Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 296
Filtrar
1.
Cell Mol Gastroenterol Hepatol ; 18(2): 101350, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38704148

RESUMEN

BACKGROUND & AIMS: Gut bacterial sphingolipids, primarily produced by Bacteroidetes, have dual roles as bacterial virulence factors and regulators of the host mucosal immune system, including regulatory T cells and invariant natural killer T cells. Patients with inflammatory bowel disease display altered sphingolipids profiles in fecal samples. However, how bacterial sphingolipids modulate mucosal homeostasis and regulate intestinal inflammation remains unclear. METHODS: We used dextran sodium sulfate (DSS)-induced colitis in mice monocolonized with Bacteroides fragilis strains expressing or lacking sphingolipids to assess the influence of bacterial sphingolipids on intestinal inflammation using transcriptional, protein, and cellular analyses. Colonic explant and organoid were used to study the function of bacterial sphingolipids. Host mucosal immune cells and cytokines were profiled and characterized using flow cytometry, enzyme-linked immunosorbent assay, and Western blot, and cytokine function in vivo was investigated by monoclonal antibody injection. RESULTS: B fragilis sphingolipids exacerbated intestinal inflammation. Mice monocolonized with B fragilis lacking sphingolipids exhibited less severe DSS-induced colitis. This amelioration of colitis was associated with increased production of interleukin (IL)-22 by ILC3. Mice colonized with B fragilis lacking sphingolipids following DSS treatment showed enhanced epithelial STAT3 activity, intestinal cell proliferation, and antimicrobial peptide production. Protection against DSS colitis associated with B fragilis lacking sphingolipids was reversed on IL22 blockade. Furthermore, bacterial sphingolipids restricted epithelial IL18 production following DSS treatment and interfered with IL22 production by a subset of ILC3 cells expressing both IL18R and major histocompatibility complex class II. CONCLUSIONS: B fragilis-derived sphingolipids exacerbate mucosal inflammation by impeding epithelial IL18 expression and concomitantly suppressing the production of IL22 by ILC3 cells.


Asunto(s)
Bacteroides fragilis , Colitis , Sulfato de Dextran , Interleucina-22 , Interleucinas , Esfingolípidos , Animales , Esfingolípidos/metabolismo , Interleucinas/metabolismo , Ratones , Colitis/inmunología , Colitis/patología , Colitis/inducido químicamente , Colitis/microbiología , Colitis/metabolismo , Sulfato de Dextran/toxicidad , Bacteroides fragilis/inmunología , Modelos Animales de Enfermedad , Humanos , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Mucosa Intestinal/microbiología , Mucosa Intestinal/metabolismo , Factor de Transcripción STAT3/metabolismo , Ratones Endogámicos C57BL
2.
Gut Microbes ; 16(1): 2350156, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38726597

RESUMEN

Extensive research has explored the role of gut microbiota in colorectal cancer (CRC). Nonetheless, metatranscriptomic studies investigating the in situ functional implications of host-microbe interactions in CRC are scarce. Therefore, we characterized the influence of CRC core pathogens and biofilms on the tumor microenvironment (TME) in 40 CRC, paired normal, and healthy tissue biopsies using fluorescence in situ hybridization (FISH) and dual-RNA sequencing. FISH revealed that Fusobacterium spp. was associated with increased bacterial biomass and inflammatory response in CRC samples. Dual-RNA sequencing demonstrated increased expression of pro-inflammatory cytokines, defensins, matrix-metalloproteases, and immunomodulatory factors in CRC samples with high bacterial activity. In addition, bacterial activity correlated with the infiltration of several immune cell subtypes, including M2 macrophages and regulatory T-cells in CRC samples. Specifically, Bacteroides fragilis and Fusobacterium nucleatum correlated with the infiltration of neutrophils and CD4+ T-cells, respectively. The collective bacterial activity/biomass appeared to exert a more significant influence on the TME than core pathogens, underscoring the intricate interplay between gut microbiota and CRC. These results emphasize how biofilms and core pathogens shape the immune phenotype and TME in CRC while highlighting the need to extend the bacterial scope beyond CRC pathogens to advance our understanding and identify treatment targets.


Asunto(s)
Biopelículas , Neoplasias Colorrectales , Microbioma Gastrointestinal , Microambiente Tumoral , Neoplasias Colorrectales/microbiología , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , Humanos , Biopelículas/crecimiento & desarrollo , Microambiente Tumoral/inmunología , Masculino , Femenino , Bacterias/clasificación , Bacterias/genética , Bacterias/inmunología , Persona de Mediana Edad , Hibridación Fluorescente in Situ , Anciano , Fusobacterium nucleatum/inmunología , Citocinas/metabolismo , Macrófagos/inmunología , Macrófagos/microbiología , Fenotipo , Bacteroides fragilis/inmunología , Bacteroides fragilis/fisiología , Bacteroides fragilis/genética
3.
Pathology ; 56(4): 528-539, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38609782

RESUMEN

This study explored the relationship between faecal microbiota distribution and local or systemic immune response in patients with colorectal cancer (CRC). The study population included 114 surgically treated CRC patients. Faeces were analysed using 16S rRNA gene sequencing. The immune score in tumour microenvironment was evaluated using CD3 and CD8 immunohistochemistry. Genetic alterations, microsatellite instability status and five systemic inflammatory markers were also analysed. Thirty of 114 (26.3%) CRC patients were categorised as the 'immune type' with a high density of T-cells. The immune type CRC cases showed lower angiolymphatic invasion and longer overall survival. Of the 123 selected bacterial species, Bacteroides fragilis and Collinsella aerofaciens were prevalent in immune CRC cases, whereas Odoribacter splanchnicus and Phascolarctobacterium succinatutens were prevalent in non-immune CRC patients. Bacteroides fragilis was associated with shorter disease free survival in univariable and multivariable survival analyses. Regarding systemic immunity, a high prevalence of C. aerofaciens was associated with a high modified Glasgow prognostic score. This study revealed a potential relationship among the gut microbiome, immune microenvironment, and disease progression in patients with CRC. Our findings suggest that abundant B. fragilis in patients with CRC is associated with a 'cold immune' tumour microenvironment.


Asunto(s)
Neoplasias Colorrectales , Microbioma Gastrointestinal , Microambiente Tumoral , Humanos , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/microbiología , Neoplasias Colorrectales/cirugía , Microambiente Tumoral/inmunología , Masculino , Femenino , Persona de Mediana Edad , Anciano , Heces/microbiología , Adulto , Anciano de 80 o más Años , ARN Ribosómico 16S/genética , Pronóstico , Bacteroides fragilis/inmunología
4.
Nature ; 622(7984): 826-833, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37853119

RESUMEN

CRISPR systems are widespread in the prokaryotic world, providing adaptive immunity against mobile genetic elements1,2. Type III CRISPR systems, with the signature gene cas10, use CRISPR RNA to detect non-self RNA, activating the enzymatic Cas10 subunit to defend the cell against mobile genetic elements either directly, via the integral histidine-aspartate (HD) nuclease domain3-5 or indirectly, via synthesis of cyclic oligoadenylate second messengers to activate diverse ancillary effectors6-9. A subset of type III CRISPR systems encode an uncharacterized CorA-family membrane protein and an associated NrN family phosphodiesterase that are predicted to function in antiviral defence. Here we demonstrate that the CorA-associated type III-B (Cmr) CRISPR system from Bacteroides fragilis provides immunity against mobile genetic elements when expressed in Escherichia coli. However, B. fragilis Cmr does not synthesize cyclic oligoadenylate species on activation, instead generating S-adenosyl methionine (SAM)-AMP (SAM is also known as AdoMet) by conjugating ATP to SAM via a phosphodiester bond. Once synthesized, SAM-AMP binds to the CorA effector, presumably leading to cell dormancy or death by disruption of the membrane integrity. SAM-AMP is degraded by CRISPR-associated phosphodiesterases or a SAM-AMP lyase, potentially providing an 'off switch' analogous to cyclic oligoadenylate-specific ring nucleases10. SAM-AMP thus represents a new class of second messenger for antiviral signalling, which may function in different roles in diverse cellular contexts.


Asunto(s)
Adenosina Trifosfato , Bacteroides fragilis , Sistemas CRISPR-Cas , Escherichia coli , S-Adenosilmetionina , Sistemas de Mensajero Secundario , Adenosina Trifosfato/metabolismo , Bacteroides fragilis/enzimología , Bacteroides fragilis/genética , Bacteroides fragilis/inmunología , Proteínas Asociadas a CRISPR/genética , Proteínas Asociadas a CRISPR/metabolismo , Sistemas CRISPR-Cas/genética , Sistemas CRISPR-Cas/inmunología , Sistemas CRISPR-Cas/fisiología , Endonucleasas/química , Endonucleasas/metabolismo , Escherichia coli/genética , Escherichia coli/crecimiento & desarrollo , Escherichia coli/inmunología , Escherichia coli/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Hidrolasas Diéster Fosfóricas/genética , Hidrolasas Diéster Fosfóricas/metabolismo , ARN/inmunología , ARN/metabolismo , S-Adenosilmetionina/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo
5.
Front Immunol ; 12: 662807, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34025663

RESUMEN

The symbiotic relationship between animals and their resident microorganisms has profound effects on host immunity. The human microbiota comprises bacteria that reside in the gastrointestinal tract and are involved in a range of inflammatory and autoimmune diseases. The gut microbiota's immunomodulatory effects extend to extraintestinal tissues, including the central nervous system (CNS). Specific symbiotic antigens responsible for inducing immunoregulation have been isolated from different bacterial species. Polysaccharide A (PSA) of Bacteroides fragilis is an archetypical molecule for host-microbiota interactions. Studies have shown that PSA has beneficial effects in experimental disease models, including experimental autoimmune encephalomyelitis (EAE), the most widely used animal model for multiple sclerosis (MS). Furthermore, in vitro stimulation with PSA promotes an immunomodulatory phenotype in human T cells isolated from healthy and MS donors. In this review, we discuss the current understanding of the interactions between gut microbiota and the host in the context of CNS inflammatory demyelination, the immunomodulatory roles of gut symbionts. More specifically, we also discuss the immunomodulatory effects of B. fragilis PSA in the gut-brain axis and its therapeutic potential in MS. Elucidation of the molecular mechanisms responsible for the microbiota's impact on host physiology offers tremendous promise for discovering new therapies.


Asunto(s)
Encéfalo/metabolismo , Enfermedades Desmielinizantes/etiología , Enfermedades Desmielinizantes/metabolismo , Susceptibilidad a Enfermedades , Retroalimentación Fisiológica , Tracto Gastrointestinal/metabolismo , Animales , Bacteroides fragilis/inmunología , Enfermedades Desmielinizantes/patología , Encefalomielitis Autoinmune Experimental/inmunología , Microbioma Gastrointestinal/inmunología , Humanos , Inmunomodulación , Polisacáridos Bacterianos/inmunología
6.
J Immunol ; 206(10): 2441-2452, 2021 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-33941658

RESUMEN

Intestinal barrier is essential for dietary products and microbiota compartmentalization and therefore gut homeostasis. When this barrier is broken, cecal content overflows into the peritoneal cavity, leading to local and systemic robust inflammatory response, characterizing peritonitis and sepsis. It has been shown that IL-1ß contributes with inflammatory storm during peritonitis and sepsis and its inhibition has beneficial effects to the host. Therefore, we investigated the mechanisms underlying IL-1ß secretion using a widely adopted murine model of experimental peritonitis. The combined injection of sterile cecal content (SCC) and the gut commensal bacteria Bacteroides fragilis leads to IL-1ß-dependent peritonitis, which was mitigated in mice deficient in NLRP3 (nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3) inflammasome components. Typically acting as a damage signal, SCC, but not B. fragilis, activates canonical pathway of NLRP3 promoting IL-1ß secretion in vitro and in vivo. Strikingly, absence of fiber in the SCC drastically reduces IL-1ß production, whereas high-fiber SCC conversely increases this response in an NLRP3-dependent manner. In addition, NLRP3 was also required for IL-1ß production induced by purified dietary fiber in primed macrophages. Extending to the in vivo context, IL-1ß-dependent peritonitis was worsened in mice injected with B. fragilis and high-fiber SCC, whereas zero-fiber SCC ameliorates the pathology. Corroborating with the proinflammatory role of dietary fiber, IL-1R-deficient mice were protected from peritonitis induced by B. fragilis and particulate bran. Overall, our study highlights a function, previously unknown, for dietary fibers in fueling peritonitis through NLRP3 activation and IL-1ß secretion outside the gut.


Asunto(s)
Infecciones por Bacteroides/inmunología , Bacteroides fragilis/inmunología , Fibras de la Dieta/efectos adversos , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/deficiencia , Peritonitis/inmunología , Animales , Infecciones por Bacteroides/microbiología , Dieta , Fibras de la Dieta/administración & dosificación , Modelos Animales de Enfermedad , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Peritonitis/microbiología , Receptores de Interleucina-1/deficiencia , Receptores de Interleucina-1/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología
7.
Immunol Lett ; 233: 48-56, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33741378

RESUMEN

It has been reported that patients with rheumatoid arthritis (RA) have significantly less bacteria belonging to the Bacteroides group in their microbiota. We speculate that inhibition of cytokine production is impaired in patients with RA owing to their low levels of intestinal bacteria belonging to the Bacteroidetes group. Here we investigated the effect of Bacteroides fragilis lipopolysaccharide (B-LPS) on cytokine production in vitro and on the development of collagen antibody-induced arthritis (CAIA) in DBA/1 mice, an animal model of RA. in vitro culture experiments showed that Escherichia coli LPS (E-LPS)-induced cytokine production from THP-1 monocytic cells and peripheral blood mononuclear cells was significantly suppressed by B-LPS in a dose-dependent manner. A decrease in TNF-α and IL-1ß production was also observed in LPS-tolerized macrophages induced by B-LPS at concentrations equal to and higher than that of E-LPS. Similar results were obtained when autoclaved feces were used to induce cytokine production instead of E-LPS. In in vivo experiments using CAIA models, B-LPS had no adverse effects even when administered at 10 times the concentration of E-LPS, which elicits severe arthritis. In addition, simultaneous administration of high dose B-LPS with E-LPS or administration of B-LPS prior to E-LPS significantly suppressed arthritis development in CAIA model animals when compared with administration of E-LPS alone. These results suggest that increasing certain bacterial groups such as Bacteroides is an effective strategy for preventing arthritis development in patients with RA.


Asunto(s)
Artritis Reumatoide/etiología , Artritis Reumatoide/metabolismo , Bacteroides fragilis/inmunología , Escherichia coli/inmunología , Lipopolisacáridos/inmunología , Monocitos/inmunología , Monocitos/metabolismo , Animales , Artritis Experimental , Artritis Reumatoide/diagnóstico por imagen , Artritis Reumatoide/patología , Línea Celular , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Relación Dosis-Respuesta Inmunológica , Endotoxinas/inmunología , Humanos , Tolerancia Inmunológica , Masculino , Ratones , Monocitos/patología , Índice de Severidad de la Enfermedad , Microtomografía por Rayos X
8.
JCI Insight ; 6(3)2021 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-33554953

RESUMEN

Graft-versus-host disease (GVHD) is a pathological process caused by an exaggerated donor lymphocyte response to host antigens after allogeneic hematopoietic cell transplantation (allo-HCT). Donor T cells undergo extensive clonal expansion and differentiation, which culminate in damage to recipient target organs. Damage to the gastrointestinal tract is a main contributor to morbidity and mortality. The loss of diversity among intestinal bacteria caused by pretransplant conditioning regimens leads to an outgrowth of opportunistic pathogens and exacerbated GVHD after allo-HCT. Using murine models of allo-HCT, we found that an increase of Bacteroides in the intestinal microbiota of the recipients was associated with reduced GVHD in mice given fecal microbial transplantation. Administration of Bacteroides fragilis through oral gavage increased gut microbiota diversity and beneficial commensal bacteria and significantly ameliorated acute and chronic GVHD development. Preservation of gut integrity following B. fragilis exposure was likely attributed to increased short chain fatty acids, IL-22, and regulatory T cells, which in turn improved gut tight junction integrity and reduced inflammatory cytokine production of pathogenic T cells. The current study provides a proof of concept that a single strain of commensal bacteria can be a safe and effective means to protect gut integrity and ameliorate GVHD after allo-HCT.


Asunto(s)
Bacteroides fragilis/inmunología , Microbioma Gastrointestinal/inmunología , Enfermedad Injerto contra Huésped/prevención & control , Aloinjertos , Animales , Modelos Animales de Enfermedad , Trasplante de Microbiota Fecal , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/microbiología , Efecto Injerto vs Leucemia/inmunología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Isoantígenos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Linfocitos T/inmunología , Células Tumorales Cultivadas
9.
Infect Immun ; 88(8)2020 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-32457103

RESUMEN

The human intestinal anaerobic commensal and opportunistic pathogen Bacteroides fragilis does not synthesize the tetrapyrrole protoporphyrin IX in order to form heme that is required for growth stimulation and survival in vivo Consequently, B. fragilis acquires essential heme from host tissues during extraintestinal infection. The absence of several genes necessary for de novo heme biosynthesis is a common characteristic of many anaerobic bacteria; however, the uroS gene, encoding a uroporphyrinogen III synthase for an early step of heme biosynthesis, is conserved among the heme-requiring Bacteroidales that inhabit the mammalian gastrointestinal tract. In this study, we show that the ability of B. fragilis to utilize heme or protoporphyrin IX for growth was greatly reduced in a ΔuroS mutant. This growth defect appears to be linked to the suppression of reverse chelatase and ferrochelatase activities in the absence of uroS In addition, this ΔuroS suppressive effect was enhanced by the deletion of the yifB gene, which encodes an Mg2+-chelatase protein belonging to the ATPases associated with various cellular activities (AAA+) superfamily of proteins. Furthermore, the ΔuroS mutant and the ΔuroS ΔyifB double mutant had a severe survival defect compared to the parent strain in competitive infection assays using animal models of intra-abdominal infection and intestinal colonization. This shows that the presence of the uroS and yifB genes in B. fragilis seems to be linked to pathophysiological and nutritional competitive fitness for survival in host tissues. Genetic complementation studies and enzyme kinetics assays indicate that B. fragilis UroS is functionally different from canonical bacterial UroS proteins. Taken together, these findings show that heme assimilation and metabolism in the anaerobe B. fragilis have diverged from those of aerobic and facultative anaerobic pathogenic bacteria.


Asunto(s)
Proteínas Bacterianas/genética , Infecciones por Bacteroides/microbiología , Bacteroides fragilis/genética , Bacteroides fragilis/patogenicidad , Ferroquelatasa/genética , Hemo/metabolismo , Uroporfirinógeno III Sintetasa/genética , Animales , Proteínas Bacterianas/inmunología , Infecciones por Bacteroides/inmunología , Infecciones por Bacteroides/metabolismo , Infecciones por Bacteroides/patología , Bacteroides fragilis/inmunología , Unión Competitiva , Transporte Biológico , Ferroquelatasa/inmunología , Eliminación de Gen , Regulación de la Expresión Génica , Prueba de Complementación Genética , Hemo/inmunología , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Infecciones Intraabdominales/inmunología , Infecciones Intraabdominales/metabolismo , Infecciones Intraabdominales/microbiología , Infecciones Intraabdominales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Unión Proteica , Ratas Sprague-Dawley , Uroporfirinógeno III Sintetasa/inmunología , Virulencia
10.
Sci Rep ; 9(1): 19780, 2019 12 24.
Artículo en Inglés | MEDLINE | ID: mdl-31874963

RESUMEN

Enterotoxigenic Bacteroides fragilis is an enteric pathogen which is described as a causative agent of various intestinal infections and inflammatory diseases. Moreover, various research studies have reported it to be a leading factor in the development of colorectal cancer. As a part of the normal human microbiome, its treatment has become quite a challenge due to the alarming resistance against the available antibiotics. Although, this particular strain of B. fragilis shows susceptibility to few antibiotics, it is pertinent to devise an effective vaccine strategy for its elimination. There is no vaccine available against this pathogen up to date; therefore, we systematically ventured the outer membrane toxin producing proteins found exclusively in the toxigenic B. fragilis through the in-silico approaches to predict a multi-epitopic chimeric vaccine construct. The designed protein constitutes of epitopes which are predicted for linear B cells, Helper and T cells of outer membrane proteins expected to be putative vaccine candidates. The finalized proteins are only expressed in the enterotoxigenic B. fragilis, thus proving them to be exclusive. The 3D structure of the protein was first predicted followed by its refinement and validation via utilizing the bioinformatic approaches. Docking of the designed protein with the TLR2 receptor forecasted apt binding. Upon immune simulation, notable levels were observed in the expression of the immune cells.


Asunto(s)
Proteínas Bacterianas , Vacunas Bacterianas , Bacteroides fragilis , Biología Computacional , Epítopos , Simulación del Acoplamiento Molecular , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Vacunas Bacterianas/química , Vacunas Bacterianas/genética , Vacunas Bacterianas/inmunología , Bacteroides fragilis/química , Bacteroides fragilis/genética , Bacteroides fragilis/inmunología , Epítopos/química , Epítopos/genética , Epítopos/inmunología , Humanos , Receptor Toll-Like 2/química , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/inmunología
11.
J Lipid Res ; 60(11): 1892-1904, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31484693

RESUMEN

The glycosphingolipid, α-galactosylceramide (αGalCer), when presented by CD1d on antigen-presenting cells, efficiently activates invariant natural killer T (iNKT) cells. Thereby, it modulates immune responses against tumors, microbial and viral infections, and autoimmune diseases. Recently, the production of αGalCer by Bacteroidetes from the human gut microbiome was elucidated. Using hydrophilic interaction chromatography coupled to MS2, we screened murine intestinal tracts to identify and quantify αGalCers, and we investigated the αGalCer response to different dietary and physiologic conditions. In both the cecum and the colon of mice, we found 1-15 pmol of αGalCer per milligram of protein; in contrast, mice lacking microbiota (germ-free mice) and fed identical diet did not harbor αGalCer. The identified αGalCer contained a ß(R)-hydroxylated hexadecanoyl chain N-linked to C18-sphinganine, which differed from what has been reported with Bacteroides fragilis Unlike ß-anomeric structures, but similar to αGalCers from B. fragilis, the synthetic form of the murine αGalCer induced iNKT cell activation in vitro. Last, we observed a decrease in αGalCer production in mice exposed to conditions that alter the composition of the gut microbiota, including Western type diet, colitis, and influenza A virus infection. Collectively, this study suggests that αGalCer is produced by commensals in the mouse intestine and reveals that stressful conditions causing dysbiosis alter its synthesis. The consequences of this altered production on iNKT cell-mediated local and systemic immune responses are worthy of future studies.


Asunto(s)
Bacteroides fragilis/química , Bacteroides fragilis/inmunología , Dieta , Galactosilceramidas/inmunología , Inflamación/inmunología , Intestino Grueso/inmunología , Intestino Grueso/metabolismo , Animales , Galactosilceramidas/genética , Inflamación/microbiología , Intestino Grueso/microbiología , Ratones , Ratones Endogámicos
12.
J Leukoc Biol ; 106(5): 1079-1088, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31166618

RESUMEN

The intestinal microbiota has several effects on host physiology. Previous work from our laboratory demonstrated that the microbiota influences systemic iron homeostasis in mouse colitis models by altering inflammation-induced expression of the iron-regulating hormone hepcidin. In the present study, we examined the impact of the gut commensal bacterium Bacteroides fragilis on the expression of the iron exporter ferroportin, the target of hepcidin action, in macrophages, the cell type that plays a pivotal role in iron recycling. Mouse bone marrow-derived macrophages were exposed to B. fragilis and were analyzed by quantitative real-time polymerase chain reaction and Western blotting. We found that B. fragilis down-regulated ferroportin transcription independently of bacterial viability. Medium conditioned by the bacteria also reduced ferroportin expression, indicating the involvement of soluble factors, possibly Toll-like receptor ligands. Consistent with this idea, several of these ligands were able to down-regulate ferroportin. The B. fragilis-induced decrease in ferroportin was functionally important since it produced a significant increase in intracellular iron concentrations that prevented the effects of the iron chelator deferoxamine on Salmonella-induced IL-6 and IL-1ß production. Our results thus reveal that B. fragilis can influence macrophage iron handling and inflammatory responses by modulating ferroportin expression.


Asunto(s)
Bacteroides fragilis/inmunología , Proteínas de Transporte de Catión/inmunología , Regulación hacia Abajo/inmunología , Homeostasis/inmunología , Hierro/inmunología , Macrófagos/inmunología , Animales , Macrófagos/microbiología , Ratones
13.
Nat Commun ; 10(1): 2153, 2019 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-31089128

RESUMEN

The gut commensal Bacteroides fragilis or its capsular polysaccharide A (PSA) can prevent various peripheral and CNS sterile inflammatory disorders. Fatal herpes simplex encephalitis (HSE) results from immune pathology caused by uncontrolled invasion of the brainstem by inflammatory monocytes and neutrophils. Here we assess the immunomodulatory potential of PSA in HSE by infecting PSA or PBS treated 129S6 mice with HSV1, followed by delayed Acyclovir (ACV) treatment as often occurs in the clinical setting. Only PSA-treated mice survived, with dramatically reduced brainstem inflammation and altered cytokine and chemokine profiles. Importantly, PSA binding by B cells is essential for induction of regulatory CD4+ and CD8+ T cells secreting IL-10 to control innate inflammatory responses, consistent with the lack of PSA mediated protection in Rag-/-, B cell- and IL-10-deficient mice. Our data reveal the translational potential of PSA as an immunomodulatory symbiosis factor to orchestrate robust protective anti-inflammatory responses during viral infections.


Asunto(s)
Bacteroides fragilis/inmunología , Encefalitis por Herpes Simple/inmunología , Microbioma Gastrointestinal/inmunología , Herpesvirus Humano 1/inmunología , Polisacáridos Bacterianos/inmunología , Aciclovir/uso terapéutico , Animales , Antivirales/uso terapéutico , Linfocitos B/inmunología , Linfocitos B/metabolismo , Bacteroides fragilis/metabolismo , Chlorocebus aethiops , Modelos Animales de Enfermedad , Encefalitis por Herpes Simple/tratamiento farmacológico , Encefalitis por Herpes Simple/virología , Femenino , Herpesvirus Humano 1/patogenicidad , Interacciones Microbiota-Huesped/inmunología , Humanos , Interleucina-10/genética , Interleucina-10/inmunología , Interleucina-10/metabolismo , Masculino , Ratones , Ratones Noqueados , Polisacáridos Bacterianos/metabolismo , Simbiosis/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Células Vero
14.
Transfusion ; 59(5): 1651-1656, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30811034

RESUMEN

BACKGROUND: It is well known that specific groups of patients immunologically respond more readily than others to red blood cell (RBC) antigens. While allogeneic RBC antigen exposure is the primary determinant of alloantibody formation, other variables are also involved. Given the significant primary sequence identity between common RBC and microbial antigens, we hypothesized that certain individuals may be immunologically primed to form RBC alloantibodies via environmental exposure to cross-reactive microbial epitopes, and that such a correlation may be linked to blood group antigen immunogenicity. STUDY DESIGN AND METHODS: We examined the relationship between RBC-microbe peptide homology and the formation of alloantibodies to the most immunogenic RBC antigens, using the BLASTp homology database. Thirteen-residue peptides centered on the polymorphic amino acids of K, Jka , Lua , E, c, M, C, and S antigens were queried for identity with microbial peptides using the BLASTp database. Results were restricted to bacteria and fungi, with a selective threshold of >80% identity for inclusion, to allow for minor peptide variability. RESULTS: Significant peptide identity was found between RBC antigens and pathogenic organisms including B. fragilis, P. aeruginosa, and Candida spp., among others. Linear regression and k-medoids clustering analysis of the microbial genera meeting the inclusion criteria showed a statistically significant inverse correlation with RBC immunogenicity (b = -0.0017, r2 = 0.624 & p = 0.0197), with lower immunogenicity antigens associated with larger number of genera. CONCLUSIONS: Our findings raise a potential relationship between microbial exposure and alloantibody formation, and lead to interesting questions regarding the potential relationship between RBC antigen immunogenicity and microbial prevalence.


Asunto(s)
Antígenos de Grupos Sanguíneos/inmunología , Eritrocitos/metabolismo , Isoanticuerpos/inmunología , Aminoácidos/genética , Bacteroides fragilis/inmunología , Candida/inmunología , Humanos , Pseudomonas aeruginosa/inmunología
15.
Immunopharmacol Immunotoxicol ; 41(2): 207-213, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30706742

RESUMEN

Objective: Inflammatory bowel disease (IBD) is generally considered as a major risk factor in the progression of colitis-associated colorectal cancer (CAC). Previous studies have indicated that the composition of gut microflora may be involved in CAC induction and progress. Bacteroides fragilis (BF) is a Gram-negative anaerobe belonging to colonic symbiotic bacteria of the host. This study was aimed to investigate the protective role of BF in a colorectal cancer (CRC) model induced by azoxymethane (AOM) and dextran sulfate sodium (DSS) in germ-free (GF) mice. Materials and methods: Total 22 GF mice were divided into two groups: GF and BF group. Half of the GF mice were colonized with BF for 28 days before CRC induction by AOM/DSS. Results: BF colonization increased animal survival (100%). Cecum weight and cecum/body weight ratio significantly decreased in BF/AOM/DSS group. Interestingly, there was a significant decrease in tumor number and tumor incidence in the BF/AOM/DSS group as compared to the GF/AOM/DSS group. The adenocarcinoma/adenoma incidence and histologic score were also decreased in the BF/AOM/DSS group. In addition, immunohistochemistry staining found decreased numbers of cell proliferation (PCNA) and inflammatory cell (granulocytes) infiltration in the colon mucosa of the BF group. The ß-catenin staining in the BF/AOM/DSS group had fewer and weaker positive signal expressions. Taking together, the BF colonization significantly ameliorated AOM/DSS-induced CRC by suppressing the activity of cell proliferation-related molecules and reducing the number of inflammatory cells. Conclusions: Symbiotic BF may play a pivotal role in maintaining the gastrointestinal immunophysiologic balance and regulating anti-tumorigenesis responses.


Asunto(s)
Azoximetano/toxicidad , Bacteroides fragilis/inmunología , Colitis , Neoplasias Colorrectales , Sulfato de Dextran/toxicidad , Vida Libre de Gérmenes , Animales , Colitis/inducido químicamente , Colitis/inmunología , Colitis/patología , Colitis/prevención & control , Neoplasias Colorrectales/inducido químicamente , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/prevención & control , Masculino , Ratones
16.
Appl Microbiol Biotechnol ; 103(5): 2353-2365, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30666361

RESUMEN

Lipopolysaccharide (LPS) can promote the expression of pro-inflammatory cytokines, damage the tight junction of epithelial walls, and thereby lead to chronic low-grade intestinal inflammatory disorders. Evidences of many beneficial functions from Bacteroides strains suggest their intervention capabilities in LPS-induced inflammation. In the present study, both healthy and LPS-treated mice were consistently treated with Bacteroides strains for 5 days. The intestinal microbiota alteration, epithelial permeability, cytokine expression, and autoimmune and innate immune responses were analyzed. B. fragilis HCK-B3 and B. ovatus ELH-B2 from our laboratory collection were demonstrated to assist intestinal equilibrium by maintaining the diversity of gut microbiota and relieve LPS-induced inflammation by either modulating cytokine production or restoring the Treg/Th-17 balance. Our research indicated that the Bacteroides strains with capabilities of alleviating inflammation have the potential as therapeutics to prevent intestinal inflammatory disorders and provided scientific supports for discovering next-generation probiotics.


Asunto(s)
Bacteroides fragilis/inmunología , Bacteroides fragilis/metabolismo , Probióticos/metabolismo , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Animales , Bacteroides fragilis/clasificación , Citocinas/biosíntesis , Citocinas/inmunología , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Inflamación/terapia , Lipopolisacáridos/toxicidad , Ratones , Ratones Endogámicos C57BL
17.
Mucosal Immunol ; 12(1): 164-177, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30279518

RESUMEN

Polysaccharide A (PSA), an immunogenic capsular component of non-toxigenic Bacteroides fragilis (NTBF) strain NCTC 9343, is reported to promote mucosal immune development and suppress colitis. Contrastingly, enterotoxigenic Bacteroides fragilis (ETBF) is highly associated with inflammatory bowel disease (IBD) and colorectal cancer (CRC), rapidly inducing IL-17-dependent murine colitis and tumorigenesis. In specific-pathogen-free (SPF) C57BL/6 wild-type (WT) and multiple intestinal neoplasia (MinApc716+/-) mice, we show that sequential treatment of the NTBF strain, 9343, followed by the ETBF strain, 86-5443-2-2 (86), diminished colitis and tumorigenesis. Mice treated simultaneously with 9343 and 86 exhibited both severe colitis and tumorigenesis. Abrogated disease severity in sequentially treated mice was attributed to 9343 strain dominance and decreased IL-17A, but 86 colonization prior to or simultaneous with 9343 mitigated the anti-inflammatory effect of 9343. Remarkably, 9343-mediated protection was independent of PSA, as sequentially treated mice receiving ΔPSA 9343 exhibited similar protection. Further, SPF WT and Min mice colonized with PSA-competent or PSA-deficient 9343 exhibited similar IL-10, IL-17, and IFN-γ responses. Treatment of 86-colonized mice with 9343 failed to disrupt 86 pathogenesis. Our findings demonstrate that 9343 colonization, independent of PSA, offers prophylaxis against colitis-inducing 86 but may not be a valid therapy once colitis is established.


Asunto(s)
Bacteroides fragilis/inmunología , Colitis/inmunología , Neoplasias Colorrectales/inmunología , Enfermedades Inflamatorias del Intestino/inmunología , Mucosa Intestinal/inmunología , Células Th17/inmunología , Proteína de la Poliposis Adenomatosa del Colon/genética , Animales , Bacteroides fragilis/patogenicidad , Carcinogénesis , Células Cultivadas , Colitis/inducido químicamente , Modelos Animales de Enfermedad , Humanos , Interleucina-17/metabolismo , Lipopolisacáridos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ácido Trinitrobencenosulfónico
18.
Clin Exp Immunol ; 194(2): 153-165, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30076785

RESUMEN

Ubiquitin is highly conserved across eukaryotes and is essential for normal eukaryotic cell function. The bacterium Bacteroides fragilis is a member of the normal human gut microbiota, and the only bacterium known to encode a homologue of eukaryotic ubiquitin. The B. fragilis gene sequence indicates a past horizontal gene transfer event from a eukaryotic source. It encodes a protein (BfUbb) with 63% identity to human ubiquitin which is exported from the bacterial cell. The aim of this study was (i) to determine if there was antigenic cross-reactivity between B. fragilis ubiquitin and human ubiquitin and (ii) to determine if humans produced antibodies to BfUbb. Molecular model comparisons of BfUbb and human ubiquitin predicted a high level (99·8% confidence) of structural similarity. Linear epitope mapping identified epitopes in BfUbb and human ubiquitin that cross-react. BfUbb also has epitope(s) that do not cross-react with human ubiquitin. The reaction of human serum (n = 474) to BfUbb and human ubiquitin from the following four groups of subjects was compared by enzyme-linked immunosorbent assay (ELISA): (1) newly autoantibody-positive patients, (2) allergen-specific immunoglobulin (Ig)E-negative patients, (3) ulcerative colitis patients and (4) healthy volunteers. We show that the immune system of some individuals has been exposed to BfUbb which has resulted in the generation of IgG antibodies. Serum from patients referred for first-time testing to an immunology laboratory for autoimmune disease are more likely to have a high level of antibodies to BfUbb than healthy volunteers. Molecular mimicry of human ubiquitin by BfUbb could be a trigger for autoimmune disease.


Asunto(s)
Especificidad de Anticuerpos/inmunología , Antígenos Bacterianos/inmunología , Enfermedades Autoinmunes/inmunología , Bacteroides fragilis/inmunología , Microbioma Gastrointestinal/inmunología , Ubiquitina/inmunología , Adulto , Especificidad de Anticuerpos/genética , Antígenos Bacterianos/química , Antígenos Bacterianos/genética , Autoanticuerpos/sangre , Enfermedades Autoinmunes/microbiología , Autoinmunidad , Reacciones Cruzadas , Transferencia de Gen Horizontal , Humanos , Persona de Mediana Edad , Modelos Moleculares , Conformación Molecular , Imitación Molecular , Relación Estructura-Actividad , Ubiquitina/química , Ubiquitina/genética
19.
Science ; 360(6390): 795-800, 2018 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-29724905

RESUMEN

The immune system responds vigorously to microbial infection while permitting lifelong colonization by the microbiome. Mechanisms that facilitate the establishment and stability of the gut microbiota remain poorly described. We found that a regulatory system in the prominent human commensal Bacteroides fragilis modulates its surface architecture to invite binding of immunoglobulin A (IgA) in mice. Specific immune recognition facilitated bacterial adherence to cultured intestinal epithelial cells and intimate association with the gut mucosal surface in vivo. The IgA response was required for B. fragilis (and other commensal species) to occupy a defined mucosal niche that mediates stable colonization of the gut through exclusion of exogenous competitors. Therefore, in addition to its role in pathogen clearance, we propose that IgA responses can be co-opted by the microbiome to engender robust host-microbial symbiosis.


Asunto(s)
Bacteroides fragilis/inmunología , Microbioma Gastrointestinal/inmunología , Inmunoglobulina A/inmunología , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Animales , Adhesión Bacteriana/inmunología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Bacteroides fragilis/genética , Bacteroides fragilis/ultraestructura , Células Cultivadas , Humanos , Ratones , Polisacáridos Bacterianos/inmunología , Simbiosis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...