1
|
Chan Y, Martin D, Mace KE, Jean SE, Stresman G, Drakeley C, Chang MA, Lemoine JF, Udhayakumar V, Lammie PJ, Priest JW, Rogier EW. Multiplex Serology for Measurement of IgG Antibodies Against Eleven Infectious Diseases in a National Serosurvey: Haiti 2014-2015. Front Public Health 2022; 10:897013. [PMID: 35757611 PMCID: PMC9218545 DOI: 10.3389/fpubh.2022.897013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/18/2022] [Indexed: 11/13/2022] Open
Abstract
Background Integrated surveillance for multiple diseases can be an efficient use of resources and advantageous for national public health programs. Detection of IgG antibodies typically indicates previous exposure to a pathogen but can potentially also serve to assess active infection status. Serological multiplex bead assays have recently been developed to simultaneously evaluate exposure to multiple antigenic targets. Haiti is an island nation in the Caribbean region with multiple endemic infectious diseases, many of which have a paucity of data for population-level prevalence or exposure. Methods A nationwide serosurvey occurred in Haiti from December 2014 to February 2015. Filter paper blood samples (n = 4,438) were collected from participants in 117 locations and assayed for IgG antibodies on a multiplex bead assay containing 15 different antigens from 11 pathogens: Plasmodium falciparum, Toxoplasma gondii, lymphatic filariasis roundworms, Strongyloides stercoralis, chikungunya virus, dengue virus, Chlamydia trachomatis, Treponema pallidum, enterotoxigenic Escherichia coli, Entamoeba histolytica, and Cryptosporidium parvum. Results Different proportions of the Haiti study population were IgG seropositive to the different targets, with antigens from T. gondii, C. parvum, dengue virus, chikungunya virus, and C. trachomatis showing the highest rates of seroprevalence. Antibody responses to T. pallidum and lymphatic filariasis were the lowest, with <5% of all samples IgG seropositive to antigens from these pathogens. Clear trends of increasing seropositivity and IgG levels with age were seen for all antigens except those from chikungunya virus and E. histolytica. Parametric models were able to estimate the rate of seroconversion and IgG acquisition per year for residents of Haiti. Conclusions Multiplex serological assays can provide a wealth of information about population exposure to different infectious diseases. This current Haitian study included IgG targets for arboviral, parasitic, and bacterial infectious diseases representing multiple different modes of host transmission. Some of these infectious diseases had a paucity or complete absence of published serological studies in Haiti. Clear trends of disease burden with respect to age and location in Haiti can be used by national programs and partners for follow-up studies, resource allocation, and intervention planning.
Collapse
Affiliation(s)
- YuYen Chan
- The London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Diana Martin
- Division of Parasitic Diseases and Malaria, Center for Global Health, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Kimberly E Mace
- Division of Parasitic Diseases and Malaria, Center for Global Health, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Samuel E Jean
- Population Services International/Organization Haïtienne de Marketing Social Pour la Santé, Port-au-Prince, Haiti
| | - Gillian Stresman
- The London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Chris Drakeley
- The London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Michelle A Chang
- Division of Parasitic Diseases and Malaria, Center for Global Health, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Jean F Lemoine
- Programme National de Contrôle de la Malaria/MSPP, Port-au-Prince, Haiti
| | - Venkatachalam Udhayakumar
- Division of Parasitic Diseases and Malaria, Center for Global Health, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Patrick J Lammie
- Division of Parasitic Diseases and Malaria, Center for Global Health, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Jeffrey W Priest
- Division of Foodborne, Waterborne, and Environmental Diseases, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Eric William Rogier
- Division of Parasitic Diseases and Malaria, Center for Global Health, Centers for Disease Control and Prevention, Atlanta, GA, United States
| |
Collapse
|
2
|
Flores MS, Tamez E, Rangel R, Monjardin J, Bosques F, Obregón A, Trejo-Avila L, Quintero I, Gandarilla F, Arevalo K, Alemán E, Galán L. UBIQUITIN OF Entamoeba histolytica INDUCE ANTIBODY RESPONSE IN PATIENTS WITH INVASIVE AMOEBIASIS. Parasite Immunol 2022; 44:e12919. [PMID: 35366008 DOI: 10.1111/pim.12919] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 03/21/2022] [Accepted: 03/28/2022] [Indexed: 12/31/2022]
Abstract
Entamoeba histolytica causes amoebic liver abscess (ALA) in humans. The injury of target cells by Entamoeba histolytica includes processes controlled by the ubiquitin Ehub. Previously, we found immunodominance of Ehub glycan moieties using immunized rabbits. In this work, we analyzed dominance of antibodies to the glycoprotein Ehub in the sera from 52 patients with ALA. Controls were sera from 20 healthy people living in endemic areas with high seroprevalence of antibodies to amoebas, and 20 patients with alcoholic hepatitis (AH) to rule out the cross-reaction of Ehub with autoantibodies induced by liver damage. Antigens were trophozoite extract, glycoprotein Ehub and the recombinant protein rEhub. The sera from healthy volunteers and patients with AH do not have antibodies to glycoprotein Ehub. Surprisingly, only the antibodies from patients with ALA recognized the glycoprotein Ehub, and some sera gave a faint reaction with the recombinant protein, especially because evolutionarily, the ubiquitin is conserved between species. This is the first report demonstrating that antibodies to ubiquitin Ehub are induced exclusively in patients with invasive amoebiasis, and the antibody response is mainly to the glycoprotein, indicating glycans are immunodominant. Inhibitors of the Ehub glycans could be potential treatment for amoebiasis by selectively damaging trophozoites. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- María S Flores
- Universidad Autónoma de Nuevo León, Facultad de Ciencias Biológicas, Instituto de Biotecnología. San Nicolás de los Garza, Nuevo León, Mexico
| | - Eva Tamez
- Universidad Autónoma de Nuevo León, Facultad de Medicina, Monterrey, Nuevo León, Mexico
| | - Roberto Rangel
- Universidad Autónoma de Nuevo León, Facultad de Ciencias Biológicas, Instituto de Biotecnología. San Nicolás de los Garza, Nuevo León, Mexico
| | | | - Francisco Bosques
- Universidad Autónoma de Nuevo León, Facultad de Medicina, Monterrey, Nuevo León, Mexico
| | - Adriana Obregón
- Universidad Autónoma de Nuevo León, Facultad de Ciencias Biológicas, Instituto de Biotecnología. San Nicolás de los Garza, Nuevo León, Mexico
| | - Laura Trejo-Avila
- Universidad Autónoma de Nuevo León, Facultad de Ciencias Biológicas, Instituto de Biotecnología. San Nicolás de los Garza, Nuevo León, Mexico
| | - Isela Quintero
- Universidad Autónoma de Nuevo León, Facultad de Ciencias Biológicas, Instituto de Biotecnología. San Nicolás de los Garza, Nuevo León, Mexico
| | - Fátima Gandarilla
- Universidad Autónoma de Nuevo León, Facultad de Ciencias Biológicas, Instituto de Biotecnología. San Nicolás de los Garza, Nuevo León, Mexico
| | - Katiushka Arevalo
- Universidad Autónoma de Nuevo León, Facultad de Ciencias Biológicas, Instituto de Biotecnología. San Nicolás de los Garza, Nuevo León, Mexico
| | - Elizabeth Alemán
- Universidad Autónoma de Nuevo León, Facultad de Ciencias Biológicas, Instituto de Biotecnología. San Nicolás de los Garza, Nuevo León, Mexico
| | - Luis Galán
- Universidad Autónoma de Nuevo León, Facultad de Ciencias Biológicas, Instituto de Biotecnología. San Nicolás de los Garza, Nuevo León, Mexico
| |
Collapse
|
3
|
Aiemjoy K, Aragie S, Wittberg DM, Tadesse Z, Callahan EK, Gwyn S, Martin D, Keenan JD, Arnold BF. Seroprevalence of antibodies against Chlamydia trachomatis and enteropathogens and distance to the nearest water source among young children in the Amhara Region of Ethiopia. PLoS Negl Trop Dis 2020; 14:e0008647. [PMID: 32877398 PMCID: PMC7491729 DOI: 10.1371/journal.pntd.0008647] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 09/15/2020] [Accepted: 07/27/2020] [Indexed: 11/19/2022] Open
Abstract
The transmission of trachoma, caused by repeat infections with Chlamydia trachomatis, and many enteropathogens are linked to water quantity. We hypothesized that children living further from a water source would have higher exposure to C. trachomatis and enteric pathogens as determined by antibody responses. We used a multiplex bead assay to measure IgG antibody responses to C. trachomatis, Giardia intestinalis, Cryptosporidium parvum, Entamoeba histolytica, Salmonella enterica, Campylobacter jejuni, enterotoxigenic Escherichia coli (ETEC) and Vibrio cholerae in eluted dried blood spots collected from 2267 children ages 0-9 years in 40 communities in rural Ethiopia in 2016. Linear distance from the child's house to the nearest water source was calculated. We derived seroprevalence cutoffs using external negative control populations, if available, or by fitting finite mixture models. We used targeted maximum likelihood estimation to estimate differences in seroprevalence according to distance to the nearest water source. Seroprevalence among 1-9-year-olds was 43% for C. trachomatis, 28% for S. enterica, 70% for E. histolytica, 54% for G. intestinalis, 96% for C. jejuni, 76% for ETEC and 94% for C. parvum. Seroprevalence increased with age for all pathogens. Median distance to the nearest water source was 473 meters (IQR 268, 719). Children living furthest from a water source had a 12% (95% CI: 2.6, 21.6) higher seroprevalence of S. enterica and a 12.7% (95% CI: 2.9, 22.6) higher seroprevalence of G. intestinalis compared to children living nearest. Seroprevalence for C. trachomatis and enteropathogens was high, with marked increases for most enteropathogens in the first two years of life. Children living further from a water source had higher seroprevalence of S. enterica and G. intestinalis indicating that improving access to water in the Ethiopia's Amhara region may reduce exposure to these enteropathogens in young children.
Collapse
Affiliation(s)
- Kristen Aiemjoy
- Division of Infectious Diseases and Geographic Medicine, Stanford University School of Medicine, Stanford, California, United States of America
| | | | - Dionna M. Wittberg
- Francis I. Proctor Foundation, University of California San Francisco, San Francisco, California, United States of America
| | | | | | - Sarah Gwyn
- U.S. Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Diana Martin
- U.S. Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Jeremy D. Keenan
- Francis I. Proctor Foundation, University of California San Francisco, San Francisco, California, United States of America
| | - Benjamin F. Arnold
- Francis I. Proctor Foundation, University of California San Francisco, San Francisco, California, United States of America
| |
Collapse
|
4
|
Guzmán-Téllez P, Martínez-Castillo M, Flores-Huerta N, Rosales-Morgan G, Pacheco-Yépez J, la Garza MD, Serrano-Luna J, Shibayama M. Lectins as virulence factors in Entamoeba histolytica and free-living amoebae. Future Microbiol 2020; 15:919-936. [PMID: 32716210 DOI: 10.2217/fmb-2019-0275] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Currently, there is growing interest in the identification and purification of microbial lectins due to their involvement in the pathogenicity mechanisms of pathogens, such as Entamoeba histolytica and free-living amoebae. The Gal/GalNAc lectin from E. histolytica participates in adhesion, cytotoxicity and regulation of immune responses. Furthermore, mannose- and galactose-binding protein have been described in Acanthamoeba castellanii and Balamuthia mandrillaris, respectively and they also contribute to host damage. Finally, in Naegleria fowleri, molecules containing mannose and fucose are implicated in adhesion and cytotoxicity. Considering their relevance in the pathogenesis of the diseases caused by these protozoa, lectins appear to be promising targets in the diagnosis, vaccination and treatment of these infections.
Collapse
Affiliation(s)
- Paula Guzmán-Téllez
- Department of Infectomics & Molecular Pathogenesis, Center for Research & Advanced Studies of The National Polytechnic Institute, Av. IPN 2508, Mexico City 07360, Mexico
| | - Moisés Martínez-Castillo
- Department of Infectomics & Molecular Pathogenesis, Center for Research & Advanced Studies of The National Polytechnic Institute, Av. IPN 2508, Mexico City 07360, Mexico
- Department of Experimental Medicine, Liver, Pancreas & Motility Laboratory (HIPAM), School of Medicine, National Autonomous University of Mexico (UNAM) Mexico City, Mexico
| | - Nadia Flores-Huerta
- Department of Infectomics & Molecular Pathogenesis, Center for Research & Advanced Studies of The National Polytechnic Institute, Av. IPN 2508, Mexico City 07360, Mexico
| | - Gabriela Rosales-Morgan
- Department of Infectomics & Molecular Pathogenesis, Center for Research & Advanced Studies of The National Polytechnic Institute, Av. IPN 2508, Mexico City 07360, Mexico
| | - Judith Pacheco-Yépez
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México, México
| | - Mireya de la Garza
- Department of Cell Biology, Center for Research & Advanced Studies of The National Polytechnic Institute, Av. IPN 2508, Mexico City 07360, Mexico
| | - Jesús Serrano-Luna
- Department of Cell Biology, Center for Research & Advanced Studies of The National Polytechnic Institute, Av. IPN 2508, Mexico City 07360, Mexico
| | - Mineko Shibayama
- Department of Infectomics & Molecular Pathogenesis, Center for Research & Advanced Studies of The National Polytechnic Institute, Av. IPN 2508, Mexico City 07360, Mexico
| |
Collapse
|
5
|
Arnold BF, Martin DL, Juma J, Mkocha H, Ochieng JB, Cooley GM, Omore R, Goodhew EB, Morris JF, Costantini V, Vinjé J, Lammie PJ, Priest JW. Enteropathogen antibody dynamics and force of infection among children in low-resource settings. eLife 2019; 8:45594. [PMID: 31424386 PMCID: PMC6746552 DOI: 10.7554/elife.45594] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 08/15/2019] [Indexed: 01/22/2023] Open
Abstract
Little is known about enteropathogen seroepidemiology among children in low-resource settings. We measured serological IgG responses to eight enteropathogens (Giardia intestinalis, Cryptosporidium parvum, Entamoeba histolytica, Salmonella enterica, enterotoxigenic Escherichia coli, Vibrio cholerae, Campylobacter jejuni, norovirus) in cohorts from Haiti, Kenya, and Tanzania. We studied antibody dynamics and force of infection across pathogens and cohorts. Enteropathogens shared common seroepidemiologic features that enabled between-pathogen comparisons of transmission. Overall, exposure was intense: for most pathogens the window of primary infection was <3 years old; for highest transmission pathogens primary infection occurred within the first year. Longitudinal profiles demonstrated significant IgG boosting and waning above seropositivity cutoffs, underscoring the value of longitudinal designs to estimate force of infection. Seroprevalence and force of infection were rank-preserving across pathogens, illustrating the measures provide similar information about transmission heterogeneity. Our findings suggest antibody response can be used to measure population-level transmission of diverse enteropathogens in serologic surveillance. Diarrhea, which is caused by bacteria such as Salmonella or by viruses like norovirus, is the fourth leading cause of death among children worldwide, with children in low-resource settings being at highest risk. The pathogens that cause diarrhea spread when stool from infected people comes into contact with new hosts, for example, through inadequate sanitation or by drinking contaminated water. Currently, the best way to track these infections is to collect stool samples from people and test them for the presence of the pathogens. Unfortunately, this is costly and difficult to do on a large scale outside of clinical settings, making it hard to track the spread of diarrhea-causing pathogens. The body produces antibodies – small proteins that can detect specific pathogens – in response to an infection. These antibodies help ward off future infections by the same pathogen, so if they are present in the blood, this indicates a current or previous infection. Scientists already collect blood samples to track malaria, HIV and vaccine-preventable diseases in low-resource settings. These samples could be tested more broadly to measure the levels of antibodies against diarrhea-causing pathogens. Now, Arnold et al. have used blood samples collected from children in Haiti, Kenya, and Tanzania to measure antibody responses to 8 diarrhea-causing pathogens. The results showed that many children in these settings had been infected with all 8 pathogens before age three, and that all of the pathogens shared similar age-dependent patterns of antibody response. This finding enabled Arnold et al. to combine antibody measurements with statistical models to estimate each pathogen’s force of infection, that is, the rate at which susceptible individuals in the population become infected. This is a key step for epidemiologists to understand which pathogens cause the most infections in a population. The experiments show that testing blood samples for antibodies could provide scientists with a new tool to track the transmission of diarrhea-causing pathogens in low-resource settings. This information could help public health officials design and test efforts to prevent diarrhea, for example, by improving water treatment or developing vaccines.
Collapse
Affiliation(s)
- Benjamin F Arnold
- Division of Epidemiology and Biostatistics, University of California, Berkeley, Berkeley, United States.,Francis I. Proctor Foundation, University of California, San Francisco, San Francisco, United States.,Department of Ophthalmology, University of California, San Francisco, San Francisco, United States
| | - Diana L Martin
- Division of Parasitic Diseases and Malaria, United States Centers for Disease Control and Prevention, Atlanta, United States
| | - Jane Juma
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - Harran Mkocha
- Kongwa Trachoma Project, Kongwa, United Republic of Tanzania
| | - John B Ochieng
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - Gretchen M Cooley
- Division of Parasitic Diseases and Malaria, United States Centers for Disease Control and Prevention, Atlanta, United States
| | - Richard Omore
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - E Brook Goodhew
- Division of Parasitic Diseases and Malaria, United States Centers for Disease Control and Prevention, Atlanta, United States
| | - Jamae F Morris
- Department of African-American Studies, Georgia State University, Atlanta, United States
| | - Veronica Costantini
- Division of Viral Diseases, United States Centers for Disease Control and Prevention, Atlanta, United States
| | - Jan Vinjé
- Division of Viral Diseases, United States Centers for Disease Control and Prevention, Atlanta, United States
| | - Patrick J Lammie
- Division of Parasitic Diseases and Malaria, United States Centers for Disease Control and Prevention, Atlanta, United States.,Neglected Tropical Diseases Support Center, Task Force for Global Health, Decatur, United States
| | - Jeffrey W Priest
- Division of Foodborne, Waterborne, and Environmental Diseases, United States Centers for Disease Control and Prevention, Atlanta, United States
| |
Collapse
|
6
|
Wojcik GL, Marie C, Abhyankar MM, Yoshida N, Watanabe K, Mentzer AJ, Carstensen T, Mychaleckyj J, Kirkpatrick BD, Rich SS, Concannon P, Haque R, Tsokos GC, Petri WA, Duggal P. Genome-Wide Association Study Reveals Genetic Link between Diarrhea-Associated Entamoeba histolytica Infection and Inflammatory Bowel Disease. mBio 2018; 9:e01668-18. [PMID: 30228239 PMCID: PMC6143743 DOI: 10.1128/mbio.01668-18] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 08/09/2018] [Indexed: 12/19/2022] Open
Abstract
Entamoeba histolytica is the etiologic agent of amebic dysentery, though clinical manifestation of infection is highly variable ranging from subclinical colonization to invasive disease. We hypothesize that host genetics contribute to the variable outcomes of E. histolytica infection; thus, we conducted a genome-wide association study (GWAS) in two independent birth cohorts of Bangladeshi infants monitored for susceptibility to E. histolytica disease in the first year of life. Children with at least one diarrheal episode positive for E. histolytica (cases) were compared to children with no detectable E. histolytica infection in the same time frame (controls). Meta-analyses under a fixed-effect inverse variance weighting model identified multiple variants in a region of chromosome 10 containing loci associated with symptomatic E. histolytica infection. An intergenic insertion between CREM and CCNY (rs58000832) achieved genome-wide significance (P value from meta-analysis [Pmeta] = 6.05 × 10-9), and each additional risk allele of rs58000832 conferred 2.42 increased odds of a diarrhea-associated E. histolytica infection. The most strongly associated single nucleotide polymorphism (SNP) within a gene was in an intron of CREM (rs58468612; Pmeta = 8.94 × 10-8), which has been implicated as a susceptibility locus for inflammatory bowel disease (IBD). Gene expression resources suggest associated loci are related to the lower expression of CREM Increased CREM expression is also observed in early E. histolytica infection. Further, CREM-/- mice were more susceptible to E. histolytica amebic colitis. These genetic associations reinforce the pathological similarities observed in gut inflammation between E. histolytica infection and IBD.IMPORTANCE Diarrhea is the second leading cause of death for children globally, causing 760,000 deaths each year in children less than 5 years old. Amebic dysentery contributes significantly to this burden, especially in developing countries. The identification of host factors that control or enable enteric pathogens has the potential to transform our understanding of disease predisposition, outcomes, and treatments. Our discovery of the transcriptional regulator cAMP-responsive element modulator (CREM) as a genetic modifier of susceptibility to amebic disease has implications for understanding the pathogenesis of other diarrheal infections. Further, emerging evidence for CREM in IBD susceptibility suggests that CREM is a critical regulator of enteric inflammation and may have broad therapeutic potential as a drug target across intestinal inflammatory diseases.
Collapse
Affiliation(s)
- Genevieve L Wojcik
- Department of Genetics, Stanford University School of Medicine, Stanford, California, USA
| | - Chelsea Marie
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Mayuresh M Abhyankar
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Nobuya Yoshida
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Koji Watanabe
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Alexander J Mentzer
- Nuffield Department of Medicine, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Tommy Carstensen
- Wellcome Trust Sanger Institute, Genome Campus, Oxford, United Kingdom
- Department of Medicine, Cambridge University, Cambridge, United Kingdom
| | - Josyf Mychaleckyj
- Department of Public Health Sciences, Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Beth D Kirkpatrick
- Vaccine Testing Center, University of Vermont College of Medicine, Burlington, Vermont, USA
| | - Stephen S Rich
- Department of Public Health Sciences, Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Patrick Concannon
- Genetics Institute and Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida, USA
| | - Rashidul Haque
- International Centre for Diarrheal Disease Research, Dhaka, Bangladesh
| | - George C Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - William A Petri
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Priya Duggal
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
7
|
Abhyankar MM, Orr MT, Lin S, Suraju MO, Simpson A, Blust M, Pham T, Guderian JA, Tomai MA, Elvecrog J, Pedersen K, Petri WA, Fox CB. Adjuvant composition and delivery route shape immune response quality and protective efficacy of a recombinant vaccine for Entamoeba histolytica. NPJ Vaccines 2018; 3:22. [PMID: 29900011 PMCID: PMC5988657 DOI: 10.1038/s41541-018-0060-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Revised: 04/26/2018] [Accepted: 05/03/2018] [Indexed: 12/20/2022] Open
Abstract
Amebiasis caused by Entamoeba histolytica is the third leading cause of parasitic mortality globally, with some 100,000 deaths annually, primarily among young children. Protective immunity to amebiasis is associated with fecal IgA and IFN-γ in humans; however, no vaccine exists. We have previously identified recombinant LecA as a potential protective vaccine antigen. Here we describe the development of a stable, manufacturable PEGylated liposomal adjuvant formulation containing two synthetic Toll-like receptor (TLR) ligands: GLA (TLR4) and 3M-052 (TLR7/8). The liposomes stimulated production of monocyte/macrophage chemoattractants MCP-1 and Mip-1β, and Th1-associated cytokines IL-12p70 and IFN-γ from human whole blood dependent on TLR ligand composition and dose. The liposomes also demonstrated acceptable physicochemical compatibility with the recombinant LecA antigen. Whereas mice immunized with LecA and GLA-liposomes demonstrated enhanced antigen-specific fecal IgA titers, mice immunized with LecA and 3M-052-liposomes showed a stronger Th1 immune profile. Liposomes containing GLA and 3M-052 together elicited both LecA-specific fecal IgA and Th1 immune responses. Furthermore, the quality of the immune response could be modulated with modifications to the liposomal formulation based on PEG length. Compared to subcutaneous administration, the optimized liposome adjuvant composition with LecA antigen administered intranasally resulted in significantly enhanced fecal IgA, serum IgG2a, as well as systemic IFN-γ and IL-17A levels in mice. The optimized intranasal regimen provided greater than 80% protection from disease as measured by parasite antigen in the colon. This work demonstrates the physicochemical and immunological characterization of an optimized mucosal adjuvant system containing a combination of TLR ligands with complementary activities and illustrates the importance of adjuvant composition and route of delivery to enhance a multifaceted and protective immune response to amebiasis.
Collapse
Affiliation(s)
- Mayuresh M. Abhyankar
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia Health System, Charlottesville, VA USA
| | - Mark T. Orr
- IDRI, 1616 Eastlake Ave E, Seattle, WA USA
- Department of Global Health, University of Washington, Seattle, WA USA
| | - Susan Lin
- IDRI, 1616 Eastlake Ave E, Seattle, WA USA
| | - Mohammed O. Suraju
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia Health System, Charlottesville, VA USA
| | | | | | - Tiep Pham
- IDRI, 1616 Eastlake Ave E, Seattle, WA USA
| | | | - Mark A. Tomai
- 3M Drug Delivery Systems, 3M Center, 275-3E-10, St. Paul, MN USA
| | - James Elvecrog
- 3M Drug Delivery Systems, 3M Center, 275-3E-10, St. Paul, MN USA
| | | | - William A. Petri
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia Health System, Charlottesville, VA USA
| | - Christopher B. Fox
- IDRI, 1616 Eastlake Ave E, Seattle, WA USA
- Department of Global Health, University of Washington, Seattle, WA USA
| |
Collapse
|
8
|
Zambrano LD, Priest JW, Ivan E, Rusine J, Nagel C, Kirby M, Rosa G, Clasen TF. Use of Serologic Responses against Enteropathogens to Assess the Impact of a Point-of-Use Water Filter: A Randomized Controlled Trial in Western Province, Rwanda. Am J Trop Med Hyg 2017; 97:876-887. [PMID: 28749764 PMCID: PMC5590594 DOI: 10.4269/ajtmh.16-1006] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 05/29/2017] [Indexed: 12/28/2022] Open
Abstract
Diarrhea is a leading contributor to childhood morbidity and mortality in sub-Saharan Africa. Given the challenge of blinding most water, sanitation, and hygiene (WASH) interventions, diarrheal disease outcome measures in WASH intervention trials are subject to potential bias and misclassification. Using the platform of a cluster-randomized controlled trial of a household-based drinking water filter in western province, Rwanda, we assessed the impact of the drinking water filter on enteric seroconversion in young children as a health outcome and examined the association between serologic responses and caregiver-reported diarrhea. Among the 2,179 children enrolled in the trial, 189 children 6-12 months of age were enrolled in a nested serology study. These children had their blood drawn at baseline and 6-12 months after the intervention was distributed. Multiplex serologic assays for Giardia, Cryptosporidium, Entamoeba histolytica, norovirus, Campylobacter, enterotoxigenic Escherichia coli and Vibrio cholerae were performed. Despite imperfect uptake, receipt of the water filter was associated with a significant decrease in seroprevalence of IgG directed against Cryptosporidium parvum Cp17 and Cp23 (relative risk [RR]: 0.62, 95% confidence interval [CI]: 0.44-0.89). Serologic responses were positively associated with reported diarrhea in the previous 7 days for both Giardia intestinalis (RR: 1.94, 95% CI: 1.04-3.63) and C. parvum (RR: 2.21, 95% CI: 1.09-4.50). Serologic responses for all antigens generally increased in the follow-up round, rising sharply after 12 months of age. The water filter is associated with reduced serologic responses against C. parvum, a proxy for exposure and infection; therefore, serologic responses against protozoa may be a suitable health outcome measure for WASH trials among children with diarrhea.
Collapse
Affiliation(s)
- Laura Divens Zambrano
- Department of Environmental Health, Emory University Rollins School of Public Health, Atlanta, Georgia
| | - Jeffrey W. Priest
- Division of Foodborne, Waterborne and Environmental Diseases, National Center for Zoonotic and Emerging Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Emil Ivan
- National Reference Laboratory, Rwanda Biomedical Center, Kigali, Rwanda
| | - John Rusine
- National Reference Laboratory, Rwanda Biomedical Center, Kigali, Rwanda
| | - Corey Nagel
- OHSU/PSU School of Public Health, Oregon Health and Science University, Portland, Oregon
| | - Miles Kirby
- Department of Disease Control, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Ghislaine Rosa
- Department of Disease Control, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Thomas F. Clasen
- Department of Environmental Health, Emory University Rollins School of Public Health, Atlanta, Georgia
- Department of Disease Control, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
9
|
St-Pierre J, Moreau F, Cornick S, Quach J, Begum S, Aracely Fernandez L, Gorman H, Chadee K. The macrophage cytoskeleton acts as a contact sensor upon interaction with Entamoeba histolytica to trigger IL-1β secretion. PLoS Pathog 2017; 13:e1006592. [PMID: 28837696 PMCID: PMC5587335 DOI: 10.1371/journal.ppat.1006592] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 09/06/2017] [Accepted: 08/18/2017] [Indexed: 01/09/2023] Open
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 α5β1 integrin are absolute requirements for NLRP3 inflammasome activation and IL-1β 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-1β. 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. The protozoan parasite Entamoeba histolytica can establish an enteric infection in human hosts that leads to symptoms ranging from diarrhea to abscesses in the liver and the brain. Host susceptibility to amebic infection is in part determined by the quality and potency of the host immune response that occurs once the parasite overcomes the mucus bilayers and colonic epithelial barriers, and invades underlying tissues. At the cellular level, one of the key events that shape the inflammatory response occurs during direct parasite interaction with host macrophages via surface proteins. The ensuing cascades of intracellular signaling events have only partly been uncovered. Interestingly, only direct interaction between live parasites and macrophages, as opposed to soluble factors or dead parasites, is a prerequisite to the generation of a prompt raging pro-inflammatory response. We have sought to further elucidate the mechanisms by which macrophages distinguish live parasites and found that the macrophage cell skeleton undergoes rapid significant alteration upon Eh contact. Furthermore, we uncovered a previously unknown role for two Eh enzymes in triggering macrophage pro-inflammatory responses. Through this work, we gain a better understanding of the molecular interactions that occur at the macrophage-ameba interface that regulate host inflammatory responses.
Collapse
Affiliation(s)
- Joëlle St-Pierre
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - France Moreau
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Steve Cornick
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Jeanie Quach
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Sharmin Begum
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Luz Aracely Fernandez
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Hayley Gorman
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Kris Chadee
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- * E-mail:
| |
Collapse
|
10
|
Arnold BF, van der Laan MJ, Hubbard AE, Steel C, Kubofcik J, Hamlin KL, Moss DM, Nutman TB, Priest JW, Lammie PJ. Measuring changes in transmission of neglected tropical diseases, malaria, and enteric pathogens from quantitative antibody levels. PLoS Negl Trop Dis 2017; 11:e0005616. [PMID: 28542223 PMCID: PMC5453600 DOI: 10.1371/journal.pntd.0005616] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 06/01/2017] [Accepted: 05/01/2017] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND Serological antibody levels are a sensitive marker of pathogen exposure, and advances in multiplex assays have created enormous potential for large-scale, integrated infectious disease surveillance. Most methods to analyze antibody measurements reduce quantitative antibody levels to seropositive and seronegative groups, but this can be difficult for many pathogens and may provide lower resolution information than quantitative levels. Analysis methods have predominantly maintained a single disease focus, yet integrated surveillance platforms would benefit from methodologies that work across diverse pathogens included in multiplex assays. METHODS/PRINCIPAL FINDINGS We developed an approach to measure changes in transmission from quantitative antibody levels that can be applied to diverse pathogens of global importance. We compared age-dependent immunoglobulin G curves in repeated cross-sectional surveys between populations with differences in transmission for multiple pathogens, including: lymphatic filariasis (Wuchereria bancrofti) measured before and after mass drug administration on Mauke, Cook Islands, malaria (Plasmodium falciparum) before and after a combined insecticide and mass drug administration intervention in the Garki project, Nigeria, and enteric protozoans (Cryptosporidium parvum, Giardia intestinalis, Entamoeba histolytica), bacteria (enterotoxigenic Escherichia coli, Salmonella spp.), and viruses (norovirus groups I and II) in children living in Haiti and the USA. Age-dependent antibody curves fit with ensemble machine learning followed a characteristic shape across pathogens that aligned with predictions from basic mechanisms of humoral immunity. Differences in pathogen transmission led to shifts in fitted antibody curves that were remarkably consistent across pathogens, assays, and populations. Mean antibody levels correlated strongly with traditional measures of transmission intensity, such as the entomological inoculation rate for P. falciparum (Spearman's rho = 0.75). In both high- and low transmission settings, mean antibody curves revealed changes in population mean antibody levels that were masked by seroprevalence measures because changes took place above or below the seropositivity cutoff. CONCLUSIONS/SIGNIFICANCE Age-dependent antibody curves and summary means provided a robust and sensitive measure of changes in transmission, with greatest sensitivity among young children. The method generalizes to pathogens that can be measured in high-throughput, multiplex serological assays, and scales to surveillance activities that require high spatiotemporal resolution. Our results suggest quantitative antibody levels will be particularly useful to measure differences in exposure for pathogens that elicit a transient antibody response or for monitoring populations with very high- or very low transmission, when seroprevalence is less informative. The approach represents a new opportunity to conduct integrated serological surveillance for neglected tropical diseases, malaria, and other infectious diseases with well-defined antigen targets.
Collapse
Affiliation(s)
- Benjamin F. Arnold
- School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Mark J. van der Laan
- School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Alan E. Hubbard
- School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Cathy Steel
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Joseph Kubofcik
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Katy L. Hamlin
- Division of Parasitic Diseases and Malaria, United States Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Delynn M. Moss
- Division of Foodborne, Waterborne, and Environmental Diseases, United States Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Thomas B. Nutman
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Jeffrey W. Priest
- Division of Foodborne, Waterborne, and Environmental Diseases, United States Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Patrick J. Lammie
- Division of Parasitic Diseases and Malaria, United States Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
- Neglected Tropical Diseases Support Center, Task Force for Global Health, Decatur, Georgia, United States of America
| |
Collapse
|
11
|
Abhyankar MM, Noor Z, Tomai MA, Elvecrog J, Fox CB, Petri WA. Nanoformulation of synergistic TLR ligands to enhance vaccination against Entamoeba histolytica. Vaccine 2017; 35:916-922. [PMID: 28089548 PMCID: PMC5301946 DOI: 10.1016/j.vaccine.2016.12.057] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Revised: 11/23/2016] [Accepted: 12/26/2016] [Indexed: 02/07/2023]
Abstract
Diarrheal infectious diseases represent a major cause of global morbidity and mortality. There is an urgent need for vaccines against diarrheal pathogens, especially parasites. Modern subunit vaccines rely on combining a highly purified antigen with an adjuvant to increase their efficacy. In the present study, we evaluated the ability of a nanoliposome adjuvant system to trigger a strong mucosal immune response to the Entamoeba histolytica Gal/GalNAc lectin LecA antigen. CBA/J mice were immunized with alum, emulsion or liposome based formulations containing synthetic TLR agonists. A liposome formulation containing TLR4 and TLR7/8 agonists was selected based on its ability to generate intestinal IgA, plasma IgG2a/IgG1, IFN-γ and IL-17A. Immunization with a mucosal prime followed by a parenteral boost generated a high mucosal IgA response that inhibited adherence of parasites to mammalian cells. Inclusion of the immune potentiator all-trans retinoic acid in the regimen further improved the mucosal IgA response. Immunization protected from infection with up to 55% efficacy. Our results show that a nanoliposome delivery system containing TLR agonists is a promising prospect for the development of vaccines against enteric pathogens, especially when a multifaceted immune response is desired.
Collapse
Affiliation(s)
- Mayuresh M Abhyankar
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, United States.
| | - Zannatun Noor
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, United States
| | - Mark A Tomai
- 3M Drug Delivery Systems, 3M Center, 275-3E-10, St Paul, MN 55144, USA
| | - James Elvecrog
- 3M Drug Delivery Systems, 3M Center, 275-3E-10, St Paul, MN 55144, USA
| | - Christopher B Fox
- IDRI, Seattle, WA, USA; Department of Global Health, University of Washington, Seattle, WA 98104, USA
| | - William A Petri
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, United States.
| |
Collapse
|
12
|
Dynamic Interactions of a Conserved Enterotoxigenic Escherichia coli Adhesin with Intestinal Mucins Govern Epithelium Engagement and Toxin Delivery. Infect Immun 2016; 84:3608-3617. [PMID: 27736776 PMCID: PMC5116737 DOI: 10.1128/iai.00692-16] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 10/02/2016] [Indexed: 12/30/2022] Open
Abstract
At present, there is no vaccine for enterotoxigenic Escherichia coli (ETEC), an important cause of diarrheal illness. Nevertheless, recent microbial pathogenesis studies have identified a number of molecules produced by ETEC that contribute to its virulence and are novel antigenic targets to complement canonical vaccine approaches. EtpA is a secreted two-partner adhesin that is conserved within the ETEC pathovar. EtpA interacts with the tips of ETEC flagella to promote bacterial adhesion, toxin delivery, and intestinal colonization by forming molecular bridges between the bacteria and the epithelial surface. However, the nature of EtpA interactions with the intestinal epithelium remains poorly defined. Here, we demonstrate that EtpA interacts with glycans presented by transmembrane and secreted intestinal mucins at epithelial surfaces to facilitate pathogen-host interactions that culminate in toxin delivery. Moreover, we found that a major effector molecule of ETEC, the heat-labile enterotoxin (LT), may enhance these interactions by stimulating the production of the gel-forming mucin MUC2. Our studies suggest, however, that EtpA participates in complex and dynamic interactions between ETEC and the gastrointestinal mucosae in which host glycoproteins promote bacterial attachment while simultaneously limiting the epithelial engagement required for effective toxin delivery. Collectively, these data provide additional insight into the intricate nature of ETEC interactions with the intestinal epithelium that have potential implications for rational approaches to vaccine design.
Collapse
|
13
|
Singh RS, Walia AK, Kanwar JR, Kennedy JF. Amoebiasis vaccine development: A snapshot on E. histolytica with emphasis on perspectives of Gal/GalNAc lectin. Int J Biol Macromol 2016; 91:258-68. [DOI: 10.1016/j.ijbiomac.2016.05.043] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 05/10/2016] [Accepted: 05/11/2016] [Indexed: 01/10/2023]
|
14
|
|
15
|
Watanabe K, Petri WA. Molecular biology research to benefit patients with Entamoeba histolytica infection. Mol Microbiol 2015; 98:208-17. [PMID: 26173474 DOI: 10.1111/mmi.13131] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/13/2015] [Indexed: 12/28/2022]
Abstract
The development of molecular microbiology has made it possible for us to deepen our understanding of the pathogenesis of amebiasis. Research using the trophozoite form of Entamoeba histolytica has clearly shown us the importance of the interface between the parasite and host cells in vitro. Immuno-pathogenesis after excystation was similarly well advanced by the use of a novel murine model of amebic colitis. However, it is still challenging to apply these findings to clinical and epidemiological settings. This is mainly because of the lack of a complete infection animal model of amebiasis by oral-fecal infection. Moreover, in vitro experiments have predominantly been performed using the same axenic cultured strain HM-1: IMSS isolated about 50 years ago, whereas highly diverse strains are prevalent all over the world. Translational research informed by clinical observations has the greatest potential for the development of effective interventions. Here, we highlight discoveries of the experiments designed from cohort observation and discuss remaining problems to be solved.
Collapse
Affiliation(s)
- Koji Watanabe
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, USA.,AIDS Clinical Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - William A Petri
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
16
|
Ralston KS. Chew on this: amoebic trogocytosis and host cell killing by Entamoeba histolytica. Trends Parasitol 2015; 31:442-52. [PMID: 26070402 DOI: 10.1016/j.pt.2015.05.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Revised: 05/04/2015] [Accepted: 05/05/2015] [Indexed: 01/23/2023]
Abstract
Entamoeba histolytica was named 'histolytica' (from histo-, 'tissue'; lytic-, 'dissolving') for its ability to destroy host tissues. Direct killing of host cells by the amoebae is likely to be the driving factor that underlies tissue destruction, but the mechanism was unclear. We recently showed that, after attaching to host cells, amoebae bite off and ingest distinct host cell fragments, and that this contributes to cell killing. We review this process, termed 'amoebic trogocytosis' (trogo-, 'nibble'), and how this process interplays with phagocytosis, or whole cell ingestion, in this organism. 'Nibbling' processes have been described in other microbes and in multicellular organisms. The discovery of amoebic trogocytosis in E. histolytica may also shed light on an evolutionarily conserved process for intercellular exchange.
Collapse
Affiliation(s)
- Katherine S Ralston
- Department of Microbiology and Molecular Genetics, University of California, Davis, CA 95616, USA.
| |
Collapse
|
17
|
Protection against Amoebic Liver Abscess in Hamster by Intramuscular Immunization with an Autographa californica Baculovirus Driving the Expression of the Gal-Lectin LC3 Fragment. BIOMED RESEARCH INTERNATIONAL 2015; 2015:760598. [PMID: 26090442 PMCID: PMC4452260 DOI: 10.1155/2015/760598] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 09/23/2014] [Indexed: 12/17/2022]
Abstract
In a previous study, we demonstrated that oral immunization using Autographa californica baculovirus driving the expression of the Gal-lectin LC3 fragment (AcNPV-LC3) of Entamoeba histolytica conferred protection against ALA development in hamsters. In this study, we determined the ability of AcNPV-LC3 to protect against ALA by the intramuscular route as well as the liver immune response associated with protection. Results showed that 55% of hamsters IM immunized with AcNPV-LC3 showed sterile protection against ALA, whereas other 20% showed reduction in the size and extent of abscesses, resulting in some protection in 75% of animals compared to the sham control group. Levels of protection showed a linear correlation with the development and intensity of specific antiamoeba cellular and humoral responses, evaluated in serum and spleen of hamsters, respectively. Evaluation of the Th1/Th2 cytokine patterns expressed in the liver of hamsters showed that sterile protection was associated with the production of high levels of IFNγ and IL-4. These results suggest that the baculovirus system is equally efficient by the intramuscular as well as the oral routes for ALA protection and that the Gal-lectin LC3 fragment is a highly protective antigen against hepatic amoebiasis through the local induction of IFNγ and IL-4.
Collapse
|
18
|
Aguirre García M, Gutiérrez-Kobeh L, López Vancell R. Entamoeba histolytica: adhesins and lectins in the trophozoite surface. Molecules 2015; 20:2802-15. [PMID: 25671365 PMCID: PMC6272351 DOI: 10.3390/molecules20022802] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Accepted: 01/13/2015] [Indexed: 01/06/2023] Open
Abstract
Entamoeba histolytica is the causative agent of amebiasis in humans and is responsible for 100,000 deaths annually, making it the third leading cause of death due to a protozoan parasite. Pathogenesis appears to result from the potent cytotoxic activity of the parasite, which kills host cells within minutes. Although the mechanism is unknown, it is well established to be contact-dependent. The life cycle of the parasite alternates with two forms: the resistant cyst and the invasive trophozoite. The adhesive interactions between the parasite and surface glycoconjugates of host cells, as well as those lining the epithelia, are determinants for invasion of human tissues, for its cytotoxic activity, and finally for the outcome of the disease. In this review we present an overview of the information available on the amebic lectins and adhesins that are responsible of those adhesive interactions and we also refer to their effect on the host immune response. Finally, we present some concluding remarks and perspectives in the field.
Collapse
Affiliation(s)
- Magdalena Aguirre García
- Departmento de Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Dr. Balmis #148, Col. Doctores, C.P. 06726 Mexico, D.F., Mexico.
| | - Laila Gutiérrez-Kobeh
- Departmento de Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Dr. Balmis #148, Col. Doctores, C.P. 06726 Mexico, D.F., Mexico.
| | - Rosario López Vancell
- Departmento de Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Dr. Balmis #148, Col. Doctores, C.P. 06726 Mexico, D.F., Mexico.
| |
Collapse
|
19
|
Immunization with the Entamoeba histolytica surface metalloprotease EhMSP-1 protects hamsters from amebic liver abscess. Infect Immun 2014; 83:713-20. [PMID: 25452550 DOI: 10.1128/iai.02490-14] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Diarrhea and amebic liver abscesses due to invasive Entamoeba histolytica infections are an important cause of morbidity and mortality in the developing world. Entamoeba histolytica adherence and cell migration, two phenotypes linked to virulence, are both aberrant in trophozoites deficient in the metallosurface protease EhMSP-1, which is a homologue of the Leishmania vaccine candidate leishmanolysin (GP63). We examined the potential of EhMSP-1 for use as a vaccine antigen to protect against amebic liver abscesses. First, existing serum samples from South Africans naturally infected with E. histolytica were examined by enzyme-linked immunosorbent assay (ELISA) for the presence of EhMSP-1-specific IgG. Nine of 12 (75%) people with anti-E. histolytica IgG also had EhMSP-1-specific IgG antibodies. We next used a hamster model of amebic liver abscess to determine the effect of immunization with a mixture of four recombinant EhMSP-1 protein fragments. EhMSP-1 immunization stimulated a robust IgG antibody response. Furthermore, EhMSP-1 immunization of hamsters reduced development of severe amebic liver abscesses following intrahepatic injection of E. histolytica by a combined rate of 68% in two independent animal experiments. Purified IgG from immunized compared to control animals bound to the surface of E. histolytica trophozoites and accelerated amebic lysis via activation of the classical complement cascade. We concluded that EhMSP-1 is a promising antigen that warrants further study to determine its full potential as a target for therapy and/or prevention of invasive amebiasis.
Collapse
|
20
|
Mortimer L, Moreau F, Cornick S, Chadee K. Gal-lectin-dependent contact activates the inflammasome by invasive Entamoeba histolytica. Mucosal Immunol 2014; 7:829-41. [PMID: 24253103 DOI: 10.1038/mi.2013.100] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Revised: 10/16/2013] [Accepted: 10/21/2013] [Indexed: 02/04/2023]
Abstract
Entamoeba histolytica (Eh) is an extracellular protozoan parasite of the human colon, which occasionally breaches the intestinal barrier. Eradicating ameba that invades is essential for host survival. A defining but uncharacterized feature of amebic invasion is direct contact between ameba and host cells. This event corresponds with a massive pro-inflammatory response. To date, pathogen recognition receptors (PRRs) that are activated by contact with viable Eh are unknown. Here we show that the innate immune system responds in a qualitatively different way to contact with viable Eh vs. soluble ligands produced by viable or dead ameba. This unique Eh Gal-lectin contact-dependent response in macrophages was mediated by activation of the inflammasome. Soluble native Gal-lectin did not induce inflammasome activation, but was sufficient for transcriptional priming of the inflammasome and non-inflammasome-dependent pro-inflammatory cytokine release. We conclude the inflammasome is a pathogenicity sensor for invasive Eh and identify for the first time a PRR that specifically responds to contact with intact parasites in a manner that accords with scale immune response to parasite invasion.
Collapse
Affiliation(s)
- L Mortimer
- Faculty of Medicine, Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Gastrointestinal Research Group, University of Calgary, Calgary, Alberta, Canada
| | - F Moreau
- Faculty of Medicine, Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Gastrointestinal Research Group, University of Calgary, Calgary, Alberta, Canada
| | - S Cornick
- Faculty of Medicine, Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Gastrointestinal Research Group, University of Calgary, Calgary, Alberta, Canada
| | - K Chadee
- Faculty of Medicine, Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Gastrointestinal Research Group, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
21
|
Moss DM, Priest JW, Hamlin K, Derado G, Herbein J, Petri WA, Lammie PJ. Longitudinal evaluation of enteric protozoa in Haitian children by stool exam and multiplex serologic assay. Am J Trop Med Hyg 2014; 90:653-60. [PMID: 24591430 DOI: 10.4269/ajtmh.13-0545] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Haitian children were monitored longitudinally in a filariasis study. Included were stool samples examined for Giardia intestinalis and Entamoeba histolytica cysts, and serum specimens analyzed for immunoglobulin G (IgG) responses to eight recombinant antigens from G. intestinalis (variant-specific surface protein [VSP1-VSP5]), E. histolytica (lectin adhesion molecule [LecA]), and Cryptosporidium parvum (17- and 27-kDa) using a multiplex bead assay. The IgG responses to VSP antigens peaked at 2 years of age and then diminished and were significantly lower (P < 0.002) in children > 4.5 years than in children < 4.5 years. The IgG responses to Cryptosporidium tended to increase with age. The IgG responses to LecA and VSP antigens and the prevalence of stools positive for cysts were significantly higher (P < 0.037 and P < 0.035, respectively) in the rainy season than in the dry season. The multiplex bead assay provides a powerful tool for analyzing serologic responses to multiple pathogens.
Collapse
Affiliation(s)
- Delynn M Moss
- Division of Foodborne, Waterborne, and Environmental Diseases, National Center for Emerging and Zoonotic Infectious Diseases, Division of Parasitic Diseases and Malaria, National Center for Global Health, Centers for Disease Control and Prevention, Atlanta, Georgia; Departments of Medicine, Microbiology, and Pathology, University of Virginia, and TECHLAB, Inc., Blacksburg, Virginia
| | | | | | | | | | | | | |
Collapse
|
22
|
Quach J, St-Pierre J, Chadee K. The future for vaccine development against Entamoeba histolytica. Hum Vaccin Immunother 2014; 10:1514-21. [PMID: 24504133 DOI: 10.4161/hv.27796] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Entamoeba histolytica is the causative agent of amebiasis, one of the top three parasitic causes of mortality worldwide. In the majority of infected individuals, E. histolytica asymptomatically colonizes the large intestine, while in others, the parasite breaches the mucosal epithelial barrier to cause amebic colitis and can disseminate to soft organs to cause abscesses. Vaccinations using native and recombinant forms of the parasite Gal-lectin have been successful in protecting animals against intestinal amebiasis and amebic liver abscess. Protection against amebic liver abscesses has also been reported by targeting other E. histolytica components including the serine-rich protein and the 29-kDa-reductase antigen. To date, vaccines against the Gal-lectin hold the most promise but clinical trials will be required to validate its efficacy in humans. Here, we review the current strategies and future perspectives involved in the development of a vaccine against E. histolytica.
Collapse
Affiliation(s)
- Jeanie Quach
- Faculty of Medicine; Department of Microbiology, Immunology, and Infectious Diseases; Snyder Institute for Chronic Diseases; Gastrointestinal Research Group; University of Calgary; Calgary, AB Canada
| | - Joëlle St-Pierre
- Faculty of Medicine; Department of Microbiology, Immunology, and Infectious Diseases; Snyder Institute for Chronic Diseases; Gastrointestinal Research Group; University of Calgary; Calgary, AB Canada
| | - Kris Chadee
- Faculty of Medicine; Department of Microbiology, Immunology, and Infectious Diseases; Snyder Institute for Chronic Diseases; Gastrointestinal Research Group; University of Calgary; Calgary, AB Canada
| |
Collapse
|
23
|
Mahdavi J, Pirinccioglu N, Oldfield NJ, Carlsohn E, Stoof J, Aslam A, Self T, Cawthraw SA, Petrovska L, Colborne N, Sihlbom C, Borén T, Wooldridge KG, Ala'Aldeen DAA. A novel O-linked glycan modulates Campylobacter jejuni major outer membrane protein-mediated adhesion to human histo-blood group antigens and chicken colonization. Open Biol 2014; 4:130202. [PMID: 24451549 PMCID: PMC3909276 DOI: 10.1098/rsob.130202] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Campylobacter jejuni is an important cause of human foodborne gastroenteritis; strategies to prevent infection are hampered by a poor understanding of the complex interactions between host and pathogen. Previous work showed that C. jejuni could bind human histo-blood group antigens (BgAgs) in vitro and that BgAgs could inhibit the binding of C. jejuni to human intestinal mucosa ex vivo. Here, the major flagella subunit protein (FlaA) and the major outer membrane protein (MOMP) were identified as BgAg-binding adhesins in C. jejuni NCTC11168. Significantly, the MOMP was shown to be O-glycosylated at Thr268; previously only flagellin proteins were known to be O-glycosylated in C. jejuni. Substitution of MOMP Thr268 led to significantly reduced binding to BgAgs. The O-glycan moiety was characterized as Gal(β1–3)-GalNAc(β1–4)-GalNAc(β1–4)-GalNAcα1-Thr268; modelling suggested that O-glycosylation has a notable effect on the conformation of MOMP and this modulates BgAg-binding capacity. Glycosylation of MOMP at Thr268 promoted cell-to-cell binding, biofilm formation and adhesion to Caco-2 cells, and was required for the optimal colonization of chickens by C. jejuni, confirming the significance of this O-glycosylation in pathogenesis.
Collapse
Affiliation(s)
- Jafar Mahdavi
- School of Life Sciences, University of Nottingham, Nottingham NG7 2RD, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Shimokawa C, Culleton R, Imai T, Suzue K, Hirai M, Taniguchi T, Kobayashi S, Hisaeda H, Hamano S. Species-specific immunity induced by infection with Entamoeba histolytica and Entamoeba moshkovskii in mice. PLoS One 2013; 8:e82025. [PMID: 24312397 PMCID: PMC3843725 DOI: 10.1371/journal.pone.0082025] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 10/24/2013] [Indexed: 11/18/2022] Open
Abstract
Entamoeba histolytica, the parasitic amoeba responsible for amoebiasis, causes approximately 100,000 deaths every year. There is currently no vaccine against this parasite. We have previously shown that intracecal inoculation of E. histolytica trophozoites leads to chronic and non-healing cecitis in mice. Entamoeba moshkovskii, a closely related amoeba, also causes diarrhea and other intestinal disorders in this model. Here, we investigated the effect of infection followed by drug-cure of these species on the induction of immunity against homologous or heterologous species challenge. Mice were infected with E. histolytica or E. moshkovskii and treated with metronidazole 14 days later. Re-challenge with E. histolytica or E. moshkovskii was conducted seven or 28 days following confirmation of the clearance of amoebae, and the degree of protection compared to non-exposed control mice was evaluated. We show that primary infection with these amoebae induces a species-specific immune response which protects against challenge with the homologous, but not a heterologous species. These findings pave the way, therefore, for the identification of novel amoebae antigens that may become the targets of vaccines and provide a useful platform to investigate host protective immunity to Entamoeba infections.
Collapse
Affiliation(s)
| | - Richard Culleton
- Malaria Unit, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
| | - Takashi Imai
- Department of Parasitology, Graduate School of Medicine, Gunma University, Gunma, Japan
| | - Kazutomo Suzue
- Department of Parasitology, Graduate School of Medicine, Gunma University, Gunma, Japan
| | - Makoto Hirai
- Department of Parasitology, Graduate School of Medicine, Gunma University, Gunma, Japan
| | - Tomoyo Taniguchi
- Department of Parasitology, Graduate School of Medicine, Gunma University, Gunma, Japan
| | - Seiki Kobayashi
- Department of Tropical Medicine and Parasitology, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Hajime Hisaeda
- Department of Parasitology, Graduate School of Medicine, Gunma University, Gunma, Japan
| | - Shinjiro Hamano
- Department of Parasitology, Nagasaki University, Nagasaki, Japan
- Global COE Program, Nagasaki University, Nagasaki, Japan
- * E-mail:
| |
Collapse
|
25
|
Kaur U, Khurana S, Saikia UN, Dubey ML. Immunogenicity and protective efficacy of heparan sulphate binding proteins of Entamoeba histolytica in a guinea pig model of intestinal amoebiasis. Exp Parasitol 2013; 135:486-96. [PMID: 24007700 DOI: 10.1016/j.exppara.2013.08.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Revised: 08/07/2013] [Accepted: 08/12/2013] [Indexed: 10/26/2022]
Abstract
Entamoeba histolytica infection is associated with considerable morbidity and mortality in the form of intestinal and extraintestinal amoebiasis. No vaccine is yet available for amoebiasis. Heparan Sulphate Binding Proteins (HSBPs) from E. histolytica were evaluated for immunogenicity and protective efficacy in a Guinea pig model. Animals were immunized subcutaneously with 30μg of HSBP by three weekly inoculations. The immunogenicity of HSBP was determined by antibody response (IgG, IgM and IgA), splenocyte proliferation assay and in vitro direct amoebicidal assay with splenic lymphocytes and monocytes from vaccinated and control animals. The efficacy of the vaccine was evaluated by challenge infection to vaccinated and control animals by intra-caecal inoculation of E. histolytica trophozoites and comparing gross and histopathological findings in caeca of these animals. HSBP was found to induce specific anti-amoebic response as seen by specific antibody production and direct amoebicidal activity of splenocytes. The vaccine also showed partial protection against challenge infection in vaccinated animals as shown by mild/absent lesions and histopathological findings.
Collapse
Affiliation(s)
- Upninder Kaur
- Departments of Parasitology, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | | | | | | |
Collapse
|
26
|
New concepts in diagnostics for infectious diarrhea. Mucosal Immunol 2013; 6:876-85. [PMID: 23881355 DOI: 10.1038/mi.2013.50] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2013] [Accepted: 06/11/2013] [Indexed: 02/04/2023]
Abstract
Conventional approaches to the diagnosis of infectious diarrhea must include several modalities to detect an array of potential viruses, bacteria, and parasites. We will provide a general overview of the wide range of diagnostic modalities available for enteropathogens, briefly discuss some of the limitations of conventional methods, and then focus on new molecular methods, including real-time PCR and next-generation sequencing. In particular, we will discuss quantitation of pathogen load with these techniques. We will then describe examples whereby novel diagnostics may help illuminate the etiology of infectious diarrhea, where they may not, and how they may benefit studies of immunity to enteric infections.
Collapse
|
27
|
Affiliation(s)
- Shannon N. Moonah
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia Health System, Charlottesville, Virginia, United States of America
| | - Nona M. Jiang
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia Health System, Charlottesville, Virginia, United States of America
| | - William A. Petri
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia Health System, Charlottesville, Virginia, United States of America
- * E-mail:
| |
Collapse
|
28
|
Barroso L, Abhyankar M, Noor Z, Read K, Pedersen K, White R, Fox C, Petri WA, Lyerly D. Expression, purification, and evaluation of recombinant LecA as a candidate for an amebic colitis vaccine. Vaccine 2013; 32:1218-24. [PMID: 23827311 DOI: 10.1016/j.vaccine.2013.06.056] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Revised: 06/06/2013] [Accepted: 06/19/2013] [Indexed: 11/25/2022]
Abstract
Entamoeba histolytica, which causes amebic colitis and liver abscess, is considered a major enteric pathogen in residents and travelers to developing countries where the disease is endemic. Interaction of this protozoan parasite with the intestine is mediated through the binding of the trophozoite stage to intestinal mucin and epithelium via a galactose and N-acetyl-d-galactosamine (Gal/GalNAc) lectin comprised of a disulfide linked heavy (ca. 180 kDa) and light chain (ca. 35 kDa) and a noncovalently bound intermediate subunit (ca. 150 kDa). Our efforts to develop a vaccine against this pathogen have focused on an internal 578 amino acid fragment, designated LecA, located within the cysteine-rich region of the heavy chain subunit because: (i) it is a major target of adherence-blocking antibodies of seropositive individuals and (ii) vaccination with his-tagged LecA provides protection in animal models. We developed a purification process for preparing highly purified non-tagged LecA using a codon-optimized gene expressed in Escherichia coli. The process consisted of: (i) cell lysis, collection and washing of inclusion bodies; (ii) solubilization and refolding of denatured LecA; and (iii) a polishing gel filtration step. The purified fragment existed primarily as a random coil with β-sheet structure, contained low endotoxin and nucleic acid, was highly immunoreactive, and elicited antibodies that recognized native lectin and that inhibited in vitro adherence of trophozoites to CHO cells. Immunization of CBA mice with LecA resulted in significant protection against cecal colitis. Our procedure yields sufficient amounts of highly purified LecA for future studies on stability, immunogenicity, and protection with protein-adjuvant formulations.
Collapse
Affiliation(s)
- L Barroso
- TECHLAB, Inc., 2001 Kraft Drive, Blacksburg, VA 24060-6158, USA
| | - M Abhyankar
- Division of Infectious Diseases and International Health University of Virginia Health System, Carter Harrison Building, Room 1709A, 345 Crispell Drive, P.O. Box 801340, Charlottesville, VA 22908-1340, USA
| | - Z Noor
- Division of Infectious Diseases and International Health University of Virginia Health System, Carter Harrison Building, Room 1709A, 345 Crispell Drive, P.O. Box 801340, Charlottesville, VA 22908-1340, USA
| | - K Read
- TECHLAB, Inc., 2001 Kraft Drive, Blacksburg, VA 24060-6158, USA
| | - K Pedersen
- TECHLAB, Inc., 2001 Kraft Drive, Blacksburg, VA 24060-6158, USA
| | - R White
- TECHLAB, Inc., 2001 Kraft Drive, Blacksburg, VA 24060-6158, USA
| | - C Fox
- Infectious Disease Research Institute, Seattle, WA, USA
| | - W A Petri
- Division of Infectious Diseases and International Health University of Virginia Health System, Carter Harrison Building, Room 1709A, 345 Crispell Drive, P.O. Box 801340, Charlottesville, VA 22908-1340, USA
| | - D Lyerly
- TECHLAB, Inc., 2001 Kraft Drive, Blacksburg, VA 24060-6158, USA.
| |
Collapse
|
29
|
References. Parasitology 2012. [DOI: 10.1002/9781119968986.refs] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
30
|
Abd Alla MD, Wolf R, White GL, Kosanke SD, Cary D, Verweij JJ, Zhang MJ, Ravdin JI. Efficacy of a Gal-lectin subunit vaccine against experimental Entamoeba histolytica infection and colitis in baboons (Papio sp.). Vaccine 2012; 30:3068-75. [PMID: 22406457 DOI: 10.1016/j.vaccine.2012.02.066] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Revised: 02/03/2012] [Accepted: 02/25/2012] [Indexed: 11/30/2022]
Abstract
To determine the efficacy of a Gal-lectin based intranasal synthetic peptide vaccine, we developed a new experimental primate model of Entamoeba histolytica intestinal infection. Release of xenic E. histolytica trophozoites (5×10(6)) into the small bowel of baboons (Papio sp.) resulted in a rapid intestinal anti-amebic antibody response and a brief infection; however, release of trophozoites directly into the cecum (5 baboons) elicited a sustained E. histolytica infection, as determined by quantitative fecal PCR, and an ulcerative, inflammatory colitis observed on colonoscopy and histopathology. In three controlled experiments, baboons received four immunizations at seven day intervals of 1600 μg of the vaccine/nostril, with Cholera toxin, 20 μg/nostril as adjuvant; vaccinated (n=6) and control baboons (n=6) baboons were then challenged via colonoscopy with xenic trophozoites (5×10(6)). During 90 days of follow up, 250 of 415 (60.24%) fecal samples in control baboons had a (+) PCR for E. histolytica, compared to only 36 of 423 (8.51%) samples from vaccinated baboons (P<0.001). All 6 vaccinated baboons were free of infection by the 51st day after challenge, 5 of 6 controls positive had (+) fecal PCRs for up to 126 days post-challenge (P=0.019). Inflammatory colitis developed in 4 of 6 control baboons post-challenge, with invasive E. histolytica trophozoites present in 2 of the 4 on histopathology. There was no evidence of inflammatory colitis or parasite invasion in any of the vaccinated baboons; there was a strong inverse correlation between positive ELISA OD value indicating the presence of intestinal anti-peptide IgA antibodies and baboons having a positive fecal PCR CT value, P<0.001. In conclusion, we developed a novel primate model of E. histolytica intestinal infection and demonstrated that a Gal-lectin-based intranasal synthetic peptide vaccine was highly efficacious in preventing experimental E. histolytica infection and colitis in baboons.
Collapse
|
31
|
Meneses-Ruiz DM, Laclette JP, Aguilar-Díaz H, Hernández-Ruiz J, Luz-Madrigal A, Sampieri A, Vaca L, Carrero JC. Mucosal delivery of ACNPV baculovirus driving expression of the Gal-lectin LC3 fragment confers protection against amoebic liver abscess in hamster. Int J Biol Sci 2011; 7:1345-56. [PMID: 22110386 PMCID: PMC3221370 DOI: 10.7150/ijbs.7.1345] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Accepted: 10/01/2011] [Indexed: 12/23/2022] Open
Abstract
Mucosal vaccination against amoebiasis using the Gal-lectin of E. histolytica has been proposed as one of the leading strategies for controlling this human disease. However, most mucosal adjuvants used are toxic and the identification of safe delivery systems is necessary. Here, we evaluate the potential of a recombinant Autographa californica baculovirus driving the expression of the LC3 fragment of the Gal-lectin to confer protection against amoebic liver abscess (ALA) in hamsters following oral or nasal immunization. Hamsters immunized by oral route showed complete absence (57.9%) or partial development (21%) of ALA, resulting in some protection in 78.9% of animals when compared with the wild type baculovirus and sham control groups. In contrast, nasal immunization conferred only 21% of protection efficacy. Levels of ALA protection showed lineal correlation with the development of an anti-amoebic cellular immune response evaluated in spleens, but not with the induction of seric IgG anti-amoeba antibodies. These results suggest that baculovirus driving the expression of E. histolytica vaccine candidate antigens is useful for inducing protective cellular and humoral immune responses following oral immunization, and therefore it could be used as a system for mucosal delivery of an anti-amoebic vaccine.
Collapse
Affiliation(s)
- D M Meneses-Ruiz
- Department of Immunology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México. A.P. 70228, México D.F., México
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Weedall GD, Sherrington J, Paterson S, Hall N. Evidence of gene conversion in genes encoding the Gal/GalNac lectin complex of Entamoeba. PLoS Negl Trop Dis 2011; 5:e1209. [PMID: 21738808 PMCID: PMC3125142 DOI: 10.1371/journal.pntd.0001209] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2010] [Accepted: 05/01/2011] [Indexed: 01/29/2023] Open
Abstract
The human gut parasite Entamoeba histolytica, uses a lectin complex on its cell surface to bind to mucin and to ligands on the intestinal epithelia. Binding to mucin is necessary for colonisation and binding to intestinal epithelia for invasion, therefore blocking this binding may protect against amoebiasis. Acquired protective immunity raised against the lectin complex should create a selection pressure to change the amino acid sequence of lectin genes in order to avoid future detection. We present evidence that gene conversion has occurred in lineages leading to E. histolytica strain HM1:IMSS and E. dispar strain SAW760. This evolutionary mechanism generates diversity and could contribute to immune evasion by the parasites. Gene conversion is a process of recombination that can generate diversity among genes. Gene conversion occurs in some pathogenic species of protozoa to generate diversity among gene families encoding important antigens. The process may contribute to immune evasion by the parasites. Gene conversion, or indeed recombination of any kind, has not previously been demonstrated in human intestinal parasites of the genus Entamoeba. Here, we analysed genes encoding members of an important antigenic protein complex on the surface of Entamoeba parasites which is involved in invasion of the intestinal wall. Three gene families encode heavy-, light- and intermediate-subunits of the complex. We estimated genetic divergence between related genes from two species of Entamoeba, E. histolytica and E. dispar, and compared them to divergence among neighbouring genes and to the average across the whole genome, initially looking for evidence that the genes were evolving under positive selection. However, instead we saw patterns of genetic difference between some of the light- and intermediate-subunit genes indicating the action of gene conversion among members of these gene families. This indicates that recombinational mechanisms may play a part in the molecular evolution of these parasites.
Collapse
Affiliation(s)
- Gareth D. Weedall
- Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
- * E-mail:
| | - James Sherrington
- Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Steve Paterson
- Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Neil Hall
- Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
33
|
Guo X, Barroso L, Lyerly DM, Petri WA, Houpt ER. CD4+ and CD8+ T cell- and IL-17-mediated protection against Entamoeba histolytica induced by a recombinant vaccine. Vaccine 2010; 29:772-7. [PMID: 21095257 DOI: 10.1016/j.vaccine.2010.11.013] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2010] [Revised: 11/02/2010] [Accepted: 11/06/2010] [Indexed: 01/17/2023]
Abstract
Amebiasis in the murine model can be prevented by vaccination with the Gal/GalNAc lectin through a T cell-dependent mechanism. In this work we further decipher the mechanism of this protection. Mice vaccinated with the recombinant "LecA" fragment of the Gal/GalNAc lectin with alum were capable of transferring protection to naïve recipients by both CD4+ T cells and surprisingly CD8+ T cells. We then examined the cytokine profile of these cells. CD4+ T cells from PBMC of LecA-alum vaccinated mice were observed to be a major source of IFN-γ, known to be a protective cytokine with this vaccine. In contrast, CD8+ T cells produced relatively little IFN-γ but more IL-17 than the CD4 compartment. We thus examined the role of IL-17 in vaccine mediated protection and found through neutralization experiments that this cytokine contributed to LecA-alum vaccine protection. In addition we examined whether these cells exhibited direct amebicidal activity in vitro and found that both populations had amebicidal activity at high concentrations (1000:1) but CD8+ T cells appeared more potent, capable of cytotoxicity at a 100:1 ratio. In conclusion, both CD4 and CD8 T cells exert protection with this amebiasis vaccine. The mechanism of CD8 T cell-mediated protection may include direct amebicidal activity and/or IL-17 production. Both IL-17 and IFN-γ are useful surrogates for immune protection.
Collapse
Affiliation(s)
- Xiaoti Guo
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | | | | | | | | |
Collapse
|
34
|
Carrero JC, Contreras-Rojas A, Sánchez-Hernández B, Petrosyan P, Bobes RJ, Ortiz-Ortiz L, Laclette JP. Protection against murine intestinal amoebiasis induced by oral immunization with the 29 kDa antigen of Entamoeba histolytica and cholera toxin. Exp Parasitol 2010; 126:359-65. [PMID: 20303954 DOI: 10.1016/j.exppara.2010.03.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2009] [Revised: 03/05/2010] [Accepted: 03/12/2010] [Indexed: 10/19/2022]
Abstract
Entamoeba histolytica antigens recognized by salivary IgA from infected patients include the 29 kDa antigen (Eh29), an alkyl hydroperoxide reductase. Here, we investigate the potential of recombinant Eh29 and an Eh29-cholera toxin subunit B (CTxB) fusion protein to confer protection against intestinal amoebiasis after oral immunization. The purified Eh29-CTxB fusion retained the critical ability to bind ganglioside GM(1), as determined by ELISA. Oral immunization of C3H/HeJ mice with Eh29 administered in combination with a subclinical dose of whole cholera toxin, but not as an Eh29-CTxB fusion, induced elevated levels of intestinal IgA and serum IgG anti-Eh29 antibodies that inhibited trophozoites adherence to MDCK cell monolayers. The 80% of immunized mice seen to develop IgA and IgG immune responses showed no evidence of infection in tissue sections harvested following intracecal challenge with virulent E. histolytica trophozoites. These results suggest that Eh29 is capable of inducing protective anti-amoebic immune responses in mice following oral immunization and could be used in the development of oral vaccines against amoebiasis.
Collapse
Affiliation(s)
- J C Carrero
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, A.P. 70228, 04510 México D.F., Mexico.
| | | | | | | | | | | | | |
Collapse
|
35
|
Mortimer L, Chadee K. The immunopathogenesis of Entamoeba histolytica. Exp Parasitol 2010; 126:366-80. [PMID: 20303955 DOI: 10.1016/j.exppara.2010.03.005] [Citation(s) in RCA: 137] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2009] [Revised: 03/08/2010] [Accepted: 03/12/2010] [Indexed: 12/17/2022]
Abstract
Amebiasis is the disease caused by the enteric dwelling protozoan parasite Entamoeba histolytica. The WHO considers amebiasis as one of the major health problems in developing countries; it is surpassed by only malaria and schistosomiasis for death caused by parasitic infection. E. histolytica primarily lives in the colon as a harmless commensal, but is capable of causing devastating dysentery, colitis and liver abscess. What triggers the switch to a pathogenic phenotype and the onset of disease is unknown. We are becoming increasingly aware of the complexity of the host-parasite interaction. During chronic stages of amebiasis, the host develops an immune response that is incapable of eliminating tissue resident parasites, while the parasite actively immunosuppresses the host. However, most individuals with symptomatic infections succumb only to an episode of dysentery. Why most halt invasion and a minority progress to chronic disease remains poorly understood. This review presents a current understanding of the immune processes that shape the outcome of E. histolytica infections during its different stages.
Collapse
Affiliation(s)
- Leanne Mortimer
- Faculty of Medicine, Department of Microbiology and Infectious Diseases, University of Calgary Health Sciences Centre, 3330 Hospital Dr. NW, Calgary, Alberta, Canada
| | | |
Collapse
|
36
|
Interaction of zinc or vitamin A supplementation and specific parasite infections on Mexican infants' growth: a randomized clinical trial. Eur J Clin Nutr 2009; 63:1176-84. [PMID: 19623197 DOI: 10.1038/ejcn.2009.53] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND The efficacy of micronutrient supplementation on growth may be modified by specific gastrointestinal parasite infections. METHODS We carried out a double-blind placebo-controlled trial to evaluate the effect of vitamin A and zinc supplementation on gastro-intestinal pathogen infections and growth among 584 infants in Mexico City. Children aged 5-15 months were assigned to receive either a vitamin A supplement every 2 months (20,000 IU of retinol for infants < or =; 1 year or 45,000 IU for infants >1 year), a daily supplement of 20 mg of zinc, a combined vitamin A-zinc supplement or a placebo, and were followed up for 1 year. Weight and length were measured once a month and morbidity histories were recorded twice a week for 12 months. Monthly stool samples were screened for Giardia duodenalis, Ascaris lumbricoides and Entamoeba spp. Growth velocity slopes, generated from the linear regression of individual child length, and height-for-age z-scores on time were analyzed as end points in regression models, adjusting for the presence of parasite infections. RESULTS The main effect of vitamin A supplementation was in height improvement (P<0.05), and was only found in the model evaluating infants with any parasite. There was an interaction effect of slower growth (P<0.05) found in infants infected with any parasite and supplemented with vitamin A in slower growth (P<0.05). In addition, the interaction of zinc supplementation and Giardia duodenalis or A. lumbricoides was associated with reduced growth (P<0.05). CONCLUSION Gastro-intestinal parasite infections may modify the effect that zinc or vitamin A supplementation has on childhood growth.
Collapse
|
37
|
Current and future perspectives on the chemotherapy of the parasitic protozoa Trichomonas vaginalis and Entamoeba histolytica. Future Med Chem 2009; 1:619-43. [DOI: 10.4155/fmc.09.59] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Trichomonas vaginalis and Entamoeba histolytica are clinically important protozoa that affect humans. T. vaginalis produces sexually transmitted infections and E. histolytica is the causative agent of amebic dysentery. Metronidazole, a compound first used to treat T. vaginalis in 1959, is still the main drug used worldwide to treat these pathogens. It is essential to find new biochemical differences in these organisms that could be exploited to develop new antiprotozoal chemotherapeutics. Recent findings associated with T. vaginalis and E. histolytica biochemistry and host–pathogen interactions are surveyed. Knowledge concerning the biochemistry of these parasites is serving to form the foundation for the development of new approaches to control these important human pathogens.
Collapse
|
38
|
Protection against intestinal amebiasis by a recombinant vaccine is transferable by T cells and mediated by gamma interferon. Infect Immun 2009; 77:3909-18. [PMID: 19564375 DOI: 10.1128/iai.00487-09] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
We have previously shown that vaccination with purified Entamoeba histolytica Gal/GalNAc lectin or recombinant subunits can protect mice from intestinal amebiasis upon intracecal challenge. In this study, we demonstrated with adoptive-transfer experiments that this lectin vaccine protection is mediated by T cells but not serum. The cell-mediated immune (CMI) response was characterized by significant gamma interferon (IFN-gamma), interleukin 12 (IL-12), IL-2, IL-10, and IL-17 production. To move toward a human vaccine, we switched to a recombinant protein and tested a range of adjuvants and routes appropriate for humans. We found that subcutaneous delivery of LecA with IDRI's adjuvant system EM014 elicited a potent Th1-type CMI profile and provided significant protection, as measured by culture negativity (79% efficacy); intranasal immunization with cholera toxin provided 56% efficacy; and alum induced a Th2-type response that protected 62 to 68% of mice. Several antibody and CMI cytokine responses were examined for correlates of protection, and prechallenge IFN-gamma(+) or IFN-gamma-, IL-2-, and tumor necrosis factor alpha-triple-positive CD4 cells in blood were statistically associated with protection. To test the role of IFN-gamma in LecA-mediated protection, we neutralized IFN-gamma in LecA-immunized mice and found that it abrogated the protection conferred by vaccination. These data demonstrate that CMI is sufficient for vaccine protection from intestinal amebiasis and reveal an important role for IFN-gamma, even in the setting of alum.
Collapse
|
39
|
Lejeune M, Rybicka JM, Chadee K. Recent discoveries in the pathogenesis and immune response toward Entamoeba histolytica. Future Microbiol 2009; 4:105-18. [PMID: 19207103 DOI: 10.2217/17460913.4.1.105] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Entamoeba histolytica is an enteric dwelling human protozoan parasite that causes the disease amoebiasis, which is endemic in the developing world. Over the past four decades, considerable effort has been made to understand the parasite and the disease. Improved diagnostics can now differentiate pathogenic E. histolytica from that of the related but nonpathogenic Entamoeba dispar, thus minimizing screening errors. Classically, the triad of Gal-lectin, cysteine proteinases and amoebapores of the parasite were thought to be the major proteins involved in the pathogenesis of amoebiasis. However, other amoebic molecules such as lipophosphopeptidoglycan, perioxiredoxin, arginase, and lysine and glutamic acid-rich proteins are also implicated. Recently, the genome of E. histolytica has been sequenced, which has widened our scope to study additional virulence factors. E. histolytica genome-based approaches have now confirmed the presence of Golgi apparatus-like vesicles and the machinery for glycosylation, thus improving the chances of identifying potential drug targets for chemotherapeutic intervention. Apart from Gal-lectin-based vaccines, promising vaccine targets such as serine-rich E. histolytica protein have yielded encouraging results. Considerable efforts have also been made to skew vaccination responses towards appropriate T-helper cell immunity that could augment the efficacy of vaccine candidates under study. Thus, ongoing efforts mining the information made available with the sequencing of the E. histolytica genome will no doubt identify and characterize other important potential vaccine/drug targets and lead to effective immunologic strategies for the control of amoebiasis.
Collapse
Affiliation(s)
- Manigandan Lejeune
- University of Calgary, Department of Microbiology & Infectious Diseases, Calgary, AB, T2N 4N1, Canada.
| | | | | |
Collapse
|
40
|
Characterization of Entamoeba histolytica intermediate subunit lectin-specific human monoclonal antibodies generated in transgenic mice expressing human immunoglobulin loci. Infect Immun 2008; 77:549-56. [PMID: 19001071 DOI: 10.1128/iai.01002-08] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Four fully human monoclonal antibodies (MAbs) to Entamoeba histolytica intermediate subunit lectin (Igl) were prepared in XenoMouse mice, which are transgenic mice expressing human immunoglobulin loci. Examination of the reactivities of these MAbs to recombinant Igl1 and Igl2 of E. histolytica showed that XEhI-20 {immunoglobulin G2(kappa) [IgG2(kappa)]} and XEhI-28 [IgG2(kappa)] were specific to Igl1, XEhI-B5 [IgG2(kappa)] was specific to Igl2, and XEhI-H2 [IgM(kappa)] was reactive with both Igls. Gene analyses revealed that the V(H) and V(L) germ lines were VH3-48 and L2 for XEhI-20, VH3-21 and L2 for XEhI-28, VH3-33 and B3 for XEhI-B5, and VH4-4 and A19 for XEhI-H2, respectively. Flow cytometry analyses showed that the epitopes recognized by all of these MAbs were located on the surfaces of living trophozoites. Confocal microscopy demonstrated that most Igl1 and Igl2 proteins were colocalized on the surface and in the cytoplasm, but different localization patterns in intracellular vacuoles were also present. The preincubation of trophozoites with XEhI-20, XEhI-B5, and XEhI-H2 caused significant inhibition of the adherence of trophozoites to Chinese hamster ovary cells, whereas preincubation with XEhI-28 did not do so. XEhI-20, XEhI-B5, and XEhI-H2 were injected intraperitoneally into hamsters 24 h prior to intrahepatic challenge with E. histolytica trophozoites. One week later, the mean abscess size in groups injected with one of the three MAbs was significantly smaller than that in controls injected with polyclonal IgG or IgM isolated from healthy humans. These results demonstrate that human MAbs to Igls may be applicable for immunoprophylaxis of amebiasis.
Collapse
|
41
|
Petri WA, Miller M, Binder HJ, Levine MM, Dillingham R, Guerrant RL. Enteric infections, diarrhea, and their impact on function and development. J Clin Invest 2008; 118:1277-90. [PMID: 18382740 DOI: 10.1172/jci34005] [Citation(s) in RCA: 281] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Enteric infections, with or without overt diarrhea, have profound effects on intestinal absorption, nutrition, and childhood development as well as on global mortality. Oral rehydration therapy has reduced the number of deaths from dehydration caused by infection with an enteric pathogen, but it has not changed the morbidity caused by such infections. This Review focuses on the interactions between enteric pathogens and human genetic determinants that alter intestinal function and inflammation and profoundly impair human health and development. We also discuss specific implications for novel approaches to interventions that are now opened by our rapidly growing molecular understanding.
Collapse
Affiliation(s)
- William A Petri
- Center for Global Health, Division of Infectious Diseases and International Health, University of Virginia School of Medicine, Charlottesville, Virginia 22908, USA
| | | | | | | | | | | |
Collapse
|
42
|
Abstract
Resistance to the establishment of intestinal Entamoeba histolytica infection is dependent on the inbred mouse strain. In this work we used the inbred strains B6 (resistant), CBA (susceptible), B6CBAF(1) and a backcross of B6CBAF(1) to CBA to further examine the genetic basis of resistance. Mouse genotype was assessed with single nucleotide polymorphism and microsatellite markers and infection assessed by culture 9 days after intracecal E. histolytica challenge. The backcross population showed a male predisposition to culture positivity (P<0.002). F1 genotype at two loci on chromosomes 1 and 2 exhibited suggestive linkage with resistance to infection (P=0.0007 and 0.0200). Additional suggestive quantitative trait locus were observed on chromosomes 1, 9 and 13 for cecal parasite antigen load and histologic evidence of inflammation. Infection in C3H x B6 recombinant inbred mice supported the mapping data. Candidate B6 genes on chromosomes 1 and 2 were examined by microarray analysis of epithelial tissues from B6 vs CBA mice. This work shows a male predisposition to intestinal amebiasis and suggests that relatively few B6 loci can confer resistance in inbred mice. Future identification of regional candidate genes has implications for understanding the human variability to amebic infection.
Collapse
|
43
|
Bercu TE, Petri WA, Behm JW. Amebic colitis: new insights into pathogenesis and treatment. Curr Gastroenterol Rep 2008; 9:429-33. [PMID: 17991346 DOI: 10.1007/s11894-007-0054-8] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Amebiasis, caused by the protozoan parasite Entamoeba histolytica, affects more than 50 million people worldwide, with over 100,000 deaths annually. The majority of cases are asymptomatic; however, significant morbidity and mortality are associated with illness in the remaining 10% of cases. Recent advances in the understanding of the mechanism of infection by E. histolytica, the role of the innate immune system, and the role of genetic disposition to infection will allow the development of novel detection and treatment methods. The disease mechanisms, clinical findings, therapeutic strategies, and important developments regarding amebiasis are discussed here.
Collapse
Affiliation(s)
- Tracy E Bercu
- Division of Infectious Diseases and International Health, University of Virginia Health System, P.O. Box 801340, Charlottesville, VA 22908, USA.
| | | | | |
Collapse
|
44
|
Long KZ, Rosado JL, Montoya Y, de Lourdes Solano M, Hertzmark E, DuPont HL, Santos JI. Effect of vitamin A and zinc supplementation on gastrointestinal parasitic infections among Mexican children. Pediatrics 2007; 120:e846-55. [PMID: 17908741 DOI: 10.1542/peds.2006-2187] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVE Gastrointestinal parasites continue to be an important cause of morbidity and stunting among children in developing countries. We evaluated the effect of vitamin A and zinc supplementation on infections by Giardia lamblia, Ascaris lumbricoides, and Entamoeba histolytica. METHODS A randomized, double-blind, placebo-controlled trial was conducted among 707 children who were 6 to 15 months of age and from periurban areas of Mexico City, Mexico, between January 2000 and May 2002. Children, who were assigned to receive either vitamin A every 2 months, a daily zinc supplement, a combined vitamin A and zinc supplement, or a placebo, were followed for 1 year. The primary end points were the 12-month rates and durations of infection for the 3 parasites and rates of parasite-associated diarrheal disease as determined in stools collected once a month and after diarrheal episodes. RESULTS G. lamblia infections were reduced and A. lumbricoides infections increased among children in the combined vitamin A and zinc group or the zinc alone group, respectively. Durations of Giardia infections were reduced among children in all 3 treatment arms, whereas Ascaris infections were reduced in the vitamin A and zinc group. In contrast, E. histolytica infection durations were longer among zinc-supplemented children. Finally, E. histolytica- and A. lumbricoides-associated diarrheal episodes were reduced among children who received zinc alone or a combined vitamin A and zinc supplement, respectively. CONCLUSIONS We found that vitamin A and zinc supplementation was associated with distinct parasite-specific health outcomes. Vitamin A plus zinc reduces G. lamblia incidence, whereas zinc supplementation increases A. lumbricoides incidence but decreases E. histolytica-associated diarrhea.
Collapse
Affiliation(s)
- Kurt Z Long
- Department of Nutrition, Harvard School of Public Health, Boston, Massachusetts, USA.
| | | | | | | | | | | | | |
Collapse
|
45
|
Ivory CPA, Chadee K. Intranasal immunization with Gal-inhibitable lectin plus an adjuvant of CpG oligodeoxynucleotides protects against Entamoeba histolytica challenge. Infect Immun 2007; 75:4917-22. [PMID: 17620349 PMCID: PMC2044551 DOI: 10.1128/iai.00725-07] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2007] [Accepted: 06/30/2007] [Indexed: 11/20/2022] Open
Abstract
The development of an effective amebiasis vaccine could improve child health in the developing world, reducing cases of amebic colitis and liver abscess. An ideal vaccine would be comprised of a well-characterized parasite antigen and an adjuvant, which would have high potency while driving the immune response in a Th1 direction. This study describes a mucosal vaccine composed of the Entamoeba histolytica galactose/N-acetyl-D-galactosamine-inhibitable lectin (Gal-lectin) and CpG oligodeoxynucleotides (CpG-ODN). The Gal-lectin is a protein involved in parasite virulence and adherence and is known to activate immune cells, while CpG-ODN are known to be potent inducers of type 1-like immune responses. We demonstrated that intranasal administration of the vaccine resulted in strong Gal-lectin-specific Th1 responses and humoral responses. Vaccination induced the production of Gal-lectin-specific T cells and the production of the proinflammatory cytokine gamma interferon. Vaccinated animals had detectable serum anti-Gal-lectin immunoglobulin G (IgG) and stool anti-Gal-lectin IgA capable of blocking parasite adherence to target cells in vitro. One week after immunization, gerbils were challenged intrahepatically with live trophozoites. Vaccinated gerbils had no detectable abscesses after day 5, whereas control gerbils developed larger abscesses. These results show that mucosal vaccination with Gal-lectin and CpG-ODN can induce both systemic and humoral immune responses.
Collapse
Affiliation(s)
- Catherine P A Ivory
- Faculty of Medicine, Department of Microbiology and Infectious Diseases, University of Calgary Health Sciences Centre, 3330 Hospital Dr. NW, Calgary, Alberta T2N 4N1, Canada
| | | |
Collapse
|
46
|
Abstract
Entamoeba histolytica is a eukaryotic protozoan parasite and is the causative agent of amebic colitis and amebic liver abscess. Many insights into the innate and acquired immune responses to infection with E. histolytica have been made in recent years. These findings have provided a foundation for producing a vaccine that could help to prevent the initial establishment of infection in the intestinal wall. The galactose and N-acetyl-D-galactosamine-specific lectin on the surface of the ameba is an immunodominant molecule that is highly conserved and has an integral role in the stimulation of these immune responses. The structure of the lectin has been defined, and the heavy subunit with its cysteine-rich region has been demonstrated in animal models to have some efficacy as a possible vaccine agent for prevention of amebic infection. Finding an ideal animal model of amebic intestinal infection has been difficult, but the C3H mouse and severe combined immunodeficient mouse-human intestinal xenograft models have both provided valuable insights into the first line of immune defense at the mucosal wall of the colon. Providing safe food and water to all people in the developing world is a formidable task that is not achievable in the foreseeable future. However, a vaccine for amebiasis could make a significant impact on the morbidity and mortality from the disease. Many components of the ameba are immunogenic and may serve as targets for a future vaccine, including the galactose and N-acetyl-D-galactosamine lectin, the serine-rich E. histolytica protein, cysteine proteinases, lipophosphoglycans, amebapores and the 29-kDa protein.
Collapse
Affiliation(s)
- Omer A Chaudhry
- Division of Infectious Diseases and International Medicine, Department of Internal Medicine, University of Virginia Health System, Charlottesville, VA 22908-1340, USA.
| | | |
Collapse
|
47
|
Guo X, Houpt E, Petri WA. Crosstalk at the initial encounter: interplay between host defense and ameba survival strategies. Curr Opin Immunol 2007; 19:376-84. [PMID: 17702556 PMCID: PMC2679172 DOI: 10.1016/j.coi.2007.07.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2007] [Revised: 06/14/2007] [Accepted: 07/04/2007] [Indexed: 10/22/2022]
Abstract
The host-parasite relationship is based on a series of interplays between host defense mechanisms and parasite survival strategies. Progress has been made in understanding the role of host immune response in amebiasis. While host cells elaborate diverse mechanisms for pathogen expulsion, amebae have also developed complex strategies to modulate host immune response and facilitate their own survival. This paper will give an overview of current research on the mutual interactions between host and Entamoeba histolytica in human and experimental amebiasis. Understanding this crosstalk is crucial for the effective design and implementation of new vaccines and drugs for this leading parasitic disease.
Collapse
Affiliation(s)
- Xiaoti Guo
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA 22908-1340
| | - Eric Houpt
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA 22908-1340
| | - William A. Petri
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA 22908-1340
| |
Collapse
|
48
|
Carrero JC, Cervantes-Rebolledo C, Aguilar-Díaz H, Díaz-Gallardo MY, Laclette JP, Morales-Montor J. The role of the secretory immune response in the infection by Entamoeba histolytica. Parasite Immunol 2007; 29:331-8. [PMID: 17576362 DOI: 10.1111/j.1365-3024.2007.00955.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Intestinal infection with the protozoan parasite Entamoeba histolytica elicits a local immune response with rising of specific secretory IgA (sIgA) antibodies detectable in several compartments associated to mucosa. Anti-amoebic sIgA antibodies have been reported in faeces, saliva, bile and breast milk from dysenteric patients and research trying to elucidate their role in protection has recently intensified. IgA antibodies inhibit the in vitro adherence of E. histolytica trophozoites to epithelial cell monolayers by recognizing several membrane antigens, including the galactose-binding lectin (Gal-lectin), main surface molecule involved in adherence, and the serine and cystein-rich proteins, all of them potential vaccine candidates. In fact, the presence of sIgA anti-Gal lectin in faeces of patients recovered from amoebic liver abscess (ALA) was associated with immunity to E. dispar. Moreover, the combined nasal and intraperitoneal vaccination of C3H/HeJ mice with native and recombinant Gal-lectin protected mice against an intracecal challenge with virulent E. histolytica trophozoites, protection that seemed to be associated with the induction of specific intestinal sIgA antibodies. Therefore, the stimulation of intestinal secretory response by mucosal delivery of amoebic antigens has been positioned as a promising strategy for inducing protection against human amoebiasis.
Collapse
Affiliation(s)
- J C Carrero
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México AP 70228, México.
| | | | | | | | | | | |
Collapse
|
49
|
Abd Alla MD, White GL, Rogers TB, Cary ME, Carey DW, Ravdin JI. Adherence-inhibitory intestinal immunoglobulin a antibody response in baboons elicited by use of a synthetic intranasal lectin-based amebiasis subunit vaccine. Infect Immun 2007; 75:3812-22. [PMID: 17526742 PMCID: PMC1952019 DOI: 10.1128/iai.00341-07] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We designed an amebiasis subunit vaccine that is constructed by using four peptide epitopes of the galactose-inhibitable lectin heavy subunit that were recognized by intestinal secretory immunoglobulin A (IgA) antibodies from immune human subjects. These epitopes are contained in the region encompassing amino acids 758 to 1134 of the lectin heavy subunit, designated LC3. Baboons (Papio anubis) are natural hosts for Entamoeba histolytica; naturally infected baboons raised in captivity possess serum IgA antibodies to the same four LC3 epitopes as humans. Uninfected, seronegative baboons received four intranasal immunizations at 7-day intervals with the synthetic peptide vaccine (400, 800, or 1,600 mug per nostril) with cholera toxin (20 mug) as the adjuvant. As determined by an enzyme-linked immunosorbent assay (ELISA), each dose of the peptide vaccine elicited antipeptide serum IgA and IgG and intestinal IgA antibody responses in all six immunized baboons by day 28, 7 days after the last immunization (P, <0.01 for each dose compared to the cholera toxin control). The peptide vaccine elicited serum IgG and intestinal IgA antibodies that recognized purified recombinant LC3 protein (P, <0.008 and 0.02, respectively) and native lectin protein (P < 0.01). In addition, an indirect immunofluorescence assay with whole trophozoites (P < 0.01) and Western blot analysis confirmed that serum IgG antibodies from vaccinated baboons recognized native lectin protein on the surfaces of axenic E. histolytica trophozoites or from solubilized amebae. All four synthetic peptides were immunogenic; the vaccine elicited dose- and time-dependent responses, as determined by ELISA optical density readings indicating the production of serum and intestinal antibodies (P, <0.02 for antipeptide and antilectin antibodies). As a positive control, intranasal immunization with purified recombinant LC3 protein with cholera toxin as the adjuvant elicited a serum anti-LC3 IgA and IgG antibody response (P, 0.05 and <0.0001, respectively); however, no intestinal anti-LC3 IgA antibody response was observed (P = 0.4). Of interest, serum IgA and IgG antibodies elicited by the recombinant LC3 vaccine did not recognize any of the four putatively protective LC3 peptide epitopes. Both serum and fecal antibodies elicited by the peptide vaccine exhibited neutralizing activity, as determined by their dose-dependent inhibition of the galactose-specific adherence of E. histolytica trophozoites to Chinese hamster ovary cells in vitro (P, <0.001 for each group of antibodies compared to the control). In summary, a lectin-based intranasal polylysine-linked synthetic peptide vaccine was effective in eliciting an adherence-inhibitory, intestinal antilectin IgA antibody response in baboons. Future studies with the baboon model will determine vaccine efficacy against asymptomatic E. histolytica intestinal infection.
Collapse
Affiliation(s)
- Mohamed D Abd Alla
- Department of Medicine, University of Minnesota, 14-110 Phillips Wangensteen Building, 516 Delaware Street S.E., Minneapolis, MN 55455, USA
| | | | | | | | | | | |
Collapse
|
50
|
Abstract
Amoebiasis, infection by the protozoan parasite Entamoeba histolytica, remains a global health problem, despite the availability of effective treatment. While improved sanitation could lead to the eradication of this disease, it is unlikely that this will occur worldwide in the foreseeable future; thus alternative measures must be pursued. One approach is to develop a vaccine to prevent this deadly disease. Clinical studies indicate that mucosal immunity may provide some protection against recurrent intestinal infection with E. histolytica, but there is no clear evidence that protective immunity develops after amoebic liver abscess. Over the past decade, progress in vaccine development has been facilitated by new animal models that allow better testing of potential vaccine candidates and the application of recombinant technology to vaccine design. Oral vaccines and DNA-based vaccines have been successfully tested in animals models for immunogenicity and efficacy. There has been significant progress on a number of fronts, but there are unanswered questions regarding the effectiveness of immune responses in preventing disease in man and, as yet, no testing of any of these vaccines in humans has been performed. In addition, there are strong economic barriers to developing an amoebiasis vaccine and questions about how and where an effective vaccine would be utilized.
Collapse
Affiliation(s)
- S L Stanley
- Department of Medicine, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA.
| |
Collapse
|