Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
ACR Open Rheumatol ; 4(5): 417-425, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35174673

RESUMO

OBJECTIVE: The study objective was to examine alterations in gastrointestinal (GI) microbial composition in patients with systemic sclerosis (SSc) and to investigate the relationship between SSc features and GI microbiota using two independent, international cohorts. METHODS: Prospective patients with SSc from Lund University (LU), Sweden, from the University of California, Los Angeles (UCLA), United States, and control subjects provided stool specimens for 16S ribosomal RNA sequencing. Alpha and beta diversity analyses were performed. Multivariate negative binomial models identified differentially abundant genera between groups. RESULTS: Patients from LU with SSc (n = 106) with recent SSc diagnosis (median disease duration 2.0 years) had lower abundance of commensal genera (eg, Faecalibacterium) and higher abundance of pathobiont genera (eg, Desulfovibrio) than LU-controls (n = 85). Patients from UCLA with SSc (n = 71) had a similar prevalence of females, a similar body mass index, and similar age but an increased disease duration (median 7.1 years) compared with patients from LU with SSc. Factors associated with beta diversity in patients with SSc from both LU and UCLA included disease duration (P = 0.0016), interstitial lung disease (P = 0.003), small intestinal bacterial overgrowth (P = 0.002), and immunosuppression use (P = 0.014). In multivariable analysis, the UCLA-SSc cohort had higher abundance of specific pathobiont genera (eg, Streptococcus) compared with the LU-SSc cohort. CONCLUSION: Enrichments and depletions in certain microbial genera were observed in patients recently diagnosed with SSc, suggesting that dysbiosis is present in early SSc. Specific disease features were independently associated with fecal microbial composition in both cohorts. After controlling for these factors, the abundance of several pathobiont bacteria differed between the cohorts, suggesting that environmental factors, along with disease manifestations, should be considered in future SSc studies.

2.
Int Psychogeriatr ; 34(1): 33-45, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-33757609

RESUMO

OBJECTIVES: (1) To investigate if gut microbiota can be a predictor of remission in geriatric depression and to identify features of the gut microbiota that is associated with remission. (2) To determine if changes in gut microbiota occur with remission in geriatric depression. DESIGN: Secondary analysis of a parent randomized placebo-controlled trial (NCT02466958). SETTING: Los Angeles, CA, USA (2016-2018). PARTICIPANTS: Seventeen subjects with major depressive disorder, over 60 years of age, 41.2% female. INTERVENTION: Levomilacipran (LVM) or placebo. MEASUREMENTS: Remission was defined by Hamilton Depression Rating Scale score of 6 or less at 12 weeks. 16S-ribosomal RNA sequencing based fecal microbiota composition and diversity were measured at baseline and 12 weeks. Differences in fecal microbiota were evaluated between remitters and non-remitters as well as between baseline and post-treatment samples. LVM and placebo groups were combined in all the analyses. RESULTS: Baseline microbiota showed no community level α-diversity or ß-diversity differences between remitters and non-remitters. At the individual taxa level, a random forest classifier created with nine genera from the baseline microbiota was highly accurate in predicting remission (AUC = .857). Of these, baseline enrichment of Faecalibacterium, Agathobacter and Roseburia relative to a reference frame was associated with treatment outcome of remission. Differential abundance analysis revealed significant genus level changes from baseline to post-treatment in remitters, but not in non-remitters. CONCLUSIONS: This is the first study demonstrating fecal microbiota as a potential predictor of treatment response in geriatric depression. Our findings need to be confirmed in larger prospective studies.


Assuntos
Transtorno Depressivo Maior , Microbioma Gastrointestinal , Idoso , Antidepressivos/uso terapêutico , Depressão , Transtorno Depressivo Maior/tratamento farmacológico , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Projetos Piloto , Estudos Prospectivos , Resultado do Tratamento
3.
J Clin Endocrinol Metab ; 106(12): 3464-3476, 2021 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-34343292

RESUMO

CONTEXT: Experimental studies suggest that vitamin D receptor signaling may benefit the gut microbiome. In humans, whether vitamin D supplementation directly alters the gut microbiome is not well studied. OBJECTIVE: To determine whether correcting vitamin D deficiency with cholecalciferol (vitamin D3, D3) or calcifediol (25-hydroxyvitamin D3, 25(OH)D3) changes gut microbiome composition. METHODS: 18 adults with vitamin D deficiency (25-hydroxyvitamin D [25(OH)D] <20 ng/mL) received 60 µg/day of D3 or 20 µg/day of 25(OH)D3 for 8 weeks. Changes in serum 25(OH)D, 1,25-diydroxyvitamin D (1,25(OH)2D), and 24,25-dihydroxyvitamin D (24,25(OH)2D) were assessed. We characterized composition of the fecal microbiota using 16S rRNA gene sequencing, and examined changes in α-diversity (Chao 1, Faith's Phylogenetic Diversity, Shannon Index), ß-diversity (DEICODE), and genus-level abundances (DESeq2). RESULTS: Vitamin D3 and 25(OH)D3 groups were similar. After 8 weeks of vitamin D3, mean 25(OH)D and 24,25(OH)2D increased significantly, but 1,25(OH)2D did not (25(OH)D: 17.8-30.1 ng/mL, P = .002; 24,25(OH)2D: 1.1 to 2.7 ng/mL, P =0.003; 1,25(OH)2D: 49.5-53.0 pg/mL, P = .9). After 8 weeks of 25(OH)D3, mean 25(OH)D, 24,25(OH)2D, and 1,25(OH)2D increased significantly (25(OH)D: 16.7-50.6 ng/mL, P < .0001; 24,25(OH)2D: 1.3-6.2 ng/mL, P = .0001; 1,25(OH)2D: 56.5-74.2 pg/mL, P = .05). Fecal microbial α-diversity and ß-diversity did not change with D3 or 25D3 supplementation. Mean relative abundance of Firmicutes increased and mean relative abundance of Bacterioidetes decreased from baseline to 4 weeks, but returned to baseline by study completion. DESeq2 analysis did not confirm any statistically significant taxonomic changes. CONCLUSION: In a small sample of healthy adults with vitamin D deficiency, restoration of vitamin D sufficiency with vitamin D3 or 25(OH)D3 did not lead to lasting changes in the fecal microbiota.


Assuntos
Biomarcadores/sangue , Calcifediol/administração & dosagem , Colecalciferol/administração & dosagem , Fezes/microbiologia , Microbioma Gastrointestinal , Deficiência de Vitamina D/microbiologia , Vitaminas/administração & dosagem , Adolescente , Adulto , Suplementos Nutricionais , Feminino , Seguimentos , Humanos , Masculino , Projetos Piloto , Prognóstico , Deficiência de Vitamina D/sangue , Deficiência de Vitamina D/tratamento farmacológico , Deficiência de Vitamina D/patologia , Adulto Jovem
4.
Dev Psychopathol ; : 1-12, 2021 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-34108055

RESUMO

Perturbations to the gut microbiome are implicated in altered neurodevelopmental trajectories that may shape life span risk for emotion dysregulation and affective disorders. However, the sensitive periods during which the microbiome may influence neurodevelopment remain understudied. We investigated relationships between gut microbiome composition across infancy and temperament at 12 months of age. In 67 infants, we examined if gut microbiome composition assessed at 1-3 weeks, 2, 6, and 12 months of age was associated with temperament at age 12 months. Stool samples were sequenced using the 16S Illumina MiSeq platform. Temperament was assessed using the Infant Behavior Questionnaire-Revised (IBQ-R). Beta diversity at age 1-3 weeks was associated with surgency/extraversion at age 12 months. Bifidobacterium and Lachnospiraceae abundance at 1-3 weeks of age was positively associated with surgency/extraversion at age 12 months. Klebsiella abundance at 1-3 weeks was negatively associated with surgency/extraversion at 12 months. Concurrent composition was associated with negative affectivity at 12 months, including a positive association with Ruminococcus-1 and a negative association with Lactobacillus. Our findings support a relationship between gut microbiome composition and infant temperament. While exploratory due to the small sample size, these results point to early and late infancy as sensitive periods during which the gut microbiome may exert effects on neurodevelopment.

5.
World J Biol Psychiatry ; 21(8): 634-641, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-31552779

RESUMO

Objectives: Circulating cytokines have been associated with depression, but their detection has limitations, which may be overcome by direct detection of intracellular cytokines (ICCs) after lipopolysaccharide (LPS) stimulation in vitro. This study compared circulating versus LPS-induced inflammatory markers as correlates of subthreshold depressive symptoms.Methods: Secondary data analysis of a cross-sectional insomnia study in healthy community-dwelling older adults was conducted. In 117 participants (≥55 years), plasma tumour necrosis factor-α (TNF-α), interleukin-6 (IL-6), C-reactive protein (CRP) and in vitro LPS-induced monocyte production of IL-6 and TNF-α were assayed. Depressive symptoms were assessed using the clinician-rated Inventory of Depressive Symptomatology (IDS-C). Multivariate linear regression was conducted to test the associations between inflammatory markers and subthreshold depressive symptoms in the entire sample as well as in subgroups stratified into higher and lower inflammation levels.Results: LPS-induced TNF-α (adjusted ß = 0.28, p = .04), IL-6 (0.29, p = .03) and TNF-α + IL-6 (0.43, p = .001) significantly positively correlated with subthreshold depressive symptoms only in higher inflammation subgroups. No circulating biomarkers positively correlated in any subgroups. In the entire sample, no biomarkers were significantly associated with subthreshold depressive symptoms.Conclusions: LPS-induced cytokines may be more sensitive correlates of subthreshold depressive symptoms than circulating cytokines, particularly in older adults with higher systemic inflammation.Clinical Trials Registry: ClinicalTrials.gov NCT00280020.


Assuntos
Citocinas/sangue , Citocinas/metabolismo , Depressão/sangue , Depressão/metabolismo , Inflamação/sangue , Inflamação/imunologia , Espaço Intracelular/metabolismo , Lipopolissacarídeos/imunologia , Idoso , Idoso de 80 Anos ou mais , Biomarcadores/sangue , Biomarcadores/metabolismo , Proteína C-Reativa/análise , Proteína C-Reativa/metabolismo , Estudos Transversais , Citocinas/imunologia , Depressão/complicações , Depressão/imunologia , Feminino , Humanos , Inflamação/complicações , Interleucina-6/sangue , Interleucina-6/metabolismo , Espaço Intracelular/imunologia , Masculino , Pessoa de Meia-Idade , Fator de Necrose Tumoral alfa/sangue , Fator de Necrose Tumoral alfa/imunologia
6.
Development ; 143(19): 3632-3637, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27702788

RESUMO

In situ hybridization methods are used across the biological sciences to map mRNA expression within intact specimens. Multiplexed experiments, in which multiple target mRNAs are mapped in a single sample, are essential for studying regulatory interactions, but remain cumbersome in most model organisms. Programmable in situ amplifiers based on the mechanism of hybridization chain reaction (HCR) overcome this longstanding challenge by operating independently within a sample, enabling multiplexed experiments to be performed with an experimental timeline independent of the number of target mRNAs. To assist biologists working across a broad spectrum of organisms, we demonstrate multiplexed in situ HCR in diverse imaging settings: bacteria, whole-mount nematode larvae, whole-mount fruit fly embryos, whole-mount sea urchin embryos, whole-mount zebrafish larvae, whole-mount chicken embryos, whole-mount mouse embryos and formalin-fixed paraffin-embedded human tissue sections. In addition to straightforward multiplexing, in situ HCR enables deep sample penetration, high contrast and subcellular resolution, providing an incisive tool for the study of interlaced and overlapping expression patterns, with implications for research communities across the biological sciences.


Assuntos
Hibridização In Situ/métodos , RNA Mensageiro/metabolismo , Animais , Drosophila , Embrião não Mamífero/metabolismo , Humanos , Peixe-Zebra
7.
Nat Rev Microbiol ; 14(1): 20-32, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26499895

RESUMO

Animals assemble and maintain a diverse but host-specific gut microbial community. In addition to characteristic microbial compositions along the longitudinal axis of the intestines, discrete bacterial communities form in microhabitats, such as the gut lumen, colonic mucus layers and colonic crypts. In this Review, we examine how the spatial distribution of symbiotic bacteria among physical niches in the gut affects the development and maintenance of a resilient microbial ecosystem. We consider novel hypotheses for how nutrient selection, immune activation and other mechanisms control the biogeography of bacteria in the gut, and we discuss the relevance of this spatial heterogeneity to health and disease.


Assuntos
Ecossistema , Microbioma Gastrointestinal , Trato Gastrointestinal/microbiologia , Animais , Interações Hospedeiro-Patógeno , Humanos , Simbiose
8.
Nature ; 501(7467): 426-9, 2013 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-23955152

RESUMO

Mammals harbour a complex gut microbiome, comprising bacteria that confer immunological, metabolic and neurological benefits. Despite advances in sequence-based microbial profiling and myriad studies defining microbiome composition during health and disease, little is known about the molecular processes used by symbiotic bacteria to stably colonize the gastrointestinal tract. We sought to define how mammals assemble and maintain the Bacteroides, one of the most numerically prominent genera of the human microbiome. Here we find that, whereas the gut normally contains hundreds of bacterial species, germ-free mice mono-associated with a single Bacteroides species are resistant to colonization by the same, but not different, species. To identify bacterial mechanisms for species-specific saturable colonization, we devised an in vivo genetic screen and discovered a unique class of polysaccharide utilization loci that is conserved among intestinal Bacteroides. We named this genetic locus the commensal colonization factors (ccf). Deletion of the ccf genes in the model symbiont, Bacteroides fragilis, results in colonization defects in mice and reduced horizontal transmission. The ccf genes of B. fragilis are upregulated during gut colonization, preferentially at the colonic surface. When we visualize microbial biogeography within the colon, B. fragilis penetrates the colonic mucus and resides deep within crypt channels, whereas ccf mutants are defective in crypt association. Notably, the CCF system is required for B. fragilis colonization following microbiome disruption with Citrobacter rodentium infection or antibiotic treatment, suggesting that the niche within colonic crypts represents a reservoir for bacteria to maintain long-term colonization. These findings reveal that intestinal Bacteroides have evolved species-specific physical interactions with the host that mediate stable and resilient gut colonization, and the CCF system represents a novel molecular mechanism for symbiosis.


Assuntos
Bacteroides/classificação , Bacteroides/fisiologia , Trato Gastrointestinal/microbiologia , Metagenoma/fisiologia , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Bacteroides/genética , Bacteroides/crescimento & desenvolvimento , Bacteroides fragilis/genética , Bacteroides fragilis/crescimento & desenvolvimento , Bacteroides fragilis/metabolismo , Colo/microbiologia , Sequência Conservada/genética , Evolução Molecular , Feminino , Deleção de Genes , Genes Bacterianos/genética , Vida Livre de Germes , Mucosa Intestinal/microbiologia , Masculino , Camundongos , Polissacarídeos/metabolismo , Especificidade da Espécie , Simbiose/genética
9.
Anaerobe ; 17(4): 137-41, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21664470

RESUMO

The mammalian gastrointestinal tract harbors a vast microbial ecosystem, known as the microbiota, which benefits host biology. Bacteroides fragilis is an important anaerobic gut commensal of humans that prevents and cures intestinal inflammation. We wished to elucidate aspects of gut colonization employed by B. fragilis. Fluorescence in situ hybridization was performed on colonic tissue sections from B. fragilis and Escherichia coli dual-colonized gnotobiotic mice. Epifluorescence imaging reveals that both E. coli and B. fragilis are found in the lumen of the colon, but only B. fragilis is found in the mucosal layer. This observation suggests that physical association with intestinal mucus could be a possible mechanism of gut colonization by B. fragilis. We investigated this potential interaction using an in vitro mucus binding assay and show here that B. fragilis binds to murine colonic mucus. We further demonstrate that B. fragilis specifically and quantitatively binds to highly purified mucins (the major constituent in intestinal mucus) using flow cytometry analysis of fluorescently labeled purified murine and porcine mucins. These results suggest that interactions between B. fragilis and intestinal mucin may play a critical role during host-bacterial symbiosis.


Assuntos
Bacteroides fragilis/metabolismo , Colo/microbiologia , Mucosa Intestinal/microbiologia , Mucinas/metabolismo , Animais , Bacteroides fragilis/crescimento & desenvolvimento , Colo/citologia , Escherichia coli/crescimento & desenvolvimento , Escherichia coli/fisiologia , Vida Livre de Germes , Humanos , Masculino , Metagenoma/fisiologia , Camundongos , Suínos , Simbiose/fisiologia
10.
Science ; 332(6032): 974-7, 2011 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-21512004

RESUMO

Mucosal surfaces constantly encounter microbes. Toll-like receptors (TLRs) mediate recognition of microbial patterns to eliminate pathogens. By contrast, we demonstrate that the prominent gut commensal Bacteroides fragilis activates the TLR pathway to establish host-microbial symbiosis. TLR2 on CD4(+) T cells is required for B. fragilis colonization of a unique mucosal niche in mice during homeostasis. A symbiosis factor (PSA, polysaccharide A) of B. fragilis signals through TLR2 directly on Foxp3(+) regulatory T cells to promote immunologic tolerance. B. fragilis lacking PSA is unable to restrain T helper 17 cell responses and is defective in niche-specific mucosal colonization. Therefore, commensal bacteria exploit the TLR pathway to actively suppress immunity. We propose that the immune system can discriminate between pathogens and the microbiota through recognition of symbiotic bacterial molecules in a process that engenders commensal colonization.


Assuntos
Bacteroides fragilis/crescimento & desenvolvimento , Bacteroides fragilis/imunologia , Tolerância Imunológica , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Polissacarídeos Bacterianos/metabolismo , Receptor 2 Toll-Like/metabolismo , Animais , Colo/imunologia , Colo/microbiologia , Vida Livre de Germes , Homeostase , Humanos , Imunidade nas Mucosas , Interleucina-10/metabolismo , Metagenoma , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Polissacarídeos Bacterianos/imunologia , Transdução de Sinais , Organismos Livres de Patógenos Específicos , Simbiose , Linfócitos T Reguladores/imunologia , Células Th17/imunologia , Receptor 2 Toll-Like/imunologia
11.
Adv Immunol ; 107: 243-74, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21034976

RESUMO

All animals live in symbiosis. Shaped by eons of co-evolution, host-bacterial associations have developed into prosperous relationships creating mechanisms for mutual benefits to both microbe and host. No better example exists in biology than the astounding numbers of bacteria harbored by the lower gastrointestinal tract of mammals. The mammalian gut represents a complex ecosystem consisting of an extraordinary number of resident commensal bacteria existing in homeostasis with the host's immune system. Most impressive about this relationship may be the concept that the host not only tolerates, but has evolved to require colonization by beneficial microorganisms, known as commensals, for various aspects of immune development and function. The microbiota provides critical signals that promote maturation of immune cells and tissues, leading to protection from infections by pathogens. Gut bacteria also appear to contribute to non-infectious immune disorders such as inflammatory bowel disease and autoimmunity. How the microbiota influences host immune responses is an active area of research with important implications for human health. This review synthesizes emerging findings and concepts that describe the mutualism between the microbiota and mammals, specifically emphasizing the role of gut bacteria in shaping an immune response that mediates the balance between health and disease. Unlocking how beneficial bacteria affect the development of the immune system may lead to novel and natural therapies based on harnessing the immunomodulatory properties of the microbiota.


Assuntos
Imunidade nas Mucosas/imunologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Simbiose/imunologia , Animais , Humanos , Tolerância Imunológica
12.
Proc Natl Acad Sci U S A ; 105(10): 3951-6, 2008 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-18319345

RESUMO

Microbes occupy countless ecological niches in nature. Sometimes these environments may be on or within another organism, as is the case in both microbial infections and symbiosis of mammals. Unlike pathogens that establish opportunistic infections, hundreds of human commensal bacterial species establish a lifelong cohabitation with their hosts. Although many virulence factors of infectious bacteria have been described, the molecular mechanisms used during beneficial host-symbiont colonization remain almost entirely unknown. The novel identification of multiple surface polysaccharides in the important human symbiont Bacteroides fragilis raised the critical question of how these molecules contribute to commensalism. To understand the function of the bacterial capsule during symbiotic colonization of mammals, we generated B. fragilis strains deleted in the global regulator of polysaccharide expression and isolated mutants with defects in capsule expression. Surprisingly, attempts to completely eliminate capsule production are not tolerated by the microorganism, which displays growth deficits and subsequent reversion to express capsular polysaccharides. We identify an alternative pathway by which B. fragilis is able to reestablish capsule production and modulate expression of surface structures. Most importantly, mutants expressing single, defined surface polysaccharides are defective for intestinal colonization compared with bacteria expressing a complete polysaccharide repertoire. Restoring the expression of multiple capsular polysaccharides rescues the inability of mutants to compete for commensalism. These findings suggest a model whereby display of multiple capsular polysaccharides provides essential functions for bacterial colonization during host-symbiont mutualism.


Assuntos
Bacteroides fragilis/crescimento & desenvolvimento , Simbiose , Animais , Cápsulas Bacterianas/metabolismo , Bacteroides fragilis/genética , Contagem de Colônia Microbiana , Deleção de Genes , Genes Bacterianos , Vida Livre de Germes , Humanos , Camundongos , Fenótipo , Polissacarídeos Bacterianos/metabolismo , Regiões Promotoras Genéticas/genética , Propriedades de Superfície
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...