Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
J Mol Diagn ; 21(3): 449-461, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31005411

RESUMO

Accurate diagnosis and stratification of children with irritable bowel syndrome (IBS) remain challenging. Given the central role of recurrent abdominal pain in IBS, we evaluated the relationships of pediatric IBS and abdominal pain with intestinal microbes and fecal metabolites using a comprehensive clinical characterization and multiomics strategy. Using rigorous clinical phenotyping, we identified preadolescent children (aged 7 to 12 years) with Rome III IBS (n = 23) and healthy controls (n = 22) and characterized their fecal microbial communities using whole-genome shotgun metagenomics and global unbiased fecal metabolomic profiling. Correlation-based approaches and machine learning algorithms identified associations between microbes, metabolites, and abdominal pain. IBS cases differed from controls with respect to key bacterial taxa (eg, Flavonifractor plautii and Lachnospiraceae bacterium 7_1_58FAA), metagenomic functions (eg, carbohydrate metabolism and amino acid metabolism), and higher-order metabolites (eg, secondary bile acids, sterols, and steroid-like compounds). Significant associations between abdominal pain frequency and severity and intestinal microbial features were identified. A random forest classifier built on metagenomic and metabolic markers successfully distinguished IBS cases from controls (area under the curve, 0.93). Leveraging multiple lines of evidence, intestinal microbes, genes/pathways, and metabolites were associated with IBS, and these features were capable of distinguishing children with IBS from healthy children. These multi-omics features, and their links to childhood IBS coupled with nutritional interventions, may lead to new microbiome-guided diagnostic and therapeutic strategies.


Assuntos
Síndrome do Intestino Irritável/microbiologia , Microbiota , Dor Abdominal/etiologia , Dor Abdominal/microbiologia , Bactérias/genética , Estudos de Casos e Controles , Criança , Fezes/microbiologia , Feminino , Trato Gastrointestinal/microbiologia , Genômica , Humanos , Síndrome do Intestino Irritável/complicações , Masculino , Metaboloma , Análise Multivariada , Análise de Componente Principal , Estatísticas não Paramétricas
2.
Microbiome ; 3: 36, 2015 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-26306392

RESUMO

BACKGROUND: The gut microbiome influences myriad host functions, including nutrient acquisition, immune modulation, brain development, and behavior. Although human gut microbiota are recognized to change as we age, information regarding the structure and function of the gut microbiome during childhood is limited. Using 16S rRNA gene and shotgun metagenomic sequencing, we characterized the structure, function, and variation of the healthy pediatric gut microbiome in a cohort of school-aged, pre-adolescent children (ages 7-12 years). We compared the healthy pediatric gut microbiome with that of healthy adults previously recruited from the same region (Houston, TX, USA). RESULTS: Although healthy children and adults harbored similar numbers of taxa and functional genes, their composition and functional potential differed significantly. Children were enriched in Bifidobacterium spp., Faecalibacterium spp., and members of the Lachnospiraceae, while adults harbored greater abundances of Bacteroides spp. From a functional perspective, significant differences were detected with respect to the relative abundances of genes involved in vitamin synthesis, amino acid degradation, oxidative phosphorylation, and triggering mucosal inflammation. Children's gut communities were enriched in functions which may support ongoing development, while adult communities were enriched in functions associated with inflammation, obesity, and increased risk of adiposity. CONCLUSIONS: Previous studies suggest that the human gut microbiome is relatively stable and adult-like after the first 1 to 3 years of life. Our results suggest that the healthy pediatric gut microbiome harbors compositional and functional qualities that differ from those of healthy adults and that the gut microbiome may undergo a more prolonged development than previously suspected.


Assuntos
Biodiversidade , Microbioma Gastrointestinal , Adulto , Fatores Etários , Criança , Análise por Conglomerados , Código de Barras de DNA Taxonômico , Feminino , Voluntários Saudáveis , Humanos , Masculino , Metagenoma , RNA Ribossômico 16S/genética
5.
Nature ; 466(7305): 503-7, 2010 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-20622853

RESUMO

X-linked mental retardation (XLMR) is a complex human disease that causes intellectual disability. Causal mutations have been found in approximately 90 X-linked genes; however, molecular and biological functions of many of these genetically defined XLMR genes remain unknown. PHF8 (PHD (plant homeo domain) finger protein 8) is a JmjC domain-containing protein and its mutations have been found in patients with XLMR and craniofacial deformities. Here we provide multiple lines of evidence establishing PHF8 as the first mono-methyl histone H4 lysine 20 (H4K20me1) demethylase, with additional activities towards histone H3K9me1 and me2. PHF8 is located around the transcription start sites (TSS) of approximately 7,000 RefSeq genes and in gene bodies and intergenic regions (non-TSS). PHF8 depletion resulted in upregulation of H4K20me1 and H3K9me1 at the TSS and H3K9me2 in the non-TSS sites, respectively, demonstrating differential substrate specificities at different target locations. PHF8 positively regulates gene expression, which is dependent on its H3K4me3-binding PHD and catalytic domains. Importantly, patient mutations significantly compromised PHF8 catalytic function. PHF8 regulates cell survival in the zebrafish brain and jaw development, thus providing a potentially relevant biological context for understanding the clinical symptoms associated with PHF8 patients. Lastly, genetic and molecular evidence supports a model whereby PHF8 regulates zebrafish neuronal cell survival and jaw development in part by directly regulating the expression of the homeodomain transcription factor MSX1/MSXB, which functions downstream of multiple signalling and developmental pathways. Our findings indicate that an imbalance of histone methylation dynamics has a critical role in XLMR.


Assuntos
Encéfalo/embriologia , Encéfalo/enzimologia , Cabeça/embriologia , Histona Desmetilases/metabolismo , Histonas/metabolismo , Fatores de Transcrição/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Animais , Biocatálise , Encéfalo/citologia , Domínio Catalítico , Ciclo Celular , Linhagem Celular Tumoral , Sobrevivência Celular , DNA Intergênico/genética , Regulação da Expressão Gênica , Histona Desmetilases/genética , Histonas/química , Proteínas de Homeodomínio/genética , Humanos , Arcada Osseodentária/citologia , Arcada Osseodentária/embriologia , Lisina/metabolismo , Deficiência Intelectual Ligada ao Cromossomo X/enzimologia , Deficiência Intelectual Ligada ao Cromossomo X/genética , Metilação , Neurônios/citologia , Neurônios/enzimologia , Regiões Promotoras Genéticas , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Sítio de Iniciação de Transcrição , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética
6.
Mol Biol Rep ; 36(6): 1225-9, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18649013

RESUMO

The promoter driving expression of the Mycobacterium tuberculosis gene lipF (Rv3487c) had previously been identified as being upregulated by acidic stress. Subsequently a 59 base pair (bp) acid inducible minimal promoter region was isolated in which a putative -10 region was identified. In this study we use mutational analysis to investigate the -10 region of the lipF promoter. Mutations within this region lead to a dramatic decrease of promoter activity, while a mutation outside of this region does not affect promoter activity.


Assuntos
Análise Mutacional de DNA/métodos , Esterases/genética , Regulação Bacteriana da Expressão Gênica , Mycobacterium tuberculosis/enzimologia , Regiões Promotoras Genéticas , Genes Bacterianos , Mycobacterium tuberculosis/genética
7.
Proc Natl Acad Sci U S A ; 105(41): 15908-13, 2008 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-18838694

RESUMO

Enzyme replacement therapy for lysosomal storage diseases is currently based on endocytosis of lysosomal enzymes via the mannose or mannose 6-phosphate receptors. We are developing a technology for endocytosis of lysosomal enzymes that depends on generic, chemically conjugated reagents. These reagents are aptamers (single-stranded nucleic acid molecules) selected to bind to the extracellular domain of the mouse transferrin receptor. After selection, an RNA aptamer and a DNA aptamer were modified with biotin and linked to dye-labeled streptavidin for detection by confocal microscopy. Aptamer-streptavidin conjugates showed saturable uptake into mouse fibroblasts (Ltk(-) cells), which could be inhibited by an excess of free aptamer but not by tRNA, calf thymus DNA, or transferrin. The RNA aptamer-streptavidin conjugate was mouse-specific, as human cells (293T) did not take it up unless first transfected with the mouse transferrin receptor. Some streptavidin separated from the recycling pathway of transferrin and colocalized with lysosomes. After characterization in the model system, the DNA aptamer was conjugated to a lysosomal enzyme, alpha-l-iduronidase, from which mannose 6-phosphate had been removed. The aptamer had been modified by attachment of terminal glycerol for oxidation by periodate and reaction of the resulting aldehyde with amino groups on the protein. Dephospho-alpha-L-iduronidase-aptamer conjugate was taken up in saturable manner by alpha-L-iduronidase-deficient mouse fibroblasts, with half-maximal uptake estimated as 1.6 nM. Endocytosed enzyme-aptamer conjugate corrected glycosaminoglycan accumulation, indicating that it reached lysosomes and was functional in those organelles. Both uptake and correction were inhibited by unconjugated aptamer, confirming the role of the aptamer in receptor-mediated endocytosis.


Assuntos
Aptâmeros de Nucleotídeos/farmacologia , Endocitose , Enzimas/metabolismo , Lisossomos/enzimologia , Animais , Células Cultivadas , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Iduronidase/metabolismo , Camundongos , Receptores da Transferrina/metabolismo , Especificidade da Espécie , Estreptavidina
8.
Drug Metab Dispos ; 33(5): 623-9, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15673599

RESUMO

Efalizumab (Raptiva) is a humanized monoclonal antibody specific for CD11a, the alpha-chain component of the lymphocyte function-associated antigen 1. In humans, the rate of efalizumab elimination from serum was related to the level of CD11a cell surface expression. These data suggested a role for the CD11a receptor, itself, in efalizumab clearance. Recently, we conducted a series of in vitro studies that suggested a role for CD11a-expressing T cells in efalizumab clearance as mediated by cellular internalization and lysosome-mediated degradation (Coffey et al., 2004). To further study the mechanism of anti-CD11a clearance in vivo, we assessed the tissue distribution, cellular internalization, and subcellular localization of a rat anti-mouse CD11a monoclonal antibody in various tissues in mice. Anti-CD11a antibody primarily distributed to leukocytes and macrophages in the peripheral blood, spleen, and liver, with uptake in the lymph nodes and bone marrow after 72 h. At least a portion of the antibody was internalized and cleared by peripheral blood mononuclear cells, lymphocytes, and splenocytes in a time-dependent manner in vivo. Internalized antibody costained with LysoTracker Red, suggesting that it was transported to lysosomes for degradation. Together, these data suggest that one clearance mechanism for anti-CD11a antibody in vivo is via receptor-mediated internalization and lysosomal degradation by CD11a-expressing cells and tissues.


Assuntos
Anticorpos Monoclonais/farmacocinética , Antígeno CD11a/imunologia , Animais , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais Humanizados , Autorradiografia , Medula Óssea/metabolismo , Citometria de Fluxo , Imuno-Histoquímica , Fígado/metabolismo , Linfonodos/metabolismo , Macrófagos/metabolismo , Masculino , Camundongos , Microscopia Eletrônica de Transmissão , Neutrófilos/metabolismo , Receptores de Antígenos/metabolismo , Baço/citologia , Baço/metabolismo , Frações Subcelulares/metabolismo , Distribuição Tecidual
9.
DNA Cell Biol ; 23(7): 457-62, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15294095

RESUMO

Selection of phage libraries against complex living targets such as whole cells or organs can yield valuable targeting ligands without prior knowledge of the targeted receptor. Our previous studies have shown that noninfective multivalent ligand display phagemids internalize into mammalian cells more efficiently than their monovalent counterparts suggesting that cell-based selection of internalizing ligands might be improved using multivalently displayed peptides, antibodies or cDNAs. However, alternative methods of phage recovery are needed to select phage from noninfective libraries. To this end, we reasoned that rolling circle amplification (RCA) of phage DNA could be used to recover noninfective phage. In feasibility studies, we obtained up to 1.5 million-fold enrichment of internalizing EGF-targeted phage using RCA. When RCA was applied to a large random peptide library, eight distinct human prostate carcinoma cell-internalizing peptides were isolated within three selection rounds. These data establish RCA as an alternative to infection for phage recovery that can be used to identify peptides from noninfective phage display libraries or infective libraries under conditions where there is the potential for loss of phage infectivity.


Assuntos
Bacteriófagos/genética , Biblioteca Gênica , Biologia Molecular/métodos , Carcinoma/genética , Fator de Crescimento Epidérmico/isolamento & purificação , Fator de Crescimento Epidérmico/metabolismo , Humanos , Ligantes , Masculino , Biblioteca de Peptídeos , Neoplasias da Próstata/genética , Células Tumorais Cultivadas
10.
Biotechniques ; 36(6): 1018-22, 1024, 1026-9, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15211753

RESUMO

Phage display technologies are powerful tools for selecting binding ligands against purified molecular targets, live cells, and organ vasculature. However, the selection of natural ligands using phage display has been limited because of significant problems associated with the display of complex cDNA repertoires. Here we describe the use of cDNA fragmentation and open reading frame (ORF) selection to display a human placental cDNA library on the pIII coat protein of filamentous phage. The library was enriched for ORFs by selecting cDNA-beta-lactamase fusion proteins on ampicillin, resulting in a cDNA population having 97% ORFs. The ORF-selected cDNAs were fused to pIII in the phagemid vector, pUCMG4CT-198, and the library was rescued with a pIII-deleted helper phage for multivalent display. The resulting phagemid particle library consisted of 87% ORFs, compared to only 6% ORFs when prepared without ORF selection. Western blot analysis indicated cDNA-pIII fusion protein expression in eight out of nine ORF clones tested, and seven of the ORF encoded peptides were displayed multivalently. The high level of cDNA expression obtained by ORF selection suggests that ORF-enriched phage cDNA libraries prepared by these methods will be useful as functional genomics tools for identifying natural ligands from various source tissues.


Assuntos
DNA Complementar/genética , Biblioteca Gênica , Engenharia Genética/métodos , Fases de Leitura Aberta/genética , Biblioteca de Peptídeos , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...