Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Nat Immunol ; 17(10): 1176-86, 2016 10.
Article in English | MEDLINE | ID: mdl-27548431

ABSTRACT

Inflammasomes are positioned to rapidly escalate the intensity of inflammation by activating interleukin (IL)-1ß, IL-18 and cell death by pyroptosis. However, negative regulation of inflammasomes remains poorly understood, as is the signaling cascade that dampens inflammasome activity. We found that rapid NLRP3 inflammasome activation was directly inhibited by protein kinase A (PKA), which was induced by prostaglandin E2 (PGE2) signaling via the PGE2 receptor E-prostanoid 4 (EP4). PKA directly phosphorylated the cytoplasmic receptor NLRP3 and attenuated its ATPase function. We found that Ser295 in human NLRP3 was critical for rapid inhibition and PKA phosphorylation. Mutations in NLRP3-encoding residues adjacent to Ser295 have been linked to the inflammatory disease CAPS (cryopyrin-associated periodic syndromes). NLRP3-S295A phenocopied the human CAPS mutants. These data suggest that negative regulation at Ser295 is critical for restraining the NLRP3 inflammasome and identify a molecular basis for CAPS-associated NLRP3 mutations.


Subject(s)
Cryopyrin-Associated Periodic Syndromes/immunology , Cyclic AMP-Dependent Protein Kinases/metabolism , Inflammasomes/metabolism , Monocytes/physiology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Adenosine Triphosphatases/metabolism , Animals , Cell Line , Cryopyrin-Associated Periodic Syndromes/genetics , Dinoprostone/metabolism , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mutation/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Phenotype , Phosphorylation/genetics , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Serine/genetics , Signal Transduction/genetics
2.
Gastroenterology ; 164(2): 228-240, 2023 02.
Article in English | MEDLINE | ID: mdl-36183751

ABSTRACT

BACKGROUND & AIMS: Inflammatory bowel diseases (IBD) are affected by dietary factors, including nondigestible carbohydrates (fibers), which are fermented by colonic microbes. Fibers are overall beneficial, but not all fibers are alike, and some patients with IBD report intolerance to fiber consumption. Given reproducible evidence of reduced fiber-fermenting microbes in patients with IBD, we hypothesized that fibers remain intact in select patients with reduced fiber-fermenting microbes and can then bind host cell receptors, subsequently promoting gut inflammation. METHODS: Colonic biopsies cultured ex vivo and cell lines in vitro were incubated with oligofructose (5 g/L), or fermentation supernatants (24-hour anaerobic fermentation) and immune responses (cytokine secretion [enzyme-linked immunosorbent assay/meso scale discovery] and expression [quantitative polymerase chain reaction]) were assessed. Influence of microbiota in mediating host response was examined and taxonomic classification of microbiota was conducted with Kraken2 and metabolic profiling by HUMAnN2, using R software. RESULTS: Unfermented dietary ß-fructan fibers induced proinflammatory cytokines in a subset of IBD intestinal biopsies cultured ex vivo, and immune cells (including peripheral blood mononuclear cells). Results were validated in an adult IBD randomized controlled trial examining ß-fructan supplementation. The proinflammatory response to intact ß-fructan required activation of the NLRP3 and TLR2 pathways. Fermentation of ß-fructans by human gut whole microbiota cultures reduced the proinflammatory response, but only when microbes were collected from patients without IBD or patients with inactive IBD. Fiber-induced immune responses correlated with microbe functions, luminal metabolites, and dietary fiber avoidance. CONCLUSION: Although fibers are typically beneficial in individuals with normal microbial fermentative potential, some dietary fibers have detrimental effects in select patients with active IBD who lack fermentative microbe activities. The study is publicly accessible at the U.S. National Institutes of Health database (clinicaltrials.gov identification number NCT02865707).


Subject(s)
Fructans , Inflammatory Bowel Diseases , Adult , Humans , Leukocytes, Mononuclear , Intestines , Dietary Fiber , Inflammation
3.
PLoS Pathog ; 18(3): e1010415, 2022 03.
Article in English | MEDLINE | ID: mdl-35303042

ABSTRACT

A hallmark of Entamoeba histolytica (Eh) invasion in the gut is acute inflammation dominated by the secretion of pro-inflammatory cytokines TNF-α and IL-1ß. This is initiated when Eh in contact with macrophages in the lamina propria activates caspase-1 by recruiting the NLRP3 inflammasome complex in a Gal-lectin and EhCP-A5-dependent manner resulting in the maturation and secretion of IL-1ß and IL-18. Here, we interrogated the requirements and mechanisms for Eh-induced caspase-4/1 activation in the cleavage of gasdermin D (GSDMD) to regulate bioactive IL-1ß release in the absence of cell death in human macrophages. Unlike caspase-1, caspase-4 activation occurred as early as 10 min that was dependent on Eh Gal-lectin and EhCP-A5 binding to macrophages. By utilizing CRISPR-Cas9 gene edited CASP4/1, NLRP3 KO and ASC-def cells, caspase-4 activation was found to be independent of the canonical NLRP3 inflammasomes. In CRISPR-Cas9 gene edited CASP1 macrophages, caspase-4 activation was significantly up regulated that enhanced the enzymatic cleavage of GSDMD at the same cleavage site as caspase-1 to induce GSDMD pore formation and sustained bioactive IL-1ß secretion. Eh-induced IL-1ß secretion was independent of pyroptosis as revealed by pharmacological blockade of GSDMD pore formation and in CRISPR-Cas9 gene edited GSDMD KO macrophages. This was in marked contrast to the potent positive control, lipopolysaccharide + Nigericin that induced high expression of predominantly caspase-1 that efficiently cleaved GSDMD with high IL-1ß secretion/release associated with massive cell pyroptosis. These results reveal that Eh triggered "hyperactivated macrophages" allowed caspase-4 dependent cleavage of GSDMD and IL-1ß secretion to occur in the absence of pyroptosis that may play an important role in disease pathogenesis.


Subject(s)
Entamoeba histolytica , Caspase 1/genetics , Caspase 1/metabolism , Caspases, Initiator/metabolism , Entamoeba histolytica/metabolism , Humans , Interleukin-1beta , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Phosphate-Binding Proteins/genetics , Phosphate-Binding Proteins/metabolism , Pore Forming Cytotoxic Proteins/metabolism , Pyroptosis
4.
PLoS Pathog ; 17(9): e1009936, 2021 09.
Article in English | MEDLINE | ID: mdl-34499701

ABSTRACT

While Entamoeba histolytica (Eh)-induced pro-inflammatory responses are critical in disease pathogenesis, the downstream signaling pathways that subsequently dampens inflammation and the immune response remains unclear. Eh in contact with macrophages suppresses NF-κB signaling while favoring NLRP3-dependent pro-inflammatory cytokine production by an unknown mechanism. Cullin-1 and cullin-5 (cullin-1/5) assembled into a multi-subunit RING E3 ubiquitin ligase complex are substrates for neddylation that regulates the ubiquitination pathway important in NF-κB activity and pro-inflammatory cytokine production. In this study, we showed that upon live Eh contact with human macrophages, cullin-1/4A/4B/5 but not cullin-2/3, were degraded within 10 minutes. Similar degradation of cullin-1/5 were observed from colonic epithelial cells and proximal colonic loops tissues of mice inoculated with live Eh. Degradation of cullin-1/5 was dependent on Eh-induced activation of caspase-1 via the NLRP3 inflammasome. Unlike cullin-4B, the degradation of cullin-4A was partially dependent on caspase-1 and was inhibited with a pan caspase inhibitor. Cullin-1/5 degradation was dependent on Eh cysteine proteinases EhCP-A1 and EhCP-A4, but not EhCP-A5, based on pharmacological inhibition of the cysteine proteinases and EhCP-A5 deficient parasites. siRNA silencing of cullin-1/5 decreased the phosphorylation of pIκ-Bα in response to Eh and LPS stimulation and downregulated NF-κB-dependent TNF-α mRNA expression and TNF-α and MCP-1 pro-inflammatory cytokine production. These results unravel a unique outside-in strategy employed by Eh to attenuate NF-κB-dependent pro-inflammatory responses via NLRP3 activation of caspase-1 that degraded cullin-1/5 from macrophages.


Subject(s)
Caspase 1/metabolism , Cullin Proteins/metabolism , Entamoebiasis/metabolism , Macrophages/metabolism , NF-kappa B/metabolism , Animals , Entamoeba histolytica/immunology , Entamoeba histolytica/metabolism , Entamoebiasis/immunology , Humans , Mice , Signal Transduction/physiology
5.
Am J Physiol Gastrointest Liver Physiol ; 321(5): G489-G499, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34494458

ABSTRACT

Goblet cells are specialized for the production and secretion of MUC2 glycoproteins that forms a thick layer covering the mucosal epithelium as a protective barrier against noxious substances and invading microbes. High MUC2 mucin biosynthesis induces endoplasmic reticulum (ER) stress and apoptosis in goblet cells during inflammatory and infectious diseases. Autophagy is an intracellular degradation process required for maintenance of intestinal homeostasis. In this study, we hypothesized that autophagy was triggered during high MUC2 mucin biosynthesis from colonic goblet cells to cope with metabolic stress. To interrogate this, we analyzed the autophagy process in high MUC2-producing human HT29-H and a clone HT29-L silenced for MUC2 expression by lentivirus-mediated shRNA, and WT and CRISPR/Cas9 MUC2 KO LS174T cells. Autophagy was constitutively increased in high MUC2-producing cells characterized by elevated pULK1S555 expression and increased numbers of autophagosomes as compared with MUC2 silenced or gene edited cells. Similarly, colonoids from Muc2+/+ but not Muc2-/- littermates differentiated into goblet cells showed increased autophagy. IL-22 treatment corrected misfolded MUC2 protein and alleviated the autophagy process in LS174T cells. This study highlights that autophagy plays an essential role in goblet cells to survive during high mucin biosynthesis by regulating cellular homeostasis.NEW & NOTEWORTHY It is unclear how colonic goblet cells survive by producing high output MUC2 mucin that triggers endoplasmic stress by misfolded MUC2 proteins. To cope with metabolic stress, we interrogated if autophagy played an essential role in regulating cellular homeostasis. Indeed, high MUC2 mucin biosynthesis dysregulated autophagy processes that was regulated by IL-22 to maintain gut barrier innate host defenses.


Subject(s)
Autophagy , Colon/metabolism , Endoplasmic Reticulum Stress , Energy Metabolism , Goblet Cells/metabolism , Mucin-2/biosynthesis , Animals , Autophagy/drug effects , Autophagy-Related Protein-1 Homolog/metabolism , Colon/drug effects , Colon/ultrastructure , Endoplasmic Reticulum Stress/drug effects , Energy Metabolism/drug effects , Female , Goblet Cells/drug effects , Goblet Cells/ultrastructure , HT29 Cells , Humans , Interleukins/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Mucin-2/genetics , Phosphorylation , Protein Folding , Signal Transduction , Interleukin-22
6.
PLoS Pathog ; 14(11): e1007466, 2018 11.
Article in English | MEDLINE | ID: mdl-30500860

ABSTRACT

Amebiasis is caused by the protozoan parasite Entamoeba histolytica (Eh), a potentially fatal disease occurring mainly in developing countries. How Eh interacts with innate host factors in the gut is poorly understood. Eh resides and feed in/on the outer colonic mucus layer and thus share an ecological niche with indigenous microbiota. As gut microbiota regulates innate immune responses, in this study we characterized the cooperative roles that microbiota and the mucus layer play in Eh-induced pro-inflammatory responses in the colon. To study this, we used antibiotics treated and non-treated specific pathogen free Muc2-/- and Muc2+/+ littermates and germ-free mice inoculated with Eh in colonic loops as a short infection model. In antibiotic treated Muc2-/- and Muc2+/+ littermates, Eh elicited robust mucus and water secretions, enhanced pro-inflammatory cytokines and chemokine expression with elevated MPO activity and higher pathology scores as compared to the modest response observed in non-antibiotic treated littermates. Host responses were microbiota specific as mucus secretion and pro-inflammatory responses were attenuated following homologous fecal microbial transplants in antibiotic-treated Muc2+/+ quantified by secretion of 3H-glucosamine newly synthesized mucin, Muc2 mucin immunostaining and immunohistochemistry. Eh-elicited pro-inflammatory responses and suppressed goblet cell transcription factor Math1 as revealed by in vivo imaging of Eh-colonic loops in Math1GFP mice, and in vitro using Eh-stimulated LS174T human colonic goblet cells. Eh in colonic loops increased bacterial translocation of bioluminescent E. coli and indigenous bacteria quantified by FISH and quantitative PCR. In germ-free animals, Eh-induced mucus/water secretory responses, but acute pro-inflammatory responses and MPO activity were severely impaired, allowing the parasite to bind to and disrupt mucosal epithelial cells. These findings have identified key roles for intestinal microbiota and mucus in regulating innate host defenses against Eh, and implicate dysbiosis as a risk factor for amebiasis that leads to exacerbated immune responses to cause life-threatening disease.


Subject(s)
Entamoeba histolytica/metabolism , Gastrointestinal Microbiome/immunology , Mucin-2/immunology , Animals , Cell Line , Colon/metabolism , Colon/pathology , Entamoeba histolytica/immunology , Entamoeba histolytica/pathogenicity , Epithelial Cells/metabolism , Gene Expression Regulation , Goblet Cells/metabolism , Humans , Immunity, Innate/immunology , Inflammation/pathology , Intestinal Mucosa/immunology , Mice , Mice, Inbred C57BL , Microbiota , Mucin-1 , Mucins/metabolism
7.
Infect Immun ; 87(12)2019 12.
Article in English | MEDLINE | ID: mdl-31527129

ABSTRACT

Epidemiological studies suggest frequent association of enteropathogenic bacteria with Entamoeba histolytica during symptomatic infection. In this study, we sought to determine if the interaction with enteropathogenic (EPEC) or nonpathogenic Escherichia coli (strain DH5α) could modify the virulence of E. histolytica to cause disease in animal models of amebiasis. In vitro studies showed a 2-fold increase in CaCo2 monolayer destruction when E. histolytica interacted with EPEC but not with E. coli DH5α for 2.5 h. This was associated with increased E. histolytica proteolytic activity as revealed by zymogram analysis and degradation of the E. histolytica CP-A1/5 (EhCP-A1/5) peptide substrate Z-Arg-Arg-pNC and EhCP4 substrate Z-Val-Val-Arg-AMC. Additionally, E. histolytica-EPEC interaction increased EhCP-A1, -A2, -A4, and -A5, Hgl, Apa, and Cox-1 mRNA expression. Despite the marked upregulation of E. histolytica virulence factors, nonsignificant macroscopic differences in amebic liver abscess development were observed at early stages in hamsters inoculated with either E. histolytica-EPEC or E. histolytica-E. coli DH5α. Histopathology of livers of E. histolytica-EPEC-inoculated animals revealed foci of acute inflammation 3 h postinoculation that progressively increased, producing large inflammatory reactions, ischemia, and necrosis with high expression of il-1ß, ifn-γ, and tnf-α proinflammatory cytokine genes compared with that in livers of E. histolytica-E. coli DH5α-inoculated animals. In closed colonic loops from mice, intense inflammation was observed with E. histolytica-EPEC manifested by downregulation of Math1 mRNA with a corresponding increase in the expression of Muc2 mucin and proinflammatory cytokine genes il-6, il-12, and mcp-1 These results demonstrate that E. histolytica/EPEC interaction enhanced the expression and production of key molecules associated with E. histolytica virulence, critical in pathogenesis and progression of disease.


Subject(s)
Entamoeba histolytica/pathogenicity , Entamoebiasis/pathology , Enteropathogenic Escherichia coli/physiology , Host Microbial Interactions/physiology , Animals , Caco-2 Cells , Cell Line , Cricetinae , Cysteine Proteases/metabolism , Cytokines/metabolism , Entamoeba histolytica/microbiology , HT29 Cells , Humans , Inflammation , Mesocricetus , Mice , Mice, Inbred C57BL , Mucin-2/metabolism , Virulence Factors/biosynthesis
8.
Infect Immun ; 87(11)2019 11.
Article in English | MEDLINE | ID: mdl-31427448

ABSTRACT

Entamoeba histolytica is an anaerobic parasitic protozoan and the causative agent of amoebiasis. E. histolytica expresses proteins that are structurally homologous to human proteins and uses them as virulence factors. We have previously shown that E. histolytica binds exogenous interferon gamma (IFN-γ) on its surface, and in this study, we explored whether exogenous IFN-γ could modulate parasite virulence. We identified an IFN-γ receptor-like protein on the surface of E. histolytica trophozoites by using anti-IFN-γ receptor 1 (IFN-γR1) antibody and performing immunofluorescence, Western blot, protein sequencing, and in silico analyses. Coupling of human IFN-γ to the IFN-γ receptor-like protein on live E. histolytica trophozoites significantly upregulated the expression of E. histolytica cysteine protease A1 (EhCP-A1), EhCP-A2, EhCP-A4, EhCP-A5, amebapore A (APA), cyclooxygenase 1 (Cox-1), Gal-lectin (Hgl), and peroxiredoxin (Prx) in a time-dependent fashion. IFN-γ signaling via the IFN-γ receptor-like protein enhanced E. histolytica's erythrophagocytosis of human red blood cells, which was abrogated by the STAT1 inhibitor fludarabine. Exogenous IFN-γ enhanced chemotaxis of E. histolytica, its killing of Caco-2 colonic and Hep G2 liver cells, and amebic liver abscess formation in hamsters. These results demonstrate that E. histolytica expresses a surface IFN-γ receptor-like protein that is functional and may play a role in disease pathogenesis and/or immune evasion.


Subject(s)
Entamoeba histolytica/metabolism , Protozoan Proteins/metabolism , Receptors, Interferon/chemistry , Amebiasis/immunology , Amebiasis/parasitology , Animals , Caco-2 Cells , Cell Survival , Cricetinae , Hep G2 Cells , Humans , Interferon-gamma/pharmacology , Male , Phagocytosis , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Interferon gamma Receptor
9.
PLoS Pathog ; 13(8): e1006592, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28837696

ABSTRACT

Entamoeba histolytica (Eh) is the causative agent of amebiasis, one of the major causes of dysentery-related morbidity worldwide. Recent studies have underlined the importance of the intercellular junction between Eh and host cells as a determinant in the pathogenesis of amebiasis. Despite the fact that direct contact and ligation between Eh surface Gal-lectin and EhCP-A5 with macrophage α5ß1 integrin are absolute requirements for NLRP3 inflammasome activation and IL-1ß release, many other undefined molecular events and downstream signaling occur at the interface of Eh and macrophage. In this study, we investigated the molecular events at the intercellular junction that lead to recognition of Eh through modulation of the macrophage cytoskeleton. Upon Eh contact with macrophages key cytoskeletal-associated proteins were rapidly post-translationally modified only with live Eh but not with soluble Eh proteins or fragments. Eh ligation with macrophages rapidly activated caspase-6 dependent cleavage of the cytoskeletal proteins talin, Pyk2 and paxillin and caused robust release of the pro-inflammatory cytokine, IL-1ß. Macrophage cytoskeletal cleavages were dependent on Eh cysteine proteinases EhCP-A1 and EhCP-A4 but not EhCP-A5 based on pharmacological blockade of Eh enzyme inhibitors and EhCP-A5 deficient parasites. These results unravel a model where the intercellular junction between macrophages and Eh form an area of highly interacting proteins that implicate the macrophage cytoskeleton as a sensor for Eh contact that leads downstream to subsequent inflammatory immune responses.


Subject(s)
Cytoskeleton/immunology , Entamoebiasis/immunology , Host-Parasite Interactions/immunology , Interleukin-1beta/metabolism , Macrophages/immunology , Macrophages/parasitology , Animals , Blotting, Western , Cell Line , Entamoeba histolytica/immunology , Female , Flow Cytometry , Humans , Male , Mice, Inbred C57BL , Microscopy, Confocal
10.
Am J Pathol ; 188(9): 2025-2041, 2018 09.
Article in English | MEDLINE | ID: mdl-29935164

ABSTRACT

Intestinal epithelial cell wound healing involves cell migration, proliferation, and differentiation. Although numerous studies have analyzed the migration of absorptive epithelial cells during wound healing, it remains unclear how goblet cells restitute and how MUC2 mucin production affects this process. In this study, we examined the role of high MUC2 production in goblet cell migration during wound healing and demonstrated that during high MUC2 output, goblet cells migrated slower because of impaired production of wound healing factors and endoplasmic reticulum (ER) stress. Two goblet cell lines, HT29-H and HT29-L, that produced high and low MUC2 mucin, respectively, were used. HT29-L healed wounds faster than HT29-H cells by producing significantly higher amounts of fibroblast growth factor (FGF) 1, FGF2, vascular endothelial growth factor-C, and matrix metallopeptidase 1. Predictably, treatment of HT29-H cells with recombinant FGF2 significantly enhanced migration and wound healing. High MUC2 biosynthesis in HT29-H cells induced ER stress and delayed migration that was abrogated by inhibiting ER stress with tauroursodeoxycholic acid and IL-22. FGF2- and IL-22-induced wound repair was dependent on STAT1 and STAT3 signaling. During wound healing after dextran sulfate sodium-induced colitis, restitution of Math1M1GFP+ goblet cells occurred earlier in the proximal colon, followed by the middle and then distal colon, where ulceration was severe. We conclude that high MUC2 output during colitis impairs goblet cell migration and wound healing by reducing production of growth factors critical in wound repair.


Subject(s)
Colitis/pathology , Endoplasmic Reticulum Stress , Goblet Cells/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Mucin-2/metabolism , Wound Healing , Animals , Cell Movement , Cell Proliferation , Colitis/chemically induced , Colitis/metabolism , Dextran Sulfate/toxicity , HT29 Cells , Humans , Mice
11.
Am J Pathol ; 188(6): 1354-1373, 2018 06.
Article in English | MEDLINE | ID: mdl-29545196

ABSTRACT

MUC2 mucin is a large glycoprotein produced by goblet cells that forms the protective mucus blanket overlying the intestinal epithelium as the first line of innate host defense. High MUC2 production in inflammatory bowel disease and infectious colitis depletes goblet cells and the mucus layer by an unknown mechanism. Herein, we analyzed the effect of high MUC2 biosynthesis on endoplasmic reticulum (ER) stress and apoptosis in goblet cells using a high MUC2-producing human goblet cell line (HT29-H) and an HT29-H clone (HT29-L) silenced for MUC2 expression by lentivirus-mediated shRNA. Goblet cell ER stress and apoptosis were quantified during early onset of dextran sulfate sodium-induced colitis in C57BL/6 and Math1M1GFP mice. Compared with HT29-L and MUC2 nonproducing Caco-2 cells, high MUC2-producing HT29-H cells had significantly increased ER stress and apoptosis after treatment with ER stress-inducing agents. Apoptosis was driven by increased misfolded MUC2 that triggered elevated levels of reactive oxygen species. Correcting MUC2 folding and inhibiting reactive oxygen species alleviated ER stress and rescued cells from apoptosis. During early-onset colitis, mucus hypersecretion caused severe ER stress and apoptosis of goblet cells that preceded absorptive epithelial cell damage. Thus, in gastrointestinal inflammation, high MUC2 biosynthesis and goblet cell apoptosis lead to a dysfunctional epithelial barrier. Enhancing MUC2 folding may help alleviate goblet cell depletion and maintain mucosal integrity.


Subject(s)
Apoptosis , Endoplasmic Reticulum Stress , Goblet Cells/pathology , Mucin-2/chemistry , Mucin-2/metabolism , Protein Folding , Reactive Oxygen Species/metabolism , Animals , Colitis/chemically induced , Colitis/metabolism , Colitis/pathology , Dextran Sulfate/toxicity , Goblet Cells/metabolism , HT29 Cells , Humans , Mice , Mice, Inbred C57BL , Mucin-2/genetics
12.
Infect Immun ; 86(7)2018 07.
Article in English | MEDLINE | ID: mdl-29685982

ABSTRACT

Enteric α-defensins, termed cryptdins (Crps) in mice, and lysozymes secreted by Paneth cells contribute to innate host defense in the ileum. Antimicrobial factors, including lysozymes and ß-defensins, are often embedded in luminal glycosylated colonic Muc2 mucin secreted by goblet cells that form the protective mucus layer critical for gut homeostasis and pathogen invasion. In this study, we investigated ileal innate immunity against Entamoeba histolytica, the causative agent of intestinal amebiasis, by inoculating parasites in closed ileal loops in Muc2+/+ and Muc2-/- littermates and quantifying Paneth cell localization (lysozyme expression) and function (Crp secretion). Relative to Muc2+/+ littermates, Muc2-/- littermates showed a disorganized mislocalization of Paneth cells that was diffusely distributed, with elevated lysozyme secretion in the crypts and on villi in response to E. histolytica Inhibition of E. histolytica Gal/GalNAc lectin (Gal-lectin) binding with exogenous galactose and Entamoeba histolytica cysteine proteinase 5 (EhCP5)-negative E. histolytica had no effect on parasite-induced erratic Paneth cell lysozyme synthesis. Although the basal ileal expression of Crp genes was unaffected in Muc2-/- mice in response to E. histolytica, there was a robust release of proinflammatory cytokines and Crp peptide secretions in luminal exudates that was also present in the colon. Interestingly, E. histolytica-secreted cysteine proteinases cleaved the proregion of Crp4 but not the active form. These findings define Muc2 mucin as an essential component of ileal barrier function that regulates the localization and function of Paneth cells critical for host defense against microbes.


Subject(s)
Defensins/metabolism , Entamoeba histolytica/metabolism , Entamoeba histolytica/pathogenicity , Mucins/deficiency , Mucins/metabolism , Muramidase/metabolism , Paneth Cells/metabolism , Animals , Cell Proliferation/physiology , Host-Parasite Interactions , Humans , Mice
13.
PLoS Pathog ; 12(4): e1005579, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27073869

ABSTRACT

Critical to the pathogenesis of intestinal amebiasis, Entamoeba histolytica (Eh) induces mucus hypersecretion and degrades the colonic mucus layer at the site of invasion. The parasite component(s) responsible for hypersecretion are poorly defined, as are regulators of mucin secretion within the host. In this study, we have identified the key virulence factor in live Eh that elicits the fast release of mucin by goblets cells as cysteine protease 5 (EhCP5) whereas, modest mucus secretion occurred with secreted soluble EhCP5 and recombinant CP5. Coupling of EhCP5-αvß3 integrin on goblet cells facilitated outside-in signaling by activating SRC family kinases (SFK) and focal adhesion kinase that resulted in the activation/phosphorlyation of PI3K at the site of Eh contact and production of PIP3. PKCδ was activated at the EhCP5-αvß3 integrin contact site that specifically regulated mucin secretion though the trafficking vesicle marker myristoylated alanine-rich C-kinase substrate (MARCKS). This study has identified that EhCP5 coupling with goblet cell αvß3 receptors can initiate a signal cascade involving PI3K, PKCδ and MARCKS to drive mucin secretion from goblet cells critical in disease pathogenesis.


Subject(s)
Cysteine Proteases/metabolism , Dysentery, Amebic/metabolism , Entamoeba histolytica/pathogenicity , Goblet Cells/metabolism , Integrin alphaVbeta3/metabolism , Mucins/metabolism , Animals , Blotting, Western , Cell Line , Colon/metabolism , Colon/microbiology , Colon/pathology , Disease Models, Animal , Dysentery, Amebic/pathology , Enzyme-Linked Immunosorbent Assay , Exocytosis , Flow Cytometry , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Mice , Microscopy, Confocal , Protozoan Proteins/metabolism , Signal Transduction/physiology , Virulence/physiology , Virulence Factors/metabolism
14.
Am J Pathol ; 187(11): 2486-2498, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28823873

ABSTRACT

The intestinal mucous layer provides a critical host defense against pathogen exposure and epithelial injury, yet little is known about how enteropathogens may circumvent this physiologic barrier. Giardia duodenalis is a small intestinal parasite responsible for diarrheal disease and chronic postinfectious illness. This study reveals a complex interaction at the surface of epithelial cells, between G. duodenalis and the intestinal mucous layer. Here, we reveal mechanisms whereby G. duodenalis evades and disrupts the first line of host defense by degrading human mucin-2 (MUC2), depleting mucin stores and inducing differential gene expression in the mouse small and large intestines. Human colonic biopsy specimens exposed to G. duodenalis were depleted of mucus, and in vivo mice infected with G. duodenalis had a thinner mucous layer and demonstrated differential Muc2 and Muc5ac mucin gene expression. Infection in Muc2-/- mice elevated trophozoite colonization in the small intestine and impaired weight gain. In vitro, human LS174T goblet-like cells were depleted of mucus and had elevated levels of MUC2 mRNA expression after G. duodenalis exposure. Importantly, the cysteine protease inhibitor E64 prevented mucous degradation, mucin depletion, and the increase in MUC2 expression. This article describes a novel role for Giardia's cysteine proteases in pathogenesis and how Giardia's disruptions of the mucous barrier facilitate bacterial translocation that may contribute to the onset and propagation of disease.


Subject(s)
Epithelial Cells/metabolism , Giardiasis/genetics , Mucins/genetics , Mucus/metabolism , Animals , Bacterial Translocation/genetics , Cysteine Proteases/metabolism , Female , Giardia lamblia/genetics , Giardiasis/metabolism , Humans , Intestinal Mucosa/metabolism , Male , Mice , Mucins/metabolism
15.
Infect Immun ; 85(3)2017 03.
Article in English | MEDLINE | ID: mdl-28069814

ABSTRACT

Embedded in the colonic mucus are cathelicidins, small cationic peptides secreted by colonic epithelial cells. Humans and mice have one cathelicidin-related antimicrobial peptide (CRAMP) each, LL-37/hCAP-18 and Cramp, respectively, with related structure and functions. Altered production of MUC2 mucin and antimicrobial peptides is characteristic of intestinal amebiasis. The interactions between MUC2 mucin and cathelicidins in conferring innate immunity against Entamoeba histolytica are not well characterized. In this study, we quantified whether MUC2 expression and release could regulate the expression and secretion of cathelicidin LL-37 in colonic epithelial cells and in the colon. The synthesis of LL-37 was enhanced with butyrate (a product of bacterial fermentation) and interleukin-1ß (IL-1ß) (a proinflammatory cytokine in colitis) in the presence of exogenously added purified MUC2. The LL-37 responses to butyrate and IL-1ß were higher in high-MUC2-producing cells than in lentivirus short hairpin RNA (shRNA) MUC2-silenced cells. Activation of cyclic adenylyl cyclase (AMP) and mitogen-activated protein kinase (MAPK) signaling pathways was necessary for the simultaneous expression of MUC2 and cathelicidins. In Muc2 mucin-deficient (Muc2-/-) mice, murine cathelicidin (Cramp) was significantly reduced compared to that in Muc2+/- and Muc2+/+ littermates. E. histolytica-induced acute inflammation in colonic loops stimulated high levels of cathelicidin in Muc2+/+ but not in Muc2-/- littermates. In dextran sodium sulfate (DSS)-induced colitis in Muc2+/+ mice, which depletes the mucus barrier and goblet cell mucin, Cramp expression was significantly enhanced during restitution. These studies demonstrate regulatory mechanisms between MUC2 and cathelicidins in the colonic mucosa where an intact mucus barrier is essential for expression and secretion of cathelicidins in response to E. histolytica- and DSS-induced colitis.


Subject(s)
Antimicrobial Cationic Peptides/biosynthesis , Butyrates/metabolism , Colitis/etiology , Colitis/metabolism , Mucin-2/metabolism , Animals , Antimicrobial Cationic Peptides/genetics , Cell Line , Colitis/pathology , Disease Models, Animal , Entamoeba histolytica , Entamoebiasis/metabolism , Entamoebiasis/parasitology , Entamoebiasis/pathology , Gene Expression , Humans , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/parasitology , Intestinal Mucosa/pathology , MAP Kinase Signaling System , Male , Mice , Mice, Knockout , Mitogen-Activated Protein Kinases/metabolism , Mucin-2/genetics , RNA, Messenger/genetics , Sulfates/adverse effects , Cathelicidins
16.
Am J Physiol Gastrointest Liver Physiol ; 312(1): G34-G45, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27856417

ABSTRACT

MUC2 mucin is the major glycoprotein in colonic mucus that separates intestinal microbiota from underlying host cells and serves as a food source for some eubacteria. MUC2 deficiency results in impaired epithelial barrier function, imbalance in gut microbiota, and spontaneous colitis. Probiotics have been shown to have a protective effect against colitis. In this study we used Muc2 mucin-deficient (Muc2-/-) and Muc2+/+ littermates to test whether the probiotic mixture VSL#3 requires an intact mucin barrier to exert its beneficial effect. VSL#3 alone reduced basal colonic proinflammatory cytokine levels and improved epithelial barrier function in Muc2-/- animals. Similarly, in dextran sulfate sodium-induced colitis, VSL#3 dampened the proinflammatory chemokines KC, monocyte chemoattractant protein-1, and macrophage inflammatory protein-2 and upregulated the tissue regeneration growth factors transforming growth factor-ß, fibroblast growth factor-1, and vascular endothelial growth factor-A, which accelerated resolution of colitis symptoms in Muc2-/- animals. Importantly, improved colonic health in VSL#3-treated animals was associated with attenuated reactive oxygen species production by peritoneal macrophages, restoration of antimicrobial peptide gene expression in the small intestine, and increased abundance of bacterial commensals in the gut. The beneficial effects of VSL#3 in Muc2-/- animals were mediated by acetate, an important short-chain fatty acid produced by gut bacteria. These studies provide evidence for the first time that VSL#3 can enhance epithelial barrier function by dampening the proinflammatory cytokine and chemokine response, accelerating restitution, and altering commensal microbiota in the absence of a functional mucus barrier. NEW & NOTEWORTHY: It is unclear whether probiotics require an intact mucin barrier to first colonize and/or exert their protective functions. In this study we used mucin-deficient (Muc2-/-) mice to interrogate if the multispecies probiotic mixture VSL#3 could enhance epithelial barrier function. In the absence of a mucus bilayer, VSL#3 dampened proinflammatory and chemokine production, accelerated restitution, and markedly improved gut permeability mediated by the short-chain fatty acid acetate in the colon.


Subject(s)
Colon/drug effects , Inflammation/drug therapy , Intestinal Mucosa/drug effects , Mucin-2/genetics , Probiotics/therapeutic use , Animals , Colon/metabolism , Colon/pathology , Inflammation/metabolism , Inflammation/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Mice, Knockout , Mucin-2/metabolism , Probiotics/pharmacology
17.
PLoS Pathog ; 11(5): e1004887, 2015 May.
Article in English | MEDLINE | ID: mdl-25955828

ABSTRACT

Entamoeba histolytica (Eh) is an extracellular protozoan parasite of humans that invades the colon to cause life-threatening intestinal and extra-intestinal amebiasis. Colonized Eh is asymptomatic, however, when trophozoites adhere to host cells there is a considerable inflammatory response that is critical in the pathogenesis of amebiasis. The host and/or parasite factors that trigger the inflammatory response to invading Eh are not well understood. We recently identified that Eh adherence to macrophages induces inflammasome activation and in the present study we sought to determine the molecular events upon contact that coordinates this response. Here we report that Eh contact-dependent activation of α5ß1 integrin is critical for activation of the NLRP3 inflammasome. Eh-macrophage contact triggered recruitment of α5ß1 integrin and NLRP3 into the intercellular junction, where α5ß1 integrin underwent activation by an integrin-binding cysteine protease on the parasite surface, termed EhCP5. As a result of its activation, α5ß1 integrin induced ATP release into the extracellular space through opening of pannexin-1 channels that signalled through P2X7 receptors to deliver a critical co-stimulatory signal that activated the NLRP3 inflammasome. Both the cysteine protease activity and integrin-binding domain of EhCP5 were required to trigger α5ß1 integrin that led to ATP release and NLRP3 inflammasome activation. These findings reveal engagement of α5ß1 integrin across the parasite-host junction is a key regulatory step that initiates robust inflammatory responses to Eh. We propose that α5ß1 integrin distinguishes Eh direct contact and functions with NLRP3 as pathogenicity sensor for invasive Eh infection.


Subject(s)
Carrier Proteins/metabolism , Entamoeba histolytica/immunology , Entamoebiasis/metabolism , Host-Pathogen Interactions , Inflammasomes/metabolism , Integrin alpha5beta1/agonists , Macrophages/metabolism , Animals , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Cell Adhesion , Cells, Cultured , Cysteine Proteases/chemistry , Cysteine Proteases/genetics , Cysteine Proteases/metabolism , Entamoeba histolytica/physiology , Entamoebiasis/immunology , Entamoebiasis/parasitology , Humans , Immunity, Innate , Inflammasomes/immunology , Integrin alpha5beta1/antagonists & inhibitors , Integrin alpha5beta1/genetics , Integrin alpha5beta1/metabolism , Macrophages/cytology , Macrophages/immunology , Macrophages/parasitology , Mice, Inbred C57BL , Mice, Knockout , Mutation , NLR Family, Pyrin Domain-Containing 3 Protein , Protein Interaction Domains and Motifs , Protein Transport , Proteolysis , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , RNA Interference , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
18.
Am J Pathol ; 182(3): 852-65, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23357502

ABSTRACT

Human mucin-2 (MUC-2) is the first line of innate host defense in preventing pathogen-induced epithelial injury. Entamoeba histolytica (Eh) colonizes the mucus layer by binding of the parasite's surface galactose lectin to galactose and N-acetyl-d-galactosamine residues on colonic MUC-2, preventing parasite contact-dependent cytolysis of epithelial cells. We quantified early innate responses to Eh in wild-type and MUC-2-deficient mice (Muc2(-/-)) using closed colonic loops. Eh infection in wild-type but not Muc2(-/-) mice induced a time-dependent increase in (3)H-labeled mucin and nonmucin glycoprotein secretions. Immunohistochemical staining revealed intense MUC-2 secretion, which formed a thick, protective mucus plug overlying the surface epithelium, entrapping Eh. In Muc2(-/-) mice, Eh induced a pronounced time-dependent secretory exudate with increased gross pathology scores and serum albumin leakage. Colonic pathology, secretory responses, and increased proinflammatory cytokine secretions of TNF-α, IFN-γ, and IL-13 correlated with altered expression of the tight junction proteins claudin-2, occludin, and ZO-1. We identified the putative Eh virulence factor that elicits the proinflammatory responses and alters tight junction permeability as Eh cysteine protease A5 (EhCP-A5). The present findings demonstrate that colonic mucins confer both luminal and epithelial barrier functions and that, in the absence of MUC-2, mice are more susceptible to Eh-induced secretory and proinflammatory responses mediated by EhCP-A5.


Subject(s)
Entamoeba histolytica/physiology , Epithelial Cells/pathology , Epithelial Cells/parasitology , Inflammation Mediators/metabolism , Mucin-2/deficiency , Tight Junctions/parasitology , Animals , Blood Cells/metabolism , Colon/parasitology , Colon/pathology , Entamoeba histolytica/pathogenicity , Gene Expression Regulation , Goblet Cells/parasitology , Goblet Cells/pathology , Humans , Intestines/parasitology , Intestines/pathology , Mice , Mice, Inbred C57BL , Mucin-2/metabolism , Permeability , Tight Junction Proteins/genetics , Tight Junction Proteins/metabolism , Tight Junctions/metabolism , Virulence Factors/metabolism
20.
Nat Cell Biol ; 8(8): 834-42, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16862145

ABSTRACT

Mutations in the parkin gene are responsible for a common familial form of Parkinson's disease. As parkin encodes an E3 ubiquitin ligase, defects in proteasome-mediated protein degradation are believed to have a central role in the pathogenesis of Parkinson's disease. Here, we report a novel role for parkin in a proteasome-independent ubiquitination pathway. We have identified a regulated interaction between parkin and Eps15, an adaptor protein that is involved in epidermal growth factor (EGF) receptor (EGFR) endocytosis and trafficking. Treatment of cells with EGF stimulates parkin binding to both Eps15 and the EGFR and promotes parkin-mediated ubiquitination of Eps15. Binding of the parkin ubiquitin-like (Ubl) domain to the Eps15 ubiquitin-interacting motifs (UIMs) is required for parkin-mediated Eps15 ubiquitination. Furthermore, EGFR endocytosis and degradation are accelerated in parkin-deficient cells, and EGFR signalling via the phosphoinositide 3-kinase (PI(3)K)-Akt pathway is reduced in parkin knockout mouse brain. We propose that by ubiquitinating Eps15, parkin interferes with the ability of the Eps15 UIMs to bind ubiquitinated EGFR, thereby delaying EGFR internalization and degradation, and promoting PI(3)K-Akt signalling. Considering the role of Akt in neuronal survival, our results have broad new implications for understanding the pathogenesis of Parkinson's disease.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , ErbB Receptors/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Ubiquitin-Protein Ligases/metabolism , Adaptor Proteins, Vesicular Transport/genetics , Animals , Blotting, Western , COS Cells , Cell Line , Chlorocebus aethiops , Endocytosis/drug effects , Endocytosis/physiology , Epidermal Growth Factor/pharmacology , HeLa Cells , Humans , Immunoprecipitation , Mice , Mice, Knockout , NIH 3T3 Cells , Protein Binding/drug effects , Protein Binding/physiology , Protein Transport/drug effects , Protein Transport/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , Transfection , Ubiquitin-Protein Ligases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL