Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Eur J Immunol ; 54(2): e2250248, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37957831

RESUMEN

Human intestinal organoids are an ideal model system for studying gastrointestinal physiology and immunopathology. Altered physiology and mucosal immune response are hallmarks of numerous intestinal functional and inflammatory diseases, including inflammatory bowel disease (IBD), coeliac disease, irritable bowel syndrome (IBS), and obesity. These conditions impact the normal epithelial functions of the intestine, such as absorption, barrier function, secretion, and host-microbiome communication. They are accompanied by characteristic intestinal symptoms and have significant societal, economic, and healthcare burdens. To develop new treatment options, cutting-edge research is required to investigate their etiology and pathology. Human intestinal organoids derived from patient tissue recapitulate the key physiological and immunopathological aspects of these conditions, providing a promising platform for elucidating disease mechanisms. This review will summarize recent reports on patient-derived human small intestinal and colonic organoids and highlight how these models have been used to study intestinal epithelial functions in the context of inflammation, altered physiology, and immune response. Furthermore, it will elaborate on the various organoid systems in use and the techniques/assays currently available to study epithelial functions. Finally, it will conclude by discussing the limitations and future perspectives of organoid technology.


Asunto(s)
Enfermedades Inflamatorias del Intestino , Mucosa Intestinal , Humanos , Intestinos/patología , Organoides/patología , Organoides/fisiología , Intestino Delgado/patología
2.
Am J Physiol Gastrointest Liver Physiol ; 323(5): G439-G460, 2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-36165492

RESUMEN

DNA sensor pathways can initiate inflammasome, cell death, and type I interferon (IFN) signaling in immune-mediated inflammatory diseases (IMIDs), including type I interferonopathies. We investigated the involvement of these pathways in the pathogenesis of ulcerative colitis (UC) by analyzing the expression of DNA sensor, inflammasome, and type I IFN biomarker genes in colonic mucosal biopsy tissue from control (n = 31), inactive UC (n = 31), active UC (n = 33), and a UC single-cell RNA-Seq dataset. The effects of type I IFN (IFN-ß), IFN-γ, and TNF-α on gene expression, cytokine production, and cell death were investigated in human colonic organoids. In organoids treated with cytokines alone, or in combination with NLR family pyrin domain-containing 3 (NLRP3), caspase, or JAK inhibitors, cell death was measured, and supernatants were assayed for IL-1ß/IL-18/CXCL10. The expression of DNA sensor pathway genes-PYHIN family members [absent in melanoma 2 (AIM2), IFI16, myeloid cell nuclear differentiation antigen (MNDA), and pyrin and HIN domain family member 1 (PYHIN1)- as well as Z-DNA-binding protein 1 (ZBP1), cyclic GMP-AMP synthase (cGAS), and DDX41 was increased in active UC and expressed in a cell type-restricted pattern. Inflammasome genes (CASP1, IL1B, and IL18), type I IFN inducers [stimulator of interferon response cGAMP interactor 1 (STING), TBK1, and IRF3), IFNB1, and type I IFN biomarker genes (OAS2, IFIT2, and MX2) were also increased in active UC. Cotreatment of organoids with IFN-ß or IFN-γ in combination with TNFα increased expression of IFI16, ZBP1, CASP1, cGAS, and STING induced cell death and IL-1ß/IL-18 secretion. This inflammatory cell death was blocked by the JAK inhibitor tofacitinib but not by inflammasome or caspase inhibitors. Increased type I IFN activity may drive elevated expression of DNA sensor genes and JAK-dependent but inflammasome-independent inflammatory cell death of colonic epithelial cells in UC.NEW & NOTEWORTHY This study found that patients with active UC have significantly increased colonic gene expression of cytosolic DNA sensor, inflammasome, STING, and type I IFN signaling pathways. The type I IFN, IFN-ß, in combination with TNF-α induced JAK-dependent but NLRP3 and inflammasome-independent inflammatory cell death of colonic organoids. This novel inflammatory cell death phenotype is relevant to UC immunopathology and may partially explain the efficacy of the JAKinibs tofacitinib and upadacitinib in patients with UC.


Asunto(s)
Colitis Ulcerosa , Interferón Tipo I , Inhibidores de las Cinasas Janus , Humanos , Inflamasomas/metabolismo , Interleucina-18 , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Factor de Necrosis Tumoral alfa , Inhibidores de Caspasas , Organoides/metabolismo , Pirina , Caspasa 1/metabolismo , Nucleotidiltransferasas/metabolismo , ADN , Muerte Celular , Proteínas de Unión al ADN/metabolismo , Antígenos de Diferenciación
3.
Int J Mol Sci ; 22(15)2021 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-34361102

RESUMEN

Interactions between the intestinal microbiota, immune system and nervous system are essential for homeostasis in the gut. Inflammasomes contribute to innate immunity and brain-gut interactions, but their role in microbiota-neuro-immune interactions is not clear. Therefore, we investigated the effect of the inflammasome on visceral pain and local and systemic neuroimmune responses after antibiotic-induced changes to the microbiota. Wild-type (WT) and caspase-1/11 deficient (Casp1 KO) mice were orally treated for 2 weeks with an antibiotic cocktail (Abx, Bacitracin A and Neomycin), followed by quantification of representative fecal commensals (by qPCR), cecal short chain fatty acids (by HPLC), pathways implicated in the gut-neuro-immune axis (by RT-qPCR, immunofluorescence staining, and flow cytometry) in addition to capsaicin-induced visceral pain responses. Abx-treatment in WT-mice resulted in an increase in colonic macrophages, central neuro-immune interactions, colonic inflammasome and nociceptive receptor gene expression and a reduction in capsaicin-induced visceral pain. In contrast, these responses were attenuated in Abx-treated Casp1 KO mice. Collectively, the data indicate an important role for the inflammasome pathway in functional and inflammatory gastrointestinal conditions where pain and alterations in microbiota composition are prominent.


Asunto(s)
Caspasa 1/fisiología , Microbioma Gastrointestinal , Inflamasomas/inmunología , Inflamación/complicaciones , Neuroinmunomodulación , Dolor Visceral/patología , Animales , Antibacterianos/farmacología , Encéfalo/efectos de los fármacos , Encéfalo/inmunología , Encéfalo/microbiología , Encéfalo/patología , Capsaicina/toxicidad , Colon/efectos de los fármacos , Colon/inmunología , Colon/microbiología , Colon/patología , Femenino , Inflamasomas/efectos de los fármacos , Inflamación/inmunología , Inflamación/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal , Dolor Visceral/etiología , Dolor Visceral/metabolismo
4.
J Immunol ; 191(12): 6084-92, 2013 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-24198280

RESUMEN

Listeria monocytogenes is a Gram-positive bacterium that can cause septicemia and meningitis. TLRs are central receptors of the innate immune system that drive inflammatory responses to invading microbes such as L. monocytogenes. Although intestinal epithelial cells (IECs) represent the initial point of entry used by L. monocytogenes for infection, the innate immune response to L. monocytogenes in these cells has been poorly characterized to date. The aim of this study was to determine which TLRs are involved in mediating the immune response to L. monocytogenes in IECs. We performed an RNA interference screen of TLRs 1-10 in the HT-29 IEC cell line and observed the most significant reduction in chemokine output following silencing of TLR10. This effect was also observed in the macrophage cell line THP-1. The chemokines CCL20, CCL1, and IL-8 were reduced following knockdown of TLR10. Silencing of TLR10 resulted in increased viability of L. monocytogenes in both HT-29 and THP-1 cells. TLR10 was found to be predominantly expressed intracellularly in epithelia, and activation required viable L. monocytogenes. NF-κB activation was seen to require TLR2 in addition to TLR10. Taken together, these data indicate novel roles for TLR10 in sensing pathogenic infection in both the epithelium and macrophages and have identified L. monocytogenes as a source of ligand for the orphan receptor TLR10.


Asunto(s)
Células Epiteliales/inmunología , Inmunidad Mucosa , Mucosa Intestinal/inmunología , Listeria monocytogenes/inmunología , Macrófagos/inmunología , Receptor Toll-Like 10/fisiología , Quimiocinas/biosíntesis , Quimiocinas/genética , Células Epiteliales/microbiología , Regulación de la Expresión Génica/inmunología , Células HCT116 , Células HEK293 , Células HT29 , Humanos , Inmunidad Innata , Técnicas In Vitro , Interleucinas/biosíntesis , Interleucinas/genética , Mucosa Intestinal/citología , Ligandos , Macrófagos/microbiología , FN-kappa B/metabolismo , Especificidad de Órganos , Interferencia de ARN , ARN Interferente Pequeño/farmacología , Receptor Toll-Like 10/antagonistas & inhibidores , Receptor Toll-Like 10/genética , Receptor Toll-Like 10/inmunología , Receptor Toll-Like 2/fisiología , Receptores Toll-Like/biosíntesis , Receptores Toll-Like/genética
5.
Gut Microbes ; 15(1): 2235067, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37526383

RESUMEN

The human gut microbiota is a key contributor to host metabolism and physiology, thereby impacting in various ways on host health. This complex microbial community has developed many metabolic strategies to colonize, persist and survive in the gastrointestinal environment. In this regard, intracellular glycogen accumulation has been associated with important physiological functions in several bacterial species, including gut commensals. However, the role of glycogen storage in shaping the composition and functionality of the gut microbiota offers a novel perspective in gut microbiome research. Here, we review what is known about the enzymatic machinery and regulation of glycogen metabolism in selected enteric bacteria, while we also discuss its potential impact on colonization and adaptation to the gastrointestinal tract. Furthermore, we survey the presence of such glycogen biosynthesis pathways in gut metagenomic data to highlight the relevance of this metabolic trait in enhancing survival in the highly competitive and dynamic gut ecosystem.


Asunto(s)
Microbioma Gastrointestinal , Microbiota , Humanos , Microbioma Gastrointestinal/fisiología , Tracto Gastrointestinal/microbiología , Bacterias/genética , Glucógeno/metabolismo
6.
Gut Microbes ; 15(1): 2163838, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36656595

RESUMEN

Conflicting evidence exists on the association between consumption of non-steroidal anti-inflammatory drugs (NSAIDs) and symptomatic worsening of inflammatory bowel disease (IBD). We hypothesized that the heterogeneous prevalence of pathobionts [e.g., adherent-invasive Escherichia coli (AIEC)], might explain this inconsistent NSAIDs/IBD correlation. Using IL10-/- mice, we found that NSAID aggravated colitis in AIEC-colonized animals. This was accompanied by activation of the NLRP3 inflammasome, Caspase-8, apoptosis, and pyroptosis, features not seen in mice exposed to AIEC or NSAID alone, revealing an AIEC/NSAID synergistic effect. Inhibition of NLRP3 or Caspase-8 activity ameliorated colitis, with reduction in NLRP3 inflammasome activation, cell death markers, activated T-cells and macrophages, improved histology, and increased abundance of Clostridium cluster XIVa species. Our findings provide new insights into how NSAIDs and an opportunistic gut-pathobiont can synergize to worsen IBD symptoms. Targeting the NLRP3 inflammasome or Caspase-8 could be a potential therapeutic strategy in IBD patients with gut inflammation, which is worsened by NSAIDs.


Asunto(s)
Antiinflamatorios no Esteroideos , Colitis , Microbioma Gastrointestinal , Enfermedades Inflamatorias del Intestino , Animales , Ratones , Antiinflamatorios no Esteroideos/efectos adversos , Caspasa 8/metabolismo , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Colitis/microbiología , Inflamasomas , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/microbiología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Inhibidores de Caspasas/farmacología , Escherichia coli/patogenicidad
7.
Diagnostics (Basel) ; 12(1)2022 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-35054292

RESUMEN

Immune checkpoint inhibitors are monoclonal antibodies that are used to treat over one in three cancer patients. While they have changed the natural history of disease, prolonging life and preserving quality of life, they are highly active in less than 40% of patients, even in the most responsive malignancies such as melanoma, and cause significant autoimmune side effects. Licenced biomarkers include tumour Programmed Death Ligand 1 expression by immunohistochemistry, microsatellite instability, and tumour mutational burden, none of which are particularly sensitive or specific. Emerging tumour and immune tissue biomarkers such as novel immunohistochemistry scores, tumour, stromal and immune cell gene expression profiling, and liquid biomarkers such as systemic inflammatory markers, kynurenine/tryptophan ratio, circulating immune cells, cytokines and DNA are discussed in this review. We also examine the influence of the faecal microbiome on treatment outcome and its use as a biomarker of response and toxicity.

8.
Gut Microbes ; 14(1): 2007743, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35023810

RESUMEN

In healthy hosts the gut microbiota is restricted to gut tissues by several barriers some of which require MyD88-dependent innate immune sensor pathways. Nevertheless, some gut taxa have been reported to disseminate to systemic tissues. However, the extent to which this normally occurs during homeostasis in healthy organisms is still unknown. In this study, we recovered viable gut bacteria from systemic tissues of healthy wild type (WT) and MyD88-/- mice. Shotgun metagenomic-sequencing revealed a marked increase in the relative abundance of L. johnsonii in intestinal tissues of MyD88-/- mice compared to WT mice. Lactobacillus johnsonii was detected most frequently from multiple systemic tissues and at higher levels in MyD88-/- mice compared to WT mice. Viable L. johnsonii strains were recovered from different cell types sorted from intestinal and systemic tissues of WT and MyD88-/- mice. L. johnsonii could persist in dendritic cells and may represent murine immunomodulatory endosymbionts.


Asunto(s)
Microbioma Gastrointestinal , Lactobacillus johnsonii/fisiología , Factor 88 de Diferenciación Mieloide/deficiencia , Animales , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Fenómenos Fisiológicos Bacterianos , Células Dendríticas/microbiología , Tracto Gastrointestinal/microbiología , Lactobacillus johnsonii/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética
9.
mBio ; 13(1): e0333421, 2022 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-35089052

RESUMEN

The field of metagenomics has rapidly expanded to become the go-to method for complex microbial community analyses. However, there is currently no straightforward route from metagenomics to traditional culture-based methods of strain isolation, particularly in (bacterio)phage biology, leading to an investigative bottleneck. Here, we describe a method that exploits specific phage receptor binding protein (RBP)-host cell surface receptor interaction enabling isolation of phage-host combinations from an environmental sample. The method was successfully applied to two complex sample types-a dairy-derived whey sample and an infant fecal sample, enabling retrieval of specific and culturable phage hosts. IMPORTANCE PhRACS aims to bridge the current divide between in silico genetic analyses (i.e., phageomic studies) and traditional culture-based methodology. Through the labeling of specific bacterial hosts with fluorescently tagged recombinant phage receptor binding proteins and the isolation of tagged cells using flow cytometry, PhRACS allows the full potential of phageomic data to be realized in the wet laboratory.


Asunto(s)
Bacteriófagos , Microbiota , Humanos , Bacteriófagos/genética , Suero Lácteo , Receptores de Bacteriógrafos , Bacterias/genética , Metagenómica/métodos
10.
Gut Microbes ; 14(1): 2149023, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36420990

RESUMEN

The mechanisms by which early microbial colonizers of the neonate influence gut development are poorly understood. Bacterial bile salt hydrolase (BSH) acts as a putative colonization factor that influences bile acid signatures and microbe-host signaling pathways and we considered whether this activity can influence infant gut development. In silico analysis of the human neonatal gut metagenome confirmed that BSH enzyme sequences are present as early as one day postpartum. Gastrointestinal delivery of cloned BSH to immature gnotobiotic mice accelerated shortening of the colon and regularized gene expression profiles, with monocolonised mice more closely resembling conventionally raised animals. In situ expression of BSH decreased markers of cell proliferation (Ki67, Hes2 and Ascl2) and strongly increased expression of ALPI, a marker of cell differentiation and barrier function. These data suggest an evolutionary paradigm whereby microbial BSH activity potentially influences bacterial colonization and in-turn benefits host gastrointestinal maturation.


Asunto(s)
Microbioma Gastrointestinal , Transcriptoma , Femenino , Humanos , Ratones , Animales , Amidohidrolasas/genética , Amidohidrolasas/metabolismo , Tracto Gastrointestinal/microbiología , Bacterias/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo
11.
Methods Mol Biol ; 2278: 131-139, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33649953

RESUMEN

Members of the Bifidobacterium genus are some of the earliest and most important colonizers of the human neonatal gastrointestinal tract (GIT), exerting wide-ranging effects on early development of the host. However, human isolates of bifidobacteria are very inefficient colonizers of specific-pathogen-free (SPF) mice creating a technical barrier to discovery and applied research in this area. We have developed a reproducible model to facilitate transient colonization of SPF mice with human isolates of this genus through prior depletion of the gut resident microbiota with antibiotics. This chapter outlines the technical details for performing efficient microbiota depletion with antibiotics and subsequent administration of bifidobacteria for colonization.


Asunto(s)
Bifidobacterium/fisiología , Microbioma Gastrointestinal , Ratones/microbiología , Animales , Humanos , Ratones Endogámicos C57BL , Modelos Animales , Organismos Libres de Patógenos Específicos
12.
Sci Rep ; 11(1): 5896, 2021 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-33723368

RESUMEN

The mechanisms through which cells of the host innate immune system distinguish commensal bacteria from pathogens are currently unclear. Toll-like receptors (TLRs) are a class of pattern recognition receptors (PRRs) expressed by host cells which recognize microbe-associated molecular patterns (MAMPs) common to both commensal and pathogenic bacteria. Of the different TLRs, TLR2/6 recognize bacterial lipopeptides and trigger cytokines responses, especially to Gram-positive and Gram-negative pathogens. We report here that TLR2 is dispensable for triggering macrophage cytokine responses to different strains of the Gram-positive commensal bacterial species Lactobacillus salivarius. The L. salivarius UCC118 strain strongly upregulated expression of the PRRs, Mincle (Clec4e), TLR1 and TLR2 in macrophages while downregulating other TLR pathways. Cytokine responses triggered by L. salivarius UCC118 were predominantly TLR2-independent but MyD88-dependent. However, macrophage cytokine responses triggered by another Gram-positive commensal bacteria, Bifidobacterium breve UCC2003 were predominantly TLR2-dependent. Thus, we report a differential requirement for TLR2-dependency in triggering macrophage cytokine responses to different commensal Gram-positive bacteria. Furthermore, TNF-α responses to the TLR2 ligand FSL-1 and L. salivarius UCC118 were partially Mincle-dependent suggesting that PRR pathways such as Mincle contribute to the recognition of MAMPs on distinct Gram-positive commensal bacteria. Ultimately, integration of signals from these different PRR pathways and other MyD88-dependent pathways may determine immune responses to commensal bacteria at the host-microbe interface.


Asunto(s)
Citocinas/metabolismo , Ligilactobacillus salivarius/fisiología , Macrófagos/metabolismo , Macrófagos/microbiología , Factor 88 de Diferenciación Mieloide/metabolismo , Receptor Toll-Like 2/metabolismo , Animales , Humanos , Ligandos , Masculino , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Receptores de Reconocimiento de Patrones/metabolismo , Células THP-1 , Receptor Toll-Like 2/agonistas
13.
Front Microbiol ; 12: 653587, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34220742

RESUMEN

Exopolysaccharide (EPS) is a bacterial extracellular carbohydrate moiety which has been associated with immunomodulatory activity and host protective effects of several gut commensal bacteria. Bifidobacterium breve are early colonizers of the human gastrointestinal tract (GIT) but the role of EPS in mediating their effects on the host has not been investigated for many strains. Here, we characterized EPS production by a panel of human B. breve isolates and investigated the effect of EPS status on host immune responses using human and murine cell culture-based assay systems. We report that B. breve EPS production is heterogenous across strains and that immune responses in human THP-1 monocytes are strain-specific, but not EPS status-specific. Using wild type and isogenic EPS deficient mutants of B. breve strains UCC2003 and JCM7017 we show that EPS had strain-specific divergent effects on cytokine responses from murine bone marrow derived macrophages (BMDMs) and dendritic cells (BMDCs). The B. breve UCC2003 EPS negative (EPS-) strain increased expression of cytokine genes (Tnfa, Il6, Il12a, and Il23a) relative to untreated BMDCs and BMDCs treated with wild type strain. B. breve UCC2003 and JCM7017 EPS- strains increased expression of dendritic cell (DC) activation and maturation marker genes (Cd80, Cd83, and Cd86) relative to untreated BMDCs. Consistent with this, BMDCs co-cultured with B. breve UCC2003 and JCM7017 EPS- strains engineered to express OVA antigen activated OVA-specific OT-II CD4+ T-cells in a co-culture antigen-presentation assay while EPS proficient strains did not. Collectively, these data indicate that B. breve EPS proficient strains use EPS to prevent maturation of DCs and activation of antigen specific CD4+ T cells responses to B. breve. This study identifies a new immunomodulatory role for B. breve EPS and suggests it may be important for immune evasion of adaptive immunity by B. breve and contribute to host-microbe mutualism.

14.
Front Immunol ; 12: 655960, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34394073

RESUMEN

Carcinoembryogenic antigen cellular adhesion molecules (CEACAMs) are intercellular adhesion molecules highly expressed in intestinal epithelial cells. CEACAM1, -3, -5, -6, -7 are altered in patients suffering from colon cancer and inflammatory bowel diseases (IBD), but their role in the onset and pathogenesis of IBD is not well known. Herein, we aim to correlate CEACAM1, -3, -5, -6, -7 expression to the degree of inflammation in pediatric and adult IBD colon biopsies and to examine the regulation of CEACAMs on human intestinal epithelial cell lines (C2BBe1/HT29) by different IBD-associated triggers (cytokines, bacteria/metabolites, emulsifiers) and IBD-drugs (6-Mercaptopurine, Prednisolone, Tofacitinib). Biopsies from patients with pediatric Crohn's disease (CD) and adult ulcerative colitis (UC, active/inactive disease) showed a significant increase in CEACAM3, -5, -6 expression, while CEACAM5 expression was reduced in adult CD patients (active/inactive disease). Intestinal epithelial cells cultured with a pro-inflammatory cytokine cocktail and Adherent-invasive Escherichia coli (AIEC) showed a rapid induction of CEACAM1, -5, -7 followed by a reduced RNA and protein expression overtime and a constant expression of CEACAM3, correlating with IL-8 expression. Cells cultured with the emulsifier polysorbate-80 resulted in a significant induction of CEACAM3, -5, -6, -7 at a late time point, while SCFA treatment reduced CEACAM1, -5, -7 expression. No major alterations in expression of CEACAMs were noted on cells cultured with the commensal Escherichia coli K12 or the pathogen Salmonella typhimurium. IBD drugs, particularly Tofacitinib, significantly reduced cytokine-induced CEACAM1, -3, -5, -6, -7 expression associated with a reduced IL-8 secretion. In conclusion, we provide new evidence on the regulation of CEACAMs by different IBD-associated triggers, identifying a role of CEACAMs in IBD pathogenesis.


Asunto(s)
Antígeno Carcinoembrionario/genética , Moléculas de Adhesión Celular/genética , Susceptibilidad a Enfermedades , Regulación de la Expresión Génica , Enfermedades Inflamatorias del Intestino/etiología , Enfermedades Inflamatorias del Intestino/metabolismo , Mucosa Intestinal/metabolismo , Biopsia , Antígeno Carcinoembrionario/metabolismo , Moléculas de Adhesión Celular/metabolismo , Línea Celular , Enfermedad de Crohn/etiología , Enfermedad de Crohn/metabolismo , Enfermedad de Crohn/patología , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Ácidos Grasos Volátiles/metabolismo , Humanos , Mediadores de Inflamación/metabolismo , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/patología , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Familia de Multigenes , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico
15.
Cell Death Dis ; 12(10): 864, 2021 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-34556638

RESUMEN

Rewiring of host cytokine networks is a key feature of inflammatory bowel diseases (IBD) such as Crohn's disease (CD). Th1-type cytokines-IFN-γ and TNF-α-occupy critical nodes within these networks and both are associated with disruption of gut epithelial barrier function. This may be due to their ability to synergistically trigger the death of intestinal epithelial cells (IECs) via largely unknown mechanisms. In this study, through unbiased kinome RNAi and drug repurposing screens we identified JAK1/2 kinases as the principal and nonredundant drivers of the synergistic killing of human IECs by IFN-γ/TNF-α. Sensitivity to IFN-γ/TNF-α-mediated synergistic IEC death was retained in primary patient-derived intestinal organoids. Dependence on JAK1/2 was confirmed using genetic loss-of-function studies and JAK inhibitors (JAKinibs). Despite the presence of biochemical features consistent with canonical TNFR1-mediated apoptosis and necroptosis, IFN-γ/TNF-α-induced IEC death was independent of RIPK1/3, ZBP1, MLKL or caspase activity. Instead, it involved sustained activation of JAK1/2-STAT1 signalling, which required a nonenzymatic scaffold function of caspase-8 (CASP8). Further modelling in gut mucosal biopsies revealed an intercorrelated induction of the lethal CASP8-JAK1/2-STAT1 module during ex vivo stimulation of T cells. Functional studies in CD-derived organoids using inhibitors of apoptosis, necroptosis and JAKinibs confirmed the causative role of JAK1/2-STAT1 in cytokine-induced death of primary IECs. Collectively, we demonstrate that TNF-α synergises with IFN-γ to kill IECs via the CASP8-JAK1/2-STAT1 module independently of canonical TNFR1 and cell death signalling. This non-canonical cell death pathway may underpin immunopathology driven by IFN-γ/TNF-α in diverse autoinflammatory diseases such as IBD, and its inhibition may contribute to the therapeutic efficacy of anti-TNFs and JAKinibs.


Asunto(s)
Caspasa 8/metabolismo , Células Epiteliales/patología , Interferón gamma/metabolismo , Intestinos/patología , Janus Quinasa 1/metabolismo , Factor de Transcripción STAT1/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Apoptosis , Biopsia , Muerte Celular , Línea Celular Tumoral , Colon/patología , Citoprotección , Células Epiteliales/metabolismo , Humanos , Janus Quinasa 2/metabolismo , Mitocondrias/metabolismo , Organoides/patología , Interferencia de ARN , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Transducción de Señal
16.
Cell Death Dis ; 11(1): 68, 2020 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-31988296

RESUMEN

Proteins of the BCL-2 family are evolutionarily conserved modulators of apoptosis that function as sensors of cellular integrity. Over the past three decades multiple BCL-2 family members have been identified, many of which are now fully incorporated into regulatory networks governing the mitochondrial apoptotic pathway. For some, however, an exact role in cell death signalling remains unclear. One such 'orphan' BCL-2 family member is BCL-G (or BCL2L14). In this study we analysed gastrointestinal expression of human BCL-G in health and disease states, and investigated its contribution to inflammation-induced tissue damage by exposing intestinal epithelial cells (IEC) to IFN-γ and TNF-α, two pro-inflammatory mediators associated with gut immunopathology. We found that both BCL-G splice variants - BCL-GS (short) and BCL-GL (long) - were highly expressed in healthy gut tissue, and that their mRNA levels decreased in active inflammatory bowel diseases (for BCL-GS) and colorectal cancer (for BCL-GS/L). In vitro studies revealed that IFN-γ and TNF-α synergised to upregulate BCL-GS/L and to trigger apoptosis in colonic epithelial cell lines and primary human colonic organoids. Using RNAi, we showed that synergistic induction of IEC death was STAT1-dependent while optimal expression of BCL-GS/L required STAT1, NF-κB/p65 and SWI/SNF-associated chromatin remodellers BRM and BRG1. To test the direct contribution of BCL-G to the effects of IFN-γ and TNF-α on epithelial cells, we used RNAi- and CRISPR/Cas9-based perturbations in parallel with isoform-specific overexpression of BCL-G, and found that BCL-G was dispensable for Th1 cytokine-induced apoptosis of human IEC. Instead, we discovered that depletion of BCL-G differentially affected secretion of inflammatory chemokines CCL5 and CCL20, thus uncovering a non-apoptotic immunoregulatory function of this BCL-2 family member. Taken together, our data indicate that BCL-G may be involved in shaping immune responses in the human gut in health and disease states through regulation of chemokine secretion rather than intestinal apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Quimiocinas/metabolismo , Células Epiteliales/metabolismo , Interferón gamma/farmacología , Mucosa Intestinal/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Apoptosis/genética , Sistemas CRISPR-Cas , Línea Celular Tumoral , Quimiocina CCL20/metabolismo , Quimiocina CCL5/metabolismo , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Células Epiteliales/efectos de los fármacos , Técnicas de Inactivación de Genes , Humanos , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/metabolismo , FN-kappa B/metabolismo , Organoides/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/genética , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Factores de Transcripción/metabolismo , Regulación hacia Arriba
17.
Neuronal Signal ; 3(1): NS20180177, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-32269832

RESUMEN

Microglia are an essential component of the neurogenic niche in the adult hippocampus and are involved in the control of neural precursor cell (NPC) proliferation, differentiation and the survival and integration of newborn neurons in hippocampal circuitry. Microglial and neuronal cross-talk is mediated in part by the chemokine fractalkine/chemokine (C-X3-C motif) ligand 1 (CX3CL1) released from neurons, and its receptor CX3C chemokine receptor 1 (CX3CR1) which is expressed on microglia. A disruption in this pathway has been associated with impaired neurogenesis yet the specific molecular mechanisms by which this interaction occurs remain unclear. The orphan nuclear receptor TLX (Nr2e1; homologue of the Drosophila tailless gene) is a key regulator of hippocampal neurogenesis, and we have shown that in its absence microglia exhibit a pro-inflammatory activation phenotype. However, it is unclear whether a disturbance in CX3CL1/CX3CR1 communication mediates an impairment in TLX-related pathways which may have subsequent effects on neurogenesis. To this end, we assessed miRNA expression of up- and down-stream signalling molecules of TLX in the hippocampus of mice lacking CX3CR1. Our results demonstrate that a lack of CX3CR1 is associated with altered expression of TLX and its downstream targets in the hippocampus without significantly affecting upstream regulators of TLX. Thus, TLX may be a potential participant in neural stem cell (NSC)-microglial cross-talk and may be an important target in understanding inflammatory-associated impairments in neurogenesis.

18.
Neuropharmacology ; 152: 90-101, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-30582955

RESUMEN

Oxytocin mediates its behavioural effects via the centrally expressed oxytocin receptor (OTR). Oxytocin signalling has been implicated in multiple disorders involving centrally regulated pathways, including obesity, autism, schizophrenia and depression. The OTR has been described to have a complex downstream signalling pathway and an increased understanding of oxytocinergic signalling is needed for the development of novel and better treatments for centrally regulated disorders. The ghrelin receptor (GHSR), known primarily for its role in centrally regulated energy balance and food intake, has in more recent years also been shown to play a role in mood disorders, including anxiety and depression. Although there have been suggestions of crosstalk between both signalling systems, these have largely been unexplored to date. Here we show, to our knowledge for the first-time, compelling evidence for the formation of an OTR and GHSR heterocomplex, resulting in significant modulation of OTR downstream signalling. Co-localized expression of the OTR and GHSR is shown in a heterologous cellular expression system and in primary cultures of the hypothalamus and hippocampus. A physical interaction between the OTR and GHSR is confirmed using flow-cytometry based fluorescence resonance energy transfer (fcFRET). Interestingly, co-expression of the GHSR results in a significant attenuation of OTR-mediated Gαq signalling and changes in receptor trafficking within the cell. Together, these data demonstrate a potential functional relevance of an OTR/GHSR heterocomplex and its ability to alter OTR signalling, which is poised to have important implications for future therapeutic strategies, involving oxytocinergic signalling. This article is part of the Special Issue entitled 'Receptor heteromers and their allosteric receptor-receptor interactions'.


Asunto(s)
Receptores de Ghrelina/metabolismo , Receptores de Oxitocina/metabolismo , Ghrelina/metabolismo , Células HEK293 , Humanos , Oxitocina/metabolismo , Unión Proteica , Receptor Cross-Talk
20.
Expert Opin Ther Targets ; 9(5): 1031-44, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16185156

RESUMEN

Fas is a transmembrane receptor that can induce apoptosis after cross-linking with either agonistic antibodies or with Fas ligand (FasL). Although originally described as an important regulator of peripheral immune homeostasis, accumulating evidence suggests that the Fas/FasL system plays an important role in tumour development. In addition to its proapoptotic functions, accumulating evidence demonstrates that Fas can activate numerous nonapoptotic signalling pathways, and that activation of these pathways can result in increased tumourigenicity and metastasis. This review summarises the current understanding of the Fas/FasL system in tumorigenesis and discusses attempts to utilise the Fas/FasL system in the treatment of cancer.


Asunto(s)
Antineoplásicos/farmacología , Proteína Ligando Fas/metabolismo , Neoplasias/tratamiento farmacológico , Receptor fas/metabolismo , Antineoplásicos/uso terapéutico , Humanos , Neoplasias/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA