Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
EMBO J ; 42(21): e113975, 2023 11 02.
Article in English | MEDLINE | ID: mdl-37718683

ABSTRACT

Paneth cells (PCs), a specialized secretory cell type in the small intestine, are increasingly recognized as having an essential role in host responses to microbiome and environmental stresses. Whether and how commensal and pathogenic microbes modify PC composition to modulate inflammation remain unclear. Using newly developed PC-reporter mice under conventional and gnotobiotic conditions, we determined PC transcriptomic heterogeneity in response to commensal and invasive microbes at single cell level. Infection expands the pool of CD74+ PCs, whose number correlates with auto or allogeneic inflammatory disease progressions in mice. Similar correlation was found in human inflammatory disease tissues. Infection-stimulated cytokines increase production of reactive oxygen species (ROS) and expression of a PC-specific mucosal pentraxin (Mptx2) in activated PCs. A PC-specific ablation of MyD88 reduced CD74+ PC population, thus ameliorating pathogen-induced systemic disease. A similar phenotype was also observed in mice lacking Mptx2. Thus, infection stimulates expansion of a PC subset that influences disease progression.


Subject(s)
Microbiota , Paneth Cells , Humans , Animals , Mice , Paneth Cells/metabolism , Paneth Cells/pathology , Intestine, Small , Inflammation/pathology , Cytokines/metabolism
2.
Antimicrob Agents Chemother ; 66(11): e0083222, 2022 11 15.
Article in English | MEDLINE | ID: mdl-36255253

ABSTRACT

New anti-Entamoeba histolytica multistage drugs are needed because only one drug class, nitroimidazoles, is available for treating invasive disease, and it does not effectively eradicate the infective cyst stage. Zinc ditiocarb (ZnDTC), a main metabolite of the FDA-approved drug disulfiram, was recently shown to be highly effective against the invasive trophozoite stage. In this brief report, we show that ZnDTC is active against cysts, with similar potency to first-line cysticidal drug paromomycin.


Subject(s)
Alcoholism , Cysts , Entamoeba histolytica , Parasites , Animals , Disulfiram/pharmacology , Disulfiram/therapeutic use , Ditiocarb/metabolism , Ditiocarb/pharmacology
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.
PLoS Pathog ; 16(9): e1008952, 2020 09.
Article in English | MEDLINE | ID: mdl-32960936

ABSTRACT

Understanding how the protozoan protein degradation pathway is regulated could uncover new parasite biology for drug discovery. We found the COP9 signalosome (CSN) conserved in multiple pathogens such as Leishmania, Trypanosoma, Toxoplasma, and used the severe diarrhea-causing Entamoeba histolytica to study its function in medically significant protozoa. We show that CSN is an essential upstream regulator of parasite protein degradation. Genetic disruption of E. histolytica CSN by two distinct approaches inhibited cell proliferation and viability. Both CSN5 knockdown and dominant negative mutation trapped cullin in a neddylated state, disrupting UPS activity and protein degradation. In addition, zinc ditiocarb (ZnDTC), a main metabolite of the inexpensive FDA-approved globally-available drug disulfiram, was active against parasites acting in a COP9-dependent manner. ZnDTC, given as disulfiram-zinc, had oral efficacy in clearing parasites in vivo. Our findings provide insights into the regulation of parasite protein degradation, and supports the significant therapeutic potential of COP9 inhibition.


Subject(s)
COP9 Signalosome Complex/metabolism , Entamoeba histolytica/metabolism , Proteolysis , Animals , COP9 Signalosome Complex/genetics , Disulfiram/pharmacology , Ditiocarb/pharmacology , Entamoeba histolytica/genetics , Mice , Protozoan Proteins/genetics
5.
Front Immunol ; 11: 1273, 2020.
Article in English | MEDLINE | ID: mdl-32655566

ABSTRACT

Wound healing after an injury is essential for life. An in-depth understanding of the healing process is necessary to ultimately improve the currently limited treatment options for patients suffering as a result of damage to various organs and tissues. Injuries, even the most minor, trigger an inflammatory response that protects the host and activates repair pathways. In recent years, substantial progress has been made in delineating the mechanisms by which inflammatory cytokines and their receptors facilitate tissue repair and regeneration. This mini review focuses on emerging literature on the role of the cytokine macrophage migration inhibitory factor (MIF) and its cell membrane receptor CD74, in protecting against injury and promoting healing in different parts of the body.


Subject(s)
Antigens, Differentiation, B-Lymphocyte/immunology , Histocompatibility Antigens Class II/immunology , Macrophage Migration-Inhibitory Factors/immunology , Wound Healing/immunology , Animals , Antigens, Differentiation, B-Lymphocyte/metabolism , Histocompatibility Antigens Class II/metabolism , Humans , Macrophage Migration-Inhibitory Factors/metabolism , Regeneration/immunology , Signal Transduction/physiology
6.
Cell Mol Gastroenterol Hepatol ; 10(1): 101-112, 2020.
Article in English | MEDLINE | ID: mdl-32004754

ABSTRACT

BACKGROUND & AIMS: The inflammatory response to intestinal damage promotes healing through mechanisms that are incompletely understood. Gene expression of cluster of differentiation 74 (CD74), the receptor for cytokine macrophage migration inhibitory factor, is increased in patients with inflammatory bowel disease (IBD), however, the role of CD74 signaling in intestinal inflammation remains poorly understood. The aim of this study was to determine the functional role of CD74 signaling in intestinal inflammation. METHODS: We studied the characteristics of CD74 protein expression in human IBD and experimental colitis. The functional role of CD74 signaling in the intestine was investigated using cellular models; wild-type, CD74-/-, and bone marrow chimera mice; neutralizing anti-CD74 antibodies; flow cytometry; immunohistochemistry; immunofluorescence; immunoblotting; and clustered regularly interspaced short palindromic repeats and associated protein 9 technology. RESULTS: In IBD patients and experimental colitis, CD74-receptor protein expression was increased in inflamed intestinal tissue, prominently in the crypt epithelial cells. By using distinct but complementary chemical and non-chemically induced mouse models of colitis with genetic and antibody neutralization approaches, we found that CD74 signaling was necessary for gut repair. Mechanistically, we found that the macrophage migration inhibitory factor cytokine, which also is increased in colitis, stimulated the CD74 receptor, enhancing intestinal epithelial cell proliferation through activation of the protein kinase B and the extracellular signal-regulated kinase pathways. Our data also suggest that CD74 signaling in immune cells was not essential for mucosal healing. CONCLUSIONS: CD74 signaling is strongly activated during intestinal inflammation and protects the host by promoting epithelial cell regeneration, healing, and maintaining mucosal barrier integrity. Enhancing the CD74 pathway may represent a unique therapeutic strategy for promoting healing in IBD.


Subject(s)
Antigens, Differentiation, B-Lymphocyte/metabolism , Colitis, Ulcerative/immunology , Crohn Disease/immunology , Histocompatibility Antigens Class II/metabolism , Intestinal Mucosa/pathology , Signal Transduction/immunology , Animals , Antigens, Differentiation, B-Lymphocyte/genetics , Biopsy , Bone Marrow Transplantation , Cell Line, Tumor , Colitis, Ulcerative/genetics , Colitis, Ulcerative/parasitology , Colitis, Ulcerative/pathology , Crohn Disease/pathology , Datasets as Topic , Disease Models, Animal , Entamoeba histolytica/pathogenicity , Epithelial Cells/immunology , Epithelial Cells/pathology , Gene Expression Profiling , Histocompatibility Antigens Class II/genetics , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Intramolecular Oxidoreductases/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Mice , Mice, Knockout , Permeability , Primary Cell Culture , Regeneration/immunology , Signal Transduction/genetics , Transplantation Chimera
7.
Methods Mol Biol ; 2080: 237-247, 2020.
Article in English | MEDLINE | ID: mdl-31745886

ABSTRACT

Macrophage migration inhibitory factor (MIF) is a proinflammatory and proproliferative cytokine expressed in humans. MIF homologs also exist in many pathogenic protozoans, including Entamoeba, Plasmodium, Toxoplasma, and Leishmania. Production of antibodies against parasite proteins allows for the generation of assays to measure and visualize parasite infection within hosts. In this chapter, we describe how to specifically purify antibodies against Entamoeba histolytica MIF (EhMIF), and subsequently use anti-EhMIF antibodies for ELISA on mouse and human samples and for immunohistochemistry on human tissue. These methods can be applied to any protein for high-quality antibody purification.


Subject(s)
Antibodies, Protozoan/immunology , Antibodies, Protozoan/isolation & purification , Antigens, Protozoan/immunology , Entamoeba histolytica/immunology , Macrophage Migration-Inhibitory Factors/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/isolation & purification , Enzyme-Linked Immunosorbent Assay , Feces/chemistry , Humans , Immunohistochemistry , Macrophage Migration-Inhibitory Factors/antagonists & inhibitors , Mice , Recombinant Fusion Proteins/immunology
8.
J Infect Dis ; 221(7): 1185-1193, 2020 03 16.
Article in English | MEDLINE | ID: mdl-31677380

ABSTRACT

Targeting virulence factors represents a promising alternative approach to antimicrobial therapy, through the inhibition of pathogenic pathways that result in host tissue damage. Yet, virulence inhibition remains an understudied area in parasitology. Several medically important protozoan parasites such as Plasmodium, Entamoeba, Toxoplasma, and Leishmania secrete an inflammatory macrophage migration inhibitory factor (MIF) cytokine homolog, a virulence factor linked to severe disease. The aim of this study was to investigate the effectiveness of targeting parasite-produced MIF as combination therapy with standard antibiotics to reduce disease severity. Here, we used Entamoeba histolytica as the model MIF-secreting protozoan, and a mouse model that mirrors severe human infection. We found that intestinal inflammation and tissue damage were significantly reduced in mice treated with metronidazole when combined with anti-E. histolytica MIF antibodies, compared to metronidazole alone. Thus, this preclinical study provides proof-of-concept that combining antiparasite MIF-blocking antibodies with current standard-of-care antibiotics might improve outcomes in severe protozoan infections.


Subject(s)
Antibodies, Protozoan/immunology , Antiprotozoal Agents , Host-Parasite Interactions/drug effects , Macrophage Migration-Inhibitory Factors/metabolism , Protozoan Proteins/metabolism , Animals , Antiprotozoal Agents/immunology , Antiprotozoal Agents/pharmacology , Entamoeba histolytica/drug effects , Entamoeba histolytica/immunology , Entamoeba histolytica/metabolism , Entamoeba histolytica/pathogenicity , Entamoebiasis , HCT116 Cells , Humans , Mice , Models, Molecular
9.
Front Immunol ; 10: 1995, 2019.
Article in English | MEDLINE | ID: mdl-31497025

ABSTRACT

Protozoan parasites represent a major threat to health and contribute significantly to morbidity and mortality worldwide, especially in developing countries. This is further compounded by lack of effective vaccines, drug resistance and toxicity associated with current therapies. Multiple protozoans, including Plasmodium, Entamoeba, Toxoplasma, and Leishmania produce homologs of the cytokine MIF. These parasite MIF homologs are capable of altering the host immune response during infection, and play a role in immune evasion, invasion and pathogenesis. This minireview outlines well-established and emerging literature on the role of parasite MIF homologs in disease, and their potential as targets for therapeutic and preventive interventions.


Subject(s)
Immune Evasion , Macrophage Migration-Inhibitory Factors/immunology , Parasites/immunology , Parasites/pathogenicity , Animals , Humans
10.
Sci Rep ; 8(1): 10241, 2018 07 06.
Article in English | MEDLINE | ID: mdl-29980718

ABSTRACT

Multiple protozoans produce homologs of the cytokine MIF which play a role in immune evasion, invasion and pathogenesis. However, how parasite-encoded MIF activity is controlled remains poorly understood. Cytokine activity can be inhibited by intracellular binding partners that are released in the extracellular space during cell death. We investigated the presence of an endogenous parasite protein that was capable of interacting and interfering with MIF activity. A screen for protein-protein interaction was performed using immunoaffinity purification of amebic cell lysate with specific anti-Entamoeba histolytica MIF (EhMIF) antibody followed by mass spectrometry analysis, which revealed an E. histolytica-produced JAB1 protein (EhJAB1) as a potential binding partner. JAB1 was found to be highly conserved in protozoans. Direct interaction between the EhMIF and EhJAB1 was confirmed by several independent approaches with GST pull-down, co-immunoprecipitation, and Biolayer interferometry (BLI) assays. Furthermore, the C-terminal region outside the functional JAMM deneddylase motif was required for EhMIF binding, which was consistent with the top in silico predictions. In addition, EhJAB1 binding blocked EhMIF-induced IL-8 production by human epithelial cells. We report the initial characterization of a parasite-encoded JAB1 and uncover a new binding partner for a protozoan-produced MIF protein, acting as a possible negative regulator of EhMIF.


Subject(s)
Antibodies/immunology , Entamoeba histolytica/physiology , Entamoebiasis/metabolism , Inflammation/prevention & control , Macrophage Migration-Inhibitory Factors/metabolism , Protozoan Proteins/metabolism , Amino Acid Sequence , Cytokines/metabolism , Entamoebiasis/immunology , Entamoebiasis/parasitology , HCT116 Cells , Humans , Inflammation/immunology , Inflammation/metabolism , Inflammation/parasitology , Macrophage Migration-Inhibitory Factors/immunology , Protein Interaction Domains and Motifs , Protozoan Proteins/immunology
11.
Open Forum Infect Dis ; 5(7): ofy161, 2018 Jul.
Article in English | MEDLINE | ID: mdl-30046644

ABSTRACT

Amebiasis, due to the pathogenic parasite Entamoeba histolytica, is a leading cause of diarrhea globally. Largely an infection of impoverished communities in developing countries, amebiasis has emerged as an important infection among returning travelers, immigrants, and men who have sex with men residing in developed countries. Severe cases can be associated with high case fatality. Polymerase chain reaction-based diagnosis is increasingly available but remains underutilized. Nitroimidazoles are currently recommended for treatment, but new drug development to treat parasitic agents is a high priority. Amebiasis should be considered before corticosteroid therapy to decrease complications. There is no effective vaccine, so prevention focuses on sanitation and access to clean water. Further understanding of parasite biology and pathogenesis will advance future targeted therapeutic and preventative strategies.

12.
J Infect Dis ; 215(8): 1294-1302, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28186296

ABSTRACT

Understanding the mechanisms by which Entamoeba histolytica drives gut inflammation is critical for the development of improved preventive and therapeutic strategies. E. histolytica encodes a homolog of the human cytokine macrophage migration inhibitory factor (MIF). Here, we investigated the role of E. histolytica MIF (EhMIF) during infection. We found that the concentration of fecal EhMIF correlated with the level of intestinal inflammation in persons with intestinal amebiasis. Mice treated with antibodies that specifically block EhMIF had reduced chemokine expression and neutrophil infiltration in the mucosa. In addition to antibody-mediated neutralization, we used a genetic approach to test the effect of EhMIF on mucosal inflammation. Mice infected with parasites overexpressing EhMIF had increased chemokine expression, neutrophil influx, and mucosal damage. Together, these results uncover a specific parasite protein that increases mucosal inflammation, expands our knowledge of host-parasite interaction during amebic colitis, and highlights a potential immunomodulatory target.


Subject(s)
Dysentery, Amebic/pathology , Macrophage Migration-Inhibitory Factors/immunology , Neutrophils/immunology , Protozoan Proteins/immunology , Animals , Antibodies, Blocking/pharmacology , Caco-2 Cells , Cell Culture Techniques , Child, Preschool , Dysentery, Amebic/drug therapy , Entamoeba histolytica/drug effects , Feces/chemistry , Host-Parasite Interactions , Humans , Intestinal Mucosa/parasitology , Intestinal Mucosa/pathology , Macrophage Migration-Inhibitory Factors/genetics , Male , Mice , Mice, Inbred CBA , Neutrophil Infiltration/drug effects , Protozoan Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...