Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Semin Immunol ; 59: 101599, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35304068

RESUMEN

Gut microbiota has been shown to systemically shape the immunological landscape, modulate homeostasis and play a role in both health and disease. Dysbiosis of gut microbiota promotes inflammation and contributes to the pathogenesis of several major disorders in gastrointestinal tract, metabolic, neurological and respiratory diseases. Much effort is now focused on understanding host-microbes interactions and new microbiota-targeted therapies are deeply investigated as a means to restore health or prevent disease. This review details the immunoregulatory role of the gut microbiota in health and disease and discusses the most recent strategies in manipulating individual patient's microbiota for the management and prevention of inflammatory conditions.


Asunto(s)
Microbioma Gastrointestinal , Microbiota , Humanos , Disbiosis/terapia , Inflamación/terapia , Tracto Gastrointestinal
2.
Appl Microbiol Biotechnol ; 105(8): 3277-3288, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33839797

RESUMEN

Studies so far conducted on irritable bowel syndrome (IBS) have been focused mainly on the role of gut bacterial dysbiosis in modulating the intestinal permeability, inflammation, and motility, with consequences on the quality of life. Limited evidences showed a potential involvement of gut fungal communities. Here, the gut bacterial and fungal microbiota of a cohort of IBS patients have been characterized and compared with that of healthy subjects (HS). The IBS microbial community structure differed significantly compared to HS. In particular, we observed an enrichment of bacterial taxa involved in gut inflammation, such as Enterobacteriaceae, Streptococcus, Fusobacteria, Gemella, and Rothia, as well as depletion of health-promoting bacterial genera, such as Roseburia and Faecalibacterium. Gut microbial profiles in IBS patients differed also in accordance with constipation. Sequence analysis of the gut mycobiota showed enrichment of Saccharomycetes in IBS. Culturomics analysis of fungal isolates from feces showed enrichment of Candida spp. displaying from IBS a clonal expansion and a distinct genotypic profiles and different phenotypical features when compared to HS of Candida albicans isolates. Alongside the well-characterized gut bacterial dysbiosis in IBS, this study shed light on a yet poorly explored fungal component of the intestinal ecosystem, the gut mycobiota. Our results showed a differential fungal community in IBS compared to HS, suggesting potential for new insights on the involvement of the gut mycobiota in IBS. KEY POINTS: • Comparison of gut microbiota and mycobiota between IBS and healthy subjects • Investigation of cultivable fungi in IBS and healthy subjects • Candida albicans isolates result more virulent in IBS subjects compared to healthy subjects.


Asunto(s)
Microbioma Gastrointestinal , Síndrome del Colon Irritable , Disbiosis , Ecosistema , Heces , Humanos , Calidad de Vida
3.
Int J Mol Sci ; 21(11)2020 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-32486268

RESUMEN

Invariant Natural Killer T (iNKT) cells are a non-conventional, innate-like, T cell population that recognize lipid antigens presented by the cluster of differentiation (CD)1d molecule. Although iNKT cells are mostly known for mediating several immune responses due to their massive and diverse cytokine release, these cells also work as effectors in various contexts thanks to their cytotoxic potential. In this Review, we focused on iNKT cell cytotoxicity; we provide an overview of iNKT cell subsets, their activation cues, the mechanisms of iNKT cell cytotoxicity, the specific roles and outcomes of this activity in various contexts, and how iNKT killing functions are currently activated in cancer immunotherapies. Finally, we discuss the future perspectives for the better understanding and potential uses of iNKT cell killing functions in tumor immunosurveillance.


Asunto(s)
Antígenos CD1d/metabolismo , Inmunidad Innata , Inmunoterapia/métodos , Lípidos/química , Células T Asesinas Naturales/metabolismo , Animales , Células Presentadoras de Antígenos/metabolismo , Humanos , Hipoxia , Sistema Inmunológico , Activación de Linfocitos/inmunología , Monitorización Inmunológica , Neoplasias/inmunología , Pronóstico
4.
Int J Mol Sci ; 21(15)2020 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-32751239

RESUMEN

Colorectal cancer (CRC) is a multifaceted disease influenced by both environmental and genetic factors. A large body of literature has demonstrated the role of gut microbes in promoting inflammatory responses, creating a suitable microenvironment for the development of skewed interactions between the host and the gut microbiota and cancer initiation. Even if surgery is the primary therapeutic strategy, patients with advanced disease or cancer recurrence after surgery remain difficult to cure. Therefore, the gut microbiota has been proposed as a novel therapeutic target in light of recent promising data in which it seems to modulate the response to cancer immunotherapy. The use of microbe-targeted therapies, including antibiotics, prebiotics, live biotherapeutics, and fecal microbiota transplantation, is therefore considered to support current therapies in CRC management. In this review, we will discuss the importance of host-microbe interactions in CRC and how promoting homeostatic immune responses through microbe-targeted therapies may be useful in preventing/treating CRC development.


Asunto(s)
Neoplasias Colorrectales/terapia , Disbiosis/terapia , Trasplante de Microbiota Fecal , Microbioma Gastrointestinal/inmunología , Recurrencia Local de Neoplasia/terapia , Probióticos/uso terapéutico , Antibacterianos/uso terapéutico , Productos Biológicos/uso terapéutico , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/microbiología , Neoplasias Colorrectales/patología , Disbiosis/inmunología , Disbiosis/microbiología , Disbiosis/patología , Interacciones Microbiota-Huesped/inmunología , Humanos , Recurrencia Local de Neoplasia/inmunología , Recurrencia Local de Neoplasia/microbiología , Recurrencia Local de Neoplasia/patología , Prebióticos/administración & dosificación , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
5.
BMC Gastroenterol ; 18(1): 57, 2018 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-29720131

RESUMEN

BACKGROUND: Rett syndrome (RTT) is a neurological disorder mainly caused by mutations in MeCP2 gene. It has been shown that MeCP2 impairments can lead to cytokine dysregulation due to MeCP2 regulatory role in T-helper and T-reg mediated responses, thus contributing to the pro-inflammatory status associated with RTT. Furthermore, RTT subjects suffer from an intestinal dysbiosis characterized by an abnormal expansion of the Candida population, a known factor responsible for the hyper-activation of pro-inflammatory immune responses. Therefore, we asked whether the intestinal fungal population of RTT subjects might contribute the sub-inflammatory status triggered by MeCP2 deficiency. METHODS: We evaluated the cultivable gut mycobiota from a cohort of 50 RTT patients and 29 healthy controls characterizing the faecal fungal isolates for their virulence-related traits, antifungal resistance and immune reactivity in order to elucidate the role of fungi in RTT's intestinal dysbiosis and gastrointestinal physiology. RESULTS: Candida parapsilosis, the most abundant yeast species in RTT subjects, showed distinct genotypic profiles if compared to healthy controls' isolates as measured by hierarchical clustering analysis from RAPD genotyping. Their phenotypical analysis revealed that RTT's isolates produced more biofilm and were significantly more resistant to azole antifungals compared to the isolates from the healthy controls. In addition, the high levels of IL-1ß and IL-10 produced by peripheral blood mononuclear cells and the mixed Th1/Th17 cells population induced by RTT C. parapsilosis isolates suggest the capacity of these intestinal fungi to persist within the host, being potentially involved in chronic, pro-inflammatory responses. CONCLUSIONS: Here we demonstrated that intestinal C. parapsilosis isolates from RTT subjects hold phenotypic traits that might favour the previously observed low-grade intestinal inflammatory status associated with RTT. Therefore, the presence of putative virulent, pro-inflammatory C. parapsilosis strains in RTT could represent an additional factor in RTT's gastrointestinal pathophysiology, whose mechanisms are not yet clearly understood.


Asunto(s)
Candida parapsilosis/aislamiento & purificación , Candida parapsilosis/patogenicidad , Candidiasis/microbiología , Gastroenteritis/microbiología , Síndrome de Rett/microbiología , Antifúngicos/uso terapéutico , Candida albicans/genética , Candida albicans/aislamiento & purificación , Candida parapsilosis/efectos de los fármacos , Candida parapsilosis/genética , Candidiasis/tratamiento farmacológico , Candidiasis/inmunología , Citocinas/sangre , Farmacorresistencia Fúngica , Gastroenteritis/tratamiento farmacológico , Gastroenteritis/inmunología , Microbioma Gastrointestinal , Variación Genética , Genotipo , Humanos , Interleucina-10/sangre , Leucocitos Mononucleares/metabolismo , Proteína 2 de Unión a Metil-CpG/deficiencia , Proteína 2 de Unión a Metil-CpG/genética , Mutación , Síndrome de Rett/genética , Síndrome de Rett/inmunología , Virulencia
6.
Gut Microbes ; 16(1): 2388801, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39132842

RESUMEN

The interaction between the gut microbiota and invariant Natural Killer T (iNKT) cells plays a pivotal role in colorectal cancer (CRC). The pathobiont Fusobacterium nucleatum influences the anti-tumor functions of CRC-infiltrating iNKT cells. However, the impact of other bacteria associated with CRC, like Porphyromonas gingivalis, on their activation status remains unexplored. In this study, we demonstrate that mucosa-associated P. gingivalis induces a protumour phenotype in iNKT cells, subsequently influencing the composition of mononuclear-phagocyte cells within the tumor microenvironment. Mechanistically, in vivo and in vitro experiments showed that P. gingivalis reduces the cytotoxic functions of iNKT cells, hampering the iNKT cell lytic machinery through increased expression of chitinase 3-like-1 protein (CHI3L1). Neutralization of CHI3L1 effectively restores iNKT cell cytotoxic functions suggesting a therapeutic potential to reactivate iNKT cell-mediated antitumour immunity. In conclusion, our data demonstrate how P. gingivalis accelerates CRC progression by inducing the upregulation of CHI3L1 in iNKT cells, thus impairing their cytotoxic functions and promoting host tumor immune evasion.


Asunto(s)
Proteína 1 Similar a Quitinasa-3 , Neoplasias Colorrectales , Células T Asesinas Naturales , Porphyromonas gingivalis , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/microbiología , Células T Asesinas Naturales/inmunología , Porphyromonas gingivalis/inmunología , Proteína 1 Similar a Quitinasa-3/metabolismo , Proteína 1 Similar a Quitinasa-3/genética , Humanos , Animales , Ratones , Microambiente Tumoral/inmunología , Evasión Inmune , Escape del Tumor , Microbioma Gastrointestinal/inmunología , Línea Celular Tumoral , Infecciones por Bacteroidaceae/inmunología , Infecciones por Bacteroidaceae/microbiología , Femenino , Ratones Endogámicos C57BL , Masculino
7.
Appl Environ Microbiol ; 79(14): 4304-15, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23645200

RESUMEN

Strains of Bifidobacterium animalis subsp. lactis are extensively exploited by the food industry as health-promoting bacteria, although the genetic variability of members belonging to this taxon has so far not received much scientific attention. In this article, we describe the complete genetic makeup of the B. animalis subsp. lactis Bl12 genome and discuss the genetic relatedness of this strain with other sequenced strains belonging to this taxon. Moreover, a detailed comparative genomic analysis of B. animalis subsp. lactis genomes was performed, which revealed a closely related and isogenic nature of all currently available B. animalis subsp. lactis strains, thus strongly suggesting a closed pan-genome structure of this bacterial group.


Asunto(s)
Bifidobacterium/clasificación , Bifidobacterium/genética , Genoma Bacteriano , Antibacterianos/farmacología , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Bifidobacterium/efectos de los fármacos , Bifidobacterium/metabolismo , Datos de Secuencia Molecular , Filogenia , Reacción en Cadena de la Polimerasa , Proteoma , Análisis de Secuencia de ADN , Tetraciclina/farmacología , Resistencia a la Tetraciclina
8.
Anaerobe ; 21: 9-17, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23523946

RESUMEN

Bifidobacteria are extensively exploited by the food industry as health-promoting microorganisms. However, very little is known about the molecular mechanisms responsible for these beneficial activities, or the molecular players that sustain their ability to colonize and persist within the human gut. Here, we have investigated the enteric adaptation features of the gut commensal Bifidobacterium bifidum PRL2010, originally isolated from infant feces. This strain was able to survive under gastrointestinal challenges, while it was shown to adhere to human epithelial intestinal cell monolayers (Caco 2 and HT-29), thereby inhibiting adhesion of pathogenic bacteria such as Escherichia coli and Cronobacter sakazakii.


Asunto(s)
Antibacterianos/farmacología , Antibiosis , Adhesión Bacteriana , Bifidobacterium/fisiología , Tracto Gastrointestinal/microbiología , Bifidobacterium/efectos de los fármacos , Bifidobacterium/crecimiento & desarrollo , Bifidobacterium/aislamiento & purificación , Ácidos y Sales Biliares/farmacología , Células CACO-2 , Cronobacter sakazakii/fisiología , Células Epiteliales/microbiología , Escherichia coli/fisiología , Heces/microbiología , Células HT29 , Interacciones Huésped-Patógeno , Humanos , Concentración de Iones de Hidrógeno , Lactante , Intestinos/microbiología , Pruebas de Sensibilidad Microbiana , Probióticos , Cloruro de Sodio/farmacología
9.
Microbiol Spectr ; 11(6): e0077523, 2023 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-37795996

RESUMEN

IMPORTANCE: In this paper, we demonstrated that apyrase is released within the host cell cytoplasm during infection to target the intracellular ATP pool. By degrading intracellular ATP, apyrase contributes to prevent caspases activation, thereby inhibiting the activation of pyroptosis in infected cells. Our results show, for the first time, that apyrase is involved in the modulation of host cell survival, thereby aiding this pathogen to dampen the inflammatory response. This work adds a further piece to the puzzle of Shigella pathogenesis. Due to its increased spread worldwide, prevention and controlling strategies are urgently needed. Overall, this study highlighted apyrase as a suitable target for an anti-virulence therapy to tackle this pathogen.


Asunto(s)
Proteínas Bacterianas , Factores de Virulencia , Shigella flexneri , Apirasa , Células Eucariotas , Adenosina Trifosfato
10.
EMBO Mol Med ; 15(3): e16244, 2023 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-36533294

RESUMEN

Duchenne muscular dystrophy (DMD) is a progressive severe muscle-wasting disease caused by mutations in DMD, encoding dystrophin, that leads to loss of muscle function with cardiac/respiratory failure and premature death. Since dystrophic muscles are sensed by infiltrating inflammatory cells and gut microbial communities can cause immune dysregulation and metabolic syndrome, we sought to investigate whether intestinal bacteria support the muscle immune response in mdx dystrophic murine model. We highlighted a strong correlation between DMD disease features and the relative abundance of Prevotella. Furthermore, the absence of gut microbes through the generation of mdx germ-free animal model, as well as modulation of the microbial community structure by antibiotic treatment, influenced muscle immunity and fibrosis. Intestinal colonization of mdx mice with eubiotic microbiota was sufficient to reduce inflammation and improve muscle pathology and function. This work identifies a potential role for the gut microbiota in the pathogenesis of DMD.


Asunto(s)
Microbiota , Distrofia Muscular de Duchenne , Animales , Ratones , Distrofina/genética , Ratones Endogámicos mdx , Músculo Esquelético/metabolismo , Disbiosis , Distrofia Muscular de Duchenne/genética , Sistema Inmunológico/metabolismo , Sistema Inmunológico/patología , Modelos Animales de Enfermedad
11.
Mucosal Immunol ; 16(3): 326-340, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37004750

RESUMEN

iNKT cells account for a relevant fraction of effector T-cells in the intestine and are considered an attractive platform for cancer immunotherapy. Although iNKT cells are cytotoxic lymphocytes, their functional role in colorectal cancer (CRC) is still controversial, limiting their therapeutic use. Thus, we examined the immune cell composition and iNKT cell phenotype of CRC lesions in patients (n = 118) and different murine models. High-dimensional single-cell flow-cytometry, metagenomics, and RNA sequencing experiments revealed that iNKT cells are enriched in tumor lesions. The tumor-associated pathobiont Fusobacterium nucleatum induces IL-17 and Granulocyte-macrophage colony-stimulating factor (GM-CSF) expression in iNKT cells without affecting their cytotoxic capability but promoting iNKT-mediated recruitment of neutrophils with polymorphonuclear myeloid-derived suppressor cells-like phenotype and functions. The lack of iNKT cells reduced the tumor burden and recruitment of immune suppressive neutrophils. iNKT cells in-vivo activation with α-galactosylceramide restored their anti-tumor function, suggesting that iNKT cells can be modulated to overcome CRC-associated immune evasion. Tumor co-infiltration by iNKT cells and neutrophils correlates with negative clinical outcomes, highlighting the importance of iNKT cells in the pathophysiology of CRC. Our results reveal a functional plasticity of iNKT cells in CRC, suggesting a pivotal role of iNKT cells in shaping the tumor microenvironment, with relevant implications for treatment.


Asunto(s)
Antineoplásicos , Neoplasias Colorrectales , Células T Asesinas Naturales , Ratones , Animales , Neutrófilos , Antineoplásicos/farmacología , Inmunoterapia , Neoplasias Colorrectales/patología , Microambiente Tumoral
13.
Cell Rep ; 40(3): 111112, 2022 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-35858559

RESUMEN

Secretory immunoglobulin A (SIgA) interaction with commensal bacteria conditions microbiota composition and function. However, mechanisms regulating reciprocal control of microbiota and SIgA are not defined. Bacteria-derived adenosine triphosphate (ATP) limits T follicular helper (Tfh) cells in the Peyer's patches (PPs) via P2X7 receptor (P2X7R) and thereby SIgA generation. Here we show that hydrolysis of extracellular ATP (eATP) by apyrase results in amplification of the SIgA repertoire. The enhanced breadth of SIgA in mice colonized with apyrase-releasing Escherichia coli influences topographical distribution of bacteria and expression of genes involved in metabolic versus immune functions in the intestinal epithelium. SIgA-mediated conditioning of bacteria and enterocyte function is reflected by differences in nutrient absorption in mice colonized with apyrase-expressing bacteria. Apyrase-induced SIgA improves intestinal homeostasis and attenuates barrier impairment and susceptibility to infection by enteric pathogens in antibiotic-induced dysbiosis. Therefore, amplification of SIgA by apyrase can be leveraged to restore intestinal fitness in dysbiotic conditions.


Asunto(s)
Apirasa , Inmunoglobulina A Secretora , Adenosina Trifosfato/metabolismo , Animales , Bacterias/metabolismo , Homeostasis , Inmunoglobulina A Secretora/metabolismo , Mucosa Intestinal/metabolismo , Intestinos , Ratones , Ganglios Linfáticos Agregados
14.
J Crohns Colitis ; 16(9): 1461-1474, 2022 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-35358301

RESUMEN

BACKGROUND AND AIMS: Invariant natural killer T [iNKT] cells perform pleiotropic functions in different tissues by secreting a vast array of pro-inflammatory and cytotoxic molecules. However, the presence and function of human intestinal iNKT cells capable of secreting immunomodulatory molecules such as IL-10 has never been reported so far. Here we describe for the first time the presence of IL10-producing iNKT cells [NKT10 cells] in the intestinal lamina propria of healthy individuals and of Crohn's disease [CD] patients. METHODS: Frequency and phenotype of NKT10 cells were analysed ex vivo from intestinal specimens of Crohn's disease [n = 17] and controls [n = 7]. Stable CD-derived intestinal NKT10 cell lines were used to perform in vitro suppression assays and co-cultures with patient-derived mucosa-associated microbiota. Experimental colitis models were performed by adoptive cell transfer of splenic naïve CD4+ T cells in the presence or absence of IL10-sufficient or -deficient iNKT cells. In vivo induction of NKT10 cells was performed by administration of short chain fatty acids [SCFA] by oral gavage. RESULTS: Patient-derived intestinal NKT10 cells demonstrated suppressive capabilities towards pathogenic CD4+ T cells. The presence of increased proportions of mucosal NKT10 cells associated with better clinical outcomes in CD patients. Moreover, an intestinal microbial community enriched in SCFA-producing bacteria sustained the production of IL10 by iNKT cells. Finally, IL10-deficient iNKT cells failed to control the pathogenic activity of adoptively transferred CD4+ T cells in an experimental colitis model. CONCLUSIONS: These results describe an unprecedentd IL10-mediated immunoregulatory role of intestinal iNKT cells in controlling the pathogenic functions of mucosal T helper subsets and in maintaining the intestinal immune homeostasis.


Asunto(s)
Colitis , Enfermedad de Crohn , Células T Asesinas Naturales , Linfocitos T CD4-Positivos/patología , Enfermedad de Crohn/patología , Humanos , Interleucina-10/metabolismo , Mucosa Intestinal/patología , Células T Asesinas Naturales/metabolismo
15.
J Bacteriol ; 193(22): 6387-8, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22038957

RESUMEN

Bifidobacterium animalis subsp. lactis BLC1 is a probiotic bacterium that is widely exploited by food industries as the active ingredient of various functional foods. Here we report the complete genome sequence of B. animalis subsp. lactis BLC1, which is expected to provide insights into the biology of this health-promoting microorganism and improve our understanding of its phylogenetic relatedness with other members of the B. animalis subsp. lactis taxon.


Asunto(s)
Bifidobacterium/genética , Genoma Bacteriano , Secuencia de Bases , Bifidobacterium/clasificación , Datos de Secuencia Molecular , Sistemas de Lectura Abierta , Filogenia , Análisis de Secuencia de ADN
16.
Appl Environ Microbiol ; 77(24): 8578-87, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22003014

RESUMEN

Bifidobacteria have attracted significant scientific attention due to their perceived role as health-promoting microorganisms, although the genetics of the bacterial group is still underexplored. In this study, we investigated the transcriptome of Bifidobacterium bifidum PRL2010 during in vitro growth by microarray technology. When B. bifidum PRL2010 was grown in liquid broth, 425 of the 1,644 PRL2010 genes represented on the array were expressed in at least one of the three investigated growth phases, i.e., the lag, exponential, and stationary phases. These transcriptional analyses identified a core in vitro transcriptome encompassing 150 genes that are expressed in all phases. A proportion of these genes were further investigated as potential reference genes by quantitative real-time reverse transcription-PCR (qRT-PCR) assays. Their expression stability was evaluated under different growth conditions, which included cultivation on different carbon sources, exposure to environmental stresses (thermal, acidic, and osmotic), and growth phases. Our analyses validated six reference genes suitable for normalizing mRNA expression levels in qRT-PCR experiments applied to bifidobacteria.


Asunto(s)
Bifidobacterium/genética , Perfilación de la Expresión Génica/normas , Reacción en Cadena en Tiempo Real de la Polimerasa/normas , Transcripción Genética , Transcriptoma , Análisis por Micromatrices/métodos
17.
Microbiome ; 9(1): 39, 2021 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-33549144

RESUMEN

BACKGROUND: The gut microbiota plays a central role in host physiology and in several pathological mechanisms in humans. Antibiotics compromise the composition and functions of the gut microbiota inducing long-lasting detrimental effects on the host. Recent studies suggest that the efficacy of different clinical therapies depends on the action of the gut microbiota. Here, we investigated how different antibiotic treatments affect the ability of the gut microbiota to control intestinal inflammation upon fecal microbiota transplantation in an experimental colitis model and in ex vivo experiments with human intestinal biopsies. RESULTS: Murine fecal donors were pre-treated with different antibiotics, i.e., vancomycin, streptomycin, and metronidazole before FMT administration to colitic animals. The analysis of the gut microbiome, fecal metabolome, and the immunophenotyping of colonic lamina propria immune cells revealed that antibiotic pre-treatment significantly influences the capability of the microbiota to control intestinal inflammation. Streptomycin and vancomycin-treated microbiota failed to control intestinal inflammation and were characterized by the blooming of pathobionts previously associated with IBD as well as with metabolites related to the presence of oxidative stress and metabolism of simple sugars. On the contrary, the metronidazole-treated microbiota retained its ability to control inflammation co-occurring with the enrichment of Lactobacillus and of innate immune responses involving iNKT cells. Furthermore, ex vivo cultures of human intestinal lamina propria mononuclear cells and iNKT cell clones from IBD patients with vancomycin pre-treated sterile fecal water showed a Th1/Th17 skewing in CD4+ T-cell populations; metronidazole, on the other hand, induced the polarization of iNKT cells toward the production of IL10. CONCLUSIONS: Diverse antibiotic regimens affect the ability of the gut microbiota to control intestinal inflammation in experimental colitis by altering the microbial community structure and microbiota-derived metabolites. Video Abstract.


Asunto(s)
Antibacterianos/efectos adversos , Colitis/inducido químicamente , Colitis/microbiología , Modelos Animales de Enfermedad , Disbiosis/microbiología , Trasplante de Microbiota Fecal , Microbioma Gastrointestinal/efectos de los fármacos , Animales , Antibacterianos/farmacología , Colitis/inmunología , Colitis/patología , Disbiosis/inducido químicamente , Femenino , Microbioma Gastrointestinal/inmunología , Humanos , Masculino , Metronidazol/farmacología , Ratones , Células T Asesinas Naturales/efectos de los fármacos , Células T Asesinas Naturales/inmunología , Estreptomicina/efectos adversos , Células TH1/efectos de los fármacos , Células TH1/inmunología , Células Th17/efectos de los fármacos , Células Th17/inmunología , Vancomicina/efectos adversos
18.
Sci Rep ; 11(1): 7078, 2021 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-33782436

RESUMEN

Citrus limon (L.) Burm. F. is an important evergreen fruit crop whose rhizosphere and phyllosphere microbiota  have been characterized, while seed microbiota is still unknown. Bacterial and fungal endophytes were isolated from C. limon surface-sterilized seeds. The isolated fungi-belonging to Aspergillus, Quambalaria and Bjerkandera genera-and bacteria-belonging to Staphylococcus genus-were characterized for indoleacetic acid production and phosphate solubilization. Next Generation Sequencing based approaches were then used to characterize the endophytic bacterial and fungal microbiota structures of surface-sterilized C. limon seeds and of shoots obtained under aseptic conditions from in vitro growing seedlings regenerated from surface-sterilized seeds. This analysis highlighted that Cutibacterium and Acinetobacter were the most abundant bacterial genera in both seeds and shoots, while Cladosporium and Debaryomyces were the most abundant fungal genera in seeds and shoots, respectively. The localization of bacterial endophytes in seed and shoot tissues was revealed by Fluorescence In Situ Hybridization coupled with Confocal Laser Scanning Microscopy revealing vascular bundle colonization. Thus, these results highlighted for the first time the structures of endophytic microbiota of C. limon seeds and the transmission to shoots, corroborating the idea of a vertical transmission of plant microbiota and suggesting its crucial role in seed germination and plant development.


Asunto(s)
Bacterias/aislamiento & purificación , Citrus/microbiología , Hongos/aislamiento & purificación , Microbiota , Brotes de la Planta/crecimiento & desarrollo , Semillas/crecimiento & desarrollo , Citrus/embriología , Citrus/crecimiento & desarrollo
19.
Cells ; 9(5)2020 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-32429359

RESUMEN

Alterations of the gut microbiota may cause dysregulated mucosal immune responses leading to the onset of inflammatory bowel diseases (IBD) in genetically susceptible hosts. Restoring immune homeostasis through the normalization of the gut microbiota is now considered a valuable therapeutic approach to treat IBD patients. The customization of microbe-targeted therapies, including antibiotics, prebiotics, live biotherapeutics and faecal microbiota transplantation, is therefore considered to support current therapies in IBD management. In this review, we will discuss recent advancements in the understanding of host-microbe interactions in IBD and the basis to promote homeostatic immune responses through microbe-targeted therapies. By considering gut microbiota dysbiosis as a key feature for the establishment of chronic inflammatory events, in the near future it will be suitable to design new cost-effective, physiologic, and patient-oriented therapeutic strategies for the treatment of IBD that can be applied in a personalized manner.


Asunto(s)
Microbioma Gastrointestinal , Inmunidad Mucosa , Factores Inmunológicos/metabolismo , Inflamación/inmunología , Inflamación/microbiología , Intestinos/inmunología , Intestinos/microbiología , Animales , Disbiosis/complicaciones , Disbiosis/inmunología , Disbiosis/microbiología , Humanos
20.
J Crohns Colitis ; 14(9): 1190-1201, 2020 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-32100016

RESUMEN

BACKGROUND AND AIMS: Vedolizumab [VDZ] is a monoclonal antibody directed against the α4ß7 integrin heterodimer, approved for patients with inflammatory bowel diseases [IBD]. This study aimed at identifying immunological variables associated with response to vedolizumab in patients with ulcerative colitis [UC] and Crohn's disease [CD]. METHODS: This is a phase IV explorative prospective interventional trial. IBD patients received open-label VDZ at Weeks 0, 2, 6, and 14. Patients with a clinical response at Week 14 were maintained with VDZ up to Week 54. At Weeks 0 and 14, their peripheral blood was obtained and endoscopy with biopsies was performed. The Week 14 clinical response and remission, Week 54 clinical remission, and Week 14 endoscopic response were evaluated as endpoints of the study. The expression of surface markers, chemokine receptors, and α4ß7 heterodimer in peripheral blood and lamina propria lymphocytes was assessed by flow cytometry. A panel of soluble mediators was assessed in sera at baseline and at Week 14 by 45-plex. RESULTS: A total of 38 IBD patients [20 UC, 18 CD] were included in the study. At Week 14, the clinical response and remission rates were 87% and 66%, respectively. Higher baseline levels of circulating memory Th1 cells were strongly associated with clinical response at Week 14 [p = 0.0001], whereas reduced baseline levels of lamina propria memory Th17 and Th1/17 cells were associated with endoscopic response. Immunological clusters were found to be independently associated with vedolizumab outcomes at multivariable analysis. A panel of soluble markers, including IL17A, TNF, CXCL1, CCL19 for CD and G-CSF and IL7 for UC, associated with vedolizumab-induced Week 54 clinical remission. CONCLUSIONS: The results of this exploratory study uncovered a panel of circulating and mucosal immunological variables associated with response to treatment with vedolizumab.


Asunto(s)
Anticuerpos Monoclonales Humanizados , Colitis Ulcerosa , Enfermedad de Crohn , Integrinas/antagonistas & inhibidores , Mucosa Intestinal/patología , Inducción de Remisión/métodos , Adulto , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/efectos adversos , Anticuerpos Monoclonales Humanizados/inmunología , Biopsia/métodos , Colitis Ulcerosa/tratamiento farmacológico , Colitis Ulcerosa/inmunología , Colitis Ulcerosa/patología , Colonoscopía/métodos , Enfermedad de Crohn/tratamiento farmacológico , Enfermedad de Crohn/inmunología , Enfermedad de Crohn/patología , Citocinas/análisis , Citocinas/clasificación , Duración de la Terapia , Femenino , Fármacos Gastrointestinales/administración & dosificación , Fármacos Gastrointestinales/efectos adversos , Fármacos Gastrointestinales/inmunología , Humanos , Italia , Masculino , Monitorización Inmunológica/métodos , Evaluación de Procesos y Resultados en Atención de Salud , Subgrupos de Linfocitos T/patología
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda