Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 229
Filtrar
1.
Immunity ; 49(3): 504-514.e4, 2018 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-30231984

RESUMO

The adaptor protein CARD9 links detection of fungi by surface receptors to the activation of the NF-κB pathway. Mice deficient in CARD9 exhibit dysbiosis and are more susceptible to colitis. Here we examined the impact of Card9 deficiency in the development of colitis-associated colon cancer (CAC). Treatment of Card9-/- mice with AOM-DSS resulted in increased tumor loads as compared to WT mice and in the accumulation of myeloid-derived suppressor cells (MDSCs) in tumor tissue. The impaired fungicidal functions of Card9-/- macrophages led to increased fungal loads and variation in the overall composition of the intestinal mycobiota, with a notable increase in C. tropicalis. Bone marrow cells incubated with C. tropicalis exhibited MDSC features and suppressive functions. Fluconazole treatment suppressed CAC in Card9-/- mice and was associated with decreased MDSC accumulation. The frequency of MDSCs in tumor tissues of colon cancer patients correlated positively with fungal burden, pointing to the relevance of this regulatory axis in human disease.


Assuntos
Proteínas Adaptadoras de Sinalização CARD/metabolismo , Colite/imunologia , Neoplasias do Colo/imunologia , Disbiose/imunologia , Microbioma Gastrointestinal/imunologia , Células Supressoras Mieloides/fisiologia , Animais , Proteínas Adaptadoras de Sinalização CARD/genética , Proliferação de Células , Células Cultivadas , Técnicas de Cocultura , Colite/induzido quimicamente , Colite/genética , Neoplasias do Colo/genética , Disbiose/genética , Humanos , Interferon gama/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Supressoras Mieloides/microbiologia , Regiões Promotoras Genéticas/genética
2.
Proc Natl Acad Sci U S A ; 121(10): e2217877121, 2024 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-38412124

RESUMO

Intestinal epithelial expression of the tight junction protein claudin-2, which forms paracellular cation and water channels, is precisely regulated during development and in disease. Here, we show that small intestinal epithelial claudin-2 expression is selectively upregulated in septic patients. Similar changes occurred in septic mice, where claudin-2 upregulation coincided with increased flux across the paracellular pore pathway. In order to define the significance of these changes, sepsis was induced in claudin-2 knockout (KO) and wild-type (WT) mice. Sepsis-induced increases in pore pathway permeability were prevented by claudin-2 KO. Moreover, claudin-2 deletion reduced interleukin-17 production and T cell activation and limited intestinal damage. These effects were associated with reduced numbers of neutrophils, macrophages, dendritic cells, and bacteria within the peritoneal fluid of septic claudin-2 KO mice. Most strikingly, claudin-2 deletion dramatically enhanced survival in sepsis. Finally, the microbial changes induced by sepsis were less pathogenic in claudin-2 KO mice as survival of healthy WT mice injected with cecal slurry collected from WT mice 24 h after sepsis was far worse than that of healthy WT mice injected with cecal slurry collected from claudin-2 KO mice 24 h after sepsis. Claudin-2 upregulation and increased pore pathway permeability are, therefore, key intermediates that contribute to development of dysbiosis, intestinal damage, inflammation, ineffective pathogen control, and increased mortality in sepsis. The striking impact of claudin-2 deletion on progression of the lethal cascade activated during sepsis suggests that claudin-2 may be an attractive therapeutic target in septic patients.


Assuntos
Claudina-2 , Sepse , Animais , Humanos , Camundongos , Claudina-2/genética , Claudina-2/metabolismo , Disbiose/genética , Disbiose/metabolismo , Função da Barreira Intestinal , Mucosa Intestinal/metabolismo , Permeabilidade , Sepse/metabolismo , Junções Íntimas/metabolismo , Regulação para Cima
3.
PLoS Genet ; 19(12): e1011063, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38127816

RESUMO

Mutations in the GBA1 gene cause the lysosomal storage disorder Gaucher disease (GD) and are the greatest known genetic risk factors for Parkinson's disease (PD). Communication between the gut and brain and immune dysregulation are increasingly being implicated in neurodegenerative disorders such as PD. Here, we show that flies lacking the Gba1b gene, the main fly orthologue of GBA1, display widespread NF-kB signalling activation, including gut inflammation, and brain glial activation. We also demonstrate intestinal autophagic defects, gut dysfunction, and microbiome dysbiosis. Remarkably, modulating the microbiome of Gba1b knockout flies, by raising them under germ-free conditions, partially ameliorates lifespan, locomotor and immune phenotypes. Moreover, we show that modulation of the immune deficiency (IMD) pathway is detrimental to the survival of Gba1 deficient flies. We also reveal that direct stimulation of autophagy by rapamycin treatment achieves similar benefits to germ-free conditions independent of gut bacterial load. Consistent with this, we show that pharmacologically blocking autophagosomal-lysosomal fusion, mimicking the autophagy defects of Gba1 depleted cells, is sufficient to stimulate intestinal immune activation. Overall, our data elucidate a mechanism whereby an altered microbiome, coupled with defects in autophagy, drive chronic activation of NF-kB signaling in a Gba1 loss-of-function model. It also highlights that elimination of the microbiota or stimulation of autophagy to remove immune mediators, rather than prolonged immunosuppression, may represent effective therapeutic avenues for GBA1-associated disorders.


Assuntos
Microbioma Gastrointestinal , Doença de Gaucher , Doença de Parkinson , Animais , Doença de Gaucher/genética , Doença de Gaucher/metabolismo , Glucosilceramidase/genética , Drosophila/genética , Drosophila/metabolismo , Microbioma Gastrointestinal/genética , NF-kappa B/genética , Disbiose/genética , Doença de Parkinson/genética , Autofagia/genética
4.
Immunity ; 45(2): 238-9, 2016 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-27533011

RESUMO

Type 3 innate lymphoid cells (ILC3s) and enteric glia, an essential structural component of gut innervation, are well-known regulators of intestinal homeostasis. Ibiza et al. (2016) uncover a new link between commensal bacteria, enteric glial cells, and ILC3s that is required for intestinal homeostasis and defense.


Assuntos
Disbiose/genética , Microbioma Gastrointestinal/imunologia , Imunidade Inata , Intestinos/imunologia , Linfócitos/imunologia , Neuroglia/imunologia , Proteínas Proto-Oncogênicas c-ret/metabolismo , Animais , Técnicas de Silenciamento de Genes , Fatores Neurotróficos Derivados de Linhagem de Célula Glial/metabolismo , Homeostase , Humanos , Interleucinas/metabolismo , Intestinos/inervação , Camundongos , Neuroglia/microbiologia , Proteínas Proto-Oncogênicas c-ret/genética , Simbiose , Interleucina 22
5.
Immunity ; 44(3): 659-671, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26982366

RESUMO

Interleukin-17 (IL-17) and IL-17 receptor (IL-17R) signaling are essential for regulating mucosal host defense against many invading pathogens. Commensal bacteria, especially segmented filamentous bacteria (SFB), are a crucial factor that drives T helper 17 (Th17) cell development in the gastrointestinal tract. In this study, we demonstrate that Th17 cells controlled SFB burden. Disruption of IL-17R signaling in the enteric epithelium resulted in SFB dysbiosis due to reduced expression of α-defensins, Pigr, and Nox1. When subjected to experimental autoimmune encephalomyelitis, IL-17R-signaling-deficient mice demonstrated earlier disease onset and worsened severity that was associated with increased intestinal Csf2 expression and elevated systemic GM-CSF cytokine concentrations. Conditional deletion of IL-17R in the enteric epithelium demonstrated that there was a reciprocal relationship between the gut microbiota and enteric IL-17R signaling that controlled dysbiosis, constrained Th17 cell development, and regulated the susceptibility to autoimmune inflammation.


Assuntos
Encefalomielite Autoimune Experimental/imunologia , Infecções por Bactérias Gram-Positivas/imunologia , Bactérias Gram-Positivas Formadoras de Endosporo/imunologia , Intestinos/fisiologia , Receptores de Interleucina-17/metabolismo , Células Th17/imunologia , Animais , Disbiose/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/sangue , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Interações Hospedeiro-Patógeno , Imunidade nas Mucosas/genética , Interleucina-17/metabolismo , Intestinos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microbiota , NADH NADPH Oxirredutases/genética , NADH NADPH Oxirredutases/metabolismo , NADPH Oxidase 1 , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Receptores de Interleucina-17/genética , Transdução de Sinais/genética , Células Th17/microbiologia , alfa-Defensinas/genética , alfa-Defensinas/metabolismo
6.
Clin Exp Immunol ; 217(1): 57-77, 2024 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-38507548

RESUMO

The enzymatic core component of m6A writer complex, Mettl3, plays a crucial role in facilitating the development and progress of gastric and colorectal cancer (CRC). However, its underlying mechanism in regulating intestinal inflammation remains unclear and poorly investigated. First, the characteristics of Mettl3 expression in inflammatory bowel diseases (IBD) patients were examined. Afterward, we generated the mice line with intestinal epithelial cells (IECs)-specific deletion of Mettl3 verified by various experiments. We continuously recorded and compared the physiological status including survival rate etc. between the two groups. Subsequently, we took advantage of staining assays to analyze mucosal damage and immune infiltration of Mettl3WT and Mettl3KO primary IECs. Bulk RNA sequencing was used to pursuit the differential expression of genes (DEGs) and associated signaling pathways after losing Mettl3. Pyroptosis-related proteins were to determine whether cell death was caused by pyroptosis. Eventually, CyTOF was performed to probe the difference of CD45+ cells, especially CD3e+ T-cell clusters after losing Mettl3. In IBD patients, Mettl3 was highly expressed in the inner-nucleus of IECs while significantly decreased upon acute intestinal inflammation. IECs-specific deletion of Mettl3 KO mice triggered a wasting phenotype and developed spontaneous colitis. The survival rate, body weight, and intestinal length observed from 2 to 8 weeks of Mettl3KO mice were significantly lower than Mettl3WT mice. The degree of mucosal damage and immune infiltration in Mettl3KO were even more serious than in their WT littermate. Bulk RNA sequencing demonstrated that DEGs were dramatically enriched in NOD-signaling pathways due to the loss of Mettl3. The colonic epithelium was more prone to pyroptosis after losing Mettl3. Subsequently, CyTOF revealed that T cells have altered significantly in Mettl3KO. Furthermore, there was abnormal proliferation of CD4+ T and markedly exhaustion of CD8 + T in Mettl3KO mice. In severe IBD patients, Mettl3 is located in the inner-nucleus of IECs and declined when intestinal inflammation occurs. Subsequently, Mettl3 prevented mice from developing colitis.


Assuntos
Colite , Disbiose , Metiltransferases , Animais , Camundongos , Metiltransferases/genética , Metiltransferases/metabolismo , Colite/imunologia , Colite/genética , Disbiose/genética , Mucosa Intestinal/imunologia , Mucosa Intestinal/patologia , Camundongos Knockout , Humanos , Linfócitos T/imunologia , Modelos Animais de Doenças , Doenças Inflamatórias Intestinais/imunologia , Doenças Inflamatórias Intestinais/genética , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Camundongos Endogâmicos C57BL , Masculino
7.
Immunity ; 42(4): 692-703, 2015 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-25888259

RESUMO

The interleukin-17 (IL-17) family of cytokines has emerged as a critical player in inflammatory diseases. Among them, IL-25 has been shown to be important in allergic inflammation and protection against parasitic infection. Here we have demonstrated that IL-17B, a poorly understood cytokine, functions to inhibit IL-25-driven inflammation. IL-17B and IL-25, both binding to the interleukin-17 receptor B (IL-17RB), were upregulated in their expression after acute colonic inflammation. Individual inhibition of these cytokines revealed opposing functions in colon inflammation: IL-25 was pathogenic but IL-17B was protective. Similarly opposing phenotypes were observed in Citrobacter rodentium infection and allergic asthma. Moreover, IL-25 was found to promote IL-6 production from colon epithelial cells, which was inhibited by IL-17B. Therefore, our data demonstrate that IL-17B is an anti-inflammatory cytokine in the IL-17 family.


Assuntos
Asma/imunologia , Colite/imunologia , Disbiose/imunologia , Infecções por Enterobacteriaceae/imunologia , Interleucina-17/imunologia , Interleucinas/imunologia , Mucosa Intestinal/imunologia , Animais , Antibacterianos , Asma/induzido quimicamente , Asma/genética , Asma/patologia , Linhagem Celular , Citrobacter rodentium/imunologia , Colite/induzido quimicamente , Colite/genética , Colite/patologia , Disbiose/induzido quimicamente , Disbiose/genética , Disbiose/patologia , Infecções por Enterobacteriaceae/genética , Infecções por Enterobacteriaceae/microbiologia , Infecções por Enterobacteriaceae/patologia , Células Epiteliais/imunologia , Células Epiteliais/patologia , Regulação da Expressão Gênica , Interleucina-17/deficiência , Interleucina-17/genética , Interleucina-6/genética , Interleucina-6/imunologia , Interleucinas/deficiência , Interleucinas/genética , Mucosa Intestinal/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ovalbumina , Ligação Proteica , Receptores de Interleucina-17/genética , Receptores de Interleucina-17/imunologia , Transdução de Sinais , Dodecilsulfato de Sódio
8.
Immunity ; 42(4): 756-66, 2015 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-25902485

RESUMO

Staphylococcus aureus skin colonization is universal in atopic dermatitis and common in cancer patients treated with epidermal growth factor receptor inhibitors. However, the causal relationship of dysbiosis and eczema has yet to be clarified. Herein, we demonstrate that Adam17(fl/fl)Sox9-(Cre) mice, generated to model ADAM17-deficiency in human, developed eczematous dermatitis with naturally occurring dysbiosis, similar to that observed in atopic dermatitis. Corynebacterium mastitidis, S. aureus, and Corynebacterium bovis sequentially emerged during the onset of eczematous dermatitis, and antibiotics specific for these bacterial species almost completely reversed dysbiosis and eliminated skin inflammation. Whereas S. aureus prominently drove eczema formation, C. bovis induced robust T helper 2 cell responses. Langerhans cells were required for eliciting immune responses against S. aureus inoculation. These results characterize differential contributions of dysbiotic flora during eczema formation, and highlight the microbiota-host immunity axis as a possible target for future therapeutics in eczematous dermatitis.


Assuntos
Dermatite Atópica/imunologia , Disbiose/imunologia , Eczema/imunologia , Células de Langerhans/imunologia , Pele/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Proteínas ADAM/deficiência , Proteínas ADAM/genética , Proteínas ADAM/imunologia , Proteína ADAM17 , Animais , Antibacterianos/farmacologia , Corynebacterium/imunologia , Dermatite Atópica/tratamento farmacológico , Dermatite Atópica/genética , Dermatite Atópica/microbiologia , Disbiose/tratamento farmacológico , Disbiose/genética , Disbiose/microbiologia , Eczema/tratamento farmacológico , Eczema/genética , Eczema/microbiologia , Receptores ErbB/genética , Receptores ErbB/imunologia , Regulação da Expressão Gênica , Humanos , Imunidade Inata , Inflamação/tratamento farmacológico , Inflamação/genética , Inflamação/imunologia , Inflamação/microbiologia , Integrases/genética , Integrases/imunologia , Células de Langerhans/efeitos dos fármacos , Células de Langerhans/microbiologia , Células de Langerhans/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fatores de Transcrição SOX9/genética , Fatores de Transcrição SOX9/imunologia , Transdução de Sinais , Pele/efeitos dos fármacos , Pele/microbiologia , Pele/patologia , Staphylococcus aureus/imunologia , Linfócitos T Auxiliares-Indutores/efeitos dos fármacos , Linfócitos T Auxiliares-Indutores/microbiologia , Linfócitos T Auxiliares-Indutores/patologia
9.
BMC Pulm Med ; 24(1): 235, 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38745167

RESUMO

BACKGROUND: Emerging evidences have demonstrated that gut microbiota composition is associated with pulmonary arterial hypertension (PAH). However, the underlying causality between intestinal dysbiosis and PAH remains unresolved. METHOD: An analysis using the two-sample Mendelian randomization (MR) approach was conducted to examine the potential causal relationship between gut microbiota and PAH. To assess exposure data, genetic variants associated with 196 bacterial traits were extracted from the MiBioGen consortium, which included a sample size of 18,340 individuals. As for the outcomes, summary statistics for PAH were obtained from the NHGRI-EBI GWAS Catalog, which conducted a meta-analysis of four independent studies comprising a total of 11,744 samples. Causal effects were estimated employing various methods, including inverse variance weighted (IVW), MR-Egger, weighted median, weight mode and simple mode, with sensitivity analyses also being implemented with Cochran's Q test, MR-Egger intercept test, MR-PRESSO, leave-one-out analysis, and funnel plots. RESULTS: Following false discovery rate (FDR) correction, the genetically predicted genus Eubacterium fissicatena group (odds ratio (OR) 1.471, 95% confidence interval (CI) 1.178-1.837, q = 0.076) exhibited a causal association with PAH. In addition, the genus LachnospiraceaeUCG004 (OR 1.511, 95% CI 1.048-2.177) and genus RuminococcaceaeUCG002 (OR 1.407, 95% CI 1.040-1.905) showed a suggestive increased risk of PAH, while genus Eubacterium eligens group (OR 0.563, 95% CI 0.344-0.922), genus Phascolarctobacterium (OR 0.692, 95% CI 0.487-0.982), genus Erysipelatoclostridium (OR 0.757, 95% CI 0.579-0.989) and genus T-yzzerella3 (OR 0.768, 95% CI 0.624-0.945) were found to have nominal protective effect against PAH. CONCLUSION: The findings from our MR study have revealed a potential causal relationship between gut microbiota and PAH. Specifically, we have identified four types of gut microbiota that exhibit a protective effect on PAH, as well as three types that have a detrimental impact on PAH, thereby offering valuable insights for future mechanistic and clinical investigations in the field of PAH.


Assuntos
Microbioma Gastrointestinal , Análise da Randomização Mendeliana , Humanos , Microbioma Gastrointestinal/genética , Hipertensão Arterial Pulmonar/genética , Hipertensão Arterial Pulmonar/microbiologia , Estudo de Associação Genômica Ampla , Disbiose/genética , Polimorfismo de Nucleotídeo Único
10.
Semin Cancer Biol ; 86(Pt 3): 732-742, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-34273520

RESUMO

The lung microbiome plays an essential role in maintaining healthy lung function, including host immune homeostasis. Lung microbial dysbiosis or disruption of the gut-lung axis can contribute to lung carcinogenesis by causing DNA damage, inducing genomic instability, or altering the host's susceptibility to carcinogenic insults. Thus far, most studies have reported the association of microbial composition in lung cancer. Mechanistic studies describing host-microbe interactions in promoting lung carcinogenesis are limited. Considering cancer as a multifaceted disease where epigenetic dysregulation plays a critical role, epigenetic modifying potentials of microbial metabolites and toxins and their roles in lung tumorigenesis are not well studied. The current review explains microbial dysbiosis and epigenetic aberrations in lung cancer and potential therapeutic opportunities.


Assuntos
Neoplasias Pulmonares , Microbiota , Humanos , Disbiose/complicações , Disbiose/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/terapia , Transformação Celular Neoplásica , Epigênese Genética
11.
Semin Cancer Biol ; 86(Pt 3): 666-681, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-34216789

RESUMO

An overwhelming number of research articles have reported a strong relationship of the microbiome with cancer. Microbes have been observed more commonly in the body fluids like urine, stool, mucus of people with cancer compared to the healthy controls. The microbiota is responsible for both progression and suppression activities of various diseases. Thus, to maintain healthy human physiology, host and microbiota relationship should be in a balanced state. Any disturbance in this equilibrium, referred as microbiome dysbiosis becomes a prime cause for the human body to become more prone to immunodeficiency and cancer. It is well established that some of these microbes are the causative agents, whereas others may encourage the formation of tumours, but very little is known about how these microbial communications causing change at gene and epigenome level and trigger as well as encourage the tumour growth. Various studies have reported that microbes in the gut influence DNA methylation, DNA repair and DNA damage. The genes and pathways that are altered by gut microbes are also associated with cancer advancement, predominantly those implicated in cell growth and cell signalling pathways. This study exhaustively reviews the current research advancements in understanding of dysbiosis linked with colon, lung, ovarian, breast cancers and insights into the potential molecular targets of the microbiome promoting carcinogenesis, the epigenetic alterations of various potential targets by altered microbiota, as well as the role of various chemopreventive agents for timely prevention and customized treatment against various types of cancers.


Assuntos
Microbiota , Neoplasias , Humanos , Disbiose/complicações , Disbiose/genética , Epigenômica , Epigênese Genética , Neoplasias/genética
12.
J Lipid Res ; 64(12): 100469, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37922990

RESUMO

Deletion of the nuclear hormone receptor small heterodimer partner (Shp) ameliorates the development of obesity and nonalcoholic steatohepatitis (NASH) in mice. Liver-specific SHP plays a significant role in this amelioration. The gut microbiota has been associated with these metabolic disorders, and the interplay between bile acids (BAs) and gut microbiota contributes to various metabolic disorders. Since hepatic SHP is recognized as a critical regulator in BA synthesis, we assessed the involvement of gut microbiota in the antiobesity and anti-NASH phenotype of Shp-/- mice. Shp deletion significantly altered the levels of a few conjugated BAs. Sequencing the 16S rRNA gene in fecal samples collected from separately housed mice revealed apparent dysbiosis in Shp-/- mice. Cohousing Shp-/- mice with WT mice during a Western diet regimen impaired their metabolic improvement and effectively disrupted their distinctive microbiome structure, which became indistinguishable from that of WT mice. While the Western diet challenge significantly increased lipopolysaccharide and phenylacetic acid (PAA) levels in the blood of WT mice, their levels were not increased in Shp-/- mice. PAA was strongly associated with hepatic peroxisome proliferator-activated receptor gamma isoform 2 (Pparg2) activation in mice, which may represent the basis of the molecular mechanism underlying the association of gut bacteria and hepatic steatosis. Shp deletion reshapes the gut microbiota possibly by altering BAs. While lipopolysaccharide and PAA are the major driving forces derived from gut microbiota for NASH development, Shp deletion decreases these signaling molecules via dysbiosis, thereby partially protecting mice from diet-induced metabolic disorders.


Assuntos
Doenças Metabólicas , Hepatopatia Gordurosa não Alcoólica , Animais , Camundongos , Ácidos e Sais Biliares/metabolismo , Disbiose/genética , Disbiose/metabolismo , Lipopolissacarídeos/metabolismo , Fígado/metabolismo , Doenças Metabólicas/metabolismo , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 6/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , RNA Ribossômico 16S/metabolismo
13.
Gastroenterology ; 162(2): 521-534.e8, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34627858

RESUMO

BACKGROUND & AIMS: Microbiota composition and mechanisms of host-microbiota interactions in the esophagus are unclear. We aimed to uncover fundamental information about the esophageal microbiome and its potential significance to eosinophilic esophagitis (EoE). METHODS: Microbiota composition, transplantation potential, and antibiotic responsiveness in the esophagus were established via 16S ribosomal RNA sequencing. Functional outcomes of microbiota colonization were assessed by RNA sequencing analysis of mouse esophageal epithelium and compared with the human EoE transcriptome. The impact of dysbiosis was assessed using a preclinical model of EoE. RESULTS: We found that the murine esophagus is colonized with diverse microbial communities within the first month of life. The esophageal microbiota is distinct, dominated by Lactobacillales, and demonstrates spatial heterogeneity as the proximal and distal esophagus are enriched in Bifidobacteriales and Lactobacillales, respectively. Fecal matter transplantation restores the esophageal microbiota, demonstrating that the local environment drives diversity. Microbiota colonization modifies esophageal tissue morphology and gene expression that is enriched in pathways associated with epithelial barrier function and overlapping with genes involved in EoE, including POSTN, KLK5, and HIF1A. Finally, neonatal antibiotic treatment reduces the abundance of Lactobacillales and exaggerates type 2 inflammation in the esophagus. Clinical data substantiated loss of esophageal Lactobacillales in EoE compared with controls. CONCLUSIONS: The esophagus has a unique microbiome with notable differences between its proximal and distal regions. Fecal matter transplantation restores the esophageal microbiome. Antibiotic-induced dysbiosis exacerbates disease in a murine model of EoE. Collectively, these data establish the composition, transplantation potential, antibiotic responsiveness, and host-microbiota interaction in the esophagus and have implications for gastrointestinal health and disease.


Assuntos
Disbiose/microbiologia , Esofagite Eosinofílica/microbiologia , Esôfago/microbiologia , Interações entre Hospedeiro e Microrganismos/fisiologia , Animais , Bifidobacterium/genética , Moléculas de Adesão Celular/genética , Disbiose/genética , Disbiose/metabolismo , Disbiose/patologia , Esofagite Eosinofílica/genética , Esofagite Eosinofílica/metabolismo , Esofagite Eosinofílica/patologia , Mucosa Esofágica/metabolismo , Mucosa Esofágica/microbiologia , Mucosa Esofágica/patologia , Esôfago/metabolismo , Esôfago/patologia , Firmicutes/genética , Expressão Gênica , Perfilação da Expressão Gênica , Homeostase , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Calicreínas/genética , Lactobacillales/genética , Camundongos , RNA Ribossômico 16S/genética , RNA-Seq
14.
Genome Res ; 30(2): 276-286, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31992612

RESUMO

Connections between the microbiome and health are rapidly emerging in a wide range of diseases. However, a detailed mechanistic understanding of how different microbial communities are influencing their hosts is often lacking. One method researchers have used to understand these effects are germ-free (GF) mouse models. Differences found within the organ systems of these model organisms may highlight generalizable mechanisms that microbiome dysbioses have throughout the host. Here, we applied multiplexed, quantitative proteomics on the brains, spleens, hearts, small intestines, and colons of conventionally raised and GF mice, identifying associations to colonization state in over 7000 proteins. Highly ranked associations were constructed into protein-protein interaction networks and visualized onto an interactive 3D mouse model for user-guided exploration. These results act as a resource for microbiome researchers hoping to identify host effects of microbiome colonization on a given organ of interest. Our results include validation of previously reported effects in xenobiotic metabolism, the innate immune system, and glutamate-associated proteins while simultaneously providing organism-wide context. We highlight organism-wide differences in mitochondrial proteins including consistent increases in NNT, a mitochondrial protein with essential roles in influencing levels of NADH and NADPH, in all analyzed organs of conventional mice. Our networks also reveal new associations for further exploration, including protease responses in the spleen, high-density lipoproteins in the heart, and glutamatergic signaling in the brain. In total, our study provides a resource for microbiome researchers through detailed tables and visualization of the protein-level effects of microbial colonization on several organ systems.


Assuntos
Disbiose/genética , Microbioma Gastrointestinal/genética , Interações Hospedeiro-Patógeno/genética , Proteômica , Animais , Encéfalo/metabolismo , Encéfalo/microbiologia , Colo/metabolismo , Colo/microbiologia , Disbiose/microbiologia , Coração/microbiologia , Humanos , Intestino Delgado/metabolismo , Intestino Delgado/microbiologia , Fígado/metabolismo , Fígado/microbiologia , Camundongos , Baço/metabolismo , Baço/microbiologia
15.
Rheumatology (Oxford) ; 62(9): 3179-3187, 2023 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-36692134

RESUMO

OBJECTIVES: Hand synovitis, a potentially modifiable pathological lesion, is common and associated with pain and hand OA; nevertheless, its pathogenesis remains uncertain. This study investigated the relationship between gut microbiota dysbiosis and hand synovitis prevalence and evaluated whether bile acids mediate the association. METHODS: Participants were derived from a community-based observational study. Synovitis in each hand joint was assessed using US. Gut microbiota was evaluated using 16S ribosomal RNA amplicon sequencing on faeces, and plasma bile acids were measured by HPLC mass spectrometry. We examined the relationship between gut microbiota dysbiosis and hand synovitis prevalence, as well as the extent to which bile acids were involved in the association. RESULTS: Among 1336 participants (mean age: 63.2 years; women: 58.8%), 18.3% had prevalent hand synovitis (unilateral in 13.6% and bilateral in 4.7%). ß-diversity, but not α-diversity, of gut microbiota was significantly associated with prevalent hand synovitis. Higher relative abundance of the genus Prevotella and lower relative abundance of the genus Blautia were significantly associated with the prevalence of hand synovitis. Similar associations were also observed for laterality and the number of joints affected by hand synovitis. The association between Prevotella and hand synovitis was partially mediated through its effect on tauroursodeoxycholic acid and glycoursodeoxycholic acid, the mediation proportions being 25.7% and 21.6%, respectively. CONCLUSION: Our findings suggest that gut microbiota dysbiosis is associated with the prevalence of hand synovitis. Such an association appears to be partially mediated by plasma bile acids.


Assuntos
Microbioma Gastrointestinal , Sinovite , Humanos , Feminino , Pessoa de Meia-Idade , Microbioma Gastrointestinal/genética , Ácidos e Sais Biliares , Disbiose/epidemiologia , Disbiose/genética , Prevalência , Sinovite/epidemiologia
16.
Respir Res ; 24(1): 124, 2023 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-37143066

RESUMO

BACKGROUND: People living with HIV (PLWH) are at increased risk of developing Chronic Obstructive Pulmonary Disease (COPD) independent of cigarette smoking. We hypothesized that dysbiosis in PLWH is associated with epigenetic and transcriptomic disruptions in the airway epithelium. METHODS: Airway epithelial brushings were collected from 18 COPD + HIV + , 16 COPD - HIV + , 22 COPD + HIV - and 20 COPD - HIV - subjects. The microbiome, methylome, and transcriptome were profiled using 16S sequencing, Illumina Infinium Methylation EPIC chip, and RNA sequencing, respectively. Multi 'omic integration was performed using Data Integration Analysis for Biomarker discovery using Latent cOmponents. A correlation > 0.7 was used to identify key interactions between the 'omes. RESULTS: The COPD + HIV -, COPD -HIV + , and COPD + HIV + groups had reduced Shannon Diversity (p = 0.004, p = 0.023, and p = 5.5e-06, respectively) compared to individuals with neither COPD nor HIV, with the COPD + HIV + group demonstrating the most reduced diversity. Microbial communities were significantly different between the four groups (p = 0.001). Multi 'omic integration identified correlations between Bacteroidetes Prevotella, genes FUZ, FASTKD3, and ACVR1B, and epigenetic features CpG-FUZ and CpG-PHLDB3. CONCLUSION: PLWH with COPD manifest decreased diversity and altered microbial communities in their airway epithelial microbiome. The reduction in Prevotella in this group was linked with epigenetic and transcriptomic disruptions in host genes including FUZ, FASTKD3, and ACVR1B.


Assuntos
Infecções por HIV , Doença Pulmonar Obstrutiva Crônica , Humanos , Disbiose/genética , Doença Pulmonar Obstrutiva Crônica/epidemiologia , Doença Pulmonar Obstrutiva Crônica/genética , Perfilação da Expressão Gênica , Epitélio , Infecções por HIV/epidemiologia , Infecções por HIV/genética
17.
Nat Rev Genet ; 18(11): 690-699, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28824167

RESUMO

Taxonomic and functional changes to the composition of the gut microbiome have been implicated in multiple human diseases. Recent microbiome genome-wide association studies reveal that variants in many human genes involved in immunity and gut architecture are associated with an altered composition of the gut microbiome. Although many factors can affect the microbial organisms residing in the gut, a number of recent findings support the hypothesis that certain host genetic variants predispose an individual towards microbiome dysbiosis. This condition, in which the normal microbiome population structure is disturbed, is a key feature in disorders of metabolism and immunity.


Assuntos
Disbiose , Microbioma Gastrointestinal , Variação Genética , Disbiose/genética , Disbiose/microbiologia , Microbioma Gastrointestinal/genética , Humanos
18.
J Immunol ; 206(6): 1140-1150, 2021 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-33526439

RESUMO

Intestinal microbiota are closely related to host physiology. Over the long course of evolution and interaction, both commensal bacteria and their host have evolved multiple strategies to adapt to each other. However, in invertebrates, the regulatory mechanism of intestinal microbiota homeostasis is largely unknown. In the current study, a digestive tract-specific C-type lectin, designated as CTL33, was identified because of its abundance and response to bacteria in the intestine of kuruma shrimp (Marsupenaeus japonicus). Silencing of CTL33 expression led directly to intestinal dysbiosis, tissue damage, and shrimp death. CTL33 could facilitate biofilm formation by the intestinal bacteria. This function originated from its unique architecture, with a lectin domain responsible for bacteria recognition and a coiled coil region that mediated CTL33 dimerization and cross-linked the bacteria into a biofilm-like complex. By mediating the formation of a biofilm, CTL33 promoted the establishment of intestinal bacteria in intestine and maintained the homeostasis of the microbiota. Thus, to our knowledge, we demonstrated a new mechanism of C-type lectin-mediated biofilm formation by intestinal bacteria, providing new insights into intestinal homeostasis regulation in invertebrates.


Assuntos
Proteínas de Artrópodes/metabolismo , Bactérias/imunologia , Microbioma Gastrointestinal/imunologia , Lectinas Tipo C/metabolismo , Penaeidae/imunologia , Animais , Proteínas de Artrópodes/genética , Biofilmes , Disbiose/genética , Disbiose/imunologia , Disbiose/microbiologia , Técnicas de Silenciamento de Genes , Homeostase/imunologia , Interações entre Hospedeiro e Microrganismos/imunologia , Lectinas Tipo C/genética , Penaeidae/metabolismo , Penaeidae/microbiologia , Domínios Proteicos
19.
Int J Mol Sci ; 25(1)2023 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-38203644

RESUMO

Dysbiosis of oral microbiota is associated with the initiation and progression of periodontitis. The cause-and-effect relationship between genetics, periodontitis, and oral microbiome dysbiosis is poorly understood. Here, we demonstrate the power of the collaborative cross (CC) mice model to assess the effect of the genetic background on microbiome diversity shifts during periodontal infection and host suitability status. We examined the bacterial composition in plaque samples from seven different CC lines using 16s rRNA sequencing before and during periodontal infection. The susceptibility/resistance of the CC lines to alveolar bone loss was determined using the micro-CT technique. A total of 53 samples (7 lines) were collected before and after oral infection using oral swaps followed by DNA extraction and 16 s rRNA sequencing analysis. CC lines showed a significant variation in response to the co-infection (p < 0.05). Microbiome compositions were significantly different before and after infection and between resistant and susceptible lines to periodontitis (p < 0.05). Gram-positive taxa were significantly higher at the resistant lines compared to susceptible lines (p < 0.05). Gram-positive bacteria were reduced after infection, and gram-negative bacteria, specifically anaerobic groups, increased after infection. Our results demonstrate the utility of the CC mice in exploring the interrelationship between genetic background, microbiome composition, and periodontitis.


Assuntos
Perda do Osso Alveolar , Periodontite , Animais , Camundongos , Perda do Osso Alveolar/genética , Disbiose/genética , RNA Ribossômico 16S/genética , Cognição , Periodontite/genética
20.
EMBO J ; 37(5)2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29378774

RESUMO

Intestinal homeostasis relies on a continuous dialogue between the commensal bacteria and the immune system. Natural killer T (NKT) cells, which recognize CD1d-restricted microbial lipids and self-lipids, contribute to the regulation of mucosal immunity, yet the mechanisms underlying their functions remain poorly understood. Here, we demonstrate that NKT cells respond to intestinal lipids and CD11c+ cells (including dendritic cells (DCs) and macrophages) are essential to mediate lipid presentation within the gut ultimately controlling intestinal NKT cell homeostasis and activation. Conversely, CD1d and NKT cells participate in the control of the intestinal bacteria composition and compartmentalization, in the regulation of the IgA repertoire and in the induction of regulatory T cells within the gut. These changes in intestinal homeostasis require CD1d expression on DC/macrophage populations as mice with conditional deletion of CD1d on CD11c+ cells exhibit dysbiosis and altered immune homeostasis. These results unveil the importance of CD11c+ cells in controlling lipid-dependent immunity in the intestinal compartment and reveal an NKT cell-DC crosstalk as a key mechanism for the regulation of gut homeostasis.


Assuntos
Mucosa Intestinal/imunologia , Lipídeos de Membrana/imunologia , Células T Matadoras Naturais/imunologia , Linfócitos T Reguladores/imunologia , Animais , Antígenos CD1d/biossíntese , Antígenos CD1d/genética , Antígenos CD1d/imunologia , Antígeno CD11c/metabolismo , Células Dendríticas/imunologia , Disbiose/genética , Microbioma Gastrointestinal/imunologia , Imunoglobulina A/imunologia , Interleucina-4/imunologia , Mucosa Intestinal/microbiologia , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA