Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Semin Immunol ; 73: 101885, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38788491

ABSTRACT

The gut microbiota is well known to possess immunomodulatory capacities, influencing a multitude of cellular signalling pathways to maintain host homeostasis. Although the formation of the immune system initiates before birth in a sterile environment, an emerging body of literature indicates that the neonatal immune system is influenced by a first wave of external stimuli that includes signals from the maternal microbiota. A second wave of stimulus begins after birth and must be tightly regulated during the neonatal period when colonization of the host occurs concomitantly with the maturation of the immune system, requiring a fine adjustment between establishing tolerance towards the commensal microbiota and preserving inflammatory responses against pathogenic invaders. Besides integrating cues from commensal microbes, the neonatal immune system must also regulate responses triggered by other environmental signals, such as dietary antigens, which become more complex with the introduction of solid food during the weaning period. This "window of opportunity" in early life is thought to be crucial for the proper development of the immune system, setting the tone of subsequent immune responses in adulthood and modulating the risk of developing chronic and metabolic inflammatory diseases. Here we review the importance of host-microbiota interactions for the development and maturation of the immune system, particularly in the early-life period, highlighting the known mechanisms involved in such communication. This discussion is focused on recent data demonstrating microbiota-mediated education of innate immune cells and its role in the development of lymphoid tissues.

2.
J Leukoc Biol ; 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38652699

ABSTRACT

The Mammalian Target of Rapamycin (mTOR) pathway plays a key role in determining immune cells function through modulation of their metabolic status. By specific deletion of Rictor in CD11c+ myeloid cells (referred to here as CD11cRicΔ/Δ), this study investigated the role of mTOR complex 2 (mTORC2) signalling in dendritic cells (DCs) function in mice. We showed that upon DSS-induced colitis, lack of mTORC2 signalling CD11c+ cells diminishes colitis score, and abrogates dendritic cell (DC) migration to the mesenteric lymph nodes (MLN), thereby diminishing the infiltration of T helper (Th) 17 cells in the lamina propria (LP) and subsequent inflammation. These findings corroborate with abrogation of cytoskeleton organization and decreased activation of Rac1 and Cdc42 GTPases observed in CD11c+-mTORC2-deficient cells. Meta-analysis on colonic samples from ulcerative colitis (UC) patients revealed increased gene expression of pro-inflammatory cytokines which coincided with augmented expression of mTOR pathway, positive correlation between the DC marker ITGAX and IL-6, the expression of RICTOR, and CDC42. Together, this work proposes that targeting mTORC2 on DCs offers a key to hamper inflammatory responses and this way, ameliorates the progression and severity of intestinal inflammatory diseases.

3.
Cell Rep ; 42(5): 112507, 2023 05 30.
Article in English | MEDLINE | ID: mdl-37195866

ABSTRACT

During bloodstream infections, neutrophils home to the liver as part of an intravascular immune response to eradicate blood-borne pathogens, but the mechanisms regulating this crucial response are unknown. Using in vivo imaging of neutrophil trafficking in germ-free and gnotobiotic mice, we demonstrate that the intestinal microbiota guides neutrophil homing to the liver in response to infection mediated by the microbial metabolite D-lactate. Commensal-derived D-lactate augments neutrophil adhesion in the liver independent of granulopoiesis in bone marrow or neutrophil maturation and activation in blood. Instead, gut-to-liver D-lactate signaling primes liver endothelial cells to upregulate adhesion molecule expression in response to infection and promote neutrophil adherence. Targeted correction of microbiota D-lactate production in a model of antibiotic-induced dysbiosis restores neutrophil homing to the liver and reduces bacteremia in a model of Staphylococcus aureus infection. These findings reveal long-distance traffic control of neutrophil recruitment to the liver by microbiota-endothelium crosstalk.


Subject(s)
Endothelial Cells , Microbiota , Animals , Mice , Neutrophil Infiltration , Neutrophils/metabolism , Liver/metabolism , Endothelium , Lactates/metabolism
4.
Immunity ; 55(7): 1250-1267.e12, 2022 07 12.
Article in English | MEDLINE | ID: mdl-35709757

ABSTRACT

The intestine harbors a large population of resident eosinophils, yet the function of intestinal eosinophils has not been explored. Flow cytometry and whole-mount imaging identified eosinophils residing in the lamina propria along the length of the intestine prior to postnatal microbial colonization. Microscopy, transcriptomic analysis, and mass spectrometry of intestinal tissue revealed villus blunting, altered extracellular matrix, decreased epithelial cell turnover, increased gastrointestinal motility, and decreased lipid absorption in eosinophil-deficient mice. Mechanistically, intestinal epithelial cells released IL-33 in a microbiota-dependent manner, which led to eosinophil activation. The colonization of germ-free mice demonstrated that eosinophil activation in response to microbes regulated villous size alterations, macrophage maturation, epithelial barrier integrity, and intestinal transit. Collectively, our findings demonstrate a critical role for eosinophils in facilitating the mutualistic interactions between the host and microbiota and provide a rationale for the functional significance of their early life recruitment in the small intestine.


Subject(s)
Communicable Diseases , Microbiota , Animals , Eosinophils , Homeostasis , Intestinal Mucosa , Intestine, Small , Mice
5.
J Immunol ; 207(2): 626-639, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34261666

ABSTRACT

Sepsis is a complex infectious syndrome in which neutrophil participation is crucial for patient survival. Neutrophils quickly sense and eliminate the pathogen by using different effector mechanisms controlled by metabolic processes. The mammalian target of rapamycin (mTOR) pathway is an important route for metabolic regulation, and its role in neutrophil metabolism has not been fully understood yet, especially the importance of mTOR complex 2 (mTORC2) in the neutrophil effector functions. In this study, we observed that the loss of Rictor (mTORC2 scaffold protein) in primary mouse-derived neutrophils affects their chemotaxis by fMLF and their microbial killing capacity, but not the phagocytic capacity. We found that the microbicidal capacity was impaired in Rictor-deleted neutrophils because of an improper fusion of granules, reducing the hypochlorous acid production. The loss of Rictor also led to metabolic alterations in isolated neutrophils, increasing aerobic glycolysis. Finally, myeloid-Rictor-deleted mice (LysMRic Δ/Δ) also showed an impairment of the microbicidal capacity, increasing the bacterial burden in the Escherichia coli sepsis model. Overall, our results highlight the importance of proper mTORC2 activation for neutrophil effector functions and metabolism during sepsis.


Subject(s)
Mechanistic Target of Rapamycin Complex 2/metabolism , Neutrophils/metabolism , Sepsis/metabolism , Sepsis/microbiology , Animals , Chemotaxis/physiology , Escherichia coli/metabolism , Female , Glycolysis/physiology , Humans , Hypochlorous Acid/metabolism , Mice , Mice, Inbred C57BL , Phagocytosis/physiology , Signal Transduction/physiology
6.
Mucosal Immunol ; 13(4): 574-583, 2020 07.
Article in English | MEDLINE | ID: mdl-32157190

ABSTRACT

Eosinophils are granulocytes, typically implicated as end-stage effector cells in type-II immune responses. They are capable of producing a wide array of pre-formed molecules which render them with vast potential to influence a wide variety of processes. Nonetheless, eosinophil research has traditionally focused on their role in anti-helminthic responses and pathophysiological processes in type-II immune disorders, such as allergy and asthma, where eosinophilia is a hallmark phenotype. However, a number of key studies over the past decade have placed this restricted view of eosinophil function into question, presenting additional evidence for eosinophils as critical regulators of various homeostatic processes including immune maintenance, organ development, and tissue regeneration.


Subject(s)
Eosinophils/immunology , Eosinophils/metabolism , Homeostasis , Immunity, Mucosal , Mucous Membrane/cytology , Mucous Membrane/physiology , Animals , Cell Differentiation/immunology , Cellular Microenvironment/immunology , Chemotaxis/immunology , Disease Susceptibility , Eosinophils/cytology , Humans , Organ Specificity/immunology
7.
Inflamm Bowel Dis ; 26(5): 697-708, 2020 04 11.
Article in English | MEDLINE | ID: mdl-31819985

ABSTRACT

BACKGROUND: The gut microbiota is a key element to support host homeostasis and the development of the immune system. The relationship between the microbiota and immunity is a 2-way road, in which the microbiota contributes to the development/function of immune cells and immunity can affect the composition of microbes. In this context, natural killer T cells (NKT cells) are distinct T lymphocytes that play a role in gut immunity and are influenced by gut microbes. In our work, we investigated the involvement of invariant NKT cells (iNKT) in intestinal homeostasis. RESULTS: We found that iNKT-deficient mice (iNKT-KO) had reduced levels of fecal IgA and an altered composition of the gut microbiota, with increased Bacteroidetes. The absence of iNKT cells also affected TGF-ß1 levels and plasma cells, which were significantly reduced in knockout (KO) mice. In addition, when submitted to dextran sodium sulfate colitis, iNKT-KO mice had worsening of colitis when compared with wild-type (WT) mice. To further address iNKT cell contribution to intestinal homeostasis, we adoptively transferred iNKT cells to KO mice, and they were submitted to colitis. Transfer of iNKT cells improved colitis and restored fecal IgA levels and gut microbiota. CONCLUSIONS: Our results indicate that intestinal NKT cells are important modulators of intestinal homeostasis and that gut microbiota composition may be a potential target in the management of inflammatory bowel diseases.


Subject(s)
Gastrointestinal Microbiome/immunology , Homeostasis/immunology , Immunoglobulin A/analysis , Intestines/immunology , Natural Killer T-Cells/immunology , Animals , Colitis/chemically induced , Colitis/immunology , Colitis/microbiology , Dextran Sulfate , Disease Models, Animal , Feces/chemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
8.
Toxicol Rep ; 5: 512-520, 2018.
Article in English | MEDLINE | ID: mdl-29854623

ABSTRACT

Environmental and Occupational pollution has been extensively studied because of its serious implications on the human health. Formaldehyde (FA) is a pollutant widely employed in several industries and also in anatomy, pathology and histology laboratories. Studies have shown the correlation between FA exposure and development or worsening of asthma. However, the effect of FA exposure on the pulmonary fibrosis (PF) is unknown. PF is a progressive and chronic lung disease with high incidence and considerable morbidity and mortality. Few studies have shown a worsening of PF after pollutants exposure such as ozone and nitrogen dioxide. Therefore, our objective was to assess the effects of FA on the PF. Male mice C57BL6 were treated or not with bleomycin (1,5 U/kg) and exposed or not to FA inhalation (0.92 mg/m3, 1 h/day, 5 days/week during 2 weeks). Non-manipulated mice were used as control. Our data showed that FA exposure in fibrotic mice increased the number of granulocytes in the bronchoalveolar lavage followed by elevated levels of interleukin 1 beta and interleukin 17. In addition, FA exposure in fibrotic mice enhanced the gene expression of C-X-C motif chemokine ligand 1 (CXCL1) and tumor necrosis factor alpha (TNF-α) in the lung. We also showed an increase in the collagen production, while lung elastance was reduced. No differences were found in the mucus production, oedema and interstitial thickening in the lung tissue of fibrotic mice after FA exposure. In conclusion our study showed that FA exposure aggravates the lung neutrophils influx and collagen production, but did not alter the lung elastance, mucus production, oedema and interstitial tickening. This work contributes to understand the effects of pollution in the development of PF.

9.
Curr Opin Immunol ; 54: 20-27, 2018 10.
Article in English | MEDLINE | ID: mdl-29864675

ABSTRACT

The trillions of microbes that colonize mucosal surfaces are critical for educating the immune system and microbial-derived signals continually shape and set the tone of immune responses. Although Type 2 immune responses are important for mediating protection from helminth infection they also underlie atopy and allergy. Microbes modulate Type 2 immune responses through effects on Type 2 cytokines, dendritic cells and regulatory T cells. Microbial colonization in the gut, the lung and the skin during an early and critical time period in immune development appears to be of particular importance for tolerance induction and regulation of aberrant Type 2 immune responses. This is illustrated by studies showing microbial alterations in early life that are associated with allergies later in life.


Subject(s)
Hypersensitivity/immunology , Hypersensitivity/microbiology , Microbiota/immunology , Th2 Cells/immunology , Animals , Cytokines/immunology , Dendritic Cells/immunology , Humans , T-Lymphocytes, Regulatory/immunology
10.
Inflammopharmacology ; 26(1): 251-260, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29063489

ABSTRACT

Inflammatory bowel diseases (IBDs) affect millions of people worldwide and their frequencies in developed countries have increased since the twentieth century. In this context, there is an intensive search for therapies that modulate inflammation and provide tissue regeneration in IBDs. Recently, the immunomodulatory activity of adipose tissue-derived mesenchymal stromal cells (ADMSCs) has been demonstrated to play an important role on several immune cells in different conditions of inflammatory and autoimmune diseases. In this study, we explored the immunomodulatory potential of ADMSC in a classical model of DSS-induced colitis. First, we found that treatment of mice with ADMSC ameliorated the severity of DSS-induced colitis, reducing colitis pathological score and preventing colon shortening. Moreover, a prominent reduction of pro-inflammatory cytokines levels (i.e., IFN-γ, TNF-α, IL-6 and MCP-1) was observed in the colon of animals treated with ADMSC. We also observed a significant reduction in the frequencies of macrophages (F4/80+CD11b+) and dendritic cells (CD11c+CD103+) in the intestinal lamina propria of ADMSC-treated mice. Finally, we detected the up-regulation of immunoregulatory-associated molecules in intestine of mice treated with ADMSCs (i.e., elevated arginase-1 and IL-10). Thus, this present study demonstrated that ADMSC modulates the overall gut inflammation (cell activation and recruitment) in experimental colitis, providing support to the further development of new strategies in the treatment of intestinal diseases.


Subject(s)
Colitis/metabolism , Colitis/pathology , Inflammation Mediators/metabolism , Inflammation/metabolism , Inflammation/pathology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Colon/metabolism , Colon/pathology , Cytokines/metabolism , Dendritic Cells/metabolism , Dendritic Cells/pathology , Disease Models, Animal , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL
11.
Sci Rep ; 7(1): 12670, 2017 10 04.
Article in English | MEDLINE | ID: mdl-28978926

ABSTRACT

Sepsis is a severe disease with a high mortality index and it is responsible for the development of acute lung injury (ALI). We evaluated the effects of light-emitting diode (LED) on ALI induced by sepsis. Balb-c mice were injected with lipopolysaccharide or saline and then irradiated or not with red LED on their tracheas and lungs for 150 s, 2 and 6 h after LPS injections. The parameters were investigated 24 h after the LPS injections. Red LED treatment reduced neutrophil influx and the levels of interleukins 1ß, 17 A and, tumor necrosis factor-α; in addition to enhanced levels of interferon γ in the bronchoalveolar fluid. Moreover, red LED treatment enhanced the RNAm levels of IL-10 and IFN-γ. It also partially reduced the elevated oxidative burst and enhanced apoptosis, but it did not alter the translocation of nuclear factor κB, the expression of toll-like receptor 4 (TLR4), as well as, oedema or mucus production in their lung tissues. Together, our data has shown the beneficial effects of short treatment with LED on ALI that are caused by gram negative bacterial infections. It is suggested that LED applications are an inexpensive and non-invasive additional treatment for sepsis.


Subject(s)
Acute Lung Injury/therapy , Light , Sepsis/therapy , Acute Lung Injury/etiology , Animals , Bronchoalveolar Lavage Fluid , Disease Models, Animal , Gene Expression Regulation/radiation effects , Humans , Interleukin-17/genetics , Interleukin-1beta/genetics , Lung/metabolism , Lung/pathology , Mice , Mice, Inbred BALB C , NF-kappa B/genetics , Sepsis/chemically induced , Sepsis/complications , Signal Transduction/radiation effects , Toll-Like Receptor 4/genetics , Tumor Necrosis Factor-alpha/genetics
12.
World J Hepatol ; 9(23): 979-989, 2017 Aug 18.
Article in English | MEDLINE | ID: mdl-28878863

ABSTRACT

Innate lymphoid cells (ILCs) are the most recently discovered family of innate immune cells. They are a part of the innate immune system, but develop from the lymphoid lineage. They lack pattern-recognition receptors and rearranged receptors, and therefore cannot directly mediate antigen specific responses. The progenitors specifically associated with the ILCs lineage have been uncovered, enabling the distinction between ILCs and natural killer cells. Based on the requirement of specific transcription factors and their patterns of cytokine production, ILCs are categorized into three subsets (ILC1, ILC2 and ILC3). First observed in mucosal surfaces, these cell populations interact with hematopoietic and non-hematopoietic cells throughout the body during homeostasis and diseases, promoting immunity, commensal microbiota tolerance, tissue repair and inflammation. Over the last 8 years, ILCs came into the spotlight as an essential cell type able to integrate diverse host immune responses. Recently, it became known that ILC subsets play a key role in immune responses at barrier surfaces, interacting with the microbiota, nutrients and metabolites. Since the liver receives the venous blood directly from the intestinal vein, the intestine and liver are essential to maintain tolerance and can rapidly respond to infections or tissue damage. Therefore, in this review, we discuss recent findings regarding ILC functions in homeostasis and disease, with a focus on the intestine and liver.

13.
Cell Rep ; 19(11): 2272-2288, 2017 06 13.
Article in English | MEDLINE | ID: mdl-28614714

ABSTRACT

The underlying mechanism by which MyD88 regulates the development of obesity, metainflammation, and insulin resistance (IR) remains unknown. Global deletion of MyD88 in high-fat diet (HFD)-fed mice resulted in increased weight gain, impaired glucose homeostasis, elevated Dectin-1 expression in adipose tissue (AT), and proinflammatory CD11c+ AT macrophages (ATMs). Dectin-1 KO mice were protected from diet-induced obesity (DIO) and IR and had reduced CD11c+ AT macrophages. Dectin-1 antagonist improved glucose homeostasis and decreased CD11c+ AT macrophages in chow- and HFD-fed MyD88 KO mice. Dectin-1 agonist worsened glucose homeostasis in MyD88 KO mice. Dectin-1 expression is increased in AT from obese individuals. Together, our data indicate that Dectin-1 regulates AT inflammation by promoting CD11c+ AT macrophages in the absence of MyD88 and identify a role for Dectin-1 in chronic inflammatory states, such as obesity. This suggests that Dectin-1 may have therapeutic implications as a biomarker for metabolic dysregulation in humans.


Subject(s)
Adipose Tissue/metabolism , Insulin Resistance/genetics , Lectins, C-Type/metabolism , Macrophages/metabolism , Obesity/genetics , Animals , Humans , Male , Mice
14.
Microb Drug Resist ; 23(1): 56-62, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27328205

ABSTRACT

The administration of antimicrobial agents leads to an ecological imbalance of the host-microorganisms relationship, and it causes a rapid and significant reduction in the microbial diversity. The aim of the current study was to evaluate the impact of antibiotic therapy on intestinal microbiota of children between 3 and 12 years of age. The fecal samples were collected from hospitalized children (n = 31) and from healthy untreated children (n = 30). The presence of bacteria and their quantities were assessed by culture-based methods and quantitative polymerase chain reaction (qPCR). By culture method, in the children receiving antibiotics, a low recovery of Bifidobacterium spp. (54.8%), Bacteroides spp./Parabacteroides spp. (54.8%), Clostridium spp. (35.5%), and Escherichia coli (74.2%) was observed compared with the children without antibiotic therapy (100%, 80%, 63.3%, and 86.6%, respectively). By qPCR, the children receiving antibiotics showed a lower copy number for all microorganisms, except to Lactobacillus spp. (p = 0.0092). In comparison to the nontreated children, the antibiotic-treated children showed a significantly lower copy number of Bifidobacterium spp. (p = 0.0002), Clostridium perfringens (p < 0.0001), E. coli (p = 0.0268), Methanobrevibacter smithii (p = 0.0444), and phylum Firmicutes (p = 0.0009). In conclusion, our results obtained through qualitative and quantitative analyses, demonstrate that antibiotic therapy affect the intestinal microbiome of children.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Infections/drug therapy , DNA, Bacterial/genetics , Gastrointestinal Microbiome/drug effects , Bacterial Infections/microbiology , Bacterial Typing Techniques , Bacteroides/drug effects , Bacteroides/genetics , Bacteroides/growth & development , Bacteroides/isolation & purification , Bifidobacterium/drug effects , Bifidobacterium/genetics , Bifidobacterium/growth & development , Bifidobacterium/isolation & purification , Case-Control Studies , Child , Child, Preschool , Clostridium/drug effects , Clostridium/genetics , Clostridium/growth & development , Clostridium/isolation & purification , Escherichia coli/drug effects , Escherichia coli/genetics , Escherichia coli/growth & development , Escherichia coli/isolation & purification , Feces/microbiology , Female , Firmicutes/drug effects , Firmicutes/genetics , Firmicutes/growth & development , Firmicutes/isolation & purification , Gastrointestinal Microbiome/genetics , Humans , Lactobacillus/drug effects , Lactobacillus/genetics , Lactobacillus/growth & development , Lactobacillus/isolation & purification , Male , Methanobrevibacter/drug effects , Methanobrevibacter/genetics , Methanobrevibacter/growth & development , Methanobrevibacter/isolation & purification
15.
Cell Reports ; 19(11): 2272-2288, 2017.
Article in English | Sec. Est. Saúde SP, SESSP-IBPROD, Sec. Est. Saúde SP | ID: but-ib15153

ABSTRACT

The underlying mechanism by which MyD88 regulates the development of obesity, metainflammation, and insulin resistance (IR) remains unknown. Global deletion of MyD88 in high-fat diet (HFD)fed mice resulted in increased weight gain, impaired glucose homeostasis, elevated Dectin-1 expression in adipose tissue (AT), and proinflammatory CD11c+ AT macrophages (ATMs). Dectin-1 KO mice were protected from diet-induced obesity (DIO) and IR and had reduced CD11c+ AT macrophages. Dectin-1 antagonist improved glucose homeostasis and decreased CD11c+ AT macrophages in chow-and HFD-fed MyD88 KO mice. Dectin-1 agonist worsened glucose homeostasis in MyD88 KO mice. Dectin-1 expression is increased in AT from obese individuals. Together, our data indicate that Dectin-1 regulates AT inflammation by promoting CD11c+ AT macrophages in the absence of MyD88 and identify a role for Dectin-1 in chronic inflammatory states, such as obesity. This suggests that Dectin-1 may have ther-apeutic implications as a biomarker for metabolic dysregulation in humans.

16.
J Exp Med ; 213(7): 1223-39, 2016 06 27.
Article in English | MEDLINE | ID: mdl-27325889

ABSTRACT

Type 1 diabetes (T1D) is an autoimmune disease that is triggered by both genetic and environmental factors, resulting in the destruction of pancreatic ß cells. The disruption of the intestinal epithelial barrier and consequent escape of microbial products may be one of these environmental triggers. However, the immune receptors that are activated in this context remain elusive. We show here that during streptozotocin (STZ)-induced T1D, the nucleotide-binding oligomerization domain containing 2 (NOD2), but not NOD1, participates in the pathogenesis of the disease by inducing T helper 1 (Th1) and Th17 cells in the pancreatic LNs (PLNs) and pancreas. Additionally, STZ-injected wild-type (WT) diabetic mice displayed an altered gut microbiota compared with vehicle-injected WT mice, together with the translocation of bacteria to the PLNs. Interestingly, WT mice treated with broad-spectrum antibiotics (Abx) were fully protected from STZ-induced T1D, which correlated with the abrogation of bacterial translocation to the PLNs. Notably, when Abx-treated STZ-injected WT mice received the NOD2 ligand muramyl dipeptide, both hyperglycemia and the proinflammatory immune response were restored. Our results demonstrate that the recognition of bacterial products by NOD2 inside the PLNs contributes to T1D development, establishing a new putative target for intervention during the early stages of the disease.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1 , Gastrointestinal Microbiome , Lymph Nodes , Nod2 Signaling Adaptor Protein/immunology , Pancreas , Animals , Bacterial Translocation/genetics , Bacterial Translocation/immunology , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/microbiology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/microbiology , Diabetes Mellitus, Type 1/pathology , Lymph Nodes/immunology , Lymph Nodes/microbiology , Lymph Nodes/pathology , Male , Mice , Mice, Knockout , Nod2 Signaling Adaptor Protein/genetics , Pancreas/immunology , Pancreas/microbiology , Pancreas/pathology
17.
Front Immunol ; 7: 54, 2016.
Article in English | MEDLINE | ID: mdl-26925061

ABSTRACT

The mammalian gastrointestinal tract harbors a diverse microbial community with which dynamic interactions have been established over millennia of coevolution. Commensal bacteria and their products are sensed by innate receptors expressed in gut epithelia and in gut-associated immune cells, thereby promoting the proper development of mucosal immune system and host homeostasis. Many studies have demonstrated that host-microbiota interactions play a key role during local and systemic immunity. Therefore, this review will focus on how innate sensing of the gut microbiota and their metabolites through inflammasome and toll-like receptors impact the modulation of a distinct set of inflammatory and autoimmune diseases. We believe that a better understanding of the fine-tuning that governs host-microbiota interactions will further improve common prophylactic and therapeutic applications.

18.
Braz J Microbiol ; 46(4): 1135-40, 2015.
Article in English | MEDLINE | ID: mdl-26691472

ABSTRACT

Colorectal carcinoma is considered the fourth leading cause of cancer deaths worldwide. Several microorganisms have been associated with carcinogenesis, including Enterococcus spp., Helicobacter pylori, enterotoxigenic Bacteroides fragilis, pathogenic E. coli strains and oral Fusobacterium. Here we qualitatively and quantitatively evaluated the presence of oral and intestinal microorganisms in the fecal microbiota of colorectal cancer patients and healthy controls. Seventeen patients (between 49 and 70 years-old) visiting the Cancer Institute of the Sao Paulo State were selected, 7 of whom were diagnosed with colorectal carcinoma. Bacterial detection was performed by qRT-PCR. Although all of the tested bacteria were detected in the majority of the fecal samples, quantitative differences between the Cancer Group and healthy controls were detected only for F. nucleatum and C. difficile. The three tested oral microorganisms were frequently observed, suggesting a need for furthers studies into a potential role for these bacteria during colorectal carcinoma pathogenesis. Despite the small number of patients included in this study, we were able to detect significantly more F. nucleatum and C. difficile in the Cancer Group patients compared to healthy controls, suggesting a possible role of these bacteria in colon carcinogenesis. This finding should be considered when screening for colorectal cancer.


Subject(s)
Clostridioides difficile/isolation & purification , Clostridium Infections/complications , Colorectal Neoplasms/complications , Fusobacterium Infections/complications , Fusobacterium nucleatum/isolation & purification , Gastrointestinal Microbiome , Aged , Brazil/epidemiology , Clostridium Infections/epidemiology , Clostridium Infections/microbiology , Female , Fusobacterium Infections/epidemiology , Fusobacterium Infections/microbiology , Humans , Male , Middle Aged , Real-Time Polymerase Chain Reaction
19.
Braz J Microbiol ; 46(4): 1141-5, 2015.
Article in English | MEDLINE | ID: mdl-26691473

ABSTRACT

Enterotoxigenic Bacteroides fragilis (ETBF) is an important part of the human and animal intestinal microbiota and is commonly associated with diarrhea. ETBF strains produce an enterotoxin encoded by the bft gene located in the B. fragilis pathogenicity island (BfPAI). Non-enterotoxigenic B. fragilis (NTBF) strains lack the BfPAI and usually show two different genetic patterns, II and III, based on the absence or presence of a BfPAI-flanking region, respectively. The incidence of ETBF and NTBF strains in fecal samples isolated from children without acute diarrhea or any other intestinal disorders was determined. All 84 fecal samples evaluated were B. fragilis-positive by PCR, four of them harbored the bft gene, 27 contained the NTBF pattern III DNA sequence, and 52 were considered to be NTBF pattern II samples. One sample was positive for both ETBF and NTBF pattern III DNA sequences. All 19 B. fragilis strains isolated by the culture method were bft-negative, 9 belonged to pattern III and 10 to pattern II. We present an updated overview of the ETBF and NTBF incidence in the fecal microbiota of children from Sao Paulo City, Brazil.


Subject(s)
Bacterial Toxins/genetics , Bacteroides Infections/microbiology , Bacteroides fragilis/genetics , Bacteroides fragilis/isolation & purification , Feces/microbiology , Genotype , Metalloendopeptidases/genetics , Animals , Bacteroides Infections/epidemiology , Bacteroides fragilis/classification , Brazil/epidemiology , Child , Child, Preschool , DNA, Bacterial/genetics , Female , Humans , Incidence , Male , Molecular Typing , Polymerase Chain Reaction
20.
Braz. j. microbiol ; 46(4): 1141-1145, Oct.-Dec. 2015. tab
Article in English | LILACS | ID: lil-769648

ABSTRACT

Enterotoxigenic Bacteroides fragilis (ETBF) is an important part of the human and animal intestinal microbiota and is commonly associated with diarrhea. ETBF strains produce an enterotoxin encoded by the bft gene located in the B. fragilis pathogenicity island (BfPAI). Non-enterotoxigenic B. fragilis (NTBF) strains lack the BfPAI and usually show two different genetic patterns, II and III, based on the absence or presence of a BfPAI-flanking region, respectively. The incidence of ETBF and NTBF strains in fecal samples isolated from children without acute diarrhea or any other intestinal disorders was determined. All 84 fecal samples evaluated were B. fragilis-positive by PCR, four of them harbored the bft gene, 27 contained the NTBF pattern III DNA sequence, and 52 were considered to be NTBF pattern II samples. One sample was positive for both ETBF and NTBF pattern III DNA sequences. All 19 B. fragilis strains isolated by the culture method were bft-negative, 9 belonged to pattern III and 10 to pattern II. We present an updated overview of the ETBF and NTBF incidence in the fecal microbiota of children from Sao Paulo City, Brazil.


Subject(s)
Animals , Child , Child, Preschool , Female , Humans , Male , Bacterial Toxins/genetics , Bacteroides Infections/microbiology , Bacteroides fragilis/genetics , Bacteroides fragilis/isolation & purification , Feces/microbiology , Genotype , Metalloendopeptidases/genetics , Bacteroides Infections/epidemiology , Bacteroides fragilis/classification , Brazil/epidemiology , DNA, Bacterial/genetics , Incidence , Molecular Typing , Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...