Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Cell ; 174(6): 1388-1405.e21, 2018 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-30193112

RESUMO

Empiric probiotics are commonly consumed by healthy individuals as means of life quality improvement and disease prevention. However, evidence of probiotic gut mucosal colonization efficacy remains sparse and controversial. We metagenomically characterized the murine and human mucosal-associated gastrointestinal microbiome and found it to only partially correlate with stool microbiome. A sequential invasive multi-omics measurement at baseline and during consumption of an 11-strain probiotic combination or placebo demonstrated that probiotics remain viable upon gastrointestinal passage. In colonized, but not germ-free mice, probiotics encountered a marked mucosal colonization resistance. In contrast, humans featured person-, region- and strain-specific mucosal colonization patterns, hallmarked by predictive baseline host and microbiome features, but indistinguishable by probiotics presence in stool. Consequently, probiotics induced a transient, individualized impact on mucosal community structure and gut transcriptome. Collectively, empiric probiotics supplementation may be limited in universally and persistently impacting the gut mucosa, meriting development of new personalized probiotic approaches.


Assuntos
Microbioma Gastrointestinal , Probióticos/administração & dosagem , Adolescente , Adulto , Idoso , Animais , Bactérias/genética , Bactérias/isolamento & purificação , Fezes/microbiologia , Feminino , Mucosa Gástrica/microbiologia , Humanos , Mucosa Intestinal/microbiologia , Masculino , Metagenômica , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Efeito Placebo , Análise de Componente Principal , RNA Ribossômico 16S/genética , RNA Ribossômico 16S/metabolismo , Transcriptoma , Adulto Jovem
2.
Cell ; 174(6): 1406-1423.e16, 2018 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-30193113

RESUMO

Probiotics are widely prescribed for prevention of antibiotics-associated dysbiosis and related adverse effects. However, probiotic impact on post-antibiotic reconstitution of the gut mucosal host-microbiome niche remains elusive. We invasively examined the effects of multi-strain probiotics or autologous fecal microbiome transplantation (aFMT) on post-antibiotic reconstitution of the murine and human mucosal microbiome niche. Contrary to homeostasis, antibiotic perturbation enhanced probiotics colonization in the human mucosa but only mildly improved colonization in mice. Compared to spontaneous post-antibiotic recovery, probiotics induced a markedly delayed and persistently incomplete indigenous stool/mucosal microbiome reconstitution and host transcriptome recovery toward homeostatic configuration, while aFMT induced a rapid and near-complete recovery within days of administration. In vitro, Lactobacillus-secreted soluble factors contributed to probiotics-induced microbiome inhibition. Collectively, potential post-antibiotic probiotic benefits may be offset by a compromised gut mucosal recovery, highlighting a need of developing aFMT or personalized probiotic approaches achieving mucosal protection without compromising microbiome recolonization in the antibiotics-perturbed host.


Assuntos
Antibacterianos/farmacologia , Microbioma Gastrointestinal/efeitos dos fármacos , Probióticos/administração & dosagem , Adolescente , Adulto , Idoso , Animais , Transplante de Microbiota Fecal , Fezes/microbiologia , Feminino , Humanos , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/microbiologia , Lactobacillus/efeitos dos fármacos , Lactobacillus/genética , Lactobacillus/isolamento & purificação , Lactococcus/genética , Lactococcus/isolamento & purificação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , RNA Ribossômico 16S/análise , RNA Ribossômico 16S/genética , RNA Ribossômico 16S/metabolismo , Adulto Jovem
3.
Cell ; 167(6): 1495-1510.e12, 2016 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-27912059

RESUMO

The intestinal microbiota undergoes diurnal compositional and functional oscillations that affect metabolic homeostasis, but the mechanisms by which the rhythmic microbiota influences host circadian activity remain elusive. Using integrated multi-omics and imaging approaches, we demonstrate that the gut microbiota features oscillating biogeographical localization and metabolome patterns that determine the rhythmic exposure of the intestinal epithelium to different bacterial species and their metabolites over the course of a day. This diurnal microbial behavior drives, in turn, the global programming of the host circadian transcriptional, epigenetic, and metabolite oscillations. Surprisingly, disruption of homeostatic microbiome rhythmicity not only abrogates normal chromatin and transcriptional oscillations of the host, but also incites genome-wide de novo oscillations in both intestine and liver, thereby impacting diurnal fluctuations of host physiology and disease susceptibility. As such, the rhythmic biogeography and metabolome of the intestinal microbiota regulates the temporal organization and functional outcome of host transcriptional and epigenetic programs.


Assuntos
Ritmo Circadiano , Colo/microbiologia , Microbioma Gastrointestinal , Transcriptoma , Animais , Cromatina/metabolismo , Colo/metabolismo , Vida Livre de Germes , Fígado/metabolismo , Camundongos , Microscopia Eletrônica de Varredura
4.
Cell ; 166(5): 1231-1246.e13, 2016 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-27545347

RESUMO

Innate lymphoid cells (ILCs) are critical modulators of mucosal immunity, inflammation, and tissue homeostasis, but their full spectrum of cellular states and regulatory landscapes remains elusive. Here, we combine genome-wide RNA-seq, ChIP-seq, and ATAC-seq to compare the transcriptional and epigenetic identity of small intestinal ILCs, identifying thousands of distinct gene profiles and regulatory elements. Single-cell RNA-seq and flow and mass cytometry analyses reveal compartmentalization of cytokine expression and metabolic activity within the three classical ILC subtypes and highlight transcriptional states beyond the current canonical classification. In addition, using antibiotic intervention and germ-free mice, we characterize the effect of the microbiome on the ILC regulatory landscape and determine the response of ILCs to microbial colonization at the single-cell level. Together, our work characterizes the spectrum of transcriptional identities of small intestinal ILCs and describes how ILCs differentially integrate signals from the microbial microenvironment to generate phenotypic and functional plasticity.


Assuntos
Microbioma Gastrointestinal , Imunidade Inata/genética , Intestinos/imunologia , Intestinos/microbiologia , Linfócitos/imunologia , Linfócitos/microbiologia , Animais , Sequência de Bases , Cromatina/metabolismo , Citocinas/imunologia , Epigênese Genética , Regulação da Expressão Gênica , Camundongos , Camundongos Endogâmicos C57BL , Análise de Célula Única , Transcrição Gênica
5.
Cell ; 163(6): 1428-43, 2015 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-26638072

RESUMO

Host-microbiome co-evolution drives homeostasis and disease susceptibility, yet regulatory principles governing the integrated intestinal host-commensal microenvironment remain obscure. While inflammasome signaling participates in these interactions, its activators and microbiome-modulating mechanisms are unknown. Here, we demonstrate that the microbiota-associated metabolites taurine, histamine, and spermine shape the host-microbiome interface by co-modulating NLRP6 inflammasome signaling, epithelial IL-18 secretion, and downstream anti-microbial peptide (AMP) profiles. Distortion of this balanced AMP landscape by inflammasome deficiency drives dysbiosis development. Upon fecal transfer, colitis-inducing microbiota hijacks this microenvironment-orchestrating machinery through metabolite-mediated inflammasome suppression, leading to distorted AMP balance favoring its preferential colonization. Restoration of the metabolite-inflammasome-AMP axis reinstates a normal microbiota and ameliorates colitis. Together, we identify microbial modulators of the NLRP6 inflammasome and highlight mechanisms by which microbiome-host interactions cooperatively drive microbial community stability through metabolite-mediated innate immune modulation. Therefore, targeted "postbiotic" metabolomic intervention may restore a normal microenvironment as treatment or prevention of dysbiosis-driven diseases.


Assuntos
Colo/imunologia , Colo/microbiologia , Inflamassomos/imunologia , Microbiota , Receptores de Superfície Celular/metabolismo , Transdução de Sinais , Animais , Peptídeos Catiônicos Antimicrobianos , Colite/induzido quimicamente , Colite/tratamento farmacológico , Colo/metabolismo , Disbiose/metabolismo , Vida Livre de Germes , Doenças Inflamatórias Intestinais/induzido quimicamente , Doenças Inflamatórias Intestinais/tratamento farmacológico , Interleucina-18/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Superfície Celular/genética , Taurina/administração & dosagem
6.
Cell ; 163(5): 1079-1094, 2015 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-26590418

RESUMO

Elevated postprandial blood glucose levels constitute a global epidemic and a major risk factor for prediabetes and type II diabetes, but existing dietary methods for controlling them have limited efficacy. Here, we continuously monitored week-long glucose levels in an 800-person cohort, measured responses to 46,898 meals, and found high variability in the response to identical meals, suggesting that universal dietary recommendations may have limited utility. We devised a machine-learning algorithm that integrates blood parameters, dietary habits, anthropometrics, physical activity, and gut microbiota measured in this cohort and showed that it accurately predicts personalized postprandial glycemic response to real-life meals. We validated these predictions in an independent 100-person cohort. Finally, a blinded randomized controlled dietary intervention based on this algorithm resulted in significantly lower postprandial responses and consistent alterations to gut microbiota configuration. Together, our results suggest that personalized diets may successfully modify elevated postprandial blood glucose and its metabolic consequences. VIDEO ABSTRACT.


Assuntos
Algoritmos , Glicemia/análise , Diabetes Mellitus Tipo 2/sangue , Período Pós-Prandial , Diabetes Mellitus Tipo 2/dietoterapia , Diabetes Mellitus Tipo 2/microbiologia , Dieta para Diabéticos , Microbioma Gastrointestinal , Humanos , Smartphone
7.
Nature ; 555(7695): 210-215, 2018 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-29489753

RESUMO

Human gut microbiome composition is shaped by multiple factors but the relative contribution of host genetics remains elusive. Here we examine genotype and microbiome data from 1,046 healthy individuals with several distinct ancestral origins who share a relatively common environment, and demonstrate that the gut microbiome is not significantly associated with genetic ancestry, and that host genetics have a minor role in determining microbiome composition. We show that, by contrast, there are significant similarities in the compositions of the microbiomes of genetically unrelated individuals who share a household, and that over 20% of the inter-person microbiome variability is associated with factors related to diet, drugs and anthropometric measurements. We further demonstrate that microbiome data significantly improve the prediction accuracy for many human traits, such as glucose and obesity measures, compared to models that use only host genetic and environmental data. These results suggest that microbiome alterations aimed at improving clinical outcomes may be carried out across diverse genetic backgrounds.


Assuntos
Dieta/estatística & dados numéricos , Meio Ambiente , Características da Família , Microbioma Gastrointestinal/genética , Estilo de Vida , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Estudos de Coortes , Feminino , Interação Gene-Ambiente , Glucose/metabolismo , Voluntários Saudáveis , Hereditariedade/genética , Humanos , Israel , Masculino , Pessoa de Meia-Idade , Obesidade/metabolismo , Fenótipo , Polimorfismo de Nucleotídeo Único/genética , RNA Bacteriano/análise , RNA Bacteriano/genética , RNA Ribossômico 16S/análise , Reprodutibilidade dos Testes , Estudos em Gêmeos como Assunto , Gêmeos/genética , Adulto Jovem
8.
PLoS Pathog ; 14(10): e1007406, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30365535

RESUMO

Infection with Citrobacter rodentium triggers robust tissue damage repair responses, manifested by secretion of IL-22, in the absence of which mice succumbed to the infection. Of the main hallmarks of C. rodentium infection are colonic crypt hyperplasia (CCH) and dysbiosis. In order to colonize the host and compete with the gut microbiota, C. rodentium employs a type III secretion system (T3SS) that injects effectors into colonic intestinal epithelial cells (IECs). Once injected, the effectors subvert processes involved in innate immune responses, cellular metabolism and oxygenation of the mucosa. Importantly, the identity of the effector/s triggering the tissue repair response is/are unknown. Here we report that the effector EspO ,an orthologue of OspE found in Shigella spp, affects proliferation of IECs 8 and 14 days post C. rodentium infection as well as secretion of IL-22 from colonic explants. While we observed no differences in the recruitment of group 3 innate lymphoid cells (ILC3s) and T cells, which are the main sources of IL-22 at the early and late stages of C. rodentium infection respectively, infection with ΔespO was characterized by diminished recruitment of sub-mucosal neutrophils, which coincided with lower abundance of Mmp9 and chemokines (e.g. S100a8/9) in IECs. Moreover, mice infected with ΔespO triggered significantly lesser nutritional immunity (e.g. calprotectin, Lcn2) and expression of antimicrobial peptides (Reg3ß, Reg3γ) compared to mice infected with WT C. rodentium. This overlapped with a decrease in STAT3 phosphorylation in IECs. Importantly, while the reduced CCH and abundance of antimicrobial proteins during ΔespO infection did not affect C. rodentium colonization or the composition of commensal Proteobacteria, they had a subtle consequence on Firmicutes subpopulations. EspO is the first bacterial virulence factor that affects neutrophil recruitment and secretion of IL-22, as well as expression of antimicrobial and nutritional immunity proteins in IECs.


Assuntos
Peptídeos Catiônicos Antimicrobianos/metabolismo , Citrobacter rodentium/metabolismo , Infecções por Enterobacteriaceae/imunologia , Imunidade Inata/imunologia , Mucosa Intestinal/imunologia , Sistemas de Secreção Tipo III/metabolismo , Animais , Infecções por Enterobacteriaceae/metabolismo , Infecções por Enterobacteriaceae/microbiologia , Feminino , Mucosa Intestinal/lesões , Mucosa Intestinal/microbiologia , Camundongos , Camundongos Endogâmicos C57BL
9.
Proc Natl Acad Sci U S A ; 114(3): E337-E346, 2017 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-28049839

RESUMO

The oral epithelium contributes to innate immunity and oral mucosal homeostasis, which is critical for preventing local inflammation and the associated adverse systemic conditions. Nevertheless, the mechanisms by which the oral epithelium maintains homeostasis are poorly understood. Here, we studied the role of growth arrest specific 6 (GAS6), a ligand of the TYRO3-AXL-MERTK (TAM) receptor family, in regulating oral mucosal homeostasis. Expression of GAS6 was restricted to the outer layers of the oral epithelium. In contrast to protein S, the other TAM ligand, which was constitutively expressed postnatally, expression of GAS6 initiated only 3-4 wk after birth. Further analysis revealed that GAS6 expression was induced by the oral microbiota in a myeloid differentiation primary response gene 88 (MyD88)-dependent fashion. Mice lacking GAS6 presented higher levels of inflammatory cytokines, elevated frequencies of neutrophils, and up-regulated activity of enzymes, generating reactive nitrogen species. We also found an imbalance in Th17/Treg ratio known to control tissue homeostasis, as Gas6-deficient dendritic cells preferentially secreted IL-6 and induced Th17 cells. As a result of this immunological shift, a significant microbial dysbiosis was observed in Gas6-/- mice, because anaerobic bacteria largely expanded by using inflammatory byproducts for anaerobic respiration. Using chimeric mice, we found a critical role for GAS6 in epithelial cells in maintaining oral homeostasis, whereas its absence in hematopoietic cells synergized the level of dysbiosis. We thus propose GAS6 as a key immunological regulator of host-commensal interactions in the oral epithelium.


Assuntos
Homeostase/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Mucosa Bucal/metabolismo , Animais , Disbiose/metabolismo , Células Epiteliais/metabolismo , Imunidade Inata/imunologia , Inflamação/metabolismo , Interleucina-6 , Camundongos , Camundongos Endogâmicos C57BL , Células Mieloides/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Neutrófilos/metabolismo , Proteína S/metabolismo , Espécies Reativas de Nitrogênio/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , c-Mer Tirosina Quinase/metabolismo
10.
Immunol Cell Biol ; 96(3): 284-297, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29356071

RESUMO

Inflammation plays pivotal roles in different stages of tumor development. Screening for predisposing genetic abnormalities and understanding the roles these genes play in the crosstalk between immune and cancer cells will provide new targets for cancer therapy and prevention. The interferon inducible transmembrane (IFITM) genes are involved in pathogenesis of the gastro-intestinal tract. We aimed at delineating the role of IFITM3 in colonic epithelial homeostasis, inflammation and colitis-associated tumorigenesis using IFITM3-deficient mice. Chemical induction of colitis in IFITM3-deficient mice results in significantly increased clinical signs of inflammation and induction of invasive tumorigenesis. Bone marrow transplantation showed that cells of the hematopoietic system are responsible for colitis deterioration. In these mice, impaired cytokine expression skewed inflammatory response toward pathogenic Th17 with reduced expression of the anti-inflammatory cytokine IL10 during the recovery phase. Intriguingly, mice lacking the entire IFITM locus developed spontaneous chronic colitis from the age of 14 weeks. Sequencing the 16S rRNA of naïve mice lacking IFITM3 gene, or the entire locus containing five IFITM genes, revealed these mice had significant bacterial differences from their wild-type littermates. Our novel results provide strong evidence for the essential role of IFITM genes in ameliorating colitis and colitis-associated tumorigenesis.


Assuntos
Carcinogênese/genética , Carcinogênese/patologia , Colite/imunologia , Colite/microbiologia , Imunidade , Inflamação/genética , Proteínas de Membrana/genética , Microbiota , Animais , Colite/genética , Colite/patologia , Colo/patologia , Sulfato de Dextrana , Modelos Animais de Doenças , Progressão da Doença , Suscetibilidade a Doenças , Hematopoese , Imunidade/genética , Proteínas de Membrana/deficiência , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microbiota/genética , Células Mieloides/patologia
11.
Curr Opin Nephrol Hypertens ; 26(1): 1-8, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27798455

RESUMO

PURPOSE OF REVIEW: The mammalian mucosal surfaces are densely inhabited by a diverse microbial ecosystem termed the microbiota. Among these highly heterogeneous populations, the largest and richest is the gut microbiota, recently suggested to affect various physiological traits and susceptibility to disease. Novel metagenomic and metabolomic approaches, which have been developed in the past decade, have enabled the elucidation of the contribution of the microbiota to metabolic, immunologic, neurologic and endocrine homeostasis. RECENT FINDINGS: Dysbiosis, the alteration in the gut microbiota composition and function, has been lately associated with the pathogenesis of multifactorial diseases such as obesity, diabetes and cardiovascular disorders. Recent studies have also suggested associations between dysbiosis and essential hypertension, a common chronic medical condition affecting 20% or more of the adult population worldwide, which is considered a major causative factor for heart disease, stroke, chronic renal failure, blindness and dementia. SUMMARY: In this review, we discuss the accumulating research pointing to possible interplays between the gut microbiome and hypertension and highlight future prospects by which utilization of microbiome-related techniques may be incorporated into the diagnosis and therapeutic arsenal of hypertension management.


Assuntos
Pressão Sanguínea , Disbiose/fisiopatologia , Microbioma Gastrointestinal/fisiologia , Hipertensão/fisiopatologia , Animais , Humanos , Hipertensão/tratamento farmacológico
12.
Stem Cells ; 32(8): 2008-20, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24715711

RESUMO

Mesenchymal stromal cell populations include a fraction, termed mesenchymal stem cells, exhibiting multipotency. Other cells within this population possess a lesser differentiation range. This was assumed to be due to a mesenchymal cellular cascade topped by a multipotent cell, which gives rise to progeny with diminishing differentiation potentials. Here, we show that mesenchymal cells, a priori exhibiting a limited differentiation potential, may gain new capacities and become multipotent following single-cell isolation. These fate changes were accompanied by upregulation of differentiation promoting genes, many of which also became H4K20me1 methylated. Early events in the process included TGFß and Wnt modulation, and downregulation of hypoxia signaling. Indeed, hypoxic conditions inhibited the observed cell changes. Overall, cell isolation from neighboring partners caused major molecular changes and particularly, a newly established epigenetic state, ultimately leading to the acquisition of new differentiation potentials and an altered cell fate.


Assuntos
Diferenciação Celular/fisiologia , Separação Celular , Células-Tronco Mesenquimais/citologia , Animais , Células da Medula Óssea/citologia , Técnicas de Cultura de Células , Linhagem da Célula , Imunoprecipitação da Cromatina , Células Clonais/citologia , Citometria de Fluxo , Camundongos , Camundongos Endogâmicos C57BL , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase em Tempo Real
13.
Nat Commun ; 14(1): 5810, 2023 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-37726308

RESUMO

The tumor microenvironment (TME) is comprised of non-malignant cells that interact with each other and with cancer cells, critically impacting cancer biology. The TME is complex, and understanding it requires simplifying approaches. Here we provide an experimental-mathematical approach to decompose the TME into small circuits of interacting cell types. We find, using female breast cancer single-cell-RNA-sequencing data, a hierarchical network of interactions, with cancer-associated fibroblasts (CAFs) at the top secreting factors primarily to tumor-associated macrophages (TAMs). This network is composed of repeating circuit motifs. We isolate the strongest two-cell circuit motif by culturing fibroblasts and macrophages in-vitro, and analyze their dynamics and transcriptomes. This isolated circuit recapitulates the hierarchy of in-vivo interactions, and enables testing the effect of ligand-receptor interactions on cell dynamics and function, as we demonstrate by identifying a mediator of CAF-TAM interactions - RARRES2, and its receptor CMKLR1. Thus, the complexity of the TME may be simplified by identifying small circuits, facilitating the development of strategies to modulate the TME.


Assuntos
Fibroblastos Associados a Câncer , Microambiente Tumoral , Feminino , Humanos , Fibroblastos , Transporte Biológico , Comunicação Celular
14.
Nat Commun ; 13(1): 6513, 2022 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-36316305

RESUMO

Tumors initiate by mutations in cancer cells, and progress through interactions of the cancer cells with non-malignant cells of the tumor microenvironment. Major players in the tumor microenvironment are cancer-associated fibroblasts (CAFs), which support tumor malignancy, and comprise up to 90% of the tumor mass in pancreatic cancer. CAFs are transcriptionally rewired by cancer cells. Whether this rewiring is differentially affected by different mutations in cancer cells is largely unknown. Here we address this question by dissecting the stromal landscape of BRCA-mutated and BRCA Wild-type pancreatic ductal adenocarcinoma. We comprehensively analyze pancreatic cancer samples from 42 patients, revealing different CAF subtype compositions in germline BRCA-mutated vs. BRCA Wild-type tumors. In particular, we detect an increase in a subset of immune-regulatory clusterin-positive CAFs in BRCA-mutated tumors. Using cancer organoids and mouse models we show that this process is mediated through activation of heat-shock factor 1, the transcriptional regulator of clusterin. Our findings unravel a dimension of stromal heterogeneity influenced by germline mutations in cancer cells, with direct implications for clinical research.


Assuntos
Fibroblastos Associados a Câncer , Carcinoma Ductal Pancreático , Clusterina , Fatores de Transcrição de Choque Térmico , Neoplasias Pancreáticas , Animais , Camundongos , Fibroblastos Associados a Câncer/metabolismo , Carcinoma Ductal Pancreático/patologia , Clusterina/genética , Clusterina/metabolismo , Fatores de Transcrição de Choque Térmico/genética , Fatores de Transcrição de Choque Térmico/metabolismo , Neoplasias Pancreáticas/patologia , Microambiente Tumoral/genética , Humanos , Neoplasias Pancreáticas
15.
J Immunol ; 183(2): 890-6, 2009 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-19561102

RESUMO

We recently reported that heat shock protein 60 (HSP60) via TLR4 signaling activates B cells and induces them to proliferate and secrete IL-10. We now report that HSP60 inhibits mouse B cell apoptosis, spontaneous or induced by dexamethasone or anti-IgM activation. Unlike HSP60 enhancement of B cell proliferation and IL-10 secretion, TLR4 signaling was not required for the inhibition of apoptosis by HSP60; nevertheless, MyD88 was essential. Inhibition of apoptosis by HSP60 was associated with up-regulation of the antiapoptotic molecules Bcl-2, Bcl-x(L), and survivin, maintenance of the mitochondrial transmembrane potential, and inhibition of caspase-3 activation. Moreover, B cells incubated with HSP60 manifested prolonged survival following transfer into recipient mice. These results extend the varied role of HSP60 in the innate regulation of the adaptive immune response.


Assuntos
Apoptose , Linfócitos B/citologia , Chaperonina 60/fisiologia , Fator 88 de Diferenciação Mieloide/metabolismo , Transdução de Sinais , Animais , Formação de Anticorpos , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/biossíntese , Linfócitos B/transplante , Inibidores de Caspase , Sobrevivência Celular , Transplante de Células , Dexametasona/farmacologia , Imunoglobulina M/imunologia , Potencial da Membrana Mitocondrial , Camundongos , Regulação para Cima
16.
Cancer Res ; 81(7): 1639-1653, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33547159

RESUMO

Gastric cancer is the third most lethal cancer worldwide, and evaluation of the genomic status of gastric cancer cells has not translated into effective prognostic or therapeutic strategies. We therefore hypothesize that outcomes may depend on the tumor microenvironment (TME), in particular, cancer-associated fibroblasts (CAF). However, very little is known about the role of CAFs in gastric cancer. To address this, we mapped the transcriptional landscape of human gastric cancer stroma by microdissection and RNA sequencing of CAFs from patients with gastric cancer. A stromal gene signature was associated with poor disease outcome, and the transcription factor heat shock factor 1 (HSF1) regulated the signature. HSF1 upregulated inhibin subunit beta A and thrombospondin 2, which were secreted in CAF-derived extracellular vesicles to the TME to promote cancer. Together, our work provides the first transcriptional map of human gastric cancer stroma and highlights HSF1 and its transcriptional targets as potential diagnostic and therapeutic targets in the genomically stable tumor microenvironment. SIGNIFICANCE: This study shows how HSF1 regulates a stromal transcriptional program associated with aggressive gastric cancer and identifies multiple proteins within this program as candidates for therapeutic intervention. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/7/1639/F1.large.jpg.


Assuntos
Fibroblastos Associados a Câncer/fisiologia , Vesículas Extracelulares/metabolismo , Fatores de Transcrição de Choque Térmico/metabolismo , Neoplasias Gástricas/patologia , Animais , Fibroblastos Associados a Câncer/patologia , Células Cultivadas , Estudos de Coortes , Progressão da Doença , Vesículas Extracelulares/patologia , Fatores de Transcrição de Choque Térmico/genética , Humanos , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Invasividade Neoplásica , Fenótipo , Prognóstico , Via Secretória/fisiologia , Neoplasias Gástricas/diagnóstico , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/mortalidade , Análise de Sobrevida , Microambiente Tumoral/fisiologia
17.
Circulation ; 120(25): 2559-66, 2009 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-19996019

RESUMO

BACKGROUND: The identification and isolation of human cardiac progenitor cells (hCPCs) offer new approaches for myocardial regeneration and repair. Still, the optimal source of human cardiac progenitor cells and the influence of patient characteristics on their number remain unclear. Using a novel method to isolate human cardiac progenitor cells, we aimed to define the optimal source and association between their number and patient characteristics. METHODS AND RESULTS: We developed a novel isolation method that produced viable cells (7 x 10(6)+/-6.53 x 10(5)/g) from various tissue samples obtained during heart surgery or endomyocardial biopsies (113 samples from 94 patients 23 to 80 years of age). The isolated cardiac cells were grown in culture with a stem cell expansion medium. According to fluorescence-activated cell sorting analysis, cultured cells derived from the right atrium generated higher amounts of c-kit(+) (24+/-2.5%) and Islet-1(+) cells (7%) in culture (mean of passages 1, 2, and 3) than did cultured cells from the left atrium (7.3+/-3.5%), right ventricle (4.1+/-1.6%), and left ventricle (9.7+/-3%; P=0.001). According to multivariable analysis, the right atrium as the cell source and female sex were associated with a higher number of c-kit(+) cells. There was no overlap between c-kit(+) and Islet-1 expression. In vitro assays of differentiation into osteoblasts, adipocytes, and myogenic lineage showed that the isolated human cardiac progenitor cells were multipotent. Finally, the cells were transplanted into infarcted myocardium of rats and generated myocardial grafts. CONCLUSIONS: Our results show that the right atrium is the best source for c-kit(+) and Islet-1 progenitors, with higher percentages of c-kit(+) cells being produced by women.


Assuntos
Átrios do Coração/citologia , Ventrículos do Coração/citologia , Células-Tronco Multipotentes/citologia , Miócitos Cardíacos/citologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Biópsia , Diferenciação Celular/fisiologia , Células Cultivadas , Modelos Animais de Doenças , Feminino , Coração/fisiologia , Átrios do Coração/metabolismo , Ventrículos do Coração/metabolismo , Proteínas de Homeodomínio/metabolismo , Humanos , Proteínas com Homeodomínio LIM , Masculino , Pessoa de Meia-Idade , Células-Tronco Multipotentes/metabolismo , Infarto do Miocárdio/patologia , Infarto do Miocárdio/cirurgia , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Ratos , Ratos Nus , Regeneração/fisiologia , Caracteres Sexuais , Transplante de Células-Tronco , Fatores de Transcrição
18.
Exp Cell Res ; 315(11): 1904-13, 2009 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-19328780

RESUMO

Multiple myeloma is characterized by the malignant growth of immunoglobulin producing plasma cells, predominantly in the bone marrow. The effects of primary human mesenchymal stromal cells on the differentiation phenotype of multiple myeloma cells were studied by co-culture experiments. The incubation of multiple myeloma cells with bone marrow-derived mesenchymal stromal cells resulted in significant reduction of the expression of the predominant plasma cell differentiation markers CD38 and CD138, and cell surface immunoglobulin light chain. While the down-regulation of CD138 by stromal cells was completely dependent on their adhesive interactions with the multiple myeloma cells, interleukin-6 induced specific down-regulation of CD38. Mesenchymal stromal cells or their conditioned media inhibited the growth of multiple myeloma cell line, thereby reducing the overall amounts of secreted light chains. Analysis of primary multiple myeloma bone marrow samples reveled that the expression of CD38 on multiple myeloma cells was not affected by adhesive interactions. The ex vivo propagation of primary multiple myeloma cells resulted in significant increase in their differentiation markers. Overall, the data indicate that the bone marrow-derived mesenchymal stromal cells revert multiple myeloma cells to less differentiated phenotype by the combined activities of adhesive interactions and interleukin-6.


Assuntos
Interleucina-6/fisiologia , Mesoderma/citologia , Mesoderma/fisiologia , Mieloma Múltiplo/patologia , Mieloma Múltiplo/fisiopatologia , Células Estromais/citologia , Células Estromais/fisiologia , ADP-Ribosil Ciclase 1/metabolismo , Anticorpos Monoclonais/farmacologia , Biomarcadores Tumorais/metabolismo , Células da Medula Óssea/citologia , Células da Medula Óssea/fisiologia , Adesão Celular , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Linhagem Celular Tumoral , Técnicas de Cocultura , Regulação para Baixo , Humanos , Cadeias kappa de Imunoglobulina/metabolismo , Interleucina-6/antagonistas & inibidores , Interleucina-6/farmacologia , Glicoproteínas de Membrana/metabolismo , Mesoderma/efeitos dos fármacos , Testes de Neutralização , Fenótipo , Plasmócitos/citologia , Plasmócitos/fisiologia , Proteínas Recombinantes/farmacologia , Células Estromais/efeitos dos fármacos , Sindecana-1/metabolismo
19.
Nat Cancer ; 1(7): 692-708, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-35122040

RESUMO

Tumors are supported by cancer-associated fibroblasts (CAFs). CAFs are heterogeneous and carry out distinct cancer-associated functions. Understanding the full repertoire of CAFs and their dynamic changes as tumors evolve could improve the precision of cancer treatment. Here we comprehensively analyze CAFs using index and transcriptional single-cell sorting at several time points along breast tumor progression in mice, uncovering distinct subpopulations. Notably, the transcriptional programs of these subpopulations change over time and in metastases, transitioning from an immunoregulatory program to wound-healing and antigen-presentation programs, indicating that CAFs and their functions are dynamic. Two main CAF subpopulations are also found in human breast tumors, where their ratio is associated with disease outcome across subtypes and is particularly correlated with BRCA mutations in triple-negative breast cancer. These findings indicate that the repertoire of CAF changes over time in breast cancer progression, with direct clinical implications.


Assuntos
Fibroblastos Associados a Câncer , Neoplasias de Mama Triplo Negativas , Animais , Fibroblastos Associados a Câncer/metabolismo , Humanos , Glicoproteínas de Membrana/genética , Camundongos , Proteína A4 de Ligação a Cálcio da Família S100/genética , Neoplasias de Mama Triplo Negativas/genética
20.
Nat Commun ; 11(1): 6245, 2020 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-33288768

RESUMO

In the colon, long-term exposure to chronic inflammation drives colitis-associated colon cancer (CAC) in patients with inflammatory bowel disease. While the causal and clinical links are well established, molecular understanding of how chronic inflammation leads to the development of colon cancer is lacking. Here we deconstruct the evolving microenvironment of CAC by measuring proteomic changes and extracellular matrix (ECM) organization over time in a mouse model of CAC. We detect early changes in ECM structure and composition, and report a crucial role for the transcriptional regulator heat shock factor 1 (HSF1) in orchestrating these events. Loss of HSF1 abrogates ECM assembly by colon fibroblasts in cell-culture, prevents inflammation-induced ECM remodeling in mice and inhibits progression to CAC. Establishing relevance to human disease, we find high activation of stromal HSF1 in CAC patients, and detect the HSF1-dependent proteomic ECM signature in human colorectal cancer. Thus, HSF1-dependent ECM remodeling plays a crucial role in mediating inflammation-driven colon cancer.


Assuntos
Neoplasias Associadas a Colite/metabolismo , Matriz Extracelular/metabolismo , Fatores de Transcrição de Choque Térmico/metabolismo , Proteoma/metabolismo , Proteômica/métodos , Animais , Linhagem Celular Tumoral , Células Cultivadas , Neoplasias Associadas a Colite/genética , Modelos Animais de Doenças , Fatores de Transcrição de Choque Térmico/genética , Humanos , Espectrometria de Massas/métodos , Camundongos da Linhagem 129 , Camundongos Endogâmicos BALB C , Camundongos Knockout , Proteoma/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA