Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
iScience ; 27(4): 109480, 2024 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-38715940

RESUMEN

Ischemic stroke is the second leading cause of death and disability worldwide, and efforts to prevent stroke, mitigate secondary neurological damage, and promote neurological recovery remain paramount. Recent findings highlight the critical importance of microbiome-related metabolites, including vitamin B12 (VB12), in alleviating toxic stroke-associated neuroinflammation. Here, we showed that VB12 tonically programmed genes supporting microglial cell division and activation and critically controlled cellular fatty acid metabolism in homeostasis. Intriguingly, VB12 promoted mitochondrial transcriptional and metabolic activities and significantly restricted stroke-associated gene alterations in microglia. Furthermore, VB12 differentially altered the functions of microglial subsets during the acute phase of ischemic stroke, resulting in reduced brain damage and improved neurological function. Pharmacological depletion of microglia before ischemic stroke abolished VB12-mediated neurological improvement. Thus, our preclinical studies highlight the relevance of VB12 in the functional programming of microglia to alleviate neuroinflammation, minimize ischemic injury, and improve host neurological recovery after ischemic stroke.

2.
Sci Adv ; 10(13): eadi4310, 2024 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-38536923

RESUMEN

The maintenance of regulatory T (Treg) cells critically prevents autoimmunity. Pre-B cell leukemia transcription factor 1 (Pbx1) variants are associated with lupus susceptibility, particularly through the expression of a dominant negative isoform Pbx1-d in CD4+ T cells. Pbx1-d overexpression impaired Treg cell homeostasis and promoted inflammatory CD4+ T cells. Here, we showed a high expression of Pbx1 in human and murine Treg cells, which is decreased in lupus patients and mice. Pbx1 deficiency or Pbx1-d overexpression reduced the number, stability, and suppressive activity of Treg cells, which increased murine responses to immunization and autoimmune induction. Mechanistically, Pbx1 deficiency altered the expression of genes implicated in cell cycle and apoptosis in Treg cells. Intriguingly, Rtkn2, a Rho-GTPase previously associated with Treg homeostasis, was directly transactivated by Pbx1. Our results suggest that the maintenance of Treg cell homeostasis and stability by Pbx1 through cell cycle progression prevent the expansion of inflammatory T cells that otherwise exacerbates lupus progression in the hosts.


Asunto(s)
Lupus Eritematoso Sistémico , Linfocitos T Reguladores , Animales , Humanos , Ratones , División Celular , Factor de Transcripción 1 de la Leucemia de Células Pre-B/genética , Factor de Transcripción 1 de la Leucemia de Células Pre-B/metabolismo , Isoformas de Proteínas/genética , Lupus Eritematoso Sistémico/genética
3.
J Clin Periodontol ; 51(3): 309-318, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38088457

RESUMEN

AIM: To evaluate the effect of different oral irrigators on the sub-gingival microbiome composition in patients with naturally occurring plaque-induced gingivitis. MATERIALS AND METHODS: Sub-gingival plaque was collected from adults participating in a clinical trial assessing the efficacy of oral hygiene with two different oral irrigators (Waterpik Water Flosser [Group 1] and Oral-B Water Flosser [Group 2]) versus dental flossing (Group 3) for microbiome analysis. Plaque samples were reflective of naturally occurring plaque-induced gingivitis at baseline and of gingival health at the endpoint (4 weeks). Clinical measures of gingival inflammation were collected, and the sub-gingival microbiome was analysed by 16S rRNA sequencing to identify amplicon sequence variants. RESULTS: Oral hygiene instruction with self-performed manual toothbrushing and water-jet irrigation led to significant reductions in inflammation for all groups; both oral irrigators outperformed flossing in bleeding-on-probing reduction (p < .001). Microbiome diversity of sub-gingival plaque remained relatively stable over time, but significant changes were noted in certain taxa, consistent with increases in the relative abundance of commensals and reductions in late colonizers and periodontal pathogens in the water-jet groups. CONCLUSIONS: Reduction in gingival inflammation at 4 weeks within the water-jet groups is accompanied by slight but critical changes in microbiome composition. Although biodiversity does not substantially change within 4 weeks during the resolution of naturally induced gingivitis, significant relative increases in commensal early colonizers such as Streptococcus, Veillonella and Fusobacterium were accompanied by a shift towards a less anaerobic microbiota associated with return to health. These changes were contingent upon the type of interdental hygiene, with Group 1 exhibiting more significant alterations in microbiome composition towards a periodontal-health-compatible community.


Asunto(s)
Placa Dental , Gingivitis , Adulto , Humanos , Higiene Bucal , Dispositivos para el Autocuidado Bucal , Análisis de Datos Secundarios , ARN Ribosómico 16S , Índice de Placa Dental , Cepillado Dental , Gingivitis/prevención & control , Placa Dental/prevención & control , Inflamación , Agua , Método Simple Ciego
4.
iScience ; 26(7): 107122, 2023 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-37416482

RESUMEN

Gut dysbiosis has been associated with lupus pathogenesis, and fecal microbiota transfers (FMT) from lupus-prone mice shown to induce autoimmune activation into healthy mice. The immune cells of lupus patients exhibit an increased glucose metabolism and treatments with 2-deoxy-D-glucose (2DG), a glycolysis inhibitor, are therapeutic in lupus-prone mice. Here, we showed in two models of lupus with different etiologies that 2DG altered the composition of the fecal microbiome and associated metabolites. In both models, FMT from 2DG-treated mice protected lupus-prone mice of the same strain from the development of glomerulonephritis, reduced autoantibody production as well as the activation of CD4+ T cells and myeloid cells as compared to FMT from control mice. Thus, we demonstrated that the protective effect of glucose inhibition in lupus is transferable through the gut microbiota, directly linking alterations in immunometabolism to gut dysbiosis in the hosts.

5.
STAR Protoc ; 4(1): 101936, 2023 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-36520632

RESUMEN

Intestinal epithelium is composed of several cell types, which can be dissociated but difficult to maintain high cell viability due to anoikis. Herein, we describe a step-by-step protocol for the isolation of highly viable intestinal epithelial cells using ethylenediaminetetraacetate acid and TrypLE Express, which can subsequently be employed for multi-omic analyses, including single-cell RNA sequencing. For complete details on the use and execution of this protocol, please refer to Ge et al. (2022).1.


Asunto(s)
Intestinos , Multiómica , Animales , Ratones , Células Epiteliales , Mucosa Intestinal , Supervivencia Celular
6.
iScience ; 25(11): 105437, 2022 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-36388972

RESUMEN

Ischemic stroke critically impacts neurovascular homeostasis, potentially resulting in neurological disorders. However, the mechanisms through which stroke-induced inflammation modifies the molecular and metabolic circuits, particularly in ileal epithelial cells (iECs), currently remain elusive. Using multiomic approaches, we illustrated that stroke impaired the ileal microbiome and associated metabolites, leading to increased inflammatory signals and altered metabolites, potentially deteriorating the iEC homeostasis. Bulk transcriptomic and metabolomic profiling demonstrated that stroke enhanced fatty acid oxidation while reducing the tricarboxylic acid (TCA) cycle in iECs within the first day after stroke. Intriguingly, single-cell RNA sequencing analysis revealed that stroke dysregulated cell-type-specific gene responses within iECs and reduced frequencies of goblet and tuft cells. Additionally, stroke augmented interleukin-17A+ γδ T cells but decreased CD4+ T cells in the ileum. Collectively, our findings provide a comprehensive overview of stroke-induced intestinal dysbiosis and unveil responsive gene programming within iECs with implications for disease development.

7.
Nutrients ; 14(14)2022 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-35889782

RESUMEN

Vitamin B12 (VB12) is a micronutrient that is essential for DNA synthesis and cellular energy production. We recently demonstrated that VB12 oral supplementation coordinates ileal epithelial cells (iECs) and gut microbiota functions to resist pathogen colonization in mice, but it remains unclear whether VB12 directly modulates the cellular homeostasis of iECs derived from humans. Here, we integrated transcriptomic, metabolomic, and epigenomic analyses to identify VB12-dependent molecular and metabolic pathways in human iEC microtissue cultures. RNA sequencing (RNA-seq) revealed that VB12 notably activated genes involved in fatty acid metabolism and epithelial cell proliferation while suppressing inflammatory responses in human iECs. Untargeted metabolite profiling demonstrated that VB12 facilitated the biosynthesis of amino acids and methyl groups, particularly S-adenosylmethionine (SAM), and supported the function of the mitochondrial carnitine shuttle and TCA cycle. Further, genome-wide DNA methylation analysis illuminated a critical role of VB12 in sustaining cellular methylation programs, leading to differential CpG methylation of genes associated with intestinal barrier function and cell proliferation. Together, these findings suggest an essential involvement of VB12 in directing the fatty acid and mitochondrial metabolisms and reconfiguring the epigenome of human iECs to potentially support cellular oxygen utilization and cell proliferation.


Asunto(s)
Epigenómica , Vitamina B 12 , Animales , Epigénesis Genética , Células Epiteliales/metabolismo , Ácidos Grasos/metabolismo , Humanos , Ratones , Vitamina B 12/metabolismo
8.
J Exp Med ; 219(7)2022 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-35674742

RESUMEN

Deprivation of vitamin B12 (VB12) is linked to various diseases, but the underlying mechanisms in disease progression are poorly understood. Using multiomic approaches, we elucidated the responses of ileal epithelial cells (iECs) and gut microbiome to VB12 dietary restriction. Here, VB12 deficiency impaired the transcriptional and metabolic programming of iECs and reduced epithelial mitochondrial respiration and carnitine shuttling during intestinal Salmonella Typhimurium (STm) infection. Fecal microbial and untargeted metabolomic profiling identified marked changes related to VB12 deficiency, including reductions of metabolites potentially activating mitochondrial ß-oxidation in iECs and short-chain fatty acids (SCFAs). Depletion of SCFA-producing microbes by streptomycin treatment decreased the VB12-dependent STm protection. Moreover, compromised mitochondrial function of iECs correlated with declined cell capability to utilize oxygen, leading to uncontrolled oxygen-dependent STm expansion in VB12-deficient mice. Our findings uncovered previously unrecognized mechanisms through which VB12 coordinates ileal epithelial mitochondrial homeostasis and gut microbiota to regulate epithelial oxygenation, resulting in the control of aerobic STm infection.


Asunto(s)
Microbiota , Infecciones por Salmonella , Animales , Células Epiteliales , Ratones , Oxígeno , Vitamina B 12
9.
Gastroenterology ; 160(4): 1240-1255.e3, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33189700

RESUMEN

BACKGROUND & AIMS: The down-regulated in adenoma (DRA) protein, encoded by SLC26A3, a key intestinal chloride anion exchanger, has recently been identified as a novel susceptibility gene for inflammatory bowel disease (IBD). However, the mechanisms underlying the increased susceptibility to inflammation induced by the loss of DRA remain elusive. Compromised barrier is a key event in IBD pathogenesis. The current studies were undertaken to elucidate the impact of DRA deficiency on epithelial barrier integrity and to define underlying mechanisms. METHODS: Wild-type and DRA-knockout (KO) mice and crypt-derived colonoids were used as models for intestinal epithelial response. Paracellular permeability was measured by using fluorescein isothiocyanate-dextran flux. Immunoblotting, immunofluorescence, immunohistochemistry, and ribonucleoprotein immunoprecipitation assays were performed. Gut microbiome analysis was conducted to investigate the impact of DRA deficiency on gut microbial communities. RESULTS: DRA-KO mice exhibited an increased colonic paracellular permeability with significantly decreased levels of tight junction/adherens junction proteins, including ZO-1, occludin, and E-cadherin. A similar expression pattern of occludin and E-cadherin was observed in colonoids derived from DRA-KO mice and short hairpin RNA-mediated DRA knockdown in Caco-2 cells. Microbial analysis showed gut dysbiosis in DRA-KO mice. However, cohousing studies showed that dysbiosis played only a partial role in maintaining tight junction protein expression. Furthermore, our results showed increased binding of RNA-binding protein CUGBP1 with occludin and E-cadherin genes in DRA-KO mouse colon, suggesting that posttranscriptional mechanisms play a key role in gut barrier dysfunction. CONCLUSIONS: To our knowledge, our studies demonstrate a novel role of DRA in maintaining the intestinal epithelial barrier function and potential implications of its dysregulation in IBD pathogenesis.


Asunto(s)
Antiportadores/deficiencia , Antiportadores de Cloruro-Bicarbonato/deficiencia , Disbiosis/inmunología , Enfermedades Inflamatorias del Intestino/patología , Mucosa Intestinal/patología , Transportadores de Sulfato/deficiencia , Animales , Antiportadores/genética , Proteínas CELF1/metabolismo , Células CACO-2 , Cadherinas/metabolismo , Antiportadores de Cloruro-Bicarbonato/genética , Modelos Animales de Enfermedad , Disbiosis/microbiología , Disbiosis/patología , Técnicas de Silenciamiento del Gen , Humanos , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/microbiología , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Masculino , Ratones , Ratones Noqueados , Ocludina/metabolismo , Permeabilidad , Transportadores de Sulfato/genética , Uniones Estrechas/patología
10.
Sci Transl Med ; 12(551)2020 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-32641487

RESUMEN

The autoimmune disease systemic lupus erythematosus (SLE) is characterized by the production of pathogenic autoantibodies. It has been postulated that gut microbial dysbiosis may be one of the mechanisms involved in SLE pathogenesis. Here, we demonstrate that the dysbiotic gut microbiota of triple congenic (TC) lupus-prone mice (B6.Sle1.Sle2.Sle3) stimulated the production of autoantibodies and activated immune cells when transferred into germfree congenic C57BL/6 (B6) mice. Fecal transfer to B6 mice induced autoimmune phenotypes only when the TC donor mice exhibited autoimmunity. Autoimmune pathogenesis was mitigated by horizontal transfer of the gut microbiota between co-housed lupus-prone TC mice and control congenic B6 mice. Metabolomic screening identified an altered distribution of tryptophan metabolites in the feces of TC mice including an increase in kynurenine, which was alleviated after antibiotic treatment. Low dietary tryptophan prevented autoimmune pathology in TC mice, whereas high dietary tryptophan exacerbated disease. Reducing dietary tryptophan altered gut microbial taxa in both lupus-prone TC mice and control B6 mice. Consequently, fecal transfer from TC mice fed a high tryptophan diet, but not a low tryptophan diet, induced autoimmune phenotypes in germfree B6 mice. The interplay of gut microbial dysbiosis, tryptophan metabolism and host genetic susceptibility in lupus-prone mice suggest that aberrant tryptophan metabolism may contribute to autoimmune activation in this disease.


Asunto(s)
Microbioma Gastrointestinal , Lupus Eritematoso Sistémico , Animales , Autoinmunidad , Disbiosis , Ratones , Ratones Endogámicos C57BL , Triptófano
11.
Proc Natl Acad Sci U S A ; 117(1): 602-609, 2020 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-31836694

RESUMEN

Vitamin B12 (VB12) is a critical micronutrient that controls DNA metabolic pathways to maintain the host genomic stability and tissue homeostasis. We recently reported that the newly discovered commensal Propionibacterium, P. UF1, regulates the intestinal immunity to resist pathogen infection, which may be attributed in part to VB12 produced by this bacterium. Here we demonstrate that VB12 synthesized by P. UF1 is highly dependent on cobA gene-encoding uroporphyrinogen III methyltransferase, and that this vitamin distinctively regulates the cobA operon through its 5' untranslated region (5' UTR). Furthermore, conserved secondary structure and mutagenesis analyses revealed a VB12-riboswitch, cbiMCbl (140 bp), within the 5' UTR that controls the expression of downstream genes. Intriguingly, ablation of the cbiMCbl significantly dysregulates the biosynthesis of VB12, illuminating the significance of this riboswitch for bacterial VB12 biosynthesis. Collectively, our finding is an in-depth report underscoring the regulation of VB12 within the beneficial P. UF1 bacterium, through which the commensal metabolic network may improve gut bacterial cross-feeding and human health.


Asunto(s)
Regulación Bacteriana de la Expresión Génica , Propionibacterium/metabolismo , Riboswitch/genética , Vitamina B 12/biosíntesis , Regiones no Traducidas 5'/genética , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Microbioma Gastrointestinal/fisiología , Metiltransferasas/genética , Metiltransferasas/metabolismo , Mutagénesis Sitio-Dirigida , Operón/genética , Probióticos/metabolismo , Propionibacterium/genética
12.
Mucosal Immunol ; 13(1): 34-46, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31619761

RESUMEN

Microbial interaction with the host through sensing receptors, including SIGNR1, sustains intestinal homeostasis against pathogenic inflammation. The newly discovered commensal Propionibacterium strain, P. UF1, regulates the intestinal immunity against pathogen challenge. However, the molecular events driving intestinal phagocytic cell response, including colonic dendritic cells (DCs), by this bacterium are still elusive. Here, we demonstrate that the glycosylation of bacterial large surface layer protein A (LspA) by protein O-mannosyltransferase 1 (Pmt1) regulates the interaction with SIGNR1, resulting in the control of DC transcriptomic and metabolomic machineries. Programmed DCs promote protective T cell response to intestinal Listeria infection and resist chemically induced colitis in mice. Thus, our findings may highlight a novel molecular mechanism by which commensal surface glycosylation interacting with SIGNR1 directs the intestinal homeostasis to potentially protect the host against proinflammatory signals inducing colonic tissue damage.


Asunto(s)
Proteínas Bacterianas/metabolismo , Moléculas de Adhesión Celular/metabolismo , Colitis/inmunología , Colon/inmunología , Células Dendríticas/inmunología , Enfermedades Inflamatorias del Intestino/inmunología , Lectinas Tipo C/metabolismo , Listeria/fisiología , Listeriosis/inmunología , Propionibacterium/metabolismo , Receptores de Superficie Celular/metabolismo , Linfocitos T/inmunología , Animales , Proteínas Bacterianas/genética , Moléculas de Adhesión Celular/genética , Diferenciación Celular , Células Cultivadas , Colitis/inducido químicamente , Humanos , Lectinas Tipo C/genética , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Unión Proteica , Receptores de Superficie Celular/genética , Simbiosis
13.
EBioMedicine ; 44: 639-655, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31160271

RESUMEN

BACKGROUND: Malaria infection in pregnancy is a major cause of maternal and foetal morbidity and mortality worldwide. Mouse models for gestational malaria allow for the exploration of the mechanisms linking maternal malaria infection and poor pregnancy outcomes in a tractable model system. The composition of the gut microbiota has been shown to influence susceptibility to malaria infection in inbred virgin mice. In this study, we explore the ability of the gut microbiota to modulate malaria infection severity in pregnant outbred Swiss Webster mice. METHODS: In Swiss Webster mice, the composition of the gut microbiota was altered by disrupting the native gut microbes through broad-spectrum antibiotic treatment, followed by the administration of a faecal microbiota transplant derived from mice possessing gut microbes reported previously to confer susceptibility or resistance to malaria. Female mice were infected with P. chabaudi chabaudi AS in early gestation, and the progression of infection and pregnancy were tracked throughout gestation. To assess the impact of maternal infection on foetal outcomes, dams were sacrificed at term to assess foetal size and viability. Alternatively, pups were delivered by caesarean section and fostered to assess neonatal survival and pre-weaning growth in the absence of maternal morbidity. A group of dams was also euthanized at mid-gestation to assess infection and pregnancy outcomes. FINDINGS: Susceptibility to infection varied significantly as a function of source of transplanted gut microbes. Parasite burden was negatively correlated with the abundance of five specific OTUs, including Akkermansia muciniphila and OTUs classified as Allobaculum, Lactobacillus, and S24-7 species. Reduced parasite burden was associated with reduced maternal morbidity and improved pregnancy outcomes. Pups produced by dams with high parasite burdens displayed a significant reduction in survival in the first days of life relative to those from malaria-resistant dams when placed with foster dams. At midgestation, plasma cytokine levels were similar across all groups, but expression of IFNγ in the conceptus was elevated in infected dams, and IL-10 only in susceptible dams. In the latter, transcriptional and microscopic evidence of monocytic infiltration was observed with high density infection; likewise, accumulation of malaria haemozoin was enhanced in this group. These responses, combined with reduced vascularization of the placenta in this group, may contribute to poor pregnancy outcomes. Thus, high maternal parasite burden and associated maternal responses, potentially dictated by the gut microbial community, negatively impacts term foetal health and survival in the early postnatal period. INTERPRETATION: The composition of the gut microbiota in Plasmodium chabaudi chabaudi AS-infected pregnant Swiss Webster mice transcends the outbred genetics of the Swiss Webster mouse stock as a determinant of malaria infection severity, subsequently influencing pregnancy outcomes in malaria-exposed progeny. FUND: Research reported in this manuscript was supported by the University of Florida College of Veterinary Medicine (JMM, MM, and MG), the National Institute of Allergy and Infectious Diseases, the National Institute of Diabetes and Digestive and Kidney Diseases, and the Eunice Kennedy Shriver National Institute of Child Health and Human Development of the National Institutes of Health under award numbers T32AI060546 (to CDMS), R01HD46860 and R21AI111242 (to JMM), and R01 DK109560 (to MM). MG was supported by Department of Infectious Diseases and Immunology and University of Florida graduate assistantships. AA was supported by the 2017-2019 Peach State LSAMP Bridge to the Doctorate Program at the University of Georgia (National Science Foundation, Award # 1702361). The content is solely the responsibility of the authors and does not necessarily represent official views of the Eunice Kennedy Shriver National Institute of Child Health and Human Development, the National Institute of Allergy and Infectious Diseases, the National Institute of Diabetes and Digestive and Kidney Diseases, or the National Institutes of Health.


Asunto(s)
Susceptibilidad a Enfermedades , Microbioma Gastrointestinal , Predisposición Genética a la Enfermedad , Malaria/diagnóstico , Malaria/etiología , Complicaciones Parasitarias del Embarazo , Animales , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Antimaláricos/uso terapéutico , Terapia Combinada , Citocinas/metabolismo , Modelos Animales de Enfermedad , Trasplante de Microbiota Fecal , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Humanos , Malaria/terapia , Ratones , Placenta/efectos de los fármacos , Placenta/parasitología , Placenta/patología , Embarazo , Resultado del Embarazo , Pronóstico , Índice de Severidad de la Enfermedad , Resultado del Tratamiento
14.
Mucosal Immunol ; 12(2): 434-444, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30647410

RESUMEN

Newborns are highly susceptible to pathogenic infections with significant worldwide morbidity possibly due to an immature immune system. Recently, we reported that Propionibacterium strain, P. UF1, isolated from the gut microbiota of preterm infants, induced the differentiation of bacteria-specific Th17 cells. Here, we demonstrate that P. UF1 significantly increased the number of protective Th17 cells and maintained IL-10+ regulatory T cells (Tregs) in newborn mice. In addition, P. UF1 protected mice from intestinal Listeria monocytogenes (L. m) infection. P. UF1 also functionally sustained the gut microbiota and induced critical B vitamin metabolites implicated in the regulation of T cell immunity during L. m intestinal infection. Transcriptomic analysis of P. UF1-induced Th17 cells revealed genes involved in the differentiation and regulation of these cells. These results illustrate the potency of P. UF1 in the enhancement of neonatal host defense against intestinal pathogen infection.


Asunto(s)
Microbioma Gastrointestinal/inmunología , Listeria monocytogenes/fisiología , Listeriosis/inmunología , Propionibacterium/fisiología , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Animales , Animales Recién Nacidos , Diferenciación Celular/genética , Proliferación Celular , Tolerancia Inmunológica , Inmunidad Innata , Inmunomodulación , Interleucina-10/metabolismo , Interleucina-17/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Vitaminas/metabolismo
15.
Gut Microbes ; 9(3): 279-287, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29420115

RESUMEN

Regulation of Th17 and Th1 cell responses against intracellular pathogens, including Listeria monocytogenes (L. m), is critical to limit inflammation-induced tissue damage. We recently demonstrated the ability of P. UF1 bacterium derived from the intestinal bacterial commensals of preterm infants fed human breast milk to significantly mitigate pathogen-induced inflammation limiting colonic tissue damage. Here we further elucidated the potential of P. UF1 to also regulate innate and T cells, particularly Th17 and Th1 cells, against systemic L. m infection. Data demonstrate that P. UF1 not only robustly regulated protective Th17 and Th1 cells, but also sustained regulatory T cells (Treg cells) resulting in accelerated L. m clearance. Together, regulation of pathogenic inflammation by a novel probiotic bacterium such as P. UF1 may illuminate a new strategy to specifically control Th17-Th1 cells via IL-10+ Treg cells to limit systemic tissue damage induced by intracellular pathogens, including L. m.


Asunto(s)
Antibiosis , Listeria monocytogenes/inmunología , Listeriosis/prevención & control , Probióticos/administración & dosificación , Propionibacterium/inmunología , Subgrupos de Linfocitos T/inmunología , Animales , Carga Bacteriana , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Heces/microbiología , Microbioma Gastrointestinal/fisiología , Listeriosis/inmunología , Listeriosis/microbiología , Hígado/microbiología , Ratones , Ratones Endogámicos C57BL , Mutación , Propionibacterium/genética , Linfocitos T Reguladores/inmunología , Células TH1 , Células Th17
16.
Vaccine ; 36(1): 155-164, 2018 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-29180028

RESUMEN

Clostridium botulinum readily persists in the soil and secretes life-threatening botulinum neurotoxins (BoNTs) that are categorized into serotypes A to H, of which, serotype A (BoNT/A) is the most commonly occurring in nature. An efficacious vaccine with high longevity against BoNT intoxication is urgent. Herein, we developed a dual-route vaccine administered over four consecutive weeks by mucosal and parenteral routes, consisting of the heavy chain (Hc) of BoNT/A targeting dendritic cell peptide (DCpep) expressed by Lactobacillus acidophilus as a secretory immunogenic protein. The administered dual-route vaccine elicited robust and long-lasting memory B cell responses comprising germinal center (GC) B cells and follicular T cells (Tfh) that fully protected mice from lethal oral BoNT/A fatal intoxication. Additionally, passively transferring neutralizing antibodies against BoNT/A into naïve mice induced robust protection against BoNT/A lethal intoxication. Together, a targeted vaccine employing local and systemic administrative routes may represent a novel formulation eliciting protective B cell responses with remarkable longevity against threatening biologic agents such as BoNTs.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Vacunas Bacterianas/inmunología , Toxinas Botulínicas Tipo A/inmunología , Neurotoxinas/inmunología , Vacunación/métodos , Administración a través de la Mucosa , Animales , Anticuerpos Antibacterianos/inmunología , Anticuerpos Neutralizantes/administración & dosificación , Linfocitos B/inmunología , Vacunas Bacterianas/administración & dosificación , Botulismo/prevención & control , Clostridium botulinum/inmunología , Células Dendríticas/química , Células Dendríticas/inmunología , Vías de Administración de Medicamentos , Inmunización Pasiva , Memoria Inmunológica , Lactobacillus acidophilus/química , Ratones , Péptidos/administración & dosificación , Péptidos/genética , Péptidos/inmunología , Péptidos/metabolismo , Serogrupo
17.
J Clin Invest ; 127(11): 3970-3986, 2017 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-28945202

RESUMEN

Consumption of human breast milk (HBM) attenuates the incidence of necrotizing enterocolitis (NEC), which remains a leading and intractable cause of mortality in preterm infants. Here, we report that this diminution correlates with alterations in the gut microbiota, particularly enrichment of Propionibacterium species. Transfaunation of microbiota from HBM-fed preterm infants or a newly identified and cultured Propionibacterium strain, P. UF1, to germfree mice conferred protection against pathogen infection and correlated with profound increases in intestinal Th17 cells. The induction of Th17 cells was dependent on bacterial dihydrolipoamide acetyltransferase (DlaT), a major protein expressed on the P. UF1 surface layer (S-layer). Binding of P. UF1 to its cognate receptor, SIGNR1, on dendritic cells resulted in the regulation of intestinal phagocytes. Importantly, transfer of P. UF1 profoundly mitigated induced NEC-like injury in neonatal mice. Together, these results mechanistically elucidate the protective effects of HBM and P. UF1-induced immunoregulation, which safeguard against proinflammatory diseases, including NEC.


Asunto(s)
Propionibacterium/inmunología , Células Th17/fisiología , Animales , Proteínas Bacterianas/fisiología , Diferenciación Celular , Colon/inmunología , Colon/microbiología , Acetiltransferasa de Residuos Dihidrolipoil-Lisina/fisiología , Femenino , Microbioma Gastrointestinal , Genoma Bacteriano , Humanos , Inmunomodulación , Recién Nacido , Recien Nacido Prematuro , Ratones de la Cepa 129 , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Anotación de Secuencia Molecular , Propionibacterium/enzimología , Propionibacterium/genética , Análisis de Secuencia de ADN
18.
Gut Microbes ; 6(6): 392-7, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26647142

RESUMEN

Intestinal immunity is subject to complex and fine-tuned regulation dictated by interactions of the resident microbial community and their gene products with host innate cells. Deterioration of this delicate process may result in devastating autoinflammatory diseases, including inflammatory bowel disease (IBD), which primarily comprises Crohn's disease (CD) and ulcerative colitis (UC). Efficacious interventions to regulate proinflammatory signals, which play critical roles in IBD, require further scientific investigation. We recently demonstrated that rebalancing intestinal immunity via the surface layer protein A (SlpA) from Lactobacillus acidophilus NCFM potentially represents a feasible therapeutic approach to restore intestinal homeostasis. To expand on these findings, we established a new method of purifying bacterial SlpA, a new SlpA-specific monoclonal antibody, and found no SlpA-associated toxicity in mice. Thus, these data may assist in our efforts to determine the immune regulatory efficacy of SlpA in humans.


Asunto(s)
Proteínas Bacterianas/uso terapéutico , Terapia Biológica , Enfermedades Intestinales/terapia , Lactobacillus acidophilus , Secuencia de Aminoácidos , Animales , Anticuerpos Antibacterianos/inmunología , Anticuerpos Monoclonales/inmunología , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/aislamiento & purificación , Proteínas Bacterianas/toxicidad , Microbioma Gastrointestinal , Homeostasis , Intestinos/microbiología , Lactobacillus acidophilus/química , Lactobacillus acidophilus/inmunología , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular
19.
Toxins (Basel) ; 7(9): 3805-17, 2015 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-26402706

RESUMEN

Ingestion of Bacillus anthracis results in rapid gastrointestinal (GI) infection, known as GI anthrax. We previously showed that during GI anthrax, there is swift deterioration of intestinal barrier function leading to translocation of gut-associated bacteria into systemic circulation. Additionally, we described dysfunction in colonic B cells. In concordance with our previous studies, here, we report early migration of the Sterne strain of B. anthracis along with other gut-resident bacteria into the infected murine liver. Additionally, despite a global decrease in the B cell population, we observed an increase in both B-1a and marginal zone (MZ)-like B cells. Both of these cell types are capable of producing immunoglobulins against common pathogens and commensals, which act as a general antibody barrier before an antigen-specific antibody response. Accumulation of these cells in the liver was associated with an increase in chemokine expression. These data suggest that the presence of Sterne and other commensals in the liver trigger migration of MZ-like B cells from the spleen to the liver to neutralize systemic spread. Further research is required to evaluate the possible cause of their failure to clear the infection within the liver, including the potential role of dysfunctional mitogen-activated protein kinase (MAPK) signaling.


Asunto(s)
Carbunco/microbiología , Linfocitos B/microbiología , Bacillus anthracis/metabolismo , Enfermedades Gastrointestinales/microbiología , Tracto Gastrointestinal/microbiología , Animales , Carbunco/patología , Linfocitos B/metabolismo , Colon/microbiología , Modelos Animales de Enfermedad , Enfermedades Gastrointestinales/patología , Tracto Gastrointestinal/patología , Hígado/microbiología , Ratones , Bazo/microbiología , Esporas Bacterianas/metabolismo
20.
Hypertension ; 65(6): 1331-40, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25870193

RESUMEN

Emerging evidence suggests that gut microbiota is critical in the maintenance of physiological homeostasis. This study was designed to test the hypothesis that dysbiosis in gut microbiota is associated with hypertension because genetic, environmental, and dietary factors profoundly influence both gut microbiota and blood pressure. Bacterial DNA from fecal samples of 2 rat models of hypertension and a small cohort of patients was used for bacterial genomic analysis. We observed a significant decrease in microbial richness, diversity, and evenness in the spontaneously hypertensive rat, in addition to an increased Firmicutes/Bacteroidetes ratio. These changes were accompanied by decreases in acetate- and butyrate-producing bacteria. In addition, the microbiota of a small cohort of human hypertensive patients was found to follow a similar dysbiotic pattern, as it was less rich and diverse than that of control subjects. Similar changes in gut microbiota were observed in the chronic angiotensin II infusion rat model, most notably decreased microbial richness and an increased Firmicutes/Bacteroidetes ratio. In this model, we evaluated the efficacy of oral minocycline in restoring gut microbiota. In addition to attenuating high blood pressure, minocycline was able to rebalance the dysbiotic hypertension gut microbiota by reducing the Firmicutes/Bacteroidetes ratio. These observations demonstrate that high blood pressure is associated with gut microbiota dysbiosis, both in animal and human hypertension. They suggest that dietary intervention to correct gut microbiota could be an innovative nutritional therapeutic strategy for hypertension.


Asunto(s)
Disbiosis/tratamiento farmacológico , Disbiosis/epidemiología , Hipertensión/epidemiología , Minociclina/farmacología , Animales , Estudios de Cohortes , Comorbilidad , ADN Bacteriano/análisis , Modelos Animales de Enfermedad , Disbiosis/fisiopatología , Heces/microbiología , Tracto Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/microbiología , Humanos , Hipertensión/diagnóstico , Hipertensión/tratamiento farmacológico , Microbiota/efectos de los fármacos , Microbiota/fisiología , Distribución Aleatoria , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Medición de Riesgo , Especificidad de la Especie , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...