Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Semin Immunol ; 59: 101599, 2022 01.
Article in English | MEDLINE | ID: mdl-35304068

ABSTRACT

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.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Humans , Dysbiosis/therapy , Inflammation/therapy , Gastrointestinal Tract
2.
Appl Microbiol Biotechnol ; 105(8): 3277-3288, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33839797

ABSTRACT

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.


Subject(s)
Gastrointestinal Microbiome , Irritable Bowel Syndrome , Dysbiosis , Ecosystem , Feces , Humans , Quality of Life
3.
Int J Mol Sci ; 21(11)2020 May 30.
Article in English | MEDLINE | ID: mdl-32486268

ABSTRACT

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.


Subject(s)
Antigens, CD1d/metabolism , Immunity, Innate , Immunotherapy/methods , Lipids/chemistry , Natural Killer T-Cells/metabolism , Animals , Antigen-Presenting Cells/metabolism , Humans , Hypoxia , Immune System , Lymphocyte Activation/immunology , Monitoring, Immunologic , Neoplasms/immunology , Prognosis
4.
Int J Mol Sci ; 21(15)2020 Jul 29.
Article in English | MEDLINE | ID: mdl-32751239

ABSTRACT

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.


Subject(s)
Colorectal Neoplasms/therapy , Dysbiosis/therapy , Fecal Microbiota Transplantation , Gastrointestinal Microbiome/immunology , Neoplasm Recurrence, Local/therapy , Probiotics/therapeutic use , Anti-Bacterial Agents/therapeutic use , Biological Products/therapeutic use , Colorectal Neoplasms/immunology , Colorectal Neoplasms/microbiology , Colorectal Neoplasms/pathology , Dysbiosis/immunology , Dysbiosis/microbiology , Dysbiosis/pathology , Host Microbial Interactions/immunology , Humans , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/microbiology , Neoplasm Recurrence, Local/pathology , Prebiotics/administration & dosage , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
5.
BMC Gastroenterol ; 18(1): 57, 2018 May 02.
Article in English | MEDLINE | ID: mdl-29720131

ABSTRACT

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.


Subject(s)
Candida parapsilosis/isolation & purification , Candida parapsilosis/pathogenicity , Candidiasis/microbiology , Gastroenteritis/microbiology , Rett Syndrome/microbiology , Antifungal Agents/therapeutic use , Candida albicans/genetics , Candida albicans/isolation & purification , Candida parapsilosis/drug effects , Candida parapsilosis/genetics , Candidiasis/drug therapy , Candidiasis/immunology , Cytokines/blood , Drug Resistance, Fungal , Gastroenteritis/drug therapy , Gastroenteritis/immunology , Gastrointestinal Microbiome , Genetic Variation , Genotype , Humans , Interleukin-10/blood , Leukocytes, Mononuclear/metabolism , Methyl-CpG-Binding Protein 2/deficiency , Methyl-CpG-Binding Protein 2/genetics , Mutation , Rett Syndrome/genetics , Rett Syndrome/immunology , Virulence
6.
Appl Environ Microbiol ; 79(14): 4304-15, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23645200

ABSTRACT

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.


Subject(s)
Bifidobacterium/classification , Bifidobacterium/genetics , Genome, Bacterial , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bifidobacterium/drug effects , Bifidobacterium/metabolism , Molecular Sequence Data , Phylogeny , Polymerase Chain Reaction , Proteome , Sequence Analysis, DNA , Tetracycline/pharmacology , Tetracycline Resistance
7.
Anaerobe ; 21: 9-17, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23523946

ABSTRACT

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.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antibiosis , Bacterial Adhesion , Bifidobacterium/physiology , Gastrointestinal Tract/microbiology , Bifidobacterium/drug effects , Bifidobacterium/growth & development , Bifidobacterium/isolation & purification , Bile Acids and Salts/pharmacology , Caco-2 Cells , Cronobacter sakazakii/physiology , Epithelial Cells/microbiology , Escherichia coli/physiology , Feces/microbiology , HT29 Cells , Host-Pathogen Interactions , Humans , Hydrogen-Ion Concentration , Infant , Intestines/microbiology , Microbial Sensitivity Tests , Probiotics , Sodium Chloride/pharmacology
8.
Microbiol Spectr ; 11(6): e0077523, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-37795996

ABSTRACT

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.


Subject(s)
Bacterial Proteins , Virulence Factors , Shigella flexneri , Apyrase , Eukaryotic Cells , Adenosine Triphosphate
9.
EMBO Mol Med ; 15(3): e16244, 2023 03 08.
Article in English | MEDLINE | ID: mdl-36533294

ABSTRACT

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.


Subject(s)
Microbiota , Muscular Dystrophy, Duchenne , Animals , Mice , Dystrophin/genetics , Mice, Inbred mdx , Muscle, Skeletal/metabolism , Dysbiosis , Muscular Dystrophy, Duchenne/genetics , Immune System/metabolism , Immune System/pathology , Disease Models, Animal
10.
Mucosal Immunol ; 16(3): 326-340, 2023 06.
Article in English | MEDLINE | ID: mdl-37004750

ABSTRACT

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.


Subject(s)
Antineoplastic Agents , Colorectal Neoplasms , Natural Killer T-Cells , Mice , Animals , Neutrophils , Antineoplastic Agents/pharmacology , Immunotherapy , Colorectal Neoplasms/pathology , Tumor Microenvironment
11.
Appl Environ Microbiol ; 78(14): 5002-12, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22562993

ABSTRACT

The Bifidobacterium bifidum PRL2010 genome encodes a relatively small set of predicted carbohydrate transporters. Growth experiments and transcriptome analyses of B. bifidum PRL2010 revealed that carbohydrate utilization in this microorganism appears to be restricted to a relatively low number of carbohydrates.


Subject(s)
Bacterial Proteins/metabolism , Bifidobacterium/metabolism , Carbohydrate Metabolism/genetics , Gene Expression Profiling , Membrane Transport Proteins/metabolism , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Bacterial Proteins/genetics , Bifidobacterium/genetics , Bifidobacterium/growth & development , Biological Transport , Genomics , Humans , Infant , Infant, Newborn , Membrane Transport Proteins/genetics
12.
Cell Rep ; 40(3): 111112, 2022 07 19.
Article in English | MEDLINE | ID: mdl-35858559

ABSTRACT

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.


Subject(s)
Apyrase , Immunoglobulin A, Secretory , Adenosine Triphosphate/metabolism , Animals , Bacteria/metabolism , Homeostasis , Immunoglobulin A, Secretory/metabolism , Intestinal Mucosa/metabolism , Intestines , Mice , Peyer's Patches
13.
J Crohns Colitis ; 16(9): 1461-1474, 2022 Sep 08.
Article in English | MEDLINE | ID: mdl-35358301

ABSTRACT

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.


Subject(s)
Colitis , Crohn Disease , Natural Killer T-Cells , CD4-Positive T-Lymphocytes/pathology , Crohn Disease/pathology , Humans , Interleukin-10/metabolism , Intestinal Mucosa/pathology , Natural Killer T-Cells/metabolism
14.
J Bacteriol ; 193(22): 6387-8, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22038957

ABSTRACT

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.


Subject(s)
Bifidobacterium/genetics , Genome, Bacterial , Base Sequence , Bifidobacterium/classification , Molecular Sequence Data , Open Reading Frames , Phylogeny , Sequence Analysis, DNA
15.
Appl Environ Microbiol ; 77(24): 8578-87, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22003014

ABSTRACT

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.


Subject(s)
Bifidobacterium/genetics , Gene Expression Profiling/standards , Real-Time Polymerase Chain Reaction/standards , Transcription, Genetic , Transcriptome , Microarray Analysis/methods
16.
Microbiome ; 9(1): 39, 2021 02 06.
Article in English | MEDLINE | ID: mdl-33549144

ABSTRACT

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.


Subject(s)
Anti-Bacterial Agents/adverse effects , Colitis/chemically induced , Colitis/microbiology , Disease Models, Animal , Dysbiosis/microbiology , Fecal Microbiota Transplantation , Gastrointestinal Microbiome/drug effects , Animals , Anti-Bacterial Agents/pharmacology , Colitis/immunology , Colitis/pathology , Dysbiosis/chemically induced , Female , Gastrointestinal Microbiome/immunology , Humans , Male , Metronidazole/pharmacology , Mice , Natural Killer T-Cells/drug effects , Natural Killer T-Cells/immunology , Streptomycin/adverse effects , Th1 Cells/drug effects , Th1 Cells/immunology , Th17 Cells/drug effects , Th17 Cells/immunology , Vancomycin/adverse effects
17.
Sci Rep ; 11(1): 7078, 2021 03 29.
Article in English | MEDLINE | ID: mdl-33782436

ABSTRACT

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.


Subject(s)
Bacteria/isolation & purification , Citrus/microbiology , Fungi/isolation & purification , Microbiota , Plant Shoots/growth & development , Seeds/growth & development , Citrus/embryology , Citrus/growth & development
18.
Cells ; 9(5)2020 05 16.
Article in English | MEDLINE | ID: mdl-32429359

ABSTRACT

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.


Subject(s)
Gastrointestinal Microbiome , Immunity, Mucosal , Immunologic Factors/metabolism , Inflammation/immunology , Inflammation/microbiology , Intestines/immunology , Intestines/microbiology , Animals , Dysbiosis/complications , Dysbiosis/immunology , Dysbiosis/microbiology , Humans
19.
Sci Rep ; 10(1): 21443, 2020 12 08.
Article in English | MEDLINE | ID: mdl-33293569

ABSTRACT

In the present work, culture-based and culture-independent investigations were performed to determine the microbiota structure of the coelomic fluid of Mediterranean sea urchin Paracentrotus lividus individuals collected from two distinct geographical sites neighboring a high-density population bay and a nature reserve, respectively. Next Generation Sequencing analysis of 16S rRNA gene (rDNA) showed that members of the Proteobacteria, Bacteroidetes and Fusobacteria phyla, which have been previously reported to be commonly retrieved from marine invertebrates, dominate the overall population of microorganisms colonizing this liquid tissue, with minority bacterial genera exhibiting remarkable differences among individuals. Our results showed that there is a correlation between microbiota structure and geographical location of the echinoderm collection site, highlighting over-representation of metagenomic functions related to amino acid and bioactive peptides metabolism in specimens inhabiting the nature reserve. Finally, we also described the developmental delay and aberrations exhibited by sea urchin embryos exposed to distinct bacterial isolates, and showed that these defects rely upon hydrophilic compound(s) synthesized by the bacterial strains assayed. Altogether, our findings lay the groundwork to decipher the relationships of bacteria with sea urchins in their aquatic environment, also providing an additional layer of information to understand the biological roles of the coelomic fluid.


Subject(s)
Bacteria/classification , Paracentrotus/growth & development , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA/methods , Animals , Bacteria/genetics , Bacteria/growth & development , Bacteria/isolation & purification , Bacteriological Techniques , DNA, Bacterial/genetics , DNA, Ribosomal/genetics , High-Throughput Nucleotide Sequencing , Microbiota , Paracentrotus/microbiology , Phylogeny
20.
J Crohns Colitis ; 14(9): 1190-1201, 2020 Sep 16.
Article in English | MEDLINE | ID: mdl-32100016

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal, Humanized , Colitis, Ulcerative , Crohn Disease , Integrins/antagonists & inhibitors , Intestinal Mucosa/pathology , Remission Induction/methods , Adult , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/immunology , Biopsy/methods , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/immunology , Colitis, Ulcerative/pathology , Colonoscopy/methods , Crohn Disease/drug therapy , Crohn Disease/immunology , Crohn Disease/pathology , Cytokines/analysis , Cytokines/classification , Duration of Therapy , Female , Gastrointestinal Agents/administration & dosage , Gastrointestinal Agents/adverse effects , Gastrointestinal Agents/immunology , Humans , Italy , Male , Monitoring, Immunologic/methods , Outcome and Process Assessment, Health Care , T-Lymphocyte Subsets/pathology
SELECTION OF CITATIONS
SEARCH DETAIL