1
|
Pethig L, Behringer V, Kappeler PM, Fichtel C, Heistermann M. Establishment and Validation of Fecal Secretory Immunoglobulin A Measurement for Intestinal Mucosal Health Assessment in Wild Lemurs. Am J Primatol 2024; 86:e23694. [PMID: 39488843 DOI: 10.1002/ajp.23694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 10/15/2024] [Accepted: 10/19/2024] [Indexed: 11/05/2024]
Abstract
The measurement of biomarkers in blood and excreta can enable immune status assessment and provide prognostic information on individual health outcomes. In this respect, the fecal measurement of secretory immunoglobulin A (sIgA), the primary mammalian antibody for mucosal defense, has recently received increased interest in a few anthropoid primates, but a fecal sIgA assay for use in strepsirrhine primates has not yet been reported. Here, we develop and analytically validate a cost-effective in-house sandwich enzyme immunoassay for the extraction and measurement of sIgA in feces of redfronted lemurs (Eulemur rufifrons). We also tested a simple method for sIgA extraction that can be used under remote field conditions and undertook experiments to assess the robustness of sIgA concentrations to variation in processing and storage conditions of fecal extracts. Our analytical validation revealed that the assay recognizes immunoreactive sIgA in redfronted lemur feces, that sIgA can be measured accurately with no potential interference from the fecal matrix, and that assay reagents and performance are highly stable over time. The field-friendly extraction procedure produced sIgA results strongly correlated with those generated by a standard laboratory extraction method. Short-term storage at room temperature resulted in a slight decline in sIgA concentrations, whereas freezing extracts at -20°C kept sIgA levels stable for at least 3 months. Longer-term storage of >5 months, however, led to a significant decline of sIgA concentrations. Multiple freeze-thaw cycles did not affect sIgA levels. This study, therefore, provides the basis for measuring fecal sIgA in lemurs and possibly other strepsirrhines. When samples are processed properly and stored frozen, and when sIgA analysis can be performed within 3 months upon sample collection, fecal sIgA measurements can become a valuable tool for monitoring aspects of immunity and health in both zoo-housed and wild-living lemurs.
Collapse
Affiliation(s)
- Leonie Pethig
- Behavioral Ecology and Sociobiology Unit, German Primate Center, Leibniz Institute for Primate Research, Göttingen, Germany
- Department of Sociobiology/Anthropology, Johann-Friedrich-Blumenbach Institute of Zoology and Anthropology, University of Göttingen, Göttingen, Germany
| | - Verena Behringer
- Endocrinology Laboratory, German Primate Center, Leibniz Institute for Primate Research, Göttingen, Germany
| | - Peter M Kappeler
- Behavioral Ecology and Sociobiology Unit, German Primate Center, Leibniz Institute for Primate Research, Göttingen, Germany
- Department of Sociobiology/Anthropology, Johann-Friedrich-Blumenbach Institute of Zoology and Anthropology, University of Göttingen, Göttingen, Germany
| | - Claudia Fichtel
- Behavioral Ecology and Sociobiology Unit, German Primate Center, Leibniz Institute for Primate Research, Göttingen, Germany
| | - Michael Heistermann
- Endocrinology Laboratory, German Primate Center, Leibniz Institute for Primate Research, Göttingen, Germany
| |
Collapse
|
2
|
Cossarini F, Shang J, Krek A, Al-Taie Z, Hou R, Canales-Herrerias P, Tokuyama M, Tankelevich M, Tillowitz A, Jha D, Livanos AE, Leyre L, Uzzan M, Martinez-Delgado G, Taylor MD, Sharma K, Bourgonje AR, Cruz M, Ioannou G, Dawson T, D'Souza D, Kim-Schulze S, Akm A, Aberg JA, Chen BK, Kwon DS, Gnjatic S, Polydorides AD, Cerutti A, Argmann C, Vujkovic-Cvijin I, Suarez-Fariñas M, Petralia F, Faith JJ, Mehandru S. Gastrointestinal germinal center B cell depletion and reduction in IgA + plasma cells in HIV-1 infection. Sci Immunol 2024; 9:eado0090. [PMID: 39454027 PMCID: PMC11557871 DOI: 10.1126/sciimmunol.ado0090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 09/25/2024] [Indexed: 10/27/2024]
Abstract
Gastrointestinal (GI) B cells and plasma cells (PCs) are critical to mucosal homeostasis and the host response to HIV-1 infection. Here, high-resolution mapping of human B cells and PCs sampled from the colon and ileum during both viremic and suppressed HIV-1 infection identified a reduction in germinal center (GC) B cells and follicular dendritic cells (FDCs) during HIV-1 viremia. Immunoglobulin A-positive (IgA+) PCs are the major cellular output of intestinal GCs and were significantly reduced during viremic HIV-1 infection. PC-associated transcriptional perturbations, including type I interferon signaling, persisted in antiretroviral therapy (ART)-treated individuals, suggesting ongoing disruption of the intestinal immune milieu during ART. GI humoral immune perturbations were associated with changes in the intestinal microbiome composition and systemic inflammation. These findings highlight a key immune defect in the GI mucosa due to HIV-1 viremia.
Collapse
Affiliation(s)
- Francesca Cossarini
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joan Shang
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Azra Krek
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Zainab Al-Taie
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ruixue Hou
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Pablo Canales-Herrerias
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Minami Tokuyama
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Michael Tankelevich
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Adam Tillowitz
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Divya Jha
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexandra E. Livanos
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Louise Leyre
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mathieu Uzzan
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Gastroenterology Department, Hôpital Henri Mondor, APHP, Créteil, France
| | - Gustavo Martinez-Delgado
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthew D. Taylor
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Keshav Sharma
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Arno R. Bourgonje
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Michael Cruz
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Giorgio Ioannou
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Travis Dawson
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Darwin D'Souza
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Seunghee Kim-Schulze
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ahmed Akm
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Judith A. Aberg
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Benjamin K. Chen
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Douglas S. Kwon
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, USA
| | - Sacha Gnjatic
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexandros D. Polydorides
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Andrea Cerutti
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Translational Clinical Research Program, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
- Catalan Institute for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Carmen Argmann
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ivan Vujkovic-Cvijin
- F. Widjaja IBD Institute, Division of Gastroenterology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Mayte Suarez-Fariñas
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Francesca Petralia
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jeremiah J. Faith
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Saurabh Mehandru
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
3
|
Donaldson GP, Reis GL, Saad M, Wichmann C, Mamede I, Chen G, DelGaudio NL, Zhang D, Aydin B, Harrer CE, Castro TBR, Grivennikov S, Reis BS, Stadtmueller BM, Victora GD, Mucida D. Suppression of epithelial proliferation and tumourigenesis by immunoglobulin A. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.06.561290. [PMID: 37873082 PMCID: PMC10592636 DOI: 10.1101/2023.10.06.561290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Immunoglobulin A (IgA) is the most abundant antibody isotype produced across mammals and plays a specialized role in mucosal homeostasis 1 . Constantly secreted into the lumen of the intestine, IgA binds commensal microbiota to regulate their colonization and function 2,3 with unclear implications for health. IgA deficiency is common in humans but is difficult to study due to its complex aetiology and comorbidities 4-8 . Using genetically and environmentally controlled mice, here we show that IgA-deficient animals have increased susceptibility to endogenous colorectal tumours. Cellular and molecular analyses revealed that, in the absence of IgA, colonic epithelial cells induce antibacterial factors and accelerate cell cycling in response to the microbiota. Oral treatment with IgA was sufficient to both reduce steady-state proliferation and protect mice from tumours, but this function was due to antibody structure rather than binding specificity. In both organoid and monolayer culture systems, IgA directly suppressed epithelial growth. Co-immunoprecipitation mass spectrometry and a targeted CRISPR screen identified DMBT1 as an IgA-binding epithelial surface protein required for IgA-mediated suppression of proliferation. Together, IgA and DMBT1 regulate Notch signalling and tune the normal cycling of absorptive colonocyte progenitors. In mice, deleting the transmembrane and cytoplasmic signalling portions of DMBT1 or blocking Notch signalling was sufficient to reverse both the increased proliferation and tumour susceptibility of IgA knockouts. These experiments establish a homeostatic function for IgA in tempering physiological epithelial responses to microbiota to maintain mucosal health.
Collapse
|
4
|
Klimczak S, Packi K, Rudek A, Wenclewska S, Kurowski M, Kurczabińska D, Śliwińska A. The Influence of the Protozoan Giardia lamblia on the Modulation of the Immune System and Alterations in Host Glucose and Lipid Metabolism. Int J Mol Sci 2024; 25:8627. [PMID: 39201314 PMCID: PMC11354543 DOI: 10.3390/ijms25168627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/28/2024] [Accepted: 08/01/2024] [Indexed: 09/02/2024] Open
Abstract
Giardia lamblia, the cause of giardiasis, significantly impacts patients with metabolic disorders related to insulin resistance (IR). Both giardiasis and metabolic disorders share elements such as chronic inflammation and intestinal dysbiosis, which substantially affect the metabolic and cytokine profiles of patients. This review discusses the mechanisms of virulence of G. lamblia, its influence on the immune system, and its association with metabolic disorders. The review aims to show how G. lamblia invasion acts on the immune system and the glucose and lipid metabolism. Key findings reveal that G. lamblia infection, by disrupting intestinal permeability, alters microbiota composition and immune responses, potentially impairing metabolic status. Future research should focus on elucidating the specific mechanisms by which G. lamblia influences the metabolism, exploring the long-term consequences of chronic infection, and developing targeted therapeutic strategies that include both parasitic and metabolic aspects. These insights underscore the need for a multidisciplinary approach to the treatment of giardiasis in patients with metabolic disorders.
Collapse
Affiliation(s)
- Sylwia Klimczak
- Department of Nucleic Acid Biochemistry, Medical University of Lodz, 92-213 Lodz, Poland
- AllerGen Center of Personalized Medicine, 97-300 Piotrkow Trybunalski, Poland
| | - Kacper Packi
- AllerGen Center of Personalized Medicine, 97-300 Piotrkow Trybunalski, Poland
| | - Alicja Rudek
- AllerGen Center of Personalized Medicine, 97-300 Piotrkow Trybunalski, Poland
| | - Sylwia Wenclewska
- Diabetology and Internal Medicine Department, Provincial Hospital in Sieradz, 98-200 Sieradz, Poland
| | - Marcin Kurowski
- Department of Immunology and Allergy, Medical University of Lodz, 92-213 Lodz, Poland
| | | | - Agnieszka Śliwińska
- Department of Nucleic Acid Biochemistry, Medical University of Lodz, 92-213 Lodz, Poland
| |
Collapse
|
5
|
Ihara S, Nguyen BV, Miyamoto Y, Eckmann L. Mucosal vaccination in a murine gnotobiotic model of Giardia lamblia infection. Infect Immun 2024; 92:e0006524. [PMID: 38722167 PMCID: PMC11237505 DOI: 10.1128/iai.00065-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 04/10/2024] [Indexed: 06/12/2024] Open
Abstract
Giardia lamblia is an important protozoan cause of diarrheal disease worldwide, delayed development and cognitive impairment in children in low- and middle-income countries, and protracted post-infectious syndromes in developed regions. G. lamblia resides in the lumen and at the epithelial surface of the proximal small intestine but is not mucosa invasive. The protozoan parasite is genetically diverse with significant genome differences across strains and assemblages. Animal models, particularly murine models, have been instrumental in defining mechanisms of host defense against G. lamblia, but mice cannot be readily infected with most human pathogenic strains. Antibiotic pretreatment can increase susceptibility, suggesting that the normal microbiota plays a role in controlling G. lamblia infection in mice, but the broader implications on susceptibility to diverse strains are not known. Here, we have used gnotobiotic mice to demonstrate that robust intestinal infection can be achieved for a broad set of human-pathogenic strains of the genetic assemblages A and B. Furthermore, gnotobiotic mice were able to eradicate infection with a similar kinetics to conventional mice after trophozoite challenge. Germ-free mice could also be effectively immunized by the mucosal route with a protective antigen, α1-giardin, in a manner dependent on CD4 T cells. These results indicate that the gnotobiotic mouse model is powerful for investigating acquired host defenses in giardiasis, as the mice are broadly susceptible to diverse G. lamblia strains yet display no apparent defects in mucosal immunity needed for controlling and eradicating this lumen-dwelling pathogen.
Collapse
Affiliation(s)
- Sozaburo Ihara
- Department of Medicine, University of California San Diego, La Jolla, California, USA
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Brian V. Nguyen
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Yukiko Miyamoto
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Lars Eckmann
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
6
|
Xu H, Wang Z, Li Y, Xu Z. The distribution and function of teleost IgT. FISH & SHELLFISH IMMUNOLOGY 2024; 144:109281. [PMID: 38092093 DOI: 10.1016/j.fsi.2023.109281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/04/2023] [Accepted: 12/06/2023] [Indexed: 12/20/2023]
Abstract
Given the uniquely close relationship between fish and aquatic environments, fish mucosal tissues are constantly exposed to a wide array of pathogenic microorganisms in the surrounding water. To maintain mucosal homeostasis, fish have evolved a distinct mucosal immune system known as mucosal-associated lymphoid tissues (MALTs). These MALTs consist of key effector cells and molecules from the adaptive immune system, such as B cells and immunoglobulins (Igs), which play crucial roles in maintaining mucosal homeostasis and defending against external pathogen infections. Until recently, three primary Ig isotypes, IgM, IgD, and IgT, have been identified in varying proportions within the mucosal secretions of teleost fish. Similar to the role of mucosal IgA in mammals and birds, teleost IgT plays a predominant role in mucosal immunity. Following the identification of the IgT gene in 2005, significant advances have been made in researching the origin, evolution, structure, and function of teleost IgT. Multiple IgT variants have been identified in various species of teleost fish, underscoring the remarkable complexity of IgT in fish. Therefore, this study provides a comprehensive review of the recent advances in various aspects of teleost IgT, including its genomic and structural features, the diverse distribution patterns within various fish mucosal tissues (the skin, gills, gut, nasal, buccal, pharyngeal, and swim bladder mucosa), its interaction with mucosal symbiotic microorganisms, and its immune responses towards diverse pathogens, including bacteria, viruses, and parasites. We also highlight the existing research gaps in the study of teleost IgT, suggesting the need for further investigation into the functional aspects of IgT and IgT+ B cells. This research is aimed at providing valuable insights into the immune functions of IgT and the mechanisms underlying the immune responses of fish against infections.
Collapse
Affiliation(s)
- Haoyue Xu
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zixuan Wang
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuqing Li
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhen Xu
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
7
|
Fontenele ALA, de Oliveira Nóbrega CG, do Nascimento WRC, Santos PDA, de Lorena VMB, Peixoto DM, de Azevedo Albuquerque MCP, Solé D, Costa VMA, Sarinho ESC, de Souza VMO. Respiratory allergy symptoms and cytokine profiles in the presence of anti-Ascaris antibody in Giardia lamblia-infected children. Parasitol Res 2023; 122:3147-3158. [PMID: 37875615 DOI: 10.1007/s00436-023-08005-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 10/09/2023] [Indexed: 10/26/2023]
Abstract
Anti-Ascaris lumbricoides (Asc) IgE and IgG can immunomodulate the allergy; however, the influence of these isotypes has not been investigated in the giardiasis and allergy. Therefore, the frequency of respiratory allergy (RA) symptoms in Giardia lamblia-infected children, with or without anti-Asc IgE, IgG1, or IgG4 and Th1, Th2/Treg, and Th17 cytokine production, was evaluated. We performed a case-control study with children aged 2-10 years old selected by questionnaire and stool exams to form the groups: infected or uninfected with RA (G-RA, n = 55; nG-RA, n = 43); infected and uninfected without RA (G-nRA, n = 59; nG-nRA, n = 54). We performed blood leukocyte counts and in vitro culture. Cytokine levels in the supernatants (CBA), serum total IgE and anti-Asc IgE (ImmunoCAP), IgG1, IgG4, and total IgA (ELISA) were measured. Infection was not associated with allergy. Infected children showed increased levels of anti-Asc IgG1, IL-2, IFN-γ, IL-4, and IL-10. There was a lower frequency of allergy-related symptoms in anti-Asc IgG1-positive children than IgG1-negative (OR = 0.38; CI = 0.17-0.90, p = 0.027) and few eosinophils in G-RA than in G-nRA and more in G-nRA than in nG-nRA, whereas TNF-α levels were higher in the G-RA than in the nG-nRA group. For infected and positive anti-Asc IgG1, there was higher TNF-α and IL-10 production than G/-IgG1. IL-10 levels were lower in nG/ + IgG1 than in infected or non-infected, and both were negative for anti-Asc IgG1. Th1/Th2/IL-10 profiles were stimulated in the infected patients, and in those with circulating anti-Asc IgG1, the TNF-α production was strengthened with a lower risk for respiratory allergy symptoms.
Collapse
Affiliation(s)
- Ana Lúcia Arruda Fontenele
- Setor de Imunologia, Laboratório de Imunopatologia Keizo Asami, Universidade Federal de Pernambuco, Recife, Brazil
| | | | | | | | | | - Décio Medeiros Peixoto
- Centro de Pesquisa em Alergia e Imunologia Clínica, Hospital das Clínicas, Universidade Federal de Pernambuco, Recife, Brazil
| | | | - Dirceu Solé
- Divisão de Alergia, Imunologia Clínica e Reumatologia, Departamento de Pediatria da, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | | | - Valdenia Maria Oliveira de Souza
- Setor de Imunologia, Laboratório de Imunopatologia Keizo Asami, Universidade Federal de Pernambuco, Recife, Brazil.
- Departamento de Ciências Farmacêuticas, Universidade Federal de Pernambuco, Av. Prof. Moraes Rego, S/N. Cidade Universitária, 50.670-901, Recife, PE, Brazil.
| |
Collapse
|
8
|
Popovic A, Cao EY, Han J, Nursimulu N, Alves-Ferreira EVC, Burrows K, Kennard A, Alsmadi N, Grigg ME, Mortha A, Parkinson J. The commensal protist Tritrichomonas musculus exhibits a dynamic life cycle that induces extensive remodeling of the gut microbiota. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.06.528774. [PMID: 37090671 PMCID: PMC10120700 DOI: 10.1101/2023.03.06.528774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Commensal protists and gut bacterial communities exhibit complex relationships, mediated at least in part through host immunity. To improve our understanding of this tripartite interplay, we investigated community and functional dynamics between the murine protist Tritrichomonas musculus ( T. mu ) and intestinal bacteria in healthy and B cell-deficient mice. We identified dramatic, protist-driven remodeling of resident microbiome growth and activities, in parallel with T. mu functional changes, accelerated in the absence of B cells. Metatranscriptomic data revealed nutrient-based competition between bacteria and the protist. Single cell transcriptomics identified distinct T. mu life stages, providing new evidence for trichomonad sexual replication and the formation of pseudocysts. Unique cell states were validated in situ through microscopy and flow cytometry. Our results reveal complex microbial dynamics during the establishment of a commensal protist in the gut, and provide valuable datasets to drive future mechanistic studies.
Collapse
|
9
|
Mathison BA, Pritt BS. The Landscape of Parasitic Infections in the United States. Mod Pathol 2023; 36:100217. [PMID: 37182583 DOI: 10.1016/j.modpat.2023.100217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 04/30/2023] [Accepted: 05/03/2023] [Indexed: 05/16/2023]
Abstract
The landscape of parasitic infections in the United States has shifted dramatically over the past century. Although infections such as malaria have been successfully eliminated, others remain endemic and pose a significant public health risk. Numerous parasitic infections are also imported each year. This article focuses on endemic parasitic infections that may be commonly seen in anatomical pathology preparations and discusses their biology, diagnostic histopathological features, and epidemiology.
Collapse
Affiliation(s)
- Blaine A Mathison
- Institute for Clinical and Experimental Pathology, ARUP Laboratories, Salt Lake City, Utah
| | - Bobbi S Pritt
- Department of Laboratory Medicine and Pathology, Divisions of Clinical Microbiology and Anatomic Pathology, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
10
|
Hassan ZR, Salama DEA, Ibrahim HF, Ahmed SG. Ultrastructural changes and IgA modulatory effect of commercial prebiotic and probiotic in murine giardiasis. J Parasit Dis 2023; 47:224-237. [PMID: 37193505 PMCID: PMC10182204 DOI: 10.1007/s12639-022-01552-9] [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: 09/09/2022] [Accepted: 11/15/2022] [Indexed: 12/24/2022] Open
Abstract
Giardiasis, a parasitic infection of the gastrointestinal tract, is prevalent worldwide. The integrity of the intestinal epithelial barrier plays an important defensive role in giardiasis, and as Oral supplementation with prebiotics and probiotics is known to reinforce the intestinal barrier in many gastrointestinal diseases, this study assessed the effects of prebiotic and probiotic supplementation in giardiasis and compared the results with those obtained after nitazoxanide therapy. Swiss albino male lab-bred mice (n = 50) were divided into three major groups; Group I (control group), i.e., negative (noninfected nontreated) and positive controls (infected nontreated); Group II (preventive group), in which mice were provided prebiotic, probiotic, or a combination for 7 days before of infection, and Group III (therapy group), in which mice were administered prebiotic, probiotic, combined supplements and nitazoxanide from day 12 post-infection. The assessment was achieved through Giardia cyst count, histopathological examination and ultrastructure study. Also, Serological and immunohistochemical parameters were done to evaluate the modulation of IgA levels. Oral supplementation with prebiotic and probiotic, either before or after infection (in preventive or therapy groups respectively) resulted in a significant reduction in Giardia cyst shedding. Remarkable histological and ultrastructure improvement in the intestinal changes, along with a significant increase in the serological and immunohistochemical IgA levels, were seen in mice provided combined supplements and nitazoxanide (in therapy group). Thus, our results indicate that combined prebiotic and probiotic supplementation has promising anti-Giardia activity and that it can restore intestinal structures and modulate IgA response, apart from providing synergistic effects when added to nitazoxanide.
Collapse
Affiliation(s)
- Zeinab R. Hassan
- Departments of Parasitology, Faculty of Medicine for Girls, Al-Azhar University, Cairo, 11651 Egypt
| | - Doaa E. A. Salama
- Departments of Pathology, Faculty of Medicine for Girls, Al-Azhar University, Cairo, 11651 Egypt
- Department of Pathology, School of Medicine, Badr University in Cairo (BUC), Cairo, 11829 Egypt
| | - Hanan F. Ibrahim
- Departments of Microbiology and Immunology, Faculty of Medicine for Girls, Al-Azhar University, Cairo, 11651 Egypt
| | - Samah G. Ahmed
- Departments of Histology, Faculty of Medicine for Girls, Al-Azhar University, Cairo, 11651 Egypt
| |
Collapse
|
11
|
Application of Proteomics to the Study of the Therapeutics and Pathogenicity of Giardia duodenalis. Diagnostics (Basel) 2022; 12:diagnostics12112744. [DOI: 10.3390/diagnostics12112744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/21/2022] [Accepted: 11/03/2022] [Indexed: 11/12/2022] Open
Abstract
Giardia duodenalis remains a neglected tropical disease. A key feature of the sustained transmission of Giardia is the ability to form environmentally resistant cysts. For the last 38 years, proteomics has been utilised to study various aspects of the parasite across different life cycle stages. Thirty-one articles have been published in PubMed from 2012 to 2022 related to the proteomics of G. duodenalis. Currently, mass spectrometry with LC-MS/MS and MALDI-TOF/TOF has been commonly utilised in proteomic analyses of Giardia, which enables researchers to determine potential candidates for diagnostic biomarkers as well as vaccine and drug targets, in addition to allowing them to investigate the virulence of giardiasis, the pathogenicity mechanisms of G. duodenalis, and the post-translational modifications of Giardia proteins throughout encystation. Over the last decade, valuable information from proteomics analyses of G. duodenalis has been discovered in terms of the pathogenesis and virulence of Giardia, which may provide guidance for the development of better means with which to prevent and reduce the impacts of giardiasis. Nonetheless, there is room for improving proteomics analyses of G. duodenalis, since genomic sequences for additional assemblages of Giardia have uncovered previously unknown proteins associated with the Giardia proteome. Therefore, this paper aims to review the applications of proteomics for the characterisation of G. duodenalis pathogenicity and the discovery of novel vaccine as well as drug targets, in addition to proposing some general directions for future Giardia proteomic research.
Collapse
|
12
|
Ihara S, Miyamoto Y, Le CHY, Tran VN, Hanson EM, Fischer M, Hanevik K, Eckmann L. Conserved metabolic enzymes as vaccine antigens for giardiasis. PLoS Negl Trop Dis 2022; 16:e0010323. [PMID: 35468132 PMCID: PMC9037923 DOI: 10.1371/journal.pntd.0010323] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 03/12/2022] [Indexed: 11/20/2022] Open
Abstract
Giardia lamblia is a leading protozoal cause of diarrheal disease worldwide. Infection is associated with abdominal pain, malabsorption and weight loss, and protracted post-infectious syndromes. A human vaccine is not available against G. lamblia. Prior studies with human and murine immune sera have identified several parasite antigens, including surface proteins and metabolic enzymes with intracellular functions. While surface proteins have demonstrated vaccine potential, they can exhibit significant variation between G. lamblia strains. By comparison, metabolic enzymes show greater conservation but their vaccine potential has not been established. To determine whether such proteins can serve as vaccine candidates, we focused on two enzymes, α-enolase (ENO) and ornithine carbamoyl transferase (OCT), which are involved in glycolysis and arginine metabolism, respectively. We show in a cohort of patients with confirmed giardiasis that both enzymes are immunogenic. Intranasal immunization with either enzyme antigen in mice induced strong systemic IgG1 and IgG2b responses and modest mucosal IgA responses, and a marked 100- to 1,000-fold reduction in peak trophozoite load upon oral G. lamblia challenge. ENO immunization also reduced the extent and duration of cyst excretion. Examination of 44 cytokines showed only minimal intestinal changes in immunized mice, although a modest increase of CCL22 was observed in ENO-immunized mice. Spectral flow cytometry revealed increased numbers and activation state of CD4 T cells in the small intestine and an increase in α4β7-expressing CD4 T cells in mesenteric lymph nodes of ENO-immunized mice. Consistent with a key role of CD4 T cells, immunization of CD4-deficient and Rag-2 deficient mice failed to induce protection, whereas mice lacking IgA were fully protected by immunization, indicating that immunity was CD4 T cell-dependent but IgA-independent. These results demonstrate that conserved metabolic enzymes can be effective vaccine antigens for protection against G. lamblia infection, thereby expanding the repertoire of candidate antigens beyond primary surface proteins.
Collapse
Affiliation(s)
- Sozaburo Ihara
- Department of Medicine, University of California San Diego, La Jolla, California
- Division of Gastroenterology, The Institute for Adult Diseases, Asahi Life Foundation, Tokyo, Japan
| | - Yukiko Miyamoto
- Department of Medicine, University of California San Diego, La Jolla, California
| | - Christine H. Y. Le
- Department of Medicine, University of California San Diego, La Jolla, California
| | - Vivien N. Tran
- Department of Medicine, University of California San Diego, La Jolla, California
| | - Elaine M. Hanson
- Department of Medicine, University of California San Diego, La Jolla, California
| | - Marvin Fischer
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Kurt Hanevik
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Lars Eckmann
- Department of Medicine, University of California San Diego, La Jolla, California
| |
Collapse
|
13
|
Bourque DL, Neumayr A, Libman M, Chen LH. Treatment strategies for nitroimidazole-refractory giardiasis: a systematic review. J Travel Med 2022; 29:6340793. [PMID: 34350966 DOI: 10.1093/jtm/taab120] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 07/23/2021] [Accepted: 07/26/2021] [Indexed: 12/24/2022]
Abstract
RATIONALE FOR REVIEW Giardiasis is one of the most common human protozoal infections worldwide. First-line therapy of giardiasis includes nitroimidazole antibiotics. However, treatment failure with nitroimidazoles is increasingly reported, with up to 45% of patients not responding to initial treatment. There is no clear consensus on the approach to the management of nitroimidazole-refractory giardiasis. This systematic review aims to summarize the literature on pharmacotherapy for nitroimidazole-refractory giardiasis. METHODS We conducted a systematic review of the literature to determine the optimal management strategies for nitroimidazole-refractory giardiasis. We searched Pubmed/MEDLINE, Embase and Cochrane library using the following search terms 'Giardia' AND 'treatment failure' OR 'refractory giardia' OR 'resistant giardia' with date limits of 1 January 1970 to 30 June 2021. We included all reports on humans, which described clinical outcomes of individuals with treatment refractory giardiasis, including case series and case reports. A descriptive synthesis of the data was conducted with pooling of data for interventions. KEY FINDINGS Included in this review were five prospective studies, three retrospective studies, seven case series and nine case reports. Across these reports, a wide heterogeneity of treatment regimens was employed, including retreatment with an alternative nitroimidazole, combination therapy with a nitroimidazole and another agent and monotherapy with non-nitroimidazole regimens, including quinacrine, paromomycin and nitazoxanide. Retreatment with a nitroimidazole was not an effective therapy for refractory giardiasis. However, treatment with a nitroimidazole in combination with albendazole had a cure rate of 66.9%. In the included studies, quinacrine monotherapy was administered to a total of 179 patients, with a clinical cure rate of 88.8%. Overall, quinacrine was fairly well tolerated. CONCLUSIONS Reports on the treatment of nitroimidazole-refractory giardiasis demonstrate a heterogeneous approach to treatment. Of these, quinacrine appeared to be highly effective, though more data on its safety are needed.
Collapse
|
14
|
Behringer V, Müller-Klein N, Strube C, Schülke O, Heistermann M, Ostner J. Responsiveness of fecal immunoglobulin A to HPA-axis activation limits its use for mucosal immunity assessment. Am J Primatol 2021; 83:e23329. [PMID: 34554596 DOI: 10.1002/ajp.23329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 08/30/2021] [Accepted: 09/04/2021] [Indexed: 11/11/2022]
Abstract
The assessment of mucosal immunity as a component of animal health is an important aspect for the understanding of variation in host immunity, and its tradeoff against other life-history traits. We investigated immunoglobulin A (IgA), the major type of antibody associated with mucosal immunity, in relation to changes in parasitic burden following anthelminthic treatment in noninvasively collected fecal samples in a semi-free ranging group of Barbary macaques (Macaca sylvanus). We measured IgA in 340 fecal samples of fourteen females and nine males. As IgA has been found to be responsive to stressors, we also related fecal IgA (fIgA) levels to fecal glucocorticoid metabolites (fGCM) measured in the same samples as part of a previous study. We found a high variability within and between individual fIgA levels over time. Running generalized additive mixed models, we found that fIgA levels were higher in males than in females, but did not change in response to the anthelmintic treatment and the resulting reduction in worm burden. Instead, fIgA level changes were significantly correlated to changes in fGCM levels. Our findings indicate that due to the strong responsiveness of fIgA to HPA-axis activity, the measurement of fIgA may have certain limitations with respect to reflecting gastrointestinal parasitic burden. Moreover, the responsiveness of fIgA to stressors interferes with the interpretation of IgA levels in fecal samples as a measure of mucosal immunity, at least in our study population of the Barbary macaques.
Collapse
Affiliation(s)
- Verena Behringer
- Endocrinology Laboratory, German Primate Center, Leibniz Institute for Primate Research, Göttingen, Germany
| | - Nadine Müller-Klein
- Institute for Evolutionary Ecology and Conservation Genomics, Ulm University, Ulm, Germany.,Research Group Social Evolution in Primates, German Primate Center, Leibniz Institute for Primate Research, Göttingen, Germany
| | - Christina Strube
- Institute for Parasitology, Centre for Infection Medicine, University of Veterinary Medicine Hannover, Hanover, Germany
| | - Oliver Schülke
- Research Group Social Evolution in Primates, German Primate Center, Leibniz Institute for Primate Research, Göttingen, Germany.,Department of Behavioral Ecology, Johann-Friedrich-Blumenbach Institute for Zoology and Anthropology, University of Göttingen, Göttingen, Germany.,Leibniz ScienceCampus Primate Cognition, Göttingen, Germany
| | - Michael Heistermann
- Endocrinology Laboratory, German Primate Center, Leibniz Institute for Primate Research, Göttingen, Germany
| | - Julia Ostner
- Research Group Social Evolution in Primates, German Primate Center, Leibniz Institute for Primate Research, Göttingen, Germany.,Department of Behavioral Ecology, Johann-Friedrich-Blumenbach Institute for Zoology and Anthropology, University of Göttingen, Göttingen, Germany.,Leibniz ScienceCampus Primate Cognition, Göttingen, Germany
| |
Collapse
|
15
|
Age and Giardia intestinalis Infection Impact Canine Gut Microbiota. Microorganisms 2021; 9:microorganisms9091862. [PMID: 34576757 PMCID: PMC8469385 DOI: 10.3390/microorganisms9091862] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 12/16/2022] Open
Abstract
Giardia intestinalis is a flagellated protozoan responsible for giardiosis (also called giardiasis in humans), the most prevalent and widespread parasitic infection in humans and mammals worldwide. The intestinal microbiota is highly diverse and any alteration in its composition may impact on the health of the host. While studies on the mouse model of giardiosis described the role of the gut microbiota in host susceptibility to infection by the parasite, little is known about the gut microbiota during natural infections in dogs and particularly in puppies. In this study, we monitored naturally G. intestinalis-infected puppies for 3 months and quantified cyst excretion every 2 weeks. All puppies remained subclinically infected during the sampling period as confirmed by fecal examination. In parallel, we performed 16S Illumina sequencing of fecal samples from the different time points to assess the impact of G. intestinalis infection on gut microbiota development of the puppies, as well as gut health markers of immunity such as fecal IgA and calprotectin. Sequencing results revealed that the canine fecal microbiota of Giardia-infected puppies becomes more complex and less diverse with increasing age. In addition, significant differences in the structure of the microbiota were observed between puppies with high and low Giardia cyst excretion. Chronic subclinical G. intestinalis infection appears to be associated with some detrimental structural changes in the gut microbiota. G. intestinalis-associated dysbiosis is characterized by an enrichment of facultative anaerobic, mucus-degrading, pro-inflammatory species and opportunistic pathogens, as well as a reduction of Lactobacillus johnsonii at specific time points. Calprotectin levels increased with age, suggesting the establishment of chronic low-grade inflammation in puppies. Further work is needed to demonstrate whether these alterations in the canine gut microbiota could lead to a dysbiosis-related disease, such as irritable bowel syndrome (IBS) or inflammatory bowel disease (IBD).
Collapse
|
16
|
Maertens B, Gagnaire A, Paerewijck O, De Bosscher K, Geldhof P. Regulatory role of the intestinal microbiota in the immune response against Giardia. Sci Rep 2021; 11:10601. [PMID: 34011991 PMCID: PMC8134572 DOI: 10.1038/s41598-021-90261-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 05/10/2021] [Indexed: 12/15/2022] Open
Abstract
Giardia duodenalis is one of the most commonly found intestinal parasites in mammalian hosts. Infections can generally be cleared by mounting an adequate protective immune response that is orchestrated through IL-17A. This study was aimed to investigate if and how the intestinal microbiome affects the protective Th17 response against Giardia by analysing and comparing the immune response following a G. muris and G. duodenalis infection in antibiotic treated and untreated mice. Depletion of the intestinal flora by antibiotic treatment had a severe effect on the infection dynamics of both Giardia species. Not only duration of infection was affected, but also the parasite burden increased significantly. Markers associated with a protective immune response, such as IL-17A and mannose binding lectin 2 were still significantly upregulated following infection in the antibiotic-treated mice, despite the lack of protection. On the other hand, the antibiotic treatment significantly decreased the level of IgA in the intestinal lumen by affecting its transporter and by reducing the number of IgA+ B-cells at the Peyer's patches. Furthermore, the depletion of the gut microbiota by antibiotics also significantly lowered the intestinal motility. The combination of these factors likely results in a decreased clearance of the parasite from the intestinal tract.
Collapse
Affiliation(s)
- B Maertens
- Department of Virology, Parasitology and Immunology, Laboratory of Parasitology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - A Gagnaire
- Department of Virology, Parasitology and Immunology, Laboratory of Parasitology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - O Paerewijck
- Department of Virology, Parasitology and Immunology, Laboratory of Parasitology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - K De Bosscher
- VIB Department of Medical Protein Research, Translational Nuclear Receptor Research Lab, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - P Geldhof
- Department of Virology, Parasitology and Immunology, Laboratory of Parasitology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium.
| |
Collapse
|
17
|
Demirdag YY, Gupta S. Update on Infections in Primary Antibody Deficiencies. Front Immunol 2021; 12:634181. [PMID: 33643318 PMCID: PMC7905085 DOI: 10.3389/fimmu.2021.634181] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 01/07/2021] [Indexed: 11/14/2022] Open
Abstract
Bacterial respiratory tract infections are the hallmark of primary antibody deficiencies (PADs). Because they are also among the most common infections in healthy individuals, PADs are usually overlooked in these patients. Careful evaluation of the history, including frequency, chronicity, and presence of other infections, would help suspect PADs. This review will focus on infections in relatively common PADs, discussing diagnostic challenges, and some management strategies to prevent infections.
Collapse
Affiliation(s)
- Yesim Yilmaz Demirdag
- Division of Basic and Clinical Immunology, Department of Medicine, University of California, Irvine, Irvine, CA, United States
| | | |
Collapse
|
18
|
Fekete E, Allain T, Siddiq A, Sosnowski O, Buret AG. Giardia spp. and the Gut Microbiota: Dangerous Liaisons. Front Microbiol 2021; 11:618106. [PMID: 33510729 PMCID: PMC7835142 DOI: 10.3389/fmicb.2020.618106] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/15/2020] [Indexed: 12/11/2022] Open
Abstract
Alteration of the intestinal microbiome by enteropathogens is commonly associated with gastrointestinal diseases and disorders and has far-reaching consequences for overall health. Significant advances have been made in understanding the role of microbial dysbiosis during intestinal infections, including infection with the protozoan parasite Giardia duodenalis, one of the most prevalent gut protozoa. Altered species composition and diversity, functional changes in the commensal microbiota, and changes to intestinal bacterial biofilm structure have all been demonstrated during the course of Giardia infection and have been implicated in Giardia pathogenesis. Conversely, the gut microbiota has been found to regulate parasite colonization and establishment and plays a critical role in immune modulation during mono and polymicrobial infections. These disruptions to the commensal microbiome may contribute to a number of acute, chronic, and post-infectious clinical manifestations of giardiasis and may account for variations in disease presentation within and between infected populations. This review discusses recent advances in characterizing Giardia-induced bacterial dysbiosis in the gut and the roles of dysbiosis in Giardia pathogenesis.
Collapse
Affiliation(s)
- Elena Fekete
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
- Inflammation Research Network, University of Calgary, Calgary, AB, Canada
- Host-Parasite Interactions, University of Calgary, Calgary, AB, Canada
| | - Thibault Allain
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
- Inflammation Research Network, University of Calgary, Calgary, AB, Canada
- Host-Parasite Interactions, University of Calgary, Calgary, AB, Canada
| | - Affan Siddiq
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
- Inflammation Research Network, University of Calgary, Calgary, AB, Canada
- Host-Parasite Interactions, University of Calgary, Calgary, AB, Canada
| | - Olivia Sosnowski
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
- Inflammation Research Network, University of Calgary, Calgary, AB, Canada
- Host-Parasite Interactions, University of Calgary, Calgary, AB, Canada
| | - Andre G. Buret
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
- Inflammation Research Network, University of Calgary, Calgary, AB, Canada
- Host-Parasite Interactions, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
19
|
Yordanova IA, Lamatsch M, Kühl AA, Hartmann S, Rausch S. Eosinophils are dispensable for the regulation of IgA and Th17 responses in Giardia muris infection. Parasite Immunol 2020; 43:e12791. [PMID: 32918307 DOI: 10.1111/pim.12791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 09/01/2020] [Accepted: 09/02/2020] [Indexed: 11/28/2022]
Abstract
AIMS IgA and Th17 responses are pivotal for the control of Giardia infections. Eosinophils support IgA class switching, the survival of intestinal IgA+ plasma cells at steady state and can control Th17 activity in the small intestine. To see whether eosinophils regulate adaptive immune responses during giardiasis, we investigated Giardia muris infections in wild-type BALB/c and eosinophil-deficient ∆dblGATA-1 mice. METHODS AND RESULTS Infected ∆dblGATA-1 mice did not differ markedly in parasite control from wild-type mice. Confirming previous studies, naive ∆dblGATA-1 mice displayed diminished IgA+ B cell frequencies in Peyer's patches. However, IgA class switching and intestinal IgA secretion in response to G muris infection were comparable in wild-type BALB/c and ∆dblGATA-1 mice. Both strains displayed similarly low intestinal Th17 responses, accompanied by a mild expansion of type 3 innate lymphoid cells (ILC3). CONCLUSIONS Contrasting previous reports on overt small intestinal Th17 activity in eosinophil-deficient mice, IL-17A production is kept in check in the absence of eosinophils during Giardia infection. Suboptimal homeostatic IgA responses in the absence of eosinophils are transiently fostered in infected mice and the maintenance of IgA+ plasma cells appears to be restored during persisting Giardia infection.
Collapse
Affiliation(s)
- Ivet A Yordanova
- Center for Infection Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| | - Martin Lamatsch
- Department of Biology, Chemistry and Pharmacy, Institute of Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Anja A Kühl
- Core Unit for Immunopathology for Experimental Models, iPATH.Berlin, Charité - University Medicine Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Susanne Hartmann
- Center for Infection Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| | - Sebastian Rausch
- Center for Infection Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
20
|
Xu F, Jiménez-González A, Einarsson E, Ástvaldsson Á, Peirasmaki D, Eckmann L, Andersson JO, Svärd SG, Jerlström-Hultqvist J. The compact genome of Giardia muris reveals important steps in the evolution of intestinal protozoan parasites. Microb Genom 2020; 6:mgen000402. [PMID: 32618561 PMCID: PMC7641422 DOI: 10.1099/mgen.0.000402] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 06/16/2020] [Indexed: 01/05/2023] Open
Abstract
Diplomonad parasites of the genus Giardia have adapted to colonizing different hosts, most notably the intestinal tract of mammals. The human-pathogenic Giardia species, Giardia intestinalis, has been extensively studied at the genome and gene expression level, but no such information is available for other Giardia species. Comparative data would be particularly valuable for Giardia muris, which colonizes mice and is commonly used as a prototypic in vivo model for investigating host responses to intestinal parasitic infection. Here we report the draft-genome of G. muris. We discovered a highly streamlined genome, amongst the most densely encoded ever described for a nuclear eukaryotic genome. G. muris and G. intestinalis share many known or predicted virulence factors, including cysteine proteases and a large repertoire of cysteine-rich surface proteins involved in antigenic variation. Different to G. intestinalis, G. muris maintains tandem arrays of pseudogenized surface antigens at the telomeres, whereas intact surface antigens are present centrally in the chromosomes. The two classes of surface antigens engage in genetic exchange. Reconstruction of metabolic pathways from the G. muris genome suggest significant metabolic differences to G. intestinalis. Additionally, G. muris encodes proteins that might be used to modulate the prokaryotic microbiota. The responsible genes have been introduced in the Giardia genus via lateral gene transfer from prokaryotic sources. Our findings point to important evolutionary steps in the Giardia genus as it adapted to different hosts and it provides a powerful foundation for mechanistic exploration of host-pathogen interaction in the G. muris-mouse pathosystem.
Collapse
Affiliation(s)
- Feifei Xu
- Department of Cell and Molecular Biology, BMC, Box 596, Uppsala Universitet, SE-751 24 Uppsala, Sweden
| | | | - Elin Einarsson
- Department of Cell and Molecular Biology, BMC, Box 596, Uppsala Universitet, SE-751 24 Uppsala, Sweden
- Present address: Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Ásgeir Ástvaldsson
- Department of Cell and Molecular Biology, BMC, Box 596, Uppsala Universitet, SE-751 24 Uppsala, Sweden
- Present address: Department of Microbiology, National Veterinary Institute, Uppsala, Sweden
| | - Dimitra Peirasmaki
- Department of Cell and Molecular Biology, BMC, Box 596, Uppsala Universitet, SE-751 24 Uppsala, Sweden
- Present address: Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Lars Eckmann
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Jan O. Andersson
- Department of Cell and Molecular Biology, BMC, Box 596, Uppsala Universitet, SE-751 24 Uppsala, Sweden
| | - Staffan G. Svärd
- Department of Cell and Molecular Biology, BMC, Box 596, Uppsala Universitet, SE-751 24 Uppsala, Sweden
| | - Jon Jerlström-Hultqvist
- Department of Cell and Molecular Biology, BMC, Box 596, Uppsala Universitet, SE-751 24 Uppsala, Sweden
| |
Collapse
|
21
|
Jex AR, Svärd S, Hagen KD, Starcevich H, Emery-Corbin SJ, Balan B, Nosala C, Dawson SC. Recent advances in functional research in Giardia intestinalis. ADVANCES IN PARASITOLOGY 2020; 107:97-137. [PMID: 32122532 PMCID: PMC7878119 DOI: 10.1016/bs.apar.2019.12.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
This review considers current advances in tools to investigate the functional biology of Giardia, it's coding and non-coding genes, features and cellular and molecular biology. We consider major gaps in current knowledge of the parasite and discuss the present state-of-the-art in its in vivo and in vitro cultivation. Advances in in silico tools, including for the modelling non-coding RNAs and genomic elements, as well as detailed exploration of coding genes through inferred homology to model organisms, have provided significant, primary level insight. Improved methods to model the three-dimensional structure of proteins offer new insights into their function, and binding interactions with ligands, other proteins or precursor drugs, and offer substantial opportunities to prioritise proteins for further study and experimentation. These approaches can be supplemented by the growing and highly accessible arsenal of systems-based methods now being applied to Giardia, led by genomic, transcriptomic and proteomic methods, but rapidly incorporating advanced tools for detection of real-time transcription, evaluation of chromatin states and direct measurement of macromolecular complexes. Methods to directly interrogate and perturb gene function have made major leaps in recent years, with CRISPr-interference now available. These approaches, coupled with protein over-expression, fluorescent labelling and in vitro and in vivo imaging, are set to revolutionize the field and herald an exciting time during which the field may finally realise Giardia's long proposed potential as a model parasite and eukaryote.
Collapse
Affiliation(s)
- Aaron R Jex
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Melbourne, VIC, Australia; Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC, Australia.
| | - Staffan Svärd
- Centre for Biomedicine, Uppsala University, Uppsala, Sweden
| | - Kari D Hagen
- College of Biological Sciences, University of California-Davis, Davis, CA, United States
| | - Hannah Starcevich
- College of Biological Sciences, University of California-Davis, Davis, CA, United States
| | - Samantha J Emery-Corbin
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Melbourne, VIC, Australia
| | - Balu Balan
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Melbourne, VIC, Australia
| | - Chris Nosala
- College of Biological Sciences, University of California-Davis, Davis, CA, United States
| | - Scott C Dawson
- College of Biological Sciences, University of California-Davis, Davis, CA, United States
| |
Collapse
|
22
|
|
23
|
RORγt + Treg to Th17 ratios correlate with susceptibility to Giardia infection. Sci Rep 2019; 9:20328. [PMID: 31889073 PMCID: PMC6937251 DOI: 10.1038/s41598-019-56416-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 11/15/2019] [Indexed: 12/20/2022] Open
Abstract
Infections with Giardia are among the most common causes of food and water-borne diarrheal disease worldwide. Here, we investigated Th17, Treg and IgA responses, and alterations in gut microbiota in two mouse lines with varying susceptibility to Giardia muris infection. Infected BALB/c mice shed significantly more cysts compared with C57BL/6 mice. Impaired control of infection in BALB/c mice was associated with lower Th17 activity and lower IgA levels compared with C57BL/6 mice. The limited metabolic activity, proliferation and cytokine production of Th17 cells in BALB/c mice was associated with higher proportions of intestinal Foxp3+RORγt+ regulatory T cells and BALB/c mice developed increased RORγt+ Treg:Th17 ratios in response to G. muris infection. Furthermore, G. muris colonization led to a significantly reduced evenness in the gut microbial communities of BALB/c mice. Our data indicate that differential susceptibility to Giardia infections may be related to RORγt+ Treg controlling Th17 activity and that changes in the microbiota composition upon Giardia infection partially depend on the host background.
Collapse
|
24
|
Swain S, Selmi C, Gershwin ME, Teuber SS. The clinical implications of selective IgA deficiency. J Transl Autoimmun 2019; 2:100025. [PMID: 32743511 PMCID: PMC7388344 DOI: 10.1016/j.jtauto.2019.100025] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 11/21/2019] [Indexed: 01/06/2023] Open
Abstract
Selective IgA deficiency (SIgAD) is the most common primary immunodeficiency but does not always result in clinical disease. This may in part be due to the definition based on serum IgA, while most IgA is secreted at mucosal surfaces, not amenable to measurement. Clinical complications include increased risk of sinopulmonary infections with bacteria and viruses, gastrointestinal infections with a predilection for Giardia lamblia, a myriad of autoimmune diseases including systemic lupus erythematosus, hyper- and hypo-thyroidism, Type 1 diabetes, celiac disease, and rarely, malignancy. SIgAD must be differentiated from IgA deficiency that may be seen with IgG2 or IgG4 deficiency, specific antibody deficiency, or as an early manifestation prior to a diagnosis of common variable immunodeficiency. Secondary IgA deficiency is increasingly recognized and may be due to medications such as anti-epileptics, or antibiotics with disruption of the microbiome which can influence IgA levels, infections or malignancies. Patients with SIgAD should be monitored at regular intervals and educated to be aware of particular complications. There is a rare chance of development of anti-IgA IgE antibodies in patients with complete deficiency, which can result in anaphylaxis if blood products with IgA are administered. Prophylactic antibiotics may be indicated in some cases, and very rarely, supplemental IgG infusions.
Collapse
Affiliation(s)
- Samantha Swain
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, CA, USA
- Veterans Affairs Northern California Healthcare System, Mather, CA, USA
| | - Carlo Selmi
- Rheumatology and Clinical Immunology, Humanitas Research Hospital, Rozzano, Milan, Italy
- BIOMETRA Department, University of Milan, Milan, Italy
| | - M. Eric Gershwin
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, CA, USA
| | - Suzanne S. Teuber
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, CA, USA
- Veterans Affairs Northern California Healthcare System, Mather, CA, USA
| |
Collapse
|
25
|
Emery-Corbin SJ, Grüttner J, Svärd S. Transcriptomic and proteomic analyses of Giardia intestinalis: Intestinal epithelial cell interactions. ADVANCES IN PARASITOLOGY 2019; 107:139-171. [PMID: 32122528 DOI: 10.1016/bs.apar.2019.11.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Giardia intestinalis is a unicellular protozoan parasite that infects the small intestines of humans and animals. Giardiasis, the disease caused by the parasite, occurs globally across socioeconomic boundaries but is mainly endemic in developing countries and particularly within young children, where pronounced effects manifests in a failure to thrive condition. The molecular pathogenesis of Giardia has been studied using in vitro models of human and rat intestinal epithelial cells (IECs) and parasites from the two major human genotypes or assemblages (A and B). High-quality, genome sequencing of representative isolates from assemblages A (WB) and B (GS) has enabled exploration of these host-parasite models using 'omics' technologies, allowing deep and quantitative analyses of global gene expression changes in IECs and parasites during their interactions, cross-talk and competition. These include a major up-regulation of immune-related genes in the IECs early after the start of interactions, as well as competition between host cells and parasites for nutrients like sugars, amino acids and lipids, which is also reflected in their secretome interactions. Unique parasite proteins dominate these interactions, with many major up-regulated genes being either hypothetical proteins or members of Giardia-specific gene families like the high-cysteine-rich membrane proteins (HCMPs), variable surface proteins (VSPs), alpha-giardins and cysteine proteases. Furthermore, these proteins also dominate in the secretomes, suggesting that they are important virulence factors in Giardia and crucial molecular effectors at the host-parasite interface.
Collapse
Affiliation(s)
- Samantha J Emery-Corbin
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Melbourne, VIC, Australia
| | - Jana Grüttner
- Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| | - Staffan Svärd
- Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
26
|
Burrows K, Ngai L, Wong F, Won D, Mortha A. ILC2 Activation by Protozoan Commensal Microbes. Int J Mol Sci 2019; 20:ijms20194865. [PMID: 31574995 PMCID: PMC6801642 DOI: 10.3390/ijms20194865] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 09/26/2019] [Accepted: 09/27/2019] [Indexed: 12/14/2022] Open
Abstract
Group 2 innate lymphoid cells (ILC2s) are a member of the ILC family and are involved in protective and pathogenic type 2 responses. Recent research has highlighted their involvement in modulating tissue and immune homeostasis during health and disease and has uncovered critical signaling circuits. While interactions of ILC2s with the bacterial microbiome are rather sparse, other microbial members of our microbiome, including helminths and protozoans, reveal new and exciting mechanisms of tissue regulation by ILC2s. Here we summarize the current field on ILC2 activation by the tissue and immune environment and highlight particularly new intriguing pathways of ILC2 regulation by protozoan commensals in the intestinal tract.
Collapse
Affiliation(s)
- Kyle Burrows
- University of Toronto, Department of Immunology, Toronto, ON M5S 1A8, Canada.
| | - Louis Ngai
- University of Toronto, Department of Immunology, Toronto, ON M5S 1A8, Canada.
| | - Flora Wong
- University of Toronto, Department of Immunology, Toronto, ON M5S 1A8, Canada.
- Ranomics, Inc. Toronto, ON M5G 1X5, Canada.
| | - David Won
- University of Toronto, Department of Immunology, Toronto, ON M5S 1A8, Canada.
| | - Arthur Mortha
- University of Toronto, Department of Immunology, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
27
|
Identification of Conserved Candidate Vaccine Antigens in the Surface Proteome of Giardia lamblia. Infect Immun 2019; 87:IAI.00219-19. [PMID: 30962402 DOI: 10.1128/iai.00219-19] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 04/01/2019] [Indexed: 01/08/2023] Open
Abstract
Giardia lamblia, one of the most common protozoal infections of the human intestine, is an important worldwide cause of diarrheal disease, malabsorption, malnutrition, delayed cognitive development in children, and protracted postinfectious syndromes. Despite its medical importance, no human vaccine is available against giardiasis. A crude veterinary vaccine has been developed, and experimental vaccines based on expression of multiple variant-specific surface proteins have been reported, but poorly defined vaccine components and excessive antigen variability are problematic for pharmaceutical vaccine production. To expand the repertoire of antigen candidates for vaccines, we reasoned that surface proteins may provide an enriched source of such antigens since key host effectors, such as secretory IgA, can directly bind to such antigens in the intestinal lumen and interfere with epithelial attachment. Here, we have applied a proteomics approach to identify 23 novel surface antigens of G. lamblia that show >90% amino acid sequence identity between the two human-pathogenic genetic assemblages (A and B) of the parasite. Surface localization of a representative subset of these proteins was confirmed by immunostaining. Four selected proteins, uridine phosphorylase-like protein-1, protein 21.1 (GL50803_27925), α1-giardin, and α11-giardin, were subsequently produced in recombinant form and shown to be immunogenic in mice and G. lamblia-infected humans and confer protection against G. lamblia infection upon intranasal immunization in rodent models of giardiasis. These results demonstrate that identification of conserved surface antigens provides a powerful approach for overcoming a key rate-limiting step in the design and construction of an effective vaccine against giardiasis.
Collapse
|
28
|
Immune response markers in sera of children infected with Giardia duodenalis AI and AII subassemblages. Immunobiology 2019; 224:595-603. [PMID: 30962033 DOI: 10.1016/j.imbio.2019.04.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Revised: 04/01/2019] [Accepted: 04/01/2019] [Indexed: 12/29/2022]
Abstract
In this study, we evaluated serum markers of immune responses in children infected with G. duodenalis and compared them with the characterized parasite isolates. The reactivity indexes (RI) of IgG (1.503 ± 0.819) and IgA (2.308 ± 1.935) antibodies were significantly higher (P < 0.001) in infected children than in non-infected children. There were also statistically significantly higher serum levels (P < 0.05) of IFN-γ (393.10 ± 983.90 pg/mL) as well as serum (30.03 ± 10.92 μmol/L) and saliva nitric oxid derivatives (NOx) (192.4 ± 151.2 μmol/L) in children infected with G. duodenalis compared to the group of non-parasitized children (127.4 ± 274.30 pg/mL; 25.82 ± 7.74 μmol/L and 122.5 ± 105.90 μmol/L, respectively). Regarding the characterized genetic variants of G. duodenalis and the immune response profiles, no differences were observed in terms of antibody reactivity or levels of serum cytokine and NOx among children infected with AI or AII subassemblages. The elevated levels of IFN-γ and NOx indicate that G. duodenalis intestinal infection in humans induces a cellular immune response detectable at the systemic level. Moreover, no significant differences in the antibody reactivity profile or the cytokine and NOx production in the sera of children infected with AI or AII G. duodenalis variants were observed, suggesting that subtypes of the parasite do not influence the immune response profile.
Collapse
|
29
|
Liu J, Fu Z, Hellman L, Svärd SG. Cleavage specificity of recombinant Giardia intestinalis cysteine proteases: Degradation of immunoglobulins and defensins. Mol Biochem Parasitol 2019; 227:29-38. [DOI: 10.1016/j.molbiopara.2018.10.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 10/17/2018] [Accepted: 10/26/2018] [Indexed: 12/11/2022]
|
30
|
de Andrade CM, Carneiro VL, Cerqueira JV, Fonseca HF, Queiroz GA, Costa RS, Alcantara-Neves NM, Cooper P, Figueiredo CA. Parasites and allergy: Observations from Brazil. Parasite Immunol 2018; 41:e12588. [PMID: 30188574 DOI: 10.1111/pim.12588] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 07/24/2018] [Accepted: 09/04/2018] [Indexed: 11/29/2022]
Abstract
Brazil is a middle-income country undergoing the epidemiological transition. Effects of changes in daily life habits and access to clean water, sanitation and urban services on a growing urban population have contributed to a double burden of both infectious and noncommunicable chronic diseases. Studies have indicated that parasite infections may modulate the human immune system and influence the development of allergic conditions such as asthma. However, there is no consensus in the published literature on the effects of parasitic infections on allergy, perhaps as a consequence of factors determining the epidemiology of these infections that vary between populations such as age of first infection, duration and chronicity of infections, parasite burden and species, and host genetic susceptibility. In this review, we discuss the observations from Brazil concerning the relationship between parasite infections and allergy.
Collapse
Affiliation(s)
| | - Valdirene L Carneiro
- Departamento de Ciências da Vida, Universidade do Estado da Bahia, Salvador, Brazil
| | - Jéssica V Cerqueira
- Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Hellen F Fonseca
- Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Gerson A Queiroz
- Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Ryan S Costa
- Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | | | - Philip Cooper
- St. George's University of London, London, UK.,Facultad de Ciencias Medicas de la Salud y la Vida, Universidad Internacional del Ecuador, Quito, Ecuador
| | - Camila A Figueiredo
- Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| |
Collapse
|
31
|
Robak OH, Heimesaat MM, Kruglov AA, Prepens S, Ninnemann J, Gutbier B, Reppe K, Hochrein H, Suter M, Kirschning CJ, Marathe V, Buer J, Hornef MW, Schnare M, Schneider P, Witzenrath M, Bereswill S, Steinhoff U, Suttorp N, Sander LE, Chaput C, Opitz B. Antibiotic treatment-induced secondary IgA deficiency enhances susceptibility to Pseudomonas aeruginosa pneumonia. J Clin Invest 2018; 128:3535-3545. [PMID: 29771684 DOI: 10.1172/jci97065] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 05/10/2018] [Indexed: 12/13/2022] Open
Abstract
Broad-spectrum antibiotics are widely used with patients in intensive care units (ICUs), many of whom develop hospital-acquired infections with Pseudomonas aeruginosa. Although preceding antimicrobial therapy is known as a major risk factor for P. aeruginosa-induced pneumonia, the underlying mechanisms remain incompletely understood. Here we demonstrate that depletion of the resident microbiota by broad-spectrum antibiotic treatment inhibited TLR-dependent production of a proliferation-inducing ligand (APRIL), resulting in a secondary IgA deficiency in the lung in mice and human ICU patients. Microbiota-dependent local IgA contributed to early antibacterial defense against P. aeruginosa. Consequently, P. aeruginosa-binding IgA purified from lamina propria culture or IgA hybridomas enhanced resistance of antibiotic-treated mice to P. aeruginosa infection after transnasal substitute. Our study provides a mechanistic explanation for the well-documented risk of P. aeruginosa infection following antimicrobial therapy, and we propose local administration of IgA as a novel prophylactic strategy.
Collapse
Affiliation(s)
- Oliver H Robak
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Department of Medicine 1, Medical University of Vienna, Vienna, Austria
| | - Markus M Heimesaat
- Institute of Microbiology and Hygiene, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Andrey A Kruglov
- German Rheumatism Research Center, a Leibniz Institute, Berlin, Germany.,A.N. Belozersky Institute of Physico-Chemical Biology and Department of Immunology, Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Sandra Prepens
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Justus Ninnemann
- German Rheumatism Research Center, a Leibniz Institute, Berlin, Germany
| | - Birgitt Gutbier
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Katrin Reppe
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Hubertus Hochrein
- Department of Research Immunology, Bavarian Nordic GmbH, Martinsried, Germany
| | - Mark Suter
- Universität Zürich, Vetsuisse, Zurich, Switzerland
| | | | - Veena Marathe
- Institute of Medical Microbiology, University of Duisburg-Essen, Essen, Germany
| | - Jan Buer
- Institute of Medical Microbiology, University of Duisburg-Essen, Essen, Germany
| | - Mathias W Hornef
- Institute of Medical Microbiology, University Hospital, Rheinisch-Westfälische Technische Hochschule (RWTH), Aachen, Germany
| | - Markus Schnare
- Institute of Immunology, University of Marburg, Marburg, Germany
| | - Pascal Schneider
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Martin Witzenrath
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,German Center for Lung Research (DZL), Giessen, Germany
| | - Stefan Bereswill
- Institute of Microbiology and Hygiene, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Ulrich Steinhoff
- Institute of Medical Microbiology and Hygiene, University of Marburg, Marburg, Germany
| | - Norbert Suttorp
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,German Center for Lung Research (DZL), Giessen, Germany
| | - Leif E Sander
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,German Center for Lung Research (DZL), Giessen, Germany
| | - Catherine Chaput
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Bastian Opitz
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,German Center for Lung Research (DZL), Giessen, Germany
| |
Collapse
|
32
|
Cytokines, Antibodies, and Histopathological Profiles during Giardia Infection and Variant-Specific Surface Protein-Based Vaccination. Infect Immun 2018; 86:IAI.00773-17. [PMID: 29555679 DOI: 10.1128/iai.00773-17] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 03/10/2018] [Indexed: 12/13/2022] Open
Abstract
Giardiasis is one of the most common human intestinal diseases worldwide. Several experimental animal models have been used to evaluate Giardia infections, with gerbils (Meriones unguiculatus) being the most valuable model due to their high susceptibility to Giardia infection, abundant shedding of cysts, and pathophysiological alterations and signs of disease similar to those observed in humans. Here, we report cytokine and antibody profiles both during the course of Giardia infection in gerbils and after immunization with a novel oral vaccine comprising a mixture of purified variant-specific surface proteins (VSPs). Transcript levels of representative cytokines of different immune profiles as well as macro- and microtissue alterations were assessed in Peyer's patches, mesenteric lymph nodes, and spleens. During infection, cytokine responses showed a biphasic profile: an early induction of Th1 (gamma interferon [IFN-γ], interleukin-1β [IL-1β], IL-6, and tumor necrosis factor [TNF]), Th17 (IL-17), and Th2 (IL-4) cytokines, together with intestinal alterations typical of inflammation, followed by a shift toward a predominant Th2 (IL-5) response, likely associated with a counterregulatory mechanism. Conversely, immunization with an oral vaccine comprising the entire repertoire of VSPs specifically showed high levels of IL-17, IL-6, IL-4, and IL-5, without obvious signs of inflammation. Both immunized and infected animals developed local (intestinal secretory IgA [S-IgA]) and systemic (serum IgG) humoral immune responses against VSPs; however, only infected animals showed evident signs of giardiasis. This is the first comprehensive report of cytokine expression and anti-Giardia antibody production during infection and VSP vaccination in gerbils, a reliable model of the human disease.
Collapse
|
33
|
Cacciò SM, Lalle M, Svärd SG. Host specificity in the Giardia duodenalis species complex. INFECTION GENETICS AND EVOLUTION 2017; 66:335-345. [PMID: 29225147 DOI: 10.1016/j.meegid.2017.12.001] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 12/01/2017] [Accepted: 12/02/2017] [Indexed: 12/15/2022]
Abstract
Giardia duodenalis is a unicellular flagellated parasite that infects the gastrointestinal tract of a wide range of mammalian species, including humans. Investigations of protein and DNA polymorphisms revealed that G. duodenalis should be considered as a species complex, whose members, despite being morphologically indistinguishable, can be classified into eight groups, or Assemblages, separated by large genetic distances. Assemblages display various degree of host specificity, with Assemblages A and B occurring in humans and many other hosts, Assemblage C and D in canids, Assemblage E in hoofed animals, Assemblage F in cats, Assemblage G in rodents, and Assemblage H in pinnipeds. The factors determining host specificity are only partially understood, and clearly involve both the host and the parasite. Here, we review the results of in vitro and in vivo experiments, and clinical observations to highlight relevant biological and genetic differences between Assemblages, with a focus on human infection.
Collapse
Affiliation(s)
- Simone M Cacciò
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy.
| | - Marco Lalle
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Staffan G Svärd
- Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
34
|
Ashour DS, Saad AE, Dawood LM, Zamzam Y. Immunological interaction between Giardia cyst extract and experimental toxoplasmosis. Parasite Immunol 2017; 40. [PMID: 29130475 DOI: 10.1111/pim.12503] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 11/07/2017] [Indexed: 12/16/2022]
Abstract
Toxoplasmosis is mostly associated with other intestinal parasitic infections especially Giardia due to shared mode of peroral infection. Toxoplasma and Giardia induce a strong T-helper 1- immune response. Our aim was to induce a protective immune response that results in significant impact on intestinal and extra-intestinal phases of Toxoplasma infection. This study was conducted in experimental animals and assessment of Giardia cyst extract effect on Toxoplasma infection was investigated by histopathological examination of small intestine and brain, Toxoplasma cyst count and iNOS staining of the brain, measurement of IFN-γ and TGF-β in intestinal tissues. Results showed that the brain Toxoplasma cyst number was decreased in mice infected with Toxoplasma then received Giardia cyst extract as compared to mice infected with Toxoplasma only. This effect was produced because Giardia cyst extract augmented the immune response to Toxoplasma infection as evidenced by severe inflammatory reaction in the intestinal and brain tissues, increased levels of IFN-γ and TGF-β in intestinal tissues and strong iNOS staining of the brain. In conclusion, Giardia cyst extract generated a protective response against T. gondii infection. Therefore, Giardia antigen will be a suitable candidate for further researches as an immunomodulatory agent against Toxoplasma infection.
Collapse
Affiliation(s)
- D S Ashour
- Medical Parasitology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - A E Saad
- Medical Parasitology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - L M Dawood
- Biochemistry Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Y Zamzam
- Pathology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| |
Collapse
|
35
|
Fink MY, Singer SM. The Intersection of Immune Responses, Microbiota, and Pathogenesis in Giardiasis. Trends Parasitol 2017; 33:901-913. [PMID: 28830665 DOI: 10.1016/j.pt.2017.08.001] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 07/18/2017] [Accepted: 08/01/2017] [Indexed: 02/07/2023]
Abstract
Giardia lamblia is one of the most common infectious protozoans in the world. Giardia rarely causes severe life-threatening diarrhea, and may even have a slight protective effect in this regard, but it is a major contributor to malnutrition and growth faltering in children in the developing world. Giardia infection also appears to be a significant risk factor for postinfectious irritable bowel and chronic fatigue syndromes. In this review we highlight recent work focused on the impact of giardiasis and the mechanisms that contribute to the various outcomes of this infection, including changes in the composition of the microbiota, activation of immune responses, and immunopathology.
Collapse
Affiliation(s)
- Marc Y Fink
- Department of Biology, Georgetown University, Washington, DC, USA
| | - Steven M Singer
- Department of Biology, Georgetown University, Washington, DC, USA.
| |
Collapse
|
36
|
Interleukin-17 receptor A (IL-17RA) as a central regulator of the protective immune response against Giardia. Sci Rep 2017; 7:8520. [PMID: 28819174 PMCID: PMC5561107 DOI: 10.1038/s41598-017-08590-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 07/12/2017] [Indexed: 12/16/2022] Open
Abstract
The protozoan parasite Giardia is a highly prevalent intestinal pathogen with a wide host range. Data obtained in mice, cattle and humans revealed the importance of IL-17A in the development of a protective immune response against Giardia. The aim of this study was to further unravel the protective effector mechanisms triggered by IL-17A following G. muris infection in mice, by an RNA-sequencing approach. C57BL/6 WT and C57BL/6 IL-17RA KO mice were orally infected with G. muris cysts. Three weeks post infection, intestinal tissue samples were collected for RNA-sequencing, with samples from uninfected C57BL/6 WT and C57BL/6 IL-17RA KO animals serving as negative controls. Differential expression analysis showed that G. muris infection evoked the transcriptional upregulation of a wide array of genes, mainly in animals with competent IL-17RA signaling. IL-17RA signaling induced the production of various antimicrobial peptides, such as angiogenin 4 and α- and β-defensins and regulated complement activation through mannose-binding lectin 2. The expression of the receptor that regulates the secretion of IgA into the intestinal lumen, the polymeric immunoglobulin receptor, was also dependent on IL-17RA signaling. Interestingly, the transcriptome data showed for the first time the involvement of the circadian clock in the host response following Giardia infection.
Collapse
|
37
|
Quintero J, Valdez A, Samaniego B, Lopez-Romero G, Astiazaran-Garcia H, Rascon L, Breci L, Garibay-Escobar A, Robles-Zepeda R, Velazquez C. Isolation and partial characterization of an immunogenic antigen of Giardia lamblia. Parasitol Int 2017; 66:324-330. [DOI: 10.1016/j.parint.2017.01.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 11/26/2016] [Accepted: 01/13/2017] [Indexed: 11/26/2022]
|
38
|
In vitro immunomodulatory effects of microemulsions with levamisole delivery systems on blood phagocytes interacting with Giardia lamblia. Parasitol Int 2017; 66:299-304. [DOI: 10.1016/j.parint.2017.02.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 02/11/2017] [Accepted: 02/22/2017] [Indexed: 11/19/2022]
|
39
|
Shan YH, Zhang YG, Zhang JH, Wang D, Li XX, Zhang J, Wang XM, Luo SY. The physiological effects of human immunoglobulin on severe bronchiolitis patients before and after treatment. Hum Vaccin Immunother 2016; 11:2647-53. [PMID: 26308393 DOI: 10.1080/21645515.2015.1080401] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The goal of the present study is to explore the physiological effects of injected human immunoglobulin on patients with severe bronchiolitis before and after treatment. 86 young children with severe bronchiolitis were randomly divided into the observation group (43 cases) and the treatment group (43 cases). On the basis of conventional therapy, the children in the treatment group were given human immunoglobulin (400 mg/kg, 1-3 times) via intravenous injection. 60 healthy young children, as determined by a physical examination given at the Zhumadian Central Hospital, were enrolled as the control group. The T lymphocytes, cytokines, IgA, IgG, and IgM immunoglobulins in the peripheral blood of all 3 groups were measured. The clinical efficacy of the immunoglobulins to mitigate the effects of bronchiolitis and the amount of time for the reduction of symptoms to occur were observed. The serum Ca, Fe, and Zn levels of children with severe bronchiolitis were significantly lower than those of the healthy control group (p < 0.05). As such, the CD8, IgA, IgG, IgM and IFN-γ levels were also significantly lower in the children with severe bronchiolitis than in the children in the healthy control group (p < 0.05). Furthermore, the CD4, IgE, IL-4, and IL-4/IFN-γ levels and CD4/CD8 ratio were dramatically higher than in the healthy control group (p < 0.05). Serum levels of the aforementioned indicators either increased or decreased after IVIG treatment. The amount of time required for coughing, wheezing, and pulmonary rales to seize, and the duration of illness for the children with the severe bronchiolitis children was significantly shorter for those in the treatment group than for those in the observation group. Human immunoglobulin via intravenous injection showed active therapeutical effects on trace elements, T lymphocytes, and cytokines in patients with severe bronchiolitis.
Collapse
Affiliation(s)
- Yan-Hua Shan
- a Intensive Care Unit, Zhumadian Central Hospital ; Zhumadian , Henan Province , China
| | - Yong-Gang Zhang
- a Intensive Care Unit, Zhumadian Central Hospital ; Zhumadian , Henan Province , China
| | - Jian-Hua Zhang
- b School of Electric Engineering, Zhengzhou University ; Zhengzhou , Henan Province , China
| | - Dong Wang
- a Intensive Care Unit, Zhumadian Central Hospital ; Zhumadian , Henan Province , China
| | - Xiao-Xia Li
- a Intensive Care Unit, Zhumadian Central Hospital ; Zhumadian , Henan Province , China
| | - Jie Zhang
- a Intensive Care Unit, Zhumadian Central Hospital ; Zhumadian , Henan Province , China
| | - Xi-Mei Wang
- a Intensive Care Unit, Zhumadian Central Hospital ; Zhumadian , Henan Province , China
| | - Song-Yuan Luo
- a Intensive Care Unit, Zhumadian Central Hospital ; Zhumadian , Henan Province , China
| |
Collapse
|
40
|
Xiao C, Bao G, Liu Y, Wei Q, Ji Q, Liu Y, Pan L. Greater efficacy of the ECMS-oil adjuvant over other formulations on immune responses against Bordetella bronchiseptica in rabbits and the underlying mechanism. Int Immunopharmacol 2016; 38:194-203. [PMID: 27288753 DOI: 10.1016/j.intimp.2016.06.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 05/31/2016] [Accepted: 06/01/2016] [Indexed: 12/31/2022]
Abstract
In this study, the adjuvant effects of the extract of Cochinchina momordica seed ECMS+oil, oil alone, ECMS alone, conventional alum adjuvant on inactivated Bordetella bronchiseptica (Bb) vaccine or control using antigen alone without adjuvant were evaluated along with the underlying mechanism. The results in experiment A demonstrated that antibody levels in Bb whole cell protein in the ECMS800μg+oil group were significantly higher than in the other adjuvant groups (p<0.05) on day 21. The agglutination antibody titer was also higher than the other groups (p<0.05) on day 37. The ECMS800μg+oil group improved cellular immune responses compared to other adjuvant groups, including control using antigen alone without adjuvant and the PBS group (p<0.05). After Bb challenge, the ECMS800μg+oil group showed the highest protection rate, which was significantly higher than ECMS alone or control using antigen alone without adjuvant and the PBS group (p<0.05 and p<0.01). IgA cells in the ECMS800μg+oil group differed significantly from the IgA cells of other groups in the lungs (p<0.01). The results of cell recruitment showed that the number of lymphocytes in the ECMS400μg+oil were higher than the number of cells for other groups except the ECMS(100μg/800μg)+oil groups (p<0.05). Intermediate cells in the ECMS(100μg/400μg)+oil groups were higher than the number of cells for other groups, including the control using antigen alone group (p<0.05). Neutrophils in the ECMS(100μg/400μg/800μg)+oil groups were significantly higher than the ECMS 800μg and control using antigen alone groups (p<0.05). White blood cells in the ECMS100μg+oil group were significantly higher than the oil, ECMS800μg and control using antigen alone groups (p<0.05). IL-2 expression in the ECMS800μg+oil group was significantly higher than other groups, except for the ECMS400μg+oil group (p<0.05). IL-4 expression in the ECMS800μg+oil group was significantly higher than other groups (p<0.05). GATA3 in the ECMS800μg+oil groups was significantly higher than the oil, ECMS800μg and control using antigen alone group (p<0.05). ECMS-oil adjuvant mixture could most effectively protect B. bronchiseptica immunized rabbits and, therefore, could be an alternative way of improving B. bronchiseptica vaccination in rabbits.
Collapse
Affiliation(s)
- Chenwen Xiao
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang 310021, PR China
| | - Guolian Bao
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang 310021, PR China.
| | - Yan Liu
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang 310021, PR China
| | - Qiang Wei
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang 310021, PR China
| | - Quanan Ji
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang 310021, PR China
| | - Yaping Liu
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang 310021, PR China
| | - Lijun Pan
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang 310021, PR China
| |
Collapse
|
41
|
Srinivasan P, Lawa HR, Rosado JL, Al Mamun A, Khatun M, Santos JI, Utzinger J, Long KZ. Household and personal factors are sources of heterogenity in intestinal parasite clearance among Mexican children 6-15 months of age supplemented with vitamin A and zinc. Acta Trop 2016; 156:48-56. [PMID: 26772449 DOI: 10.1016/j.actatropica.2015.12.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 12/08/2015] [Accepted: 12/11/2015] [Indexed: 11/30/2022]
Abstract
A randomised, double-blind, placebo-controlled trial was carried out among Mexico children aged 6-15 months to determine how household characteristics modify vitamin A and zinc supplementation efficacy on Ascaris lumbricoides, Giardia intestinalis and Entamoeba histolytica/E. dispar infection durations. Children assigned to receive vitamin A every 2 months, a daily zinc supplement, a combined vitamin A-zinc supplement or a placebo were followed for 1 year. Parametric hazard models were fit to infection durations stratified by personal and household factors. Children supplemented with vitamin A and zinc combined from households lacking piped water and children in all three treatment arms from households with dirt floors had longer G. intestinalis and A. lumbricoides infection durations than their counterparts, respectively. Shorter E. histolytica/E.dispar durations were found among zinc-supplemented children of mothers who had <6 years of education and no indoor bathrooms. Heterogeneity in supplementation efficacy among children may reflect differences in exposure risk and baseline immune responses.
Collapse
Affiliation(s)
| | | | - Jorge L Rosado
- School of Natural Sciences, Universidad Autónoma de Querétaro, Querétaro, Mexico
| | - Abdullah Al Mamun
- University of Queensland School of Population Health, Herston, Qld, Australia
| | - Mohsina Khatun
- QIMR Berghofer Medical Research Institute, Herston, Australia
| | - José I Santos
- School of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| | - Jürg Utzinger
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel, Switzerland; University of Basel, Basel, Switzerland
| | - Kurt Z Long
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel, Switzerland; University of Basel, Basel, Switzerland.
| |
Collapse
|
42
|
Biomarkers of Gastrointestinal Host Responses to Microbial Infections. Mol Microbiol 2016. [DOI: 10.1128/9781555819071.ch46] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
43
|
Ruane D, Chorny A, Lee H, Faith J, Pandey G, Shan M, Simchoni N, Rahman A, Garg A, Weinstein EG, Oropallo M, Gaylord M, Ungaro R, Cunningham-Rundles C, Alexandropoulos K, Mucida D, Merad M, Cerutti A, Mehandru S. Microbiota regulate the ability of lung dendritic cells to induce IgA class-switch recombination and generate protective gastrointestinal immune responses. J Exp Med 2015; 213:53-73. [PMID: 26712806 PMCID: PMC4710201 DOI: 10.1084/jem.20150567] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 12/01/2015] [Indexed: 12/12/2022] Open
Abstract
Ruane et al. demonstrate a role for the microbiota in modulating protective immunity to intranasal vaccination via the ability of lung dendritic cells to induce B cell IgA class switching. Protective immunoglobulin A (IgA) responses to oral antigens are usually orchestrated by gut dendritic cells (DCs). Here, we show that lung CD103+ and CD24+CD11b+ DCs induced IgA class-switch recombination (CSR) by activating B cells through T cell–dependent or –independent pathways. Compared with lung DCs (LDC), lung CD64+ macrophages had decreased expression of B cell activation genes and induced significantly less IgA production. Microbial stimuli, acting through Toll-like receptors, induced transforming growth factor-β (TGF-β) production by LDCs and exerted a profound influence on LDC-mediated IgA CSR. After intranasal immunization with inactive cholera toxin (CT), LDCs stimulated retinoic acid–dependent up-regulation of α4β7 and CCR9 gut-homing receptors on local IgA-expressing B cells. Migration of these B cells to the gut resulted in IgA-mediated protection against an oral challenge with active CT. However, in germ-free mice, the levels of LDC-induced, CT–specific IgA in the gut are significantly reduced. Herein, we demonstrate an unexpected role of the microbiota in modulating the protective efficacy of intranasal vaccination through their effect on the IgA class-switching function of LDCs.
Collapse
Affiliation(s)
- Darren Ruane
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029 The Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Alejo Chorny
- The Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Haekyung Lee
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029 The Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Jeremiah Faith
- The Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Gaurav Pandey
- Department of Genetics and Genomic Sciences and Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Meimei Shan
- The Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Noa Simchoni
- The Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Adeeb Rahman
- The Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Aakash Garg
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Erica G Weinstein
- The Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Michael Oropallo
- The Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Michelle Gaylord
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029 The Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Ryan Ungaro
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | | | | | - Daniel Mucida
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY 10065
| | - Miriam Merad
- The Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Andrea Cerutti
- The Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Saurabh Mehandru
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029 The Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| |
Collapse
|
44
|
Miyamoto Y, Eckmann L. Drug Development Against the Major Diarrhea-Causing Parasites of the Small Intestine, Cryptosporidium and Giardia. Front Microbiol 2015; 6:1208. [PMID: 26635732 PMCID: PMC4652082 DOI: 10.3389/fmicb.2015.01208] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 10/16/2015] [Indexed: 12/23/2022] Open
Abstract
Diarrheal diseases are among the leading causes of morbidity and mortality in the world, particularly among young children. A limited number of infectious agents account for most of these illnesses, raising the hope that advances in the treatment and prevention of these infections can have global health impact. The two most important parasitic causes of diarrheal disease are Cryptosporidium and Giardia. Both parasites infect predominantly the small intestine and colonize the lumen and epithelial surface, but do not invade deeper mucosal layers. This review discusses the therapeutic challenges, current treatment options, and drug development efforts against cryptosporidiosis and giardiasis. The goals of drug development against Cryptosporidium and Giardia are different. For Cryptosporidium, only one moderately effective drug (nitazoxanide) is available, so novel classes of more effective drugs are a high priority. Furthermore, new genetic technology to identify potential drug targets and better assays for functional evaluation of these targets throughout the parasite life cycle are needed for advancing anticryptosporidial drug design. By comparison, for Giardia, several classes of drugs with good efficacy exist, but dosing regimens are suboptimal and emerging resistance begins to threaten clinical utility. Consequently, improvements in potency and dosing, and the ability to overcome existing and prevent new forms of drug resistance are priorities in antigiardial drug development. Current work on new drugs against both infections has revealed promising strategies and new drug leads. However, the primary challenge for further drug development is the underlying economics, as both parasitic infections are considered Neglected Diseases with low funding priority and limited commercial interest. If a new urgency in medical progress against these infections can be raised at national funding agencies or philanthropic organizations, meaningful and timely progress is possible in treating and possibly preventing cryptosporidiosis and giardiasis.
Collapse
Affiliation(s)
- Yukiko Miyamoto
- Department of Medicine, University of California at San Diego, La Jolla CA, USA
| | - Lars Eckmann
- Department of Medicine, University of California at San Diego, La Jolla CA, USA
| |
Collapse
|
45
|
|
46
|
Šlapeta J, Dowd SE, Alanazi AD, Westman ME, Brown GK. Differences in the faecal microbiome of non-diarrhoeic clinically healthy dogs and cats associated with Giardia duodenalis infection: impact of hookworms and coccidia. Int J Parasitol 2015; 45:585-94. [DOI: 10.1016/j.ijpara.2015.04.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 04/07/2015] [Accepted: 04/09/2015] [Indexed: 02/07/2023]
|
47
|
Nurkic J, Numanovic F, Arnautalic L, Tihic N, Halilovic D, Jahic M. Diagnostic Significance of Reduced IgA in Children. Med Arch 2015; 69:236-9. [PMID: 26543309 PMCID: PMC4610608 DOI: 10.5455/medarh.2015.69.236-239] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 07/19/2015] [Indexed: 01/06/2023] Open
Abstract
Introduction: The finding of reduced value of immunoglobulin A (IgA) in children is frequent in daily medical practice. It is important to correctly interpret the findings as adequate further diagnostic evaluation of the patient in order to make the determination on the significance of such findings. In children younger than 4 years always consider the transient impairment of immunoglobulins, maturation of child and his immune system can lead to an improvement in the clinical picture. In older children decreased IgA may lead to serious illnesses that need to be recognize and acknowledge through the appropriate diagnostic methods. At the University Clinical Center Tuzla, children with suspected deficient immune response due to reduced values of IgA, goes through further diagnostic evaluation at the Polyclinic for Laboratory Medicine, Department of Immunology and Department of Microbiology, as well as the Clinic of Radiology. Material and methods: Our study followed 91 patients, for the year 2013, through their medical charts and made evaluation of diagnostic and screening tests. Conclusion: The significance of this paper is to draw attention to the importance of diagnostic approach to IgA deficient pediatric patient and relevance of knowledge of individual diagnostic methods as well as to the proper interpretation of the results thereof.
Collapse
Affiliation(s)
- Jasmina Nurkic
- Policlinic for laboratory diagnostic. University Clinical Centre Tuzla, Tuzla, Bosnia and Herzegovina
| | - Fatima Numanovic
- Policlinic for laboratory diagnostic. University Clinical Centre Tuzla, Tuzla, Bosnia and Herzegovina
| | - Lejla Arnautalic
- Clinic for Radiology. University Clinical Centre Tuzla, Tuzla, Bosnia and Herzegovina
| | - Nijaz Tihic
- Policlinic for laboratory diagnostic. University Clinical Centre Tuzla, Tuzla, Bosnia and Herzegovina
| | - Dzenan Halilovic
- Clinic for Pulmonary Disease. University Clinical Centre Tuzla, Tuzla, Bosnia and Herzegovina
| | - Mahira Jahic
- Policlinic for laboratory diagnostic. University Clinical Centre Tuzla, Tuzla, Bosnia and Herzegovina
| |
Collapse
|
48
|
Lopez-Romero G, Quintero J, Astiazarán-García H, Velazquez C. Host defences againstGiardia lamblia. Parasite Immunol 2015; 37:394-406. [DOI: 10.1111/pim.12210] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 06/08/2015] [Indexed: 02/06/2023]
Affiliation(s)
- G. Lopez-Romero
- Coordinación de Nutrición; Centro de Investigación en Alimentación y Desarrollo A.C.; Hermosillo Sonora México
| | - J. Quintero
- Department of Chemistry-Biology; University of Sonora; Hermosillo Sonora México
| | - H. Astiazarán-García
- Coordinación de Nutrición; Centro de Investigación en Alimentación y Desarrollo A.C.; Hermosillo Sonora México
| | - C. Velazquez
- Department of Chemistry-Biology; University of Sonora; Hermosillo Sonora México
| |
Collapse
|
49
|
Dann SM, Manthey CF, Le C, Miyamoto Y, Gima L, Abrahim A, Cao AT, Hanson EM, Kolls JK, Raz E, Cong Y, Eckmann L. IL-17A promotes protective IgA responses and expression of other potential effectors against the lumen-dwelling enteric parasite Giardia. Exp Parasitol 2015; 156:68-78. [PMID: 26071205 DOI: 10.1016/j.exppara.2015.06.003] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 05/14/2015] [Accepted: 06/04/2015] [Indexed: 12/17/2022]
Abstract
Giardia lamblia is a leading protozoan cause of diarrheal disease worldwide. It colonizes the lumen and epithelial surface of the small intestine, but does not invade the mucosa. Acute infection causes only minimal mucosal inflammation. Effective immune defenses exist, yet their identity and mechanisms remain incompletely understood. Interleukin (IL)-17A has emerged as an important cytokine involved in inflammation and antimicrobial defense against bacterial pathogens at mucosal surfaces. In this study, we demonstrate that IL-17A has a crucial function in host defense against Giardia infection. Using murine infection models with G. muris and G. lamblia, we observed marked and selective induction of intestinal IL-17A with peak expression after 2 weeks. Th17 cells in the lamina propria and innate immune cells in the epithelial compartment of the small intestine were responsible for the IL-17A response. Experiments in gene-targeted mice revealed that the cytokine, and its cognate receptor IL-17RA, were required for eradication of the parasite. The actions of the cytokine were mediated by hematopoietic cells, and were required for the transport of IgA into the intestinal lumen, since IL-17A deficiency led to marked reduction of fecal IgA levels, as well as for increased intestinal expression of several other potential effectors, including β-defensin 1 and resistin-like molecule β. In contrast, intestinal hypermotility, another major antigiardial defense mechanism, was not impacted by IL-17A loss. Taken together, these findings demonstrate that IL-17A and IL-17 receptor signaling are essential for intestinal defense against the important lumen-dwelling intestinal parasite Giardia.
Collapse
Affiliation(s)
- Sara M Dann
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, USA; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Carolin F Manthey
- Department of Medicine, University of California, San Diego, CA, USA
| | - Christine Le
- Department of Medicine, University of California, San Diego, CA, USA
| | - Yukiko Miyamoto
- Department of Medicine, University of California, San Diego, CA, USA
| | - Lauren Gima
- Department of Medicine, University of California, San Diego, CA, USA
| | - Andrew Abrahim
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, USA
| | - Anthony T Cao
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Elaine M Hanson
- Department of Medicine, University of California, San Diego, CA, USA
| | - Jay K Kolls
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Eyal Raz
- Department of Medicine, University of California, San Diego, CA, USA
| | - Yingzi Cong
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA; Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Lars Eckmann
- Department of Medicine, University of California, San Diego, CA, USA.
| |
Collapse
|
50
|
Bartelt LA, Sartor RB. Advances in understanding Giardia: determinants and mechanisms of chronic sequelae. F1000PRIME REPORTS 2015; 7:62. [PMID: 26097735 PMCID: PMC4447054 DOI: 10.12703/p7-62] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Giardia lamblia is a flagellated protozoan that is the most common cause of intestinal parasitic infection in children living in resource-limited settings. The pathogenicity of Giardia has been debated since the parasite was first identified, and clinical outcomes vary across studies. Among recent perplexing findings are diametrically opposed associations between Giardia and acute versus persistent diarrhea and a poorly understood potential for long-term sequelae, including impaired child growth and cognitive development. The mechanisms driving these protean clinical outcomes remain elusive, but recent advances suggest that variability in Giardia strains, host nutritional status, the composition of microbiota, co-infecting enteropathogens, host genetically determined mucosal immune responses, and immune modulation by Giardia are all relevant factors influencing disease manifestations after Giardia infection.
Collapse
Affiliation(s)
- Luther A. Bartelt
- Division of Infectious Diseases and International Health, University of VirginiaBox 801340, Charlottesville, VA 22908USA
| | - R. Balfour Sartor
- Division of Gastroenterology, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel HillCampus Box 7032, Chapel Hill, NC 27599-7032USA
| |
Collapse
|