sequencing (scRNA-seq) is a rich resource of cellular heterogeneity, opening new avenues in the study of complex tissues. We introduce Cell Population Mapping (CPM), a deconvolution algorithm in which reference scRNA-seq profiles are leveraged to infer the composition of cell types and states from bulk transcriptome data ('scBio' CRAN R-package). Analysis of individual variations in lungs of influenza-virus-infected mice reveals that the relationship between cell abundance and clinical symptoms is a cell-state-specific property that varies gradually along the continuum of cell-activation states. The gradual change is confirmed in subsequent experiments and is further explained by a mathematical model in which clinical outcomes relate to cell-state dynamics along the activation process. Our results demonstrate the power of CPM in reconstructing the continuous spectrum of cell states within heterogeneous tissues.


Assuntos
Biologia Computacional , Genômica , Análise de Sequência de RNA , Análise de Célula Única , Algoritmos , Animais , Separação Celular , Feminino , Fibroblastos/metabolismo , Citometria de Fluxo , Perfilação da Expressão Gênica , Genoma Humano , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Pulmão/virologia , Cadeias de Markov , Camundongos , Camundongos Endogâmicos C57BL , Orthomyxoviridae , Fagócitos/metabolismo , Valores de Referência , Software , Transcriptoma
6.
Cell Syst ; 6(6): 679-691.e4, 2018 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-29886109

RESUMO

The influenza virus is a major cause of morbidity and mortality worldwide. Yet, both the impact of intracellular viral replication and the variation in host response across different cell types remain uncharacterized. Here we used single-cell RNA sequencing to investigate the heterogeneity in the response of lung tissue cells to in vivo influenza infection. Analysis of viral and host transcriptomes in the same single cell enabled us to resolve the cellular heterogeneity of bystander (exposed but uninfected) as compared with infected cells. We reveal that all major immune and non-immune cell types manifest substantial fractions of infected cells, albeit at low viral transcriptome loads relative to epithelial cells. We show that all cell types respond primarily with a robust generic transcriptional response, and we demonstrate novel markers specific for influenza-infected as opposed to bystander cells. These findings open new avenues for targeted therapy aimed exclusively at infected cells.


Assuntos
Interações Hospedeiro-Patógeno/genética , Influenza Humana/genética , Orthomyxoviridae/genética , Animais , Sequência de Bases/genética , Linhagem Celular , Células Epiteliais/imunologia , Feminino , Perfilação da Expressão Gênica/métodos , Interações Hospedeiro-Patógeno/imunologia , Humanos , Vírus da Influenza A Subtipo H1N1/imunologia , Influenza Humana/imunologia , Pulmão/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Orthomyxoviridae/metabolismo , Infecções por Orthomyxoviridae/genética , Análise de Sequência de RNA/métodos , Análise de Célula Única/métodos , Transcriptoma/genética , Replicação Viral
7.
ACS Nano ; 9(2): 1581-91, 2015 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-25558928

RESUMO

Glioblastoma multiforme (GBM) is one of the most infiltrating, aggressive, and poorly treated brain tumors. Progress in genomics and proteomics has paved the way for identifying potential therapeutic targets for treating GBM, yet the vast majority of these leading drug candidates for the treatment of GBM are ineffective, mainly due to restricted passages across the blood-brain barrier. Nanoparticles have been emerged as a promising platform to treat different types of tumors due to their ability to transport drugs to target sites while minimizing adverse effects. Herein, we devised a localized strategy to deliver RNA interference (RNAi) directly to the GBM site using hyaluronan (HA)-grafted lipid-based nanoparticles (LNPs). These LNPs having an ionized lipid were previously shown to be highly effective in delivering small interfering RNAs (siRNAs) into various cell types. LNP's surface was functionalized with hyaluronan (HA), a naturally occurring glycosaminoglycan that specifically binds the CD44 receptor expressed on GBM cells. We found that HA-LNPs can successfully bind to GBM cell lines and primary neurosphers of GBM patients. HA-LNPs loaded with Polo-Like Kinase 1 (PLK1) siRNAs (siPLK1) dramatically reduced the expression of PLK1 mRNA and cumulated in cell death even under shear flow that simulate the flow of the cerebrospinal fluid compared with control groups. Next, a human GBM U87MG orthotopic xenograft model was established by intracranial injection of U87MG cells into nude mice. Convection of Cy3-siRNA entrapped in HA-LNPs was performed, and specific Cy3 uptake was observed in U87MG cells. Moreover, convection of siPLK1 entrapped in HA-LNPs reduced mRNA levels by more than 80% and significantly prolonged survival of treated mice in the orthotopic model. Taken together, our results suggest that RNAi therapeutics could effectively be delivered in a localized manner with HA-coated LNPs and ultimately may become a therapeutic modality for GBM.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Glioblastoma/genética , Glioblastoma/terapia , Ácido Hialurônico/química , Lipídeos/química , Nanopartículas/química , Terapêutica com RNAi/métodos , Animais , Transporte Biológico , Proteínas de Ciclo Celular/deficiência , Morte Celular/genética , Linhagem Celular Tumoral , Transformação Celular Neoplásica , Portadores de Fármacos/química , Portadores de Fármacos/metabolismo , Regulação Neoplásica da Expressão Gênica/genética , Inativação Gênica , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Humanos , Receptores de Hialuronatos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Gradação de Tumores , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Proto-Oncogênicas/deficiência , Quinase 1 Polo-Like
8.
Immunology ; 120(4): 502-11, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17343612

RESUMO

Both the function and regulation of Fas expression in tumours is poorly understood. Our laboratory has reported that cultured, low Fas-expressing tumours undergo massive, yet reversible, up-regulation of cell surface Fas expression when injected into mice. The present study was aimed at determining what causes this enhanced Fas expression and whether the newly expressed Fas functions as a death receptor. Newly expressed Fas is indeed capable of inducing apoptosis. Based on our observation that Fas induction is reduced when tumour cells are injected into immune-deficient mice, we propose that Fas up-regulation in vivo involves the host's immune system. Accordingly, Fas up-regulation occurs in vitro when low Fas-expressing tumour cells are cocultured with lymphoid cells. Furthermore ascitic fluid extracted from tumour-bearing mice trigger Fas up-regulation in low Fas expressing tumours. This last finding suggests that a soluble factor(s) mediates induction of Fas expression. The best candidate for this soluble factor is nitric oxide (NO) based on the following observations: the factor in the ascites is unstable; Fas expression is induced to a lesser degree after injection into inducible NO synthase (NOS)-deficient (iNOS(-/-)) mice when compared to control mice; similarly, coculture with iNOS(-/-) splenocytes induces Fas less effectively than coculture with control splenocytes; and finally, the NO donor SNAP induces considerable Fas up-regulation in tumours in vitro. Our model is that host lymphoid cells in response to a tumour increase NO synthesis, which in turn causes enhanced Fas expression in the tumour.


Assuntos
Antígenos de Neoplasias/biossíntese , Neoplasias Experimentais/imunologia , Regulação para Cima/imunologia , Receptor fas/biossíntese , Animais , Antígenos de Neoplasias/genética , Apoptose , Citotoxicidade Imunológica , Regulação Neoplásica da Expressão Gênica , Camundongos , Camundongos Endogâmicos , Transplante de Neoplasias , Óxido Nítrico/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Baço/citologia , Baço/imunologia , Receptor fas/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA