Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters










Publication year range
1.
Oncologist ; 2024 May 20.
Article in English | MEDLINE | ID: mdl-38767987

ABSTRACT

BACKGROUND: Abemaciclib-induced diarrhea is a relevant concern in clinical practice. Postbiotics have emerged as a promising option for managing it. MATERIALS AND METHODS: We conducted a retrospective-prospective, 2-group, observational study to assess the impact of the postbiotic PostbiotiX-Restore, derived by Lactobacillus paracasei CNCM I-5220, on abemaciclib-induced diarrhea in patients with hormone receptor-positive HER2-negative breast cancer. The prospective population (Postbio group) received postbiotic during the first cycle of abemaciclib, while the retrospective one received standard care (Standard group). Diarrhea grading was defined according to the National Cancer Institute's Common Terminology Criteria for Adverse Events. RESULTS: During the first cycle, diarrhea occurred in 78.9% of patients in the Standard cohort and 97.1% in the Postbio one, with most cases being G1-G2. Severe (G3) diarrhea was significantly less frequent in the Postbio group (0%) compared to the Standard one (7.9%; P = .029). Over the entire study period, while the grading difference was not statistically significant, G3 events were less frequent in the Postbio population (5.9%) than the Standard one (15.4%). Moreover, Postbio patients required fewer dose reductions due to diarrhea compared to the Standard group (P = .002). Notably, in the Postbio population, G1 and G2 events had short median durations (3 and 1 days, respectively) and, for the 2 patients experiencing G3 events during the second abemaciclib cycle (off postbiotic), diarrhea lasted only 1 day. CONCLUSIONS: Our study demonstrates the effect of PostbiotiX-Restore in mitigating abemaciclib-induced diarrhea, resulting in reduced severity, fewer dose reductions, and shorter duration. Further exploration and validation in larger cohorts are needed.

2.
Cell Rep Med ; 4(10): 101235, 2023 10 17.
Article in English | MEDLINE | ID: mdl-37852177

ABSTRACT

The rising prevalence of obesity and metabolic disorders worldwide highlights the urgent need to find new long-term and clinically meaningful weight-loss therapies. Here, we evaluate the therapeutic potential and the mechanism of action of a biomimetic cellulose-based oral superabsorbent hydrogel (OSH). Treatment with OSH exerts effects on intestinal tissue and gut microbiota composition, functioning like a protective dynamic exoskeleton. It protects from gut barrier permeability disruption and induces rapid and consistent changes in the gut microbiota composition, specifically fostering Akkermansia muciniphila expansion. The mechanobiological, physical, and chemical structures of the gel are required for A. muciniphila growth. OSH treatment induces weight loss and reduces fat accumulation, in both preventative and therapeutic settings. OSH usage also prevents liver steatosis, immune infiltration, and fibrosis, limiting the progression of non-alcoholic fatty liver disease. Our work shows the potential of using OSH as a non-systemic mechanobiological approach to treat metabolic syndrome and its comorbidities.


Subject(s)
Exoskeleton Device , Non-alcoholic Fatty Liver Disease , Humans , Hydrogels/therapeutic use , Biomimetics , Non-alcoholic Fatty Liver Disease/prevention & control , Non-alcoholic Fatty Liver Disease/drug therapy , Obesity/prevention & control , Obesity/drug therapy
3.
Cancer Cell ; 41(10): 1717-1730.e4, 2023 Oct 09.
Article in English | MEDLINE | ID: mdl-37738976

ABSTRACT

Recent data have shown that gut microbiota has a major impact on the clinical response to immune checkpoint inhibitors (ICIs) in the context of solid tumors. ICI-based therapy acts by unlocking cognate cytotoxic T lymphocyte (CTL) effector responses, and increased sensitivity to ICIs is due to an enhancement of patients' tumor antigen (TA)-specific CTL responses. Cancer clearance by TA-specific CTL requires expression of relevant TAs on cancer cells' HLA class I molecules, and reduced HLA class I expression is a common mechanism used by cancer cells to evade the immune system. Here, we show that metabolites released by bacteria, in particular, phytosphingosine, can upregulate HLA class I expression on cancer cells, sensitizing them to TA-specific CTL lysis in vitro and in vivo, in combination with immunotherapy. This effect is mediated by postbiotic-induced upregulation of NLRC5 in response to upstream MYD88-NF-κB activation, thus significantly controlling tumor growth.

4.
Front Microbiol ; 14: 1157164, 2023.
Article in English | MEDLINE | ID: mdl-37020718

ABSTRACT

The maintenance of intestinal barrier function is essential for preventing different pathologies, such as the leaky gut syndrome (LGS), which is characterized by the passage of harmful agents, like bacteria, toxins, and viruses, into the bloodstream. Intestinal barrier integrity is controlled by several players, including the gut microbiota. Various molecules, called postbiotics, are released during the natural metabolic activity of the microbiota. Postbiotics can regulate host-microbe interactions, epithelial homeostasis, and have overall benefits for our health. In this work, we used in vitro and in vivo systems to demonstrate the role of Lactobacillus paracasei CNCM I-5220-derived postbiotic (LP-PBF) in preserving intestinal barrier integrity. We demonstrated in vitro that LP-PBF restored the morphology of tight junctions (TJs) that were altered upon Salmonella typhimurium exposure. In vivo, LP-PBF protected the gut vascular barrier and blocked S. typhimurium dissemination into the bloodstream. Interestingly, we found that LP-PBF interacts not only with the host cells, but also directly with S. typhimurium blocking its biofilm formation, partially due to the presence of biosurfactants. This study highlights that LP-PBF is beneficial in maintaining gut homeostasis due to the synergistic effect of its different components. These results suggest that LP-PBF could be utilized in managing several pathologies displaying an impaired intestinal barrier function.

5.
Gastro Hep Adv ; 1(2): 194-209, 2022.
Article in English | MEDLINE | ID: mdl-35174369

ABSTRACT

BACKGROUND AND AIMS: The SARS-CoV-2 pandemic has overwhelmed the treatment capacity of the health care systems during the highest viral diffusion rate. Patients reaching the emergency department had to be either hospitalized (inpatients) or discharged (outpatients). Still, the decision was taken based on the individual assessment of the actual clinical condition, without specific biomarkers to predict future improvement or deterioration, and discharged patients often returned to the hospital for aggravation of their condition. Here, we have developed a new combined approach of omics to identify factors that could distinguish coronavirus disease 19 (COVID-19) inpatients from outpatients. METHODS: Saliva and blood samples were collected over the course of two observational cohort studies. By using machine learning approaches, we compared salivary metabolome of 50 COVID-19 patients with that of 270 healthy individuals having previously been exposed or not to SARS-CoV-2. We then correlated the salivary metabolites that allowed separating COVID-19 inpatients from outpatients with serum biomarkers and salivary microbiota taxa differentially represented in the two groups of patients. RESULTS: We identified nine salivary metabolites that allowed assessing the need of hospitalization. When combined with serum biomarkers, just two salivary metabolites (myo-inositol and 2-pyrrolidineacetic acid) and one serum protein, chitinase 3-like-1 (CHI3L1), were sufficient to separate inpatients from outpatients completely and correlated with modulated microbiota taxa. In particular, we found Corynebacterium 1 to be overrepresented in inpatients, whereas Actinomycetaceae F0332, Candidatus Saccharimonas, and Haemophilus were all underrepresented in the hospitalized population. CONCLUSION: This is a proof of concept that a combined omic analysis can be used to stratify patients independently from COVID-19.

6.
J Immunother Cancer ; 9(11)2021 11.
Article in English | MEDLINE | ID: mdl-34824159

ABSTRACT

BACKGROUND: Natural killer (NK) cells require a functional lytic granule machinery to mediate effective antitumor responses. Evading the lytic cargo deployed at the immune synapse (IS) could be a critical step for cancer progression through yet unidentified mechanisms. METHODS: NK cell antibody-dependent cellular cytotoxicity (ADCC) is a major determinant of the clinical efficacy of some therapeutic antibodies including the anti-HER2 Trastuzumab. Thus, we screened sera of Trastuzumab-resistant HER2 +patients with breast cancer for molecules that could inhibit NK cell ADCC. We validated our findings in vitro using cytotoxicity assays and confocal imaging of the lytic granule machinery and in vivo using syngeneic and xenograft murine models. RESULTS: We found that sera from Trastuzumab-refractory patients could inhibit healthy NK cell ADCC in vitro. These sera contained high levels of the inflammatory protein chitinase 3-like 1 (CHI3L1) compared with sera from responders and healthy controls. We demonstrate that recombinant CHI3L1 inhibits both ADCC and innate NK cell cytotoxicity. Mechanistically, CHI3L1 prevents the correct polarization of the microtubule-organizing center along with the lytic granules to the IS by hindering the receptor of advanced glycation end-products and its downstream JNK signaling. In vivo, CHI3L1 administration drastically impairs the control of NK cell-sensitive tumors, while CHI3L1 blockade synergizes with ADCC to cure mice with HER2 +xenografts. CONCLUSION: Our work highlights a new paradigm of tumor immune escape mediated by CHI3L1 which acts on the cytotoxic machinery and prevents granule polarization. Targeting CHI3L1 could mitigate immune escape and potentiate antibody and cell-based immunotherapies.


Subject(s)
Chitinase-3-Like Protein 1/metabolism , Immune Evasion/immunology , Immunotherapy/methods , Killer Cells, Natural/immunology , Neoplasms/genetics , Animals , Female , Humans , Mice
7.
Science ; 374(6566): 439-448, 2021 Oct 22.
Article in English | MEDLINE | ID: mdl-34672740

ABSTRACT

Up to 40% of patients with inflammatory bowel disease present with psychosocial disturbances. We previously identified a gut vascular barrier that controls the dissemination of bacteria from the intestine to the liver. Here, we describe a vascular barrier in the brain choroid plexus (PVB) that is modulated in response to intestinal inflammation through bacteria-derived lipopolysaccharide. The inflammatory response induces PVB closure after gut vascular barrier opening by the up-regulation of the wingless-type, catenin-beta 1 (Wnt/ß-catenin) signaling pathway, rendering it inaccessible to large molecules. In a model of genetically driven closure of choroid plexus endothelial cells, we observed a deficit in short-term memory and anxiety-like behavior, suggesting that PVB closure may correlate with mental deficits. Inflammatory bowel disease­related mental symptoms may thus be the consequence of a deregulated gut­brain vascular axis.


Subject(s)
Choroid Plexus/blood supply , Choroid Plexus/physiopathology , Colitis, Ulcerative/physiopathology , Colitis, Ulcerative/psychology , Intestines/physiopathology , Memory Disorders/physiopathology , Memory, Short-Term , Animals , Anxiety/etiology , Anxiety/physiopathology , Blood-Brain Barrier/pathology , Colitis, Ulcerative/complications , Dextrans , Disease Models, Animal , Humans , Lipopolysaccharides , Memory Disorders/etiology , Mice , Mice, Inbred C57BL , Microglia/pathology , Signal Transduction , Tight Junctions/pathology , Wnt Proteins/metabolism , beta Catenin/metabolism
8.
Cell Rep ; 36(1): 109312, 2021 07 06.
Article in English | MEDLINE | ID: mdl-34233181

ABSTRACT

Efforts to overcome resistance to immune checkpoint blockade therapy have focused on vaccination strategies using neoepitopes, although they cannot be applied on a large scale due to the "private" nature of cancer mutations. Here, we show that infection of tumor cells with Salmonella induces the opening of membrane hemichannels and the extracellular release of proteasome-generated peptides by the exacerbation of endoplasmic reticulum (ER) stress. Peptides released by cancer cells foster an antitumor response in vivo, both in mice bearing B16F10 melanomas and in dogs suffering from osteosarcoma. Mass spectrometry analysis on the supernatant of human melanoma cells revealed 12 peptides capable of priming healthy-donor CD8+ T cells that recognize and kill human melanoma cells in vitro and when xenotransplanted in vivo. Hence, we identified a class of shared tumor antigens that are generated in ER-stressed cells, such as tumor cells, that do not induce tolerance and are not presented by healthy cells.


Subject(s)
Endoplasmic Reticulum Stress , Peptides , Animals , Dogs , Female , Humans , Male , Amino Acid Sequence , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cell Membrane/metabolism , Lymphocyte Activation/immunology , Melanocytes/metabolism , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice, Inbred C57BL , Neoplasm Grading , Neoplasms/immunology , Neoplasms/microbiology , Neoplasms/pathology , Osteosarcoma/immunology , Osteosarcoma/veterinary , Peptides/chemistry , Peptides/immunology , Proteasome Endopeptidase Complex/metabolism , Salmonella/physiology , Unfolded Protein Response
9.
Cancer Cell ; 39(5): 708-724.e11, 2021 05 10.
Article in English | MEDLINE | ID: mdl-33798472

ABSTRACT

Metastasis is facilitated by the formation of a "premetastatic niche," which is fostered by primary tumor-derived factors. Colorectal cancer (CRC) metastasizes mainly to the liver. We show that the premetastatic niche in the liver is induced by bacteria dissemination from primary CRC. We report that tumor-resident bacteria Escherichia coli disrupt the gut vascular barrier (GVB), an anatomical structure controlling bacterial dissemination along the gut-liver axis, depending on the virulence regulator VirF. Upon GVB impairment, bacteria disseminate to the liver, boost the formation of a premetastatic niche, and favor the recruitment of metastatic cells. In training and validation cohorts of CRC patients, we find that the increased levels of PV-1, a marker of impaired GVB, is associated with liver bacteria dissemination and metachronous distant metastases. Thus, PV-1 is a prognostic marker for CRC distant recurrence and vascular impairment, leading to liver metastases.


Subject(s)
Colorectal Neoplasms/blood supply , Colorectal Neoplasms/pathology , Liver Neoplasms/pathology , Neoplasm Metastasis/pathology , Neoplasm Recurrence, Local/pathology , Bacteria/isolation & purification , Colonic Neoplasms/blood supply , Colonic Neoplasms/pathology , Humans , Liver/pathology , Liver Neoplasms/secondary
10.
PLoS One ; 16(1): e0245903, 2021.
Article in English | MEDLINE | ID: mdl-33493208

ABSTRACT

Lactobacillus helveticus carries many properties such as the ability to survive gastrointestinal transit, modulate the host immune response, accumulate biopeptides in milk, and adhere to the epithelial cells that could contribute to improving host health. In this study, the applicability as functional cultures of four L. helveticus strains isolated from Italian hard cheeses was investigated. A preliminary strain characterization showed that the ability to produce folate was generally low while antioxidant, proteolytic, peptidase, and ß-galactosidase activities resulted high, although very variable, between strains. When stimulated moDCs were incubated in the presence of live cells, a dose-dependent release of both the pro-inflammatory cytokine IL-12p70 and the anti-inflammatory cytokine IL-10, was shown for all the four strains. In the presence of cell-free culture supernatants (postbiotics), a dose-dependent, decrease of IL-12p70 and an increase of IL-10 was generally observed. The immunomodulatory effect took place also in Caciotta-like cheese made with strains SIM12 and SIS16 as bifunctional (i.e., immunomodulant and acidifying) starter cultures, thus confirming tests in culture media. Given that the growth of bacteria in the cheese was not necessary (they were killed by pasteurization), the results indicated that some constituents of non-viable bacteria had immunomodulatory properties. This study adds additional evidence for the positive role of L. helveticus on human health and suggests cheese as a suitable food for delivering candidate strains and modulating their anti-inflammatory properties.


Subject(s)
Cheese/microbiology , Lactobacillus helveticus/isolation & purification , Food Microbiology , Humans , Italy , Lactobacillus helveticus/genetics , Lactobacillus helveticus/metabolism , Leukocytes, Mononuclear/metabolism
11.
Nat Microbiol ; 5(3): 511-524, 2020 03.
Article in English | MEDLINE | ID: mdl-31988379

ABSTRACT

The microbiota has been shown to promote intestinal tumourigenesis, but a possible anti-tumourigenic effect has also been postulated. Here, we demonstrate that changes in the microbiota and mucus composition are concomitant with tumourigenesis. We identified two anti-tumourigenic strains of the microbiota-Faecalibaculum rodentium and its human homologue, Holdemanella biformis-that are strongly under-represented during tumourigenesis. Reconstitution of ApcMin/+ or azoxymethane- and dextran sulfate sodium-treated mice with an isolate of F. rodentium (F. PB1) or its metabolic products reduced tumour growth. Both F. PB1 and H. biformis produced short-chain fatty acids that contributed to control protein acetylation and tumour cell proliferation by inhibiting calcineurin and NFATc3 activation in mouse and human settings. We have thus identified endogenous anti-tumourigenic bacterial strains with strong diagnostic, therapeutic and translational potential.


Subject(s)
Firmicutes/physiology , Gastrointestinal Microbiome/physiology , Intestinal Neoplasms/microbiology , Intestines/microbiology , Adult , Aged , Animals , Cell Proliferation/drug effects , Colonic Neoplasms/microbiology , Colonic Neoplasms/therapy , DNA, Bacterial/genetics , DNA, Bacterial/isolation & purification , Fatty Acids, Volatile/metabolism , Female , Firmicutes/isolation & purification , Humans , In Situ Hybridization, Fluorescence , Intestinal Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Middle Aged , RNA, Bacterial/genetics , RNA, Bacterial/isolation & purification
12.
J Hepatol ; 71(6): 1216-1228, 2019 12.
Article in English | MEDLINE | ID: mdl-31419514

ABSTRACT

BACKGROUND & AIMS: Fatty liver disease, including non-alcoholic fatty liver (NAFLD) and steatohepatitis (NASH), has been associated with increased intestinal barrier permeability and translocation of bacteria or bacterial products into the blood circulation. In this study, we aimed to unravel the role of both intestinal barrier integrity and microbiota in NAFLD/NASH development. METHODS: C57BL/6J mice were fed with high-fat diet (HFD) or methionine-choline-deficient diet for 1 week or longer to recapitulate aspects of NASH (steatosis, inflammation, insulin resistance). Genetic and pharmacological strategies were then used to modulate intestinal barrier integrity. RESULTS: We show that disruption of the intestinal epithelial barrier and gut vascular barrier (GVB) are early events in NASH pathogenesis. Mice fed HFD for only 1 week undergo a diet-induced dysbiosis that drives GVB damage and bacterial translocation into the liver. Fecal microbiota transplantation from HFD-fed mice into specific pathogen-free recipients induces GVB damage and epididymal adipose tissue enlargement. GVB disruption depends on interference with the WNT/ß-catenin signaling pathway, as shown by genetic intervention driving ß-catenin activation only in endothelial cells, preventing GVB disruption and NASH development. The bile acid analogue and farnesoid X receptor agonist obeticholic acid (OCA) drives ß-catenin activation in endothelial cells. Accordingly, pharmacologic intervention with OCA protects against GVB disruption, both as a preventive and therapeutic agent. Importantly, we found upregulation of the GVB leakage marker in the colon of patients with NASH. CONCLUSIONS: We have identified a new player in NASH development, the GVB, whose damage leads to bacteria or bacterial product translocation into the blood circulation. Treatment aimed at restoring ß-catenin activation in endothelial cells, such as administration of OCA, protects against GVB damage and NASH development. LAY SUMMARY: The incidence of fatty liver disease is reaching epidemic levels in the USA, with more than 30% of adults having NAFLD (non-alcoholic fatty liver disease), which can progress to more severe non-alcoholic steatohepatitis (NASH). Herein, we show that disruption of the intestinal epithelial barrier and gut vascular barrier are early events in the development of NASH. We show that the drug obeticholic acid protects against barrier disruption and thereby prevents the development of NASH, providing further evidence for its use in the prevention or treatment of NASH.


Subject(s)
Bacterial Translocation/drug effects , Capillary Permeability , Chenodeoxycholic Acid/analogs & derivatives , Gastrointestinal Microbiome/physiology , Intestinal Mucosa , Non-alcoholic Fatty Liver Disease , Animals , Capillary Permeability/drug effects , Capillary Permeability/physiology , Chenodeoxycholic Acid/pharmacology , Diet, High-Fat , Disease Models, Animal , Dysbiosis/immunology , Inflammation/metabolism , Insulin Resistance , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Liver/pathology , Mice , Non-alcoholic Fatty Liver Disease/immunology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/therapy , Protective Agents/pharmacology
13.
Nat Med ; 22(6): 624-31, 2016 06.
Article in English | MEDLINE | ID: mdl-27135741

ABSTRACT

Cetuximab is a monoclonal antibody that is effective in the treatment of metastatic colorectal cancer (mCRC). Cetuximab blocks epidermal growth factor receptor (EGFR)-ligand interaction and inhibits downstream RAS-ERK activation. However, only some activating mutations in RAS affect cetuximab efficacy, and it is not clear what else mediates treatment success. Here we hypothesized that cetuximab induces immunogenic cell death (ICD) that activates a potent antitumor response. We found that cetuximab, in combination with chemotherapy, fostered ICD in CRC cells, which we measured via the endoplasmic reticulum (ER) stress response and an increase in phagocytosis by dendritic cells. ICD induction depended on the mutational status of the EGFR signaling pathway and on the inhibition of the splicing of X-box binding protein 1 (XBP1), an unfolded protein response (UPR) mediator. We confirmed the enhanced immunogenicity elicited by cetuximab in a mouse model of human EGFR-expressing CRC. Overall, we demonstrate a new, immune-related mechanism of action of cetuximab that may help to tailor personalized medicine.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Death/drug effects , Cetuximab/pharmacology , Colorectal Neoplasms/immunology , Endoplasmic Reticulum Stress/drug effects , Phagocytosis/drug effects , Animals , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Calreticulin/drug effects , Calreticulin/metabolism , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Cell Death/immunology , Cell Line, Tumor , Colorectal Neoplasms/genetics , Dendritic Cells/drug effects , Dendritic Cells/immunology , Disease Models, Animal , Endoplasmic Reticulum Stress/immunology , Fluorouracil/administration & dosage , HCT116 Cells , HT29 Cells , Humans , Indoles/pharmacology , Irinotecan , Leucovorin/administration & dosage , Mice , Panitumumab , Phagocytosis/immunology , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Pyridones/pharmacology , Pyrimidinones/pharmacology , Sulfonamides/pharmacology , Unfolded Protein Response , Vemurafenib , X-Box Binding Protein 1/drug effects , X-Box Binding Protein 1/immunology , X-Box Binding Protein 1/metabolism
14.
Nat Commun ; 7: 11037, 2016 Mar 21.
Article in English | MEDLINE | ID: mdl-26996437

ABSTRACT

Excessive activation of blood coagulation and neutrophil accumulation have been described in several human cancers. However, whether hypercoagulation and neutrophilia are linked and involved in cancer development is currently unknown. Here we show that spontaneous intestinal tumorigenesis correlates with the accumulation of low-density neutrophils with a pro-tumorigenic N2 phenotype and unprompted neutrophil extracellular traps (NET) formation. We find that increased circulating lipopolysaccharide induces upregulation of complement C3a receptor on neutrophils and activation of the complement cascade. This leads to NETosis, induction of coagulation and N2 polarization, which prompts tumorigenesis, showing a novel link between coagulation, neutrophilia and complement activation. Finally, in a cohort of patients with small but not large intestinal cancer, we find a correlation between neutrophilia and hypercoagulation. This study provides a mechanistic explanation for the tumour-promoting effects of hypercoagulation, which could be used as a new biomarker or as a therapeutic target.


Subject(s)
Blood Coagulation , Carcinogenesis/immunology , Carcinogenesis/pathology , Extracellular Traps/metabolism , Intestine, Small/pathology , Neutrophils/metabolism , Receptors, Complement/metabolism , Adenomatous Polyposis Coli/genetics , Adult , Aged , Aged, 80 and over , Animals , Blood Coagulation/drug effects , Carcinogenesis/drug effects , Complement Activation/drug effects , Complement Pathway, Alternative/drug effects , Disease Progression , Extracellular Traps/drug effects , Hematopoiesis/drug effects , Hemostasis/drug effects , Heparin, Low-Molecular-Weight/pharmacology , Humans , Intestinal Neoplasms/pathology , Intestine, Small/drug effects , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Models, Biological , Neutrophils/drug effects , Phenotype
15.
Science ; 350(6262): 830-4, 2015 Nov 13.
Article in English | MEDLINE | ID: mdl-26564856

ABSTRACT

In healthy individuals, the intestinal microbiota cannot access the liver, spleen, or other peripheral tissues. Some pathogenic bacteria can reach these sites, however, and can induce a systemic immune response. How such compartmentalization is achieved is unknown. We identify a gut-vascular barrier (GVB) in mice and humans that controls the translocation of antigens into the blood stream and prohibits entry of the microbiota. Salmonella typhimurium can penetrate the GVB in a manner dependent on its pathogenicity island (Spi) 2-encoded type III secretion system and on decreased ß-catenin-dependent signaling in gut endothelial cells. The GVB is modified in celiac disease patients with elevated serum transaminases, which indicates that GVB dismantling may be responsible for liver damage in these patients. Understanding the GVB may provide new insights into the regulation of the gut-liver axis.


Subject(s)
Capillary Permeability/immunology , Intestines/immunology , Intestines/microbiology , Microbiota/immunology , Salmonella Infections/immunology , Salmonella typhimurium/immunology , Animals , Antigens, Bacterial/blood , Antigens, Bacterial/immunology , Celiac Disease/blood , Celiac Disease/immunology , Celiac Disease/microbiology , Genomic Islands/genetics , Genomic Islands/immunology , Humans , Ileum/blood supply , Ileum/immunology , Ileum/microbiology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestines/blood supply , Liver/immunology , Mice , Mice, Inbred C57BL , Salmonella typhimurium/genetics , Salmonella typhimurium/pathogenicity , Signal Transduction , Spleen/immunology , Transaminases/blood , Type III Secretion Systems/genetics , Type III Secretion Systems/immunology , Wnt Signaling Pathway , beta Catenin/metabolism
16.
Immunity ; 43(3): 527-40, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26362264

ABSTRACT

The interrelationship between IgAs and microbiota diversity is still unclear. Here we show that BALB/c mice had higher abundance and diversity of IgAs than C57BL/6 mice and that this correlated with increased microbiota diversity. We show that polyreactive IgAs mediated the entrance of non-invasive bacteria to Peyer's patches, independently of CX3CR1(+) phagocytes. This allowed the induction of bacteria-specific IgA and the establishment of a positive feedback loop of IgA production. Cohousing of mice or fecal transplantation had little or no influence on IgA production and had only partial impact on microbiota composition. Germ-free BALB/c, but not C57BL/6, mice already had polyreactive IgAs that influenced microbiota diversity and selection after colonization. Together, these data suggest that genetic predisposition to produce polyreactive IgAs has a strong impact on the generation of antigen-specific IgAs and the selection and maintenance of microbiota diversity.


Subject(s)
Antigens, Bacterial/immunology , Genetic Variation/immunology , Immunoglobulin A/immunology , Microbiota/immunology , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/immunology , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Feces/microbiology , Flow Cytometry , Host-Pathogen Interactions/immunology , Immunization , Immunoglobulin A/blood , Immunoglobulin A/metabolism , Metagenomics/methods , Mice, Inbred BALB C , Mice, Inbred C57BL , Microbiota/genetics , Peyer's Patches/immunology , Peyer's Patches/metabolism , Peyer's Patches/microbiology , Phylogeny , RNA, Ribosomal, 16S/genetics , Salmonella typhimurium/genetics , Salmonella typhimurium/immunology , Salmonella typhimurium/physiology , Species Specificity
17.
J Allergy Clin Immunol ; 136(2): 413-22, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26014813

ABSTRACT

BACKGROUND: Thymic stromal lymphopoietin (TSLP) is a cytokine with pleiotropic functions in the immune system. It has been associated with allergic reactions in the skin and lungs but also homeostatic tolerogenic responses in the thymus and gut. OBJECTIVE: In human subjects TSLP is present in 2 isoforms, short and long. Here we wanted to investigate the differential expression of the TSLP isoforms and discern their biological implications under homeostatic or inflammatory conditions. METHODS: We evaluated the expression of TSLPs in tissues from healthy subjects, patients with ulcerative colitis, patients with celiac disease, and patients with atopic dermatitis and on epithelial cells and keratinocytes under steady-state conditions or after stimulation. We then tested the immune activity of TSLP isoforms both in vitro and in vivo. RESULTS: We showed that TSLP isoforms are responsible for 2 opposite immune functions. The short isoform is expressed under steady-state conditions and exerts anti-inflammatory activities by affecting the capacity of PBMCs and dendritic cells to produce inflammatory cytokines. Moreover, the short isoform TSLP ameliorates experimental colitis in mice and prevents endotoxin shock. The long isoform of TSLP is proinflammatory and is only expressed during inflammation. The isoforms are differentially regulated by pathogenic bacteria, such as Salmonella species and adhesive-invasive Escherichia coli. CONCLUSIONS: We have solved the dilemma of TSLP being both homeostatic and inflammatory. The TSLP isoform ratio is altered during several inflammatory disorders, with strong implications in disease treatment and prevention. Indeed, targeting of the long isoform of TSLP at the C-terminal portion, which is common to both isoforms, might lead to unwanted side effects caused by neutralization of the homeostatic short isoform.


Subject(s)
Celiac Disease/immunology , Colitis, Ulcerative/immunology , Cytokines/immunology , Dermatitis, Atopic/immunology , Intestines/immunology , Skin/immunology , Animals , Case-Control Studies , Celiac Disease/genetics , Celiac Disease/microbiology , Celiac Disease/pathology , Colitis, Ulcerative/genetics , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/pathology , Cytokines/genetics , Dendritic Cells/immunology , Dendritic Cells/microbiology , Dendritic Cells/pathology , Dermatitis, Atopic/genetics , Dermatitis, Atopic/microbiology , Dermatitis, Atopic/pathology , Disease Models, Animal , Escherichia coli/immunology , Escherichia coli Infections/genetics , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology , Escherichia coli Infections/pathology , Female , Gene Expression Regulation/immunology , Humans , Intestines/microbiology , Intestines/pathology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/microbiology , Leukocytes, Mononuclear/pathology , Mice , Protein Isoforms/genetics , Protein Isoforms/immunology , Salmonella/immunology , Salmonella Infections/genetics , Salmonella Infections/immunology , Salmonella Infections/microbiology , Salmonella Infections/pathology , Skin/microbiology , Skin/pathology , Thymic Stromal Lymphopoietin
18.
PLoS One ; 9(2): e87615, 2014.
Article in English | MEDLINE | ID: mdl-24520333

ABSTRACT

The rapid expansion of commercially available fermented food products raises important safety issues particularly when infant food is concerned. In many cases, the activity of the microorganisms used for fermentation as well as what will be the immunological outcome of fermented food intake is not known. In this manuscript we used complex in vitro, ex-vivo and in vivo systems to study the immunomodulatory properties of probiotic-fermented products (culture supernatant and fermented milk without live bacteria to be used in infant formula). We found in vitro and ex-vivo that fermented products of Lactobacillus paracasei CBA L74 act via the inhibition of proinflammatory cytokine release leaving anti-inflammatory cytokines either unaffected or even increased in response to Salmonella typhimurium. These activities are not dependent on the inactivated bacteria but to metabolic products released during the fermentation process. We also show that our in vitro systems are predictive of an in vivo efficacy by the fermented products. Indeed CBA L74 fermented products (both culture medium and fermented milk) could protect against colitis and against an enteric pathogen infection (Salmonella typhimurium). Hence we found that fermented products can act via the inhibition of immune cell inflammation and can protect the host from pathobionts and enteric pathogens. These results open new perspectives in infant nutrition and suggest that L. paracasei CBA L74 fermented formula can provide immune benefits to formula-fed infants, without carrying live bacteria that may be potentially dangerous to an immature infant immune system.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Colitis/prevention & control , Dendritic Cells/metabolism , Fermentation/drug effects , Infant Formula/pharmacology , Lactobacillus/metabolism , Milk/metabolism , Salmonella typhimurium/drug effects , Administration, Oral , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/therapeutic use , Colitis/drug therapy , Colitis/microbiology , Dendritic Cells/drug effects , Humans , Infant , Infant Formula/administration & dosage , Inflammation Mediators/metabolism , Mice , Mice, Inbred C57BL , Protective Agents/administration & dosage , Protective Agents/pharmacology , Protective Agents/therapeutic use , Salmonella Infections, Animal/microbiology , Salmonella Infections, Animal/prevention & control , Salmonella typhimurium/physiology
19.
Immunity ; 40(2): 248-61, 2014 Feb 20.
Article in English | MEDLINE | ID: mdl-24462723

ABSTRACT

Antigen-presenting cells (APCs) in the gut are apt at oral tolerance establishment at steady state and immunity after infection; complex tasks in an environment exposed to the inflammatory burden of the microbiota. Here we show an unanticipated division of labor among APCs for the establishment of oral tolerance. Chemokine receptor CX3CR1(+) macrophages were found to take up soluble fed antigens and quickly transfer them to CD103(+) dendritic cells (DCs). Antigen transfer occurred via a mechanism that was Connexin 43-dependent and required membrane transfer, indicating a physiological role of gap junctions in antigen presentation. Deletion of Connexin 43 in APCs affected antigen transfer and resulted in the inability of CD103(+) DCs to acquire and present antigens in vivo, to drive T regulatory cell differentiation and to induce tolerance to food antigens. This functional cooperation between intestinal phagocytes might be a mechanism to avoid the exposure of tolerogenic DCs to the intestinal microbiota.


Subject(s)
Antigens, CD/metabolism , Antigens/immunology , Dendritic Cells , Food Hypersensitivity/immunology , Gap Junctions/immunology , Immune Tolerance , Integrin alpha Chains/metabolism , Macrophages/immunology , Receptors, Chemokine/metabolism , Animals , CX3C Chemokine Receptor 1 , Cells, Cultured , Dendritic Cells/metabolism , Flow Cytometry , Intestinal Mucosa/immunology , Mice , Models, Biological , Solubility
20.
Gut ; 61(7): 1007-15, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22301383

ABSTRACT

BACKGROUND AND AIMS: Probiotics and their metabolic products, here called postbiotics, have been proposed as food supplements for a healthier intestinal homeostasis, but also as therapeutic aids in inflammatory bowel disease (IBD) with, however, very little clinical benefit. This may be due to the lack of reliable preclinical models for testing the efficacy of different strains. METHODS: The activity of three probiotic strains of Lactobacillus (or a postbiotic) was analysed and compared with a pathogenic strain of Salmonella on a novel organ culture system of human healthy and IBD intestinal mucosa developed in our laboratory. The system maintains an apical to basolateral polarity during stimulation due to the presence of a glued cave cylinder. The cylinder is detached at the end of the experiment and the tissue is processed for histology and immunohistochemistry. Cytokines released from the basolateral side are analysed. RESULTS: The model system provides several physiological characteristics typical of a mucosal microenvironment including the presence of an organised mucus layer and an apical to basolateral polarity. Polarised administration of bacteria is critical to control the ensuing immune response as it mimics the physiological entrance of bacteria. The authors show that probiotics are not always beneficial for the healthy host and can also be detrimental in inflamed IBD. This study shows that a potent postbiotic can protect against the inflammatory properties of invasive Salmonella on healthy tissue and also downregulate ongoing inflammatory processes in IBD tissue. CONCLUSIONS: Probiotics can have inflammatory activities in both healthy and IBD tissue. Valid preclinical data on proper model systems should therefore be obtained before specific probiotic strains enter the clinics, especially if administered during acute inflammatory responses. Postbiotics may be a safe alternative for the treatment of patients with IBD in the acute inflammatory phase.


Subject(s)
Cytokines/metabolism , Inflammatory Bowel Diseases/therapy , Intestinal Mucosa/microbiology , Lactobacillus/metabolism , Organ Culture Techniques/methods , Probiotics/therapeutic use , Dietary Supplements , Humans , Immunohistochemistry , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Salmonella/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...