Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cell ; 182(6): 1441-1459.e21, 2020 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-32888430

RESUMO

Throughout a 24-h period, the small intestine (SI) is exposed to diurnally varying food- and microbiome-derived antigenic burdens but maintains a strict immune homeostasis, which when perturbed in genetically susceptible individuals, may lead to Crohn disease. Herein, we demonstrate that dietary content and rhythmicity regulate the diurnally shifting SI epithelial cell (SIEC) transcriptional landscape through modulation of the SI microbiome. We exemplify this concept with SIEC major histocompatibility complex (MHC) class II, which is diurnally modulated by distinct mucosal-adherent SI commensals, while supporting downstream diurnal activity of intra-epithelial IL-10+ lymphocytes regulating the SI barrier function. Disruption of this diurnally regulated diet-microbiome-MHC class II-IL-10-epithelial barrier axis by circadian clock disarrangement, alterations in feeding time or content, or epithelial-specific MHC class II depletion leads to an extensive microbial product influx, driving Crohn-like enteritis. Collectively, we highlight nutritional features that modulate SI microbiome, immunity, and barrier function and identify dietary, epithelial, and immune checkpoints along this axis to be potentially exploitable in future Crohn disease interventions.


Assuntos
Doença de Crohn/microbiologia , Células Epiteliais/metabolismo , Microbioma Gastrointestinal , Antígenos de Histocompatibilidade Classe II/metabolismo , Intestino Delgado/imunologia , Intestino Delgado/microbiologia , Transcriptoma/genética , Animais , Antibacterianos/farmacologia , Relógios Circadianos/fisiologia , Doença de Crohn/imunologia , Doença de Crohn/metabolismo , Dieta , Células Epiteliais/citologia , Células Epiteliais/imunologia , Citometria de Fluxo , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/genética , Perfilação da Expressão Gênica , Antígenos de Histocompatibilidade Classe II/genética , Homeostase , Hibridização in Situ Fluorescente , Interleucina-10/metabolismo , Interleucina-10/farmacologia , Intestino Delgado/fisiologia , Linfócitos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Periodicidade , Linfócitos T/imunologia , Transcriptoma/fisiologia
2.
J Pediatr Gastroenterol Nutr ; 69(6): 633-638, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31765333

RESUMO

The human genome has been proposed to contribute to interpersonal variability in the way we respond to nutritional intake. However, personalized diets solely based on gene-nutrient interactions have not lived up to their expectations to date. Advances in microbiome research have indicated that a science-based generation of a personalized diet based on a combination of clinical and microbial features may constitute a promising new approach enabling accurate prediction of dietary responses. In addition, scientific advances in our understanding of defined dietary components and their effects on human physiology led to the incorporation and testing of defined diets as preventive and treatment approaches for diseases, such as epilepsy, ulcerative colitis, Crohn disease, and type 1 diabetes mellitus. Additionally, exciting new studies show that tailored diet regiments have the potential to modulate pharmaceutical treatment efficacy in cancer treatment. Overall, the true therapeutic potential of nutritional interventions is coming to light but is also facing substantial challenges in understanding mechanisms of activity, optimization of dietary interventions for specific human subpopulations, and elucidation of adverse effects potentially stemming from some dietary components in a number of individuals.


Assuntos
Microbiota , Nutrigenômica/métodos , Medicina de Precisão/métodos , Diabetes Mellitus Tipo 1/dietoterapia , Diabetes Mellitus Tipo 1/microbiologia , Epilepsia/dietoterapia , Epilepsia/microbiologia , Humanos , Doenças Inflamatórias Intestinais/dietoterapia , Doenças Inflamatórias Intestinais/microbiologia , Neoplasias/dietoterapia , Neoplasias/microbiologia , Terapia Nutricional/métodos , Obesidade Infantil/dietoterapia , Obesidade Infantil/microbiologia
3.
Semin Immunol ; 32: 74-81, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28431920

RESUMO

The commensal microbiome constitutes an important modulator of host physiology and risk of disease, including cancer development and progression. Lately, the microbiome has been suggested to modulate the efficacy of anti-cancer treatment. Examples include chemotherapy and total body irradiation-induced barrier function disruption, leading to microbial efflux that drives activation of anti-tumorigenic T cells; Microbiome-driven release of reactive oxygen species contributing to the efficacy of platinum salts; and microbiome-induced immune priming promoting the anti-tumor effects of alkylating chemotherapy and immune checkpoint inhibitors. Furthermore, selected commensals are able to colonize solid tumors. This 'tumor microbiome' may further impact local tumor responses to treatment and potentially be harnessed for tumor-specific targeting and therapeutic delivery. In this review, we present recent advances in understanding of the intricate role of microbiome in modulating efficacy of a number of anti-cancer treatments, and discuss how anti-cancer treatment approaches utilizing the tumor microbiome may enhance oncological treatment efficacy.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antineoplásicos Alquilantes/uso terapêutico , Imunomodulação , Imunoterapia/métodos , Microbiota/imunologia , Neoplasias/microbiologia , Neoplasias/terapia , Animais , Carcinogênese , Receptores Coestimuladores e Inibidores de Linfócitos T/imunologia , Humanos , Neoplasias/imunologia , Simbiose , Resultado do Tratamento
4.
Cell Metab ; 25(3): 506-521, 2017 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-28273474

RESUMO

In addition to the immune system's traditional roles of conferring anti-infectious and anti-neoplastic protection, it has been recently implicated in the regulation of systemic metabolic homeostasis. This cross-talk between the immune and the metabolic systems is pivotal in promoting "metabolic health" throughout the life of an organism and plays fundamental roles in its adaptation to ever-changing environmental makeups and nutritional availability. Perturbations in this intricate immune-metabolic cross-talk contribute to the tendency to develop altered metabolic states that may culminate in metabolic disorders such as malnutrition, obesity, type 2 diabetes mellitus (T2DM), and other features of the metabolic syndrome. Regulators of immune-metabolic interactions include host genetics, nutritional status, and the intestinal microbiome. In this Perspective, we highlight current understanding of immune-metabolism interactions, illustrate differences among individuals and between populations in this respect, and point toward future avenues of research possibly enabling immune harnessing as means of personalized treatment for common metabolic disorders.


Assuntos
Saúde , Doenças Metabólicas/imunologia , Envelhecimento/fisiologia , Animais , Humanos , Doenças Metabólicas/genética , Microbiota , Modelos Biológicos , Pesquisa Translacional Biomédica
5.
Mol Metab ; 5(9): 782-94, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27617201

RESUMO

BACKGROUND: Non-alcoholic fatty liver (NAFLD) is a common, multi-factorial, and poorly understood liver disease whose incidence is globally rising. NAFLD is generally asymptomatic and associated with other manifestations of the metabolic syndrome. Yet, up to 25% of NAFLD patients develop a progressive inflammatory liver disease termed non-alcoholic steatohepatitis (NASH) that may progress towards cirrhosis, hepatocellular carcinoma, and the need for liver transplantation. In recent years, several lines of evidence suggest that the gut microbiome represents a significant environmental factor contributing to NAFLD development and its progression into NASH. Suggested microbiome-associated mechanisms contributing to NAFLD and NASH include dysbiosis-induced deregulation of the gut endothelial barrier function, which facilitates systemic bacterial translocation, and intestinal and hepatic inflammation. Furthermore, increased microbiome-modulated metabolites such as lipopolysaccharides, short chain fatty acids (SCFAs), bile acids, and ethanol, may affect liver pathology through multiple direct and indirect mechanisms. SCOPE OF REVIEW: Herein, we discuss the associations, mechanisms, and clinical implications of the microbiome's contribution to NAFLD and NASH. Understanding these contributions to the development of fatty liver pathogenesis and its clinical course may serve as a basis for development of therapeutic microbiome-targeting approaches for treatment and prevention of NAFLD and NASH. MAJOR CONCLUSIONS: Intestinal host-microbiome interactions play diverse roles in the pathogenesis and progression of NAFLD and NASH. Elucidation of the mechanisms driving these microbial effects on the pathogenesis of NAFLD and NASH may enable to identify new diagnostic and therapeutic targets of these common metabolic liver diseases. This article is part of a special issue on microbiota.

6.
BMC Med ; 14(1): 83, 2016 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-27256449

RESUMO

HIV/AIDS causes severe dysfunction of the immune system through CD4+ T cell depletion, leading to dysregulation of both the adaptive and innate immune arms. A primary target for viral infection is the gastrointestinal tract, which is a reservoir of CD4+ T cells. In addition to being a major immune hub, the human gastrointestinal tract harbors trillions of commensal microorganisms, the microbiota, which have recently been shown to play critical roles in health. Alterations in the composition and function of microbiota have been implicated in a variety of 'multi-factorial' disorders, including infectious, autoimmune, metabolic, and neoplastic disorders. It is widely accepted that, in addition to its direct role in altering the gastrointestinal CD4+ T cell compartment, HIV infection is characterized by gut microbiota compositional and functional changes. Herein, we review such alterations and discuss their potential local and systemic effects on the HIV-positive host, as well as potential roles of novel microbiota-targeting treatments in modulating HIV progression and associated adverse systemic manifestations.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Microbioma Gastrointestinal/imunologia , Infecções por HIV/microbiologia , Linfócitos T CD4-Positivos/microbiologia , Infecções por HIV/virologia , Humanos
7.
J Gene Med ; 16(11-12): 364-73, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25394283

RESUMO

BACKGROUND: Most leukodystrophies result from mutations in genes expressed in oligodendrocytes that may cause autonomous loss of function of cell structural proteins. Therefore, effective gene delivery to oligodendrocytes is necessary to develop future treatments. MATERIALS: To achieve this, we cloned a lentiviral vector in which the enhanced green fluorescent protein (EGFP) expression was driven by the oligodendrocyte specific 2,3-cyclic nucleotide 3-phosphodiesterase promoter. The vector was inserted into C57BL/6 neonatal mouse brain by combined intraventricular and parenchymal injections. RESULTS: Assessment of EGFP expression revealed a widespread distribution, specifically in cells of the oligodendrocyte linage, starting from postnatal day 6 (P6) in the subventricular zone and spreading through migrating oligodendrocyte precursors. By P30, it was detectable throughout the brain and persisted for at least 3 months, showing an increase both in the number of expressing cells and in intensity over time. EGFP expression was restricted to oligodendrocyte linage cells. On average, 20.3 ± 2.56% of all oligodendrocytes in different central nervous system areas were EGFP-positive, with regional variations. CONCLUSIONS: Lentiviral gene delivery using an oligodendrocyte-specific promoter may achieve widespread and long-lasting expression selectively in oligodendrocytes, offering a possibility for gene therapy in certain leukodystrophies, although the relatively low rates of oligodendrocyte transduction are a limitation that remains to be overcome.


Assuntos
Terapia Genética , Lentivirus/genética , Oligodendroglia/metabolismo , Animais , Encéfalo/metabolismo , Técnicas de Cocultura , Feminino , Expressão Gênica , Vetores Genéticos , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Masculino , Camundongos Endogâmicos C57BL , Células PC12 , Ratos , Nervo Isquiático/metabolismo
8.
J Infect Dev Ctries ; 7(9): 642-50, 2013 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-24042099

RESUMO

INTRODUCTION: The prevalence of H. pylori varies with geographic locations. To date there are no epidemiological data on its prevalence in Cyprus; therefore, we determined the prevalence and molecular characteristics of H. pylori infection in Cypriot patients. METHODOLOGY: DNA extracted from 103 gastric biopsies was analyzed for the presence of H. pylori by PCR using primers for ureA. H. pylori-positive biopsies were characterized by PCR using specific primers for cagA and vacA genes. The presence of clarithromycin-associated resistant mutations such as A2143G, A2142G, A2142C in 23S rRNA gene of H. pylori-positive patients was determined using a real-time PCR allelic discrimination assay. RESULTS: H. pylori was detected in 41 (39.8%) biopsies and, out of these, 17 (41.5%) tested positive for the cagA gene. The vacA alleles m1, m2, s1a, s1b, and s2 were detected in 7 (17.1%), 34 (82.9%), 12 (29.3%), 2 (4.9%), and 22 (53.7%) isolates, respectively. One (2.4%) biopsy was vacA s1a and s2-positive while one (2.4%) was positive for vacA s1a, s1b, and s2. Three (7.3%) biopsies were untypable for vacA s1, s1b, and s2. The majority (35; 85.4%) of strains were susceptible to clarithromycin while two (4.9%) had the A2143G mutation. Three (7.3%) had a mixture of an A2143G point mutant and susceptible strains while one (2.4%) had a mixture of an A2142G point mutant and susceptible strains. CONCLUSIONS: The distribution of the virulence factors cagA and vacA in the Cypriot strains resembled that of strains circulating in Middle Eastern countries geographically close to Cyprus.


Assuntos
Antígenos de Bactérias/genética , Proteínas de Bactérias/genética , Infecções por Helicobacter/epidemiologia , Infecções por Helicobacter/microbiologia , Helicobacter pylori/genética , Fatores de Virulência/genética , Adulto , Antibacterianos/farmacologia , Biópsia , Claritromicina/farmacologia , Chipre/epidemiologia , DNA Bacteriano/genética , DNA Bacteriano/isolamento & purificação , Farmacorresistência Bacteriana , Mucosa Gástrica/microbiologia , Helicobacter pylori/isolamento & purificação , Humanos , Filogeografia , Mutação Puntual , Prevalência , RNA Ribossômico 23S/genética , Urease/genética
9.
PLoS One ; 2(6): e525, 2007 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-17565378

RESUMO

Loss of inner ear sensory hair cells (HC) is a leading cause of human hearing loss and balance disorders. Unlike mammals, many lower vertebrates can regenerate these cells. We used cross-species microarrays to examine this process in the avian inner ear. Specifically, changes in expression of over 1700 transcription factor (TF) genes were investigated in hair cells of auditory and vestibular organs following treatment with two different damaging agents and regeneration in vitro. Multiple components of seven distinct known signaling pathways were clearly identifiable: TGFbeta, PAX, NOTCH, WNT, NFKappaB, INSULIN/IGF1 and AP1. Numerous components of apoptotic and cell cycle control pathways were differentially expressed, including p27(KIP) and TFs that regulate its expression. A comparison of expression trends across tissues and treatments revealed identical patterns of expression that occurred at identical times during regenerative proliferation. Network analysis of the patterns of gene expression in this large dataset also revealed the additional presence of many components (and possible network interactions) of estrogen receptor signaling, circadian rhythm genes and parts of the polycomb complex (among others). Equal numbers of differentially expressed genes were identified that have not yet been placed into any known pathway. Specific time points and tissues also exhibited interesting differences: For example, 45 zinc finger genes were specifically up-regulated at later stages of cochlear regeneration. These results are the first of their kind and should provide the starting point for more detailed investigations of the role of these many pathways in HC recovery, and for a description of their possible interactions.


Assuntos
Biomarcadores/metabolismo , Galinhas/genética , Células Epiteliais/fisiologia , Perfilação da Expressão Gênica , Células Ciliadas Auditivas Internas/fisiologia , Regeneração/fisiologia , Animais , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
10.
Hum Genet ; 115(1): 81-9, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15088139

RESUMO

Idiopathic scoliosis (IS) affects approximately 2%-3% of the population and has a heritable component. The genetics of this disorder are complex. Here, we describe a family in which a pericentric inversion of chromosome 8 co-segregates with IS. We have used fluorescence in situ hybridization to identify cloned DNAs that span the breakpoints on the two arms of the chromosome. We have identified a bacterial artificial chromosome (BAC) of 150 kb that crosses the q-arm breakpoint and a BAC of 120 kb that crosses the p-arm breakpoint. The complete genomic DNA sequence of these BACs has been analyzed to identify candidate genes and to localize further the precise breakpoints. This has revealed that the p-arm break does not interrupt any known gene and occurs in a region of highly repetitive sequence elements. On the q-arm, the break occurs between exons 10 and 11 of the gamma-1 syntrophin (SNTG1) gene. Syntrophins are a group of cytoplasmic peripheral membrane proteins that associate directly with dystrophin, the Duchenne muscular dystrophy gene; gamma1-syntrophin has been shown to be a neuronal cell-specific protein. Mutational analysis of SNTG1 exons in 152 sporadic IS patients has revealed a 6-bp deletion in exon 10 of SNTG1 in one patient and a 2-bp insertion/deletion mutation occurring in a polypyrimidine tract of intronic sequence 20 bases upstream of the SNTG1 exon 5 splice site in two patients. These changes were not seen in a screen of 480 control chromosomes. Genomic DNAs from seven affected individuals within the family of a patient carrying the 6-bp deletion were typed to determine whether the alteration co-segregated with IS. The deletion was only observed in five out of these seven individuals. Thus, although genetic heterogeneity or multiple alleles cannot be ruled out, the 6-bp deletion does not consistently co-segregate with the disease in this family.


Assuntos
Deleção de Genes , Proteínas do Tecido Nervoso/genética , Escoliose/genética , Proteínas Adaptadoras de Transdução de Sinal , Sequência de Bases , Inversão Cromossômica , Segregação de Cromossomos , Cromossomos Artificiais Bacterianos , Cromossomos Humanos Par 8 , Humanos , Hibridização in Situ Fluorescente , Masculino , Proteínas de Membrana , Linhagem , Mapeamento Físico do Cromossomo
11.
Hum Mol Genet ; 12(11): 1261-72, 2003 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-12761041

RESUMO

The sensory receptors for hearing and balance are the hair cells of the cochlea and vestibular organs of the inner ear. Permanent hearing and balance deficits can be triggered by genetic susceptibilities or environmental factors such as infection. Unlike mammalian hair cells that have a limited capacity for regeneration, the vestibular organ of the avian ear is constantly undergoing hair cell regeneration, whereas the avian cochlea undergoes regeneration only when hair cells are damaged. In order to gain insights into the genetic programs that govern the regenerative capacity of hair cells, we interrogated custom human cDNA microarrays with sensory epithelial cell targets from avian inner ears. The arrays contained probes from conserved regions of approximately 400 genes expressed primarily in the inner ear and approximately 1500 transcription factors (TF). Highly significant differences were observed for 20 inner-ear genes and more than 80 TFs. Genes up-regulated in the cochlea included BMP4, GATA3, GSN, FOXF1 and PRDM7. Genes up-regulated in the utricle included SMAD2, KIT, beta-AMYLOID, LOC51637, HMG20B and CRIP2. Many of the highly significant changes were validated by Q-PCR and in situ methods. Some of the observed changes implicated a number of known biochemical pathways including the c-kit pathway previously observed in melanogenesis. Twenty differentially expressed TFs map to chromosomal regions harboring uncloned human deafness loci, and represent novel candidates for hearing loss. The approach described here also illustrates the power of utilizing conserved human cDNA probes for cross-species comparisons.


Assuntos
Galinhas/genética , Células Epiteliais/fisiologia , Expressão Gênica , Células Ciliadas Auditivas/fisiologia , Fatores de Transcrição/genética , Animais , DNA Complementar , Orelha Interna/fisiologia , Biblioteca Gênica , Transtornos da Audição/genética , Humanos , Imuno-Histoquímica/métodos , Hibridização In Situ/métodos , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Reação em Cadeia da Polimerase/métodos , Regeneração/genética , Transtornos de Sensação/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA