Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Blood Adv ; 7(17): 4970-4982, 2023 09 12.
Article in English | MEDLINE | ID: mdl-37276450

ABSTRACT

Metabolic products of the microbiota can alter hematopoiesis. However, the contribution and site of action of bile acids is poorly understood. Here, we demonstrate that the secondary bile acids, deoxycholic acid (DCA) and lithocholic acid (LCA), increase bone marrow myelopoiesis. Treatment of bone marrow cells with DCA and LCA preferentially expanded immunophenotypic and functional colony-forming unit-granulocyte and macrophage (CFU-GM) granulocyte-monocyte progenitors (GMPs). DCA treatment of sorted hematopoietic stem and progenitor cells (HSPCs) increased CFU-GMs, indicating that direct exposure of HSPCs to DCA sufficed to increase GMPs. The vitamin D receptor (VDR) was required for the DCA-induced increase in CFU-GMs and GMPs. Single-cell RNA sequencing revealed that DCA significantly upregulated genes associated with myeloid differentiation and proliferation in GMPs. The action of DCA on HSPCs to expand GMPs in a VDR-dependent manner suggests microbiome-host interactions could directly affect bone marrow hematopoiesis and potentially the severity of infectious and inflammatory disease.


Subject(s)
Bile Acids and Salts , Myelopoiesis , Receptors, Calcitriol , Bile Acids and Salts/metabolism , Myeloid Progenitor Cells , Receptors, Calcitriol/genetics , Receptors, Calcitriol/metabolism
2.
Mucosal Immunol ; 15(1): 165-175, 2022 01.
Article in English | MEDLINE | ID: mdl-34400793

ABSTRACT

Entamoeba histolytica is a pathogenic protozoan parasite that causes intestinal colitis, diarrhea, and in some cases, liver abscess. Through transcriptomics analysis, we observed that E. histolytica infection was associated with increased expression of IL-33 mRNA in both the human and murine colon. IL-33, the IL-1 family cytokine, is released after cell injury to alert the immune system of tissue damage. Treatment with recombinant IL-33 protected mice from amebic infection and intestinal tissue damage; moreover, blocking IL-33 signaling made mice more susceptible to amebiasis. IL-33 limited the recruitment of inflammatory immune cells and decreased the pro-inflammatory cytokine IL-6 in the cecum. Type 2 immune responses were upregulated by IL-33 treatment during amebic infection. Interestingly, administration of IL-33 protected RAG2-/- mice but not RAG2-/-γc-/- mice, demonstrating that IL-33-mediated protection required the presence of innate lymphoid cells (ILCs). IL-33 induced recruitment of ILC2 but not ILC1 and ILC3 in RAG2-/- mice. At baseline and after amebic infection, there was a significantly higher IL13+ILC2s in C57BL/J mice, which are naturally resistant to amebiasis, than CBA/J mice. Adoptive transfer of ILC2s to RAG2-/-γc-/- mice restored IL-33-mediated protection. These data reveal that the IL-33-ILC2 pathway is an important host defense mechanism against amebic colitis.


Subject(s)
Colon/physiology , Dysentery, Amebic/immunology , Entamoeba histolytica/physiology , Entamoebiasis/immunology , Interleukin-33/genetics , Lymphocytes/immunology , RNA, Messenger/genetics , Animals , Cell Movement , Colon/parasitology , DNA-Binding Proteins/genetics , Disease Resistance , Gene Expression Profiling , Genetic Background , Humans , Immunity, Innate , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Signal Transduction , Th1 Cells/immunology , Th2 Cells/immunology
3.
Front Immunol ; 12: 683157, 2021.
Article in English | MEDLINE | ID: mdl-34248966

ABSTRACT

Amebiasis is a neglected tropical disease caused by Entamoeba histolytica. Although the disease burden varies geographically, amebiasis is estimated to account for some 55,000 deaths and millions of infections globally per year. Children and travelers are among the groups with the greatest risk of infection. There are currently no licensed vaccines for prevention of amebiasis, although key immune correlates for protection have been proposed from observational studies in humans. We previously described the development of a liposomal adjuvant formulation containing two synthetic TLR ligands (GLA and 3M-052) that enhanced antigen-specific fecal IgA, serum IgG2a, a mixed IFNγ and IL-17A cytokine profile from splenocytes, and protective efficacy following intranasal administration with the LecA antigen. By applying a statistical design of experiments (DOE) and desirability function approach, we now describe the optimization of the dose of each vaccine formulation component (LecA, GLA, 3M-052, and liposome) as well as the excipient composition (acyl chain length and saturation; PEGylated lipid:phospholipid ratio; and presence of antioxidant, tonicity, or viscosity agents) to maximize desired immunogenicity characteristics while maintaining physicochemical stability. This DOE/desirability index approach led to the identification of a lead candidate composition that demonstrated immune response durability and protective efficacy in the mouse model, as well as an assessment of the impact of each active vaccine formulation component on protection. Thus, we demonstrate that both GLA and 3M-052 are required for statistically significant protective efficacy. We also show that immunogenicity and efficacy results differ in female vs male mice, and the differences appear to be at least partly associated with adjuvant formulation composition.


Subject(s)
Antigens, Protozoan/immunology , Entamoeba histolytica/immunology , Entamoebiasis/immunology , Entamoebiasis/prevention & control , Protozoan Vaccines/immunology , Adjuvants, Immunologic/chemistry , Administration, Intranasal , Animals , Antibodies, Protozoan/blood , Antibodies, Protozoan/immunology , Chemical Phenomena , Cytokines/metabolism , Drug Compounding , Entamoebiasis/parasitology , Enzyme-Linked Immunosorbent Assay , Humans , Immunogenicity, Vaccine , Immunoglobulin G/immunology , Liposomes , Mice , Protozoan Vaccines/administration & dosage , Protozoan Vaccines/chemistry , Vaccination
4.
JCI Insight ; 6(15)2021 08 09.
Article in English | MEDLINE | ID: mdl-34185704

ABSTRACT

Immune dysregulation is characteristic of the more severe stages of SARS-CoV-2 infection. Understanding the mechanisms by which the immune system contributes to COVID-19 severity may open new avenues to treatment. Here, we report that elevated IL-13 was associated with the need for mechanical ventilation in 2 independent patient cohorts. In addition, patients who acquired COVID-19 while prescribed Dupilumab, a mAb that blocks IL-13 and IL-4 signaling, had less severe disease. In SARS-CoV-2-infected mice, IL-13 neutralization reduced death and disease severity without affecting viral load, demonstrating an immunopathogenic role for this cytokine. Following anti-IL-13 treatment in infected mice, hyaluronan synthase 1 (Has1) was the most downregulated gene, and accumulation of the hyaluronan (HA) polysaccharide was decreased in the lung. In patients with COVID-19, HA was increased in the lungs and plasma. Blockade of the HA receptor, CD44, reduced mortality in infected mice, supporting the importance of HA as a pathogenic mediator. Finally, HA was directly induced in the lungs of mice by administration of IL-13, indicating a new role for IL-13 in lung disease. Understanding the role of IL-13 and HA has important implications for therapy of COVID-19 and, potentially, other pulmonary diseases. IL-13 levels were elevated in patients with severe COVID-19. In a mouse model of the disease, IL-13 neutralization reduced the disease and decreased lung HA deposition. Administration of IL-13-induced HA in the lung. Blockade of the HA receptor CD44 prevented mortality, highlighting a potentially novel mechanism for IL-13-mediated HA synthesis in pulmonary pathology.


Subject(s)
COVID-19/immunology , Interleukin-13/immunology , SARS-CoV-2/immunology , Animals , COVID-19/blood , COVID-19/pathology , COVID-19/therapy , Disease Models, Animal , Disease Progression , Female , Humans , Interleukin-13/blood , Lung/immunology , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Severity of Illness Index
5.
medRxiv ; 2021 Mar 01.
Article in English | MEDLINE | ID: mdl-33688686

ABSTRACT

Immune dysregulation is characteristic of the more severe stages of SARS-CoV-2 infection. Understanding the mechanisms by which the immune system contributes to COVID-19 severity may open new avenues to treatment. Here we report that elevated interleukin-13 (IL-13) was associated with the need for mechanical ventilation in two independent patient cohorts. In addition, patients who acquired COVID-19 while prescribed Dupilumab had less severe disease. In SARS-CoV-2 infected mice, IL-13 neutralization reduced death and disease severity without affecting viral load, demonstrating an immunopathogenic role for this cytokine. Following anti-IL-13 treatment in infected mice, in the lung, hyaluronan synthase 1 (Has1) was the most downregulated gene and hyaluronan accumulation was decreased. Blockade of the hyaluronan receptor, CD44, reduced mortality in infected mice, supporting the importance of hyaluronan as a pathogenic mediator, and indicating a new role for IL-13 in lung disease. Understanding the role of IL-13 and hyaluronan has important implications for therapy of COVID-19 and potentially other pulmonary diseases.

6.
Am J Trop Med Hyg ; 103(3): 1215-1219, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32723427

ABSTRACT

Murine models of SARS-CoV-2 infection are critical for elucidating the biological pathways underlying COVID-19. Because human angiotensin-converting enzyme 2 (ACE2) is the receptor for SARS-CoV-2, mice expressing the human ACE2 gene have shown promise as a potential model for COVID-19. Five mice from the transgenic mouse strain K18-hACE2 were intranasally inoculated with SARS-CoV-2 Hong Kong/VM20001061/2020. Mice were followed twice daily for 5 days and scored for weight loss and clinical symptoms. Infected mice did not exhibit any signs of infection until day 4, when no other obvious clinical symptoms other than weight loss were observed. By day 5, all infected mice had lost around 10% of their original body weight but exhibited variable clinical symptoms. All infected mice showed high viral titers in the lungs as well as altered lung histology associated with proteinaceous debris in the alveolar space, interstitial inflammatory cell infiltration, and alveolar septal thickening. Overall, these results show that the K18-hACE2 transgenic background can be used to establish symptomatic SARS-CoV-2 infection and can be a useful mouse model for COVID-19.


Subject(s)
Betacoronavirus , Coronavirus Infections/etiology , Disease Models, Animal , Keratin-18/genetics , Peptidyl-Dipeptidase A/genetics , Pneumonia, Viral/etiology , Angiotensin-Converting Enzyme 2 , Animals , COVID-19 , Humans , Male , Mice , Mice, Transgenic , Pandemics , SARS-CoV-2
7.
J Clin Invest ; 130(8): 4019-4024, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32369444

ABSTRACT

The microbiome provides resistance to infection. However, the underlying mechanisms are poorly understood. We demonstrate that colonization with the intestinal bacterium Clostridium scindens protects from Entamoeba histolytica colitis via innate immunity. Introduction of C. scindens into the gut microbiota epigenetically altered and expanded bone marrow granulocyte-monocyte progenitors (GMPs) and resulted in increased intestinal neutrophils with subsequent challenge with E. histolytica. Introduction of C. scindens alone was sufficient to expand GMPs in gnotobiotic mice. Adoptive transfer of bone marrow from C. scindens-colonized mice into naive mice protected against amebic colitis and increased intestinal neutrophils. Children without E. histolytica diarrhea also had a higher abundance of Lachnoclostridia. Lachnoclostridia C. scindens can metabolize the bile salt cholate, so we measured deoxycholate and discovered that it was increased in the sera of C. scindens-colonized specific pathogen-free and gnotobiotic mice, as well as in children protected from amebiasis. Administration of deoxycholate alone increased GMPs and provided protection from amebiasis. We elucidated a mechanism by which C. scindens and the microbially metabolized bile salt deoxycholic acid alter hematopoietic precursors and provide innate protection from later infection with E. histolytica.


Subject(s)
Bone Marrow/immunology , Clostridiales/immunology , Dysentery, Amebic/immunology , Entamoeba histolytica/immunology , Gastrointestinal Microbiome/immunology , Animals , Bone Marrow/pathology , Disease Models, Animal , Disease Susceptibility/immunology , Disease Susceptibility/microbiology , Dysentery, Amebic/microbiology , Dysentery, Amebic/pathology , Humans , Intestines/immunology , Intestines/microbiology , Intestines/pathology , Mice
8.
PLoS Negl Trop Dis ; 13(7): e0007515, 2019 07.
Article in English | MEDLINE | ID: mdl-31260452

ABSTRACT

Giardia lamblia is a common intestinal parasitic infection that although often acutely asymptomatic, is associated with debilitating chronic intestinal and extra-intestinal sequelae. In previously healthy adults, a primary sporadic Giardia infection can lead to gut dysfunction and fatigue. These symptoms correlate with markers of inflammation that persist well after the infection is cleared. In contrast, in endemic settings, first exposure occurs in children who are frequently malnourished and also co-infected with other enteropathogens. In these children, Giardia rarely causes symptoms and associates with several decreased markers of inflammation. Mechanisms underlying these disparate and potentially enduring outcomes following Giardia infection are not presently well understood. A body of work suggests that the outcome of experimental Giardia infection is influenced by the nutritional status of the host. Here, we explore the consequences of experimental Giardia infection under conditions of protein sufficiency or deficiency on cytokine responses of ex vivo bone marrow derived dendritic cells (BMDCs) to endotoxin stimulation. We show that BMDCs from Giardia- challenged mice on a protein sufficient diet produce more IL-23 when compared to uninfected controls whereas BMDCs from Giardia challenged mice fed a protein deficient diet do not. Further, in vivo co-infection with Giardia attenuates robust IL-23 responses in endotoxin-stimulated BMDCs from protein deficient mice harboring enteroaggregative Escherichia coli. These results suggest that intestinal Giardia infection may have extra-intestinal effects on BMDC inflammatory cytokine production in a diet dependent manner, and that Giardia may influence the severity of the innate immune response to other enteropathogens. This work supports recent findings that intestinal microbial exposure may have lasting influences on systemic inflammatory responses, and may provide better understanding of potential mechanisms of post-infectious sequelae and clinical variation during Giardia and enteropathogen co-infection.


Subject(s)
Cytokines/immunology , Dendritic Cells/immunology , Diet , Endotoxins/pharmacology , Giardiasis/immunology , Animals , Bone Marrow Cells/immunology , Escherichia coli/immunology , Giardia , Immunity, Innate , Interleukin-23/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/parasitology , Male , Mice , Mice, Inbred C57BL , Protein Deficiency/immunology
9.
PLoS Pathog ; 13(8): e1006513, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28817707

ABSTRACT

The disease severity of Entamoeba histolytica infection ranges from asymptomatic to life-threatening. Recent human and animal data implicate the gut microbiome as a modifier of E. histolytica virulence. Here we have explored the association of the microbiome with susceptibility to amebiasis in infants and in the mouse model of amebic colitis. Dysbiosis occurred symptomatic E. histolytica infection in children, as evidenced by a lower Shannon diversity index of the gut microbiota. To test if dysbiosis was a cause of susceptibility, wild type C57BL/6 mice (which are innately resistant to E. histiolytica infection) were treated with antibiotics prior to cecal challenge with E. histolytica. Compared with untreated mice, antibiotic pre-treated mice had more severe colitis and delayed clearance of E. histolytica. Gut IL-25 and mucus protein Muc2, both shown to provide innate immunity in the mouse model of amebic colitis, were lower in antibiotic pre-treated mice. Moreover, dysbiotic mice had fewer cecal neutrophils and myeloperoxidase activity. Paradoxically, the neutrophil chemoattractant chemokines CXCL1 and CXCL2, as well as IL-1ß, were higher in the colon of mice with antibiotic-induced dysbiosis. Neutrophils from antibiotic pre-treated mice had diminished surface expression of the chemokine receptor CXCR2, potentially explaining their inability to migrate to the site of infection. Blockade of CXCR2 increased susceptibility of control non-antibiotic treated mice to amebiasis. In conclusion, dysbiosis increased the severity of amebic colitis due to decreased neutrophil recruitment to the gut, which was due in part to decreased surface expression on neutrophils of CXCR2.


Subject(s)
Dysentery, Amebic/microbiology , Microbiota/immunology , Neutrophil Infiltration/immunology , Animals , Child, Preschool , Disease Models, Animal , Dysentery, Amebic/immunology , Entamoeba histolytica , Feces/microbiology , Flow Cytometry , Humans , Infant , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction , Receptors, Interleukin-8B/immunology
10.
Infect Immun ; 85(8)2017 08.
Article in English | MEDLINE | ID: mdl-28584161

ABSTRACT

Parasitic protozoan infections represent a major health burden in the developing world and contribute significantly to morbidity and mortality. These infections are often associated with considerable variability in clinical presentation. An emerging body of work suggests that the intestinal microbiota may help to explain some of these differences in disease expression. The objective of this minireview is to synthesize recent progress in this rapidly advancing field. Studies of humans and animals and in vitro studies of the contribution of the intestinal microbiota to infectious disease are discussed. We hope to provide an understanding of the human-protozoal pathogen-microbiome interaction and to speculate on how that might be leveraged for treatment.


Subject(s)
Gastrointestinal Microbiome , Host-Parasite Interactions , Parasites/physiology , Animals , Humans , Parasites/pathogenicity , Protozoan Infections/therapy
11.
Clin Infect Dis ; 65(3): 414-419, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28444240

ABSTRACT

BACKGROUND: As the global polio eradication initiative prepares to cease use of oral polio vaccine (OPV) in 2020, there is increasing interest in understanding if oral vaccination provides non-specific immunity to other infections so that the consequences of this transition can be effectively planned for and mitigated. METHODS: Data were collected from infants in an urban slum in Bangladesh (Mirpur, Dhaka) as part of the performance of rotavirus and oral polio vaccines in developing countries (PROVIDE) study. Following vaccination with trivalent oral polio vaccine (tOPV) at 6, 10, and 14 weeks, infants were randomly assigned to receive tOPV (n = 315) or inactivated polio vaccine (IPV) (n = 299) at 39 weeks. Episodes of diarrhea were documented through clinic visits and twice-weekly house visits through 52 weeks. In sum, 14 pathogens associated with diarrhea were analyzed with TaqMan Array Cards. RESULTS: Although the proportion of children experiencing diarrhea was not different between the tOPV and IPV groups (P = .18), the number of days with diarrhea (P = .0037) and the number of separate diarrheal episodes (P = .054) trended lower in the OPV arm. Etiological analysis revealed that male tOPV recipients were less likely to have diarrhea of bacterial etiology (P = .0099) compared to male IPV recipients but equally likely to experience diarrhea due to viruses (P = .57) or protozoa (P = .14). Among the 6 bacterial enteric pathogens tested, only Campylobacter jejuni/coli detection was significantly reduced in the OPV arm (P = .0048). CONCLUSIONS: Our results suggest that OPV may cause nonspecific reductions in mortality, as has been studied elsewhere, by reducing etiology-specific diarrheal burden. This is likely driven by reductions in bacterial diarrhea. Further study of nonspecific OPV effects before global cessation is supported. CLINICAL TRIALS REGISTRATION: NCT01375647.


Subject(s)
Diarrhea , Poliovirus Vaccine, Oral/administration & dosage , Poliovirus Vaccine, Oral/therapeutic use , Bangladesh/epidemiology , Cross Protection , Diarrhea/epidemiology , Diarrhea/microbiology , Diarrhea/virology , Feces/virology , Female , Humans , Infant , Male
12.
mBio ; 8(1)2017 02 28.
Article in English | MEDLINE | ID: mdl-28246365

ABSTRACT

The parasite Entamoeba histolytica is a cause of diarrhea in infants in low-income countries. Previously, it was shown that tumor necrosis factor alpha (TNF-α) production was associated with increased risk of E. histolytica diarrhea in children. Interleukin-25 (IL-25) is a cytokine that is produced by intestinal epithelial cells that has a role in maintenance of gut barrier function and inhibition of TNF-α production. IL-25 expression was decreased in humans and in the mouse model of amebic colitis. Repletion of IL-25 blocked E. histolytica infection and barrier disruption in mice, increased gut eosinophils, and suppressed colonic TNF-α. Depletion of eosinophils with anti-Siglec-F antibody prevented IL-25-mediated protection. In contrast, depletion of TNF-α resulted in resistance to amebic infection. We concluded that IL-25 provides protection from amebiasis, which is dependent upon intestinal eosinophils and suppression of TNF-α.IMPORTANCE The intestinal epithelial barrier is important for protection from intestinal amebiasis. We discovered that the intestinal epithelial cytokine IL-25 was suppressed during amebic colitis in humans and that protection could be restored in the mouse model by IL-25 administration. IL-25 acted via eosinophils and suppressed TNF-α. This work illustrates a previously unrecognized pathway of innate mucosal immune response.


Subject(s)
Dysentery, Amebic/immunology , Entamoeba histolytica/immunology , Eosinophils/immunology , Interleukin-17/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Disease Models, Animal , Humans , Interleukins/metabolism , Mice
13.
Curr Trop Med Rep ; 3: 71-74, 2016.
Article in English | MEDLINE | ID: mdl-27525214

ABSTRACT

Entamoeba histolytica, the etiological agent of amebiasis, is a significant cause of pediatric diarrhea in South Asia and sub-Saharan Africa. The clinical outcome of an E. histolytica exposure varies enormously and can present as diarrhea, dysentery, or amebic liver abscess. Host and parasite factors likely contribute to the outcome of infection with the parasite, but do not explain the wide variation in presentation of disease. This suggests that other environmental factors affect disease. An emerging body of work suggests that the host intestinal bacterial microbiome may have a significant influence on the development and outcome of amebiasis.

14.
Nat Microbiol ; 1(8): 16108, 2016 07 11.
Article in English | MEDLINE | ID: mdl-27573114

ABSTRACT

Clostridium difficile is the most common hospital acquired pathogen in the USA, and infection is, in many cases, fatal. Toxins A and B are its major virulence factors, but expression of a third toxin, known as C. difficile transferase (CDT), is increasingly common. An adenosine diphosphate (ADP)-ribosyltransferase that causes actin cytoskeletal disruption, CDT is typically produced by the major, hypervirulent strains and has been associated with more severe disease. Here, we show that CDT enhances the virulence of two PCR-ribotype 027 strains in mice. The toxin induces pathogenic host inflammation via a Toll-like receptor 2 (TLR2)-dependent pathway, resulting in the suppression of a protective host eosinophilic response. Finally, we show that restoration of TLR2-deficient eosinophils is sufficient for protection from a strain producing CDT. These findings offer an explanation for the enhanced virulence of CDT-expressing C. difficile and demonstrate a mechanism by which this binary toxin subverts the host immune response.


Subject(s)
ADP Ribose Transferases/metabolism , Bacterial Proteins/metabolism , Clostridioides difficile/immunology , Clostridioides difficile/pathogenicity , Clostridium Infections/pathology , Colon/immunology , Eosinophils/immunology , Virulence Factors/metabolism , Animals , Clostridioides difficile/classification , Clostridioides difficile/genetics , Clostridium Infections/microbiology , Disease Models, Animal , Mice , Ribotyping
15.
Infect Immun ; 84(10): 2824-32, 2016 10.
Article in English | MEDLINE | ID: mdl-27456830

ABSTRACT

Intestinal segmented filamentous bacteria (SFB) protect from ameba infection, and protection is transferable with bone marrow dendritic cells (BMDCs). SFB cause an increase in serum amyloid A (SAA), suggesting that SAA might mediate SFB's effects on BMDCs. Here we further explored the role of bone marrow in SFB-mediated protection. Transient gut colonization with SFB or SAA administration alone transiently increased the H3K27 histone demethylase Jmjd3, persistently increased bone marrow Csf2ra expression and granulocyte monocyte precursors (GMPs), and protected from ameba infection. Pharmacologic inhibition of Jmjd3 H3K27 demethylase activity during SAA treatment or blockade of granulocyte-macrophage colony-stimulating factor (GM-CSF) signaling in SFB-colonized mice prevented GMP expansion, decreased gut neutrophils, and blocked protection from ameba infection. These results indicate that alteration of the microbiota and systemic exposure to SAA can influence myelopoiesis and susceptibility to amebiasis via epigenetic mechanisms. Gut microbiota-marrow communication is a previously unrecognized mechanism of innate protection from infection.


Subject(s)
Bone Marrow Cells/cytology , Entamoeba histolytica/physiology , Entamoebiasis/physiopathology , Gastrointestinal Tract/microbiology , Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Serum Amyloid A Protein/physiology , Animals , Bacteria , Bone Marrow/metabolism , Bone Marrow Cells/physiology , Dendritic Cells/metabolism , Disease Models, Animal , Granulocyte-Macrophage Progenitor Cells , Jumonji Domain-Containing Histone Demethylases/metabolism , Male , Mice , Mice, Inbred C57BL , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism
16.
Infect Immun ; 84(8): 2317-2323, 2016 08.
Article in English | MEDLINE | ID: mdl-27271747

ABSTRACT

Clostridium difficile is a major, life-threatening hospital-acquired pathogen that causes mild to severe colitis in infected individuals. The tissue destruction and inflammation which characterize C. difficile infection (CDI) are primarily due to the Rho-glucosylating toxins A and B. These toxins cause epithelial cell death and induce robust inflammatory signaling by activating the transcription factor NF-κB, leading to chemokine and cytokine secretion. The toxins also activate the inflammasome complex, which leads to secretion of the pyrogenic cytokine IL-1ß. In this study, we utilized glucosylation-deficient toxin A to show that activation of the inflammasome by this toxin is dependent on Rho glucosylation, confirming similar findings reported for toxin B. We also demonstrated that tissue destruction and in vivo inflammatory cytokine production are critically dependent on the enzymatic activity of toxin A, suggesting that inhibiting toxin glucosyltransferase activity may be effective in combating this refractory disease.


Subject(s)
Bacterial Toxins/immunology , Bacterial Toxins/metabolism , Clostridioides difficile/immunology , Clostridioides difficile/metabolism , Clostridium Infections/metabolism , Clostridium Infections/microbiology , Enterotoxins/immunology , Enterotoxins/metabolism , Immunity, Innate , Animals , Bacterial Toxins/genetics , Biomarkers , Clostridium Infections/pathology , Cytokines/metabolism , Enterotoxins/genetics , Glycosylation , Inflammation Mediators/metabolism , Male , Mice , NF-kappa B/metabolism
17.
PLoS One ; 11(4): e0153572, 2016.
Article in English | MEDLINE | ID: mdl-27089535

ABSTRACT

The intestinal surface is directly exposed to both commensal microorganisms as well as pathogens with a single layer of epithelium separating luminal microorganisms from internal tissues. Antimicrobial peptides play a crucial role in allowing epithelial cells to contain in the lumen beneficial and pathogenic microorganisms. The commensal dependent, epithelial produced, Th2 cytokine IL-25 can induce IL-13 and potentially the antimicrobial peptide angiogenin-4. Here we show that IL-13 downstream of IL-25 is required to induce angiogenin-4. IL-25 mediated induction of angiogenin-4 is furthermore not dependent on IL-22 or IL-17.


Subject(s)
Epithelial Cells/metabolism , Gene Expression Regulation/drug effects , Interleukin-13/metabolism , Interleukins/pharmacology , Intestinal Mucosa/metabolism , Ribonuclease, Pancreatic/metabolism , Animals , Blotting, Western , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/drug effects , Immunoenzyme Techniques , Interleukin-13/genetics , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Male , Mice , Mice, Inbred CBA , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Ribonuclease, Pancreatic/genetics
18.
Respir Med ; 110: 53-7, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26631486

ABSTRACT

BACKGROUND: Environmental factors that influence wheezing in early childhood in the developing world are not well understood and may be useful in predicting respiratory outcomes. Therefore, our objective was to determine the factors that can predict wheezing. METHODS: Children from Dhaka, Bangladesh were recruited at birth and episodes of wheezing were measured alongside nutritional, immunological and socioeconomic factors over a one-year period. Poisson Regression with variable selection was utilized to determine what factors were associated with wheezing. RESULTS: Elevated serum IL-10 (rate ratio (RR) = 1.51, 95% confidence interval (CI): 1.22-1.87), IL-1ß (RR = 1.55, 95% CI: 1.26-1.93) C-reactive protein (CRP) (RR = 1.41, 95% CI: 1.03-1.93) in early life, and male gender (RR = 1.52, 95% CI: 1.27-1.82) predicted increased wheezing episodes. Conversely, increased fecal alpha-1-antitrypsin (RR = 0.87, 95% CI: 0.76-1.00) and family income (RR = 0.98, 95% CI: 0.97-0.99) were associated with a decreased number of episodes of wheezing. CONCLUSIONS: Systemic inflammation early in life, poverty, and male sex placed infants at risk of more episodes of wheezing during their first year of life. These results support the hypothesis that there is a link between inflammation in infancy and the development of respiratory illness later in life and provide specific biomarkers that can predict wheezing in a low-income country.


Subject(s)
Biomarkers/analysis , Respiratory Sounds , Bangladesh/epidemiology , C-Reactive Protein/analysis , Feces/chemistry , Female , Humans , Income , Infant , Infant, Newborn , Interleukin-10/blood , Interleukin-1beta/blood , Male , Poisson Distribution , Risk Factors , alpha 1-Antitrypsin/analysis
19.
J Infect Dis ; 213(10): 1579-85, 2016 May 15.
Article in English | MEDLINE | ID: mdl-26712950

ABSTRACT

BACKGROUND: An estimated 1 million children die each year before their fifth birthday from diarrhea. Previous population-based surveys of pediatric diarrheal diseases have identified the protozoan parasite Entamoeba histolytica, the etiological agent of amebiasis, as one of the causes of moderate-to-severe diarrhea in sub-Saharan Africa and South Asia. METHODS: We prospectively studied the natural history of E. histolytica colonization and diarrhea among infants in an urban slum of Dhaka, Bangladesh. RESULTS: Approximately 80% of children were infected with E. histolytica by the age of 2 years. Fecal anti-galactose/N-acetylgalactosamine lectin immunoglobulin A was associated with protection from reinfection, while a high parasite burden and expansion of the Prevotella copri level was associated with diarrhea. CONCLUSIONS: E. histolytica infection was prevalent in this population, with most infections asymptomatic and diarrhea associated with both the amount of parasite and the composition of the microbiota.


Subject(s)
Antibodies, Protozoan/immunology , Diarrhea, Infantile/etiology , Entamoeba histolytica/immunology , Entamoebiasis/complications , Gastrointestinal Microbiome , Immunoglobulin A/immunology , Animals , Bangladesh/epidemiology , Cohort Studies , Entamoebiasis/parasitology , Feces/parasitology , Female , Follow-Up Studies , Humans , Infant , Lectins/immunology , Longitudinal Studies , Male , Poverty Areas , Prevalence , Prospective Studies
20.
mBio ; 5(6): e01817, 2014 Nov 04.
Article in English | MEDLINE | ID: mdl-25370489

ABSTRACT

UNLABELLED: There is an emerging paradigm that the human microbiome is central to many aspects of health and may have a role in preventing enteric infection. Entamoeba histolytica is a major cause of amebic diarrhea in developing countries. It colonizes the colon lumen in close proximity to the gut microbiota. Interestingly, not all individuals are equally susceptible to E. histolytica infection. Therefore, as the microbiota is highly variable within individuals, we sought to determine if a component of the microbiota could regulate susceptibility to infection. In studies utilizing a murine model, we demonstrated that colonization of the gut with the commensal Clostridia-related bacteria known as segmented filamentous bacteria (SFB) is protective during E. histolytica infection. SFB colonization in this model was associated with elevated cecal levels of interleukin 17A (IL-17A), dendritic cells, and neutrophils. Bone marrow-derived dendritic cells (BMDCs) from SFB-colonized mice had higher levels of IL-23 production in response to stimulation with trophozoites. Adoptive transfer of BMDCs from an SFB(+) to an SFB(-) mouse was sufficient to provide protection against E. histolytica. IL-17A induction during BMDC transfer was necessary for this protection. This work demonstrates that intestinal colonization with a specific commensal bacterium can provide protection during amebiasis in a murine model. Most importantly, this work demonstrates that the microbiome can mediate protection against an enteric infection via extraintestinal effects on bone marrow-derived dendritic cells. IMPORTANCE: Entamoeba histolytica is the causative agent of amebiasis, an infectious disease that contributes significantly to morbidity and mortality due to diarrhea in the developing world. We showed in a murine model that colonization with the commensal members of the Clostridia known as SFB provides protection against E. histolytica and that dendritic cells from SFB-colonized mice alone can recapitulate protection. Understanding interactions between enteropathogens, commensal intestinal bacteria, and the mucosal immune response, including dendritic cells, will help in the development of effective treatments for this disease and other infectious and inflammatory diseases. The demonstration of immune-mediated protection due to communication from the microbiome to the bone marrow represents an emerging field of study that will yield unique approaches to the development of these treatments.


Subject(s)
Clostridium/growth & development , Dendritic Cells/immunology , Dysentery, Amebic/prevention & control , Entamoeba histolytica/immunology , Gastrointestinal Tract/microbiology , Interleukin-17/metabolism , Animals , Clostridium/immunology , Clostridium/physiology , Disease Models, Animal , Mice , Neutrophils/immunology , Symbiosis
SELECTION OF CITATIONS
SEARCH DETAIL
...