1
|
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
|
2
|
Sardinha-Silva A, Gazzinelli-Guimaraes PH, Ajakaye OG, Ferreira TR, Alves-Ferreira EV, Tjhin ET, Gregg B, Fink MY, Coelho CH, Singer SM, Grigg ME. Giardia intestinalis reshapes mucosal immunity toward a Type 2 response that attenuates inflammatory bowel-like diseases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.02.583119. [PMID: 38903060 PMCID: PMC11188066 DOI: 10.1101/2024.03.02.583119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
Diarrheal diseases are the second leading cause of death in children worldwide. Epidemiological studies show that co-infection with Giardia intestinalis decreases the severity of diarrhea. Here, we show that Giardia is highly prevalent in the stools of asymptomatic school-aged children. It orchestrates a Th2 mucosal immune response, characterized by increased antigen-specific Th2 cells, IL-25, Type 2-associated cytokines, and goblet cell hyperplasia. Giardia infection expanded IL-10-producing Th2 and GATA3+ Treg cells that promoted chronic carriage, parasite transmission, and conferred protection against Toxoplasma gondii-induced lethal ileitis and DSS-driven colitis by downregulating proinflammatory cytokines, decreasing Th1/Th17 cell frequency, and preventing collateral tissue damage. Protection was dependent on STAT6 signaling, as Giardia-infected STAT6-/- mice no longer regulated intestinal bystander inflammation. Our findings demonstrate that Giardia infection reshapes mucosal immunity toward a Type 2 response, which confers a mutualistic protection against inflammatory disease processes and identifies a critical role for protists in regulating mucosal defenses.
Collapse
Affiliation(s)
- Aline Sardinha-Silva
- Molecular Parasitology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Pedro H. Gazzinelli-Guimaraes
- Helminth Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Oluwaremilekun G. Ajakaye
- Molecular Parasitology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
- Department of Animal and Environmental Biology, Adekunle Ajasin University, Akungba Akoko, Ondo State, Nigeria
| | - Tiago R. Ferreira
- Intracellular Parasite Biology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Eliza V.C. Alves-Ferreira
- Molecular Parasitology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Erick T. Tjhin
- Molecular Parasitology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Beth Gregg
- Molecular Parasitology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Marc Y. Fink
- Department of Biology, Georgetown University, Washington, DC 20057, USA
| | - Camila H. Coelho
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Steven M. Singer
- Department of Biology, Georgetown University, Washington, DC 20057, USA
| | - Michael E. Grigg
- Molecular Parasitology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
3
|
Brackman LC, Jung MS, Green EH, Joshi N, Revetta FL, McClain MS, Markham NO, Piazuelo MB, Scott Algood HM. IL-17 signaling protects against Helicobacter pylori-induced gastric cancer. Gut Microbes 2024; 16:2430421. [PMID: 39588838 PMCID: PMC11639209 DOI: 10.1080/19490976.2024.2430421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 09/19/2024] [Accepted: 11/12/2024] [Indexed: 11/27/2024] Open
Abstract
Helicobacter pylori infection is the predominant risk factor for the development of gastric cancer. Risk is enhanced by specific H. pylori virulence factors, diet, and the inflammatory response. Chronic activation of T helper (Th) 1 and Th17 pathways contributes to prolonged inflammation; yet, higher expression of IL-17 receptor (IL-17RA) is a favorable prognostic marker for survival after gastric cancer diagnosis. The protective impact of IL-17RA signaling is not understood. To investigate if IL-17RA signaling protects during H. pylori-induced carcinogenesis, the transgenic InsGAStg/tg mouse, which is prone to H. pylori-induced gastric cancer, was utilized. InsGAStg/tg mice and InsGAStg/tgIl17ra-/- mice were infected with a cag type 4 secretion system (T4SS) positive H. pylori strain for up to 6 months. Six weeks post-infection, IL-17RA deficiency led to increased bacterial burden, increased gastritis, and development of lymphoid follicles. Increased inflammation was associated with heightened cellular proliferation and earlier loss of parietal and chief cells in InsGAStg/tgIl17ra-/- mice. Gastric cancers developed more frequently by 3- and 6-months post-infection in H. pylori-infected InsGAStg/tgIl17ra-/- mice compared to InsGAStg/tg mice. Chronic inflammation was exacerbated with IL-17RA deficiency, characterized by elevated Th1/Th17 cytokines, increased B cell infiltration, and enhanced IgA production, despite reduced expression of the polymeric immunoglobulin receptor. Further, paragastric lymph nodes of InsGAStg/tgIl17ra-/- mice were enlarged relative to controls and displayed altered gene expression profiles. Increased inflammation was accompanied by a significant increase in Cybb expression, which encodes NADPH oxidase 2, suggesting that increased oxidative damage may occur in the absence of IL-17RA. Further, there is increased phosphorylation of histone 2AX in IL-17RA deficient mice, indicating that the DNA damage response is highly activated. These data suggest that IL-17RA signaling activates a protective pathway to prevent excessive inflammation which otherwise can lead to increased oxidative stress, DNA damage, and drive gastric carcinogenesis after H. pylori infection.
Collapse
Affiliation(s)
- Lee C. Brackman
- Department of Medicine, Division of Infectious Disease, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Matthew S. Jung
- Department of Medicine, Division of Infectious Disease, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Emily H. Green
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA
- Vanderbilt Institute of Infection, Immunity, and Inflammation (VI4), Vanderbilt University Medical Center, Nashville, TN, USA
| | - Nikhita Joshi
- Tennessee Valley Healthcare System, Department of Veterans Affairs, Nashville, TN, USA
- School of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Frank L. Revetta
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Mark S. McClain
- Department of Medicine, Division of Infectious Disease, Vanderbilt University School of Medicine, Nashville, TN, USA
- Vanderbilt Institute of Infection, Immunity, and Inflammation (VI4), Vanderbilt University Medical Center, Nashville, TN, USA
| | - Nicholas O. Markham
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA
- Vanderbilt Institute of Infection, Immunity, and Inflammation (VI4), Vanderbilt University Medical Center, Nashville, TN, USA
- Tennessee Valley Healthcare System, Department of Veterans Affairs, Nashville, TN, USA
- Department of Medicine, Division of Gastroenterology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - M. Blanca Piazuelo
- Department of Medicine, Division of Gastroenterology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Holly M. Scott Algood
- Department of Medicine, Division of Infectious Disease, Vanderbilt University School of Medicine, Nashville, TN, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA
- Vanderbilt Institute of Infection, Immunity, and Inflammation (VI4), Vanderbilt University Medical Center, Nashville, TN, USA
- Tennessee Valley Healthcare System, Department of Veterans Affairs, Nashville, TN, USA
| |
Collapse
|
4
|
Peroxisome Proliferator-Activated Receptor-Targeted Therapies: Challenges upon Infectious Diseases. Cells 2023; 12:cells12040650. [PMID: 36831317 PMCID: PMC9954612 DOI: 10.3390/cells12040650] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 02/22/2023] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) α, β, and γ are nuclear receptors that orchestrate the transcriptional regulation of genes involved in a variety of biological responses, such as energy metabolism and homeostasis, regulation of inflammation, cellular development, and differentiation. The many roles played by the PPAR signaling pathways indicate that PPARs may be useful targets for various human diseases, including metabolic and inflammatory conditions and tumors. Accumulating evidence suggests that each PPAR plays prominent but different roles in viral, bacterial, and parasitic infectious disease development. In this review, we discuss recent PPAR research works that are focused on how PPARs control various infections and immune responses. In addition, we describe the current and potential therapeutic uses of PPAR agonists/antagonists in the context of infectious diseases. A more comprehensive understanding of the roles played by PPARs in terms of host-pathogen interactions will yield potential adjunctive personalized therapies employing PPAR-modulating agents.
Collapse
|
5
|
Sangkanu S, Paul AK, Chuprom J, Mitsuwan W, Boonhok R, de Lourdes Pereira M, Oliveira SMR, Wilairatana P, Rahmatullah M, Wiart C, Nawaz M, Sin C, Kayesth S, Nissapatorn V. Conserved Candidate Antigens and Nanoparticles to Develop Vaccine against Giardia intestinalis. Vaccines (Basel) 2022; 11:vaccines11010096. [PMID: 36679941 PMCID: PMC9863896 DOI: 10.3390/vaccines11010096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 12/22/2022] [Accepted: 12/27/2022] [Indexed: 01/04/2023] Open
Abstract
Giardia intestinalis (Giardia lambia, Giardia duodenalis) infections in humans may be asymptomatic or symptomatic and associated with diarrhea (without blood), abdominal cramps, bloating, flatulence, and weight loss. The protozoan Giardia is the third most common cause of diarrhea and death in children under five, preceded only by rotavirus and by Cryptosporidium parvum and C. hominis infections. Antimicrobial drugs, particularly 5-nitroimidazole (5-NIs), are used to treat giardiasis in humans. Immunologically naive or immunocompromised host are more vulnerable to Giardia infection, whereas a degree of resistance to this protozoan is present in humans living in endemic areas. This suggests that vaccination may be a potential and appropriate means to control this parasitic disease outbreak and protect the human population. This review discusses Giardia antigens related to vaccine development. Additionally, based on the latest development of nanoparticle technology, a combination of methods for future research and development is proposed for the design of the next generation of powerful immunogens and an effective vaccine against Giardia.
Collapse
Affiliation(s)
- Suthinee Sangkanu
- School of Allied Health Sciences, Southeast Asia Water Team (SEA Water Team) and World Union for Herbal Drug Discovery (WUHeDD), Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Alok K. Paul
- School of Pharmacy and Pharmacology, University of Tasmania, Hobart, TAS 7001, Australia
| | - Julalak Chuprom
- School of Languages and General Education (SOLGEN), Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Watcharapong Mitsuwan
- Akkhraratchakumari Veterinary College, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Rachasak Boonhok
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Research Excellence Center for Innovation and Health Products (RECIHP), Nakhon Si Thammarat 80160, Thailand
| | - Maria de Lourdes Pereira
- CICECO-Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal
- Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Sonia Marlene Rodrigues Oliveira
- CICECO-Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal
- Hunter Medical Research Institute, New Lambton, NSW 2305, Australia
| | - Polrat Wilairatana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Mohammed Rahmatullah
- Department of Biotechnology & Genetic Engineering, University of Development Alternative, Dhaka 1209, Bangladesh
| | - Christophe Wiart
- The Institute for Tropical Biology and Conservation, University Malaysia Sabah, Jalan UMS, Kota Kinabalu 88400, Malaysia
| | - Muhammad Nawaz
- Department of Nano-Medicine Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia
| | - Chea Sin
- Faculty of Pharmacy, University of Puthisastra, Phnom Penh 12211, Cambodia
| | - Sunil Kayesth
- Department of Zoology, Deshbandhu College, University of Delhi, New Delhi 110019, India
| | - Veeranoot Nissapatorn
- School of Allied Health Sciences, Southeast Asia Water Team (SEA Water Team) and World Union for Herbal Drug Discovery (WUHeDD), Walailak University, Nakhon Si Thammarat 80160, Thailand
- Correspondence:
| |
Collapse
|
6
|
Reyes-Duarte I, Burgara-Estrella AJ, Bustamante-Córdova L, Moya-Camarena SY, Parra-Sánchez H, Álvarez-Bajo O, López-Cervantes G, Montalvo-Corral M. Conjugated linoleic acid modifies transcriptional cytokine profile and induces early specific secretory IgA response in Giardia lamblia infected mice. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2022; 25:1468-1476. [PMID: 36544518 PMCID: PMC9742564 DOI: 10.22038/ijbms.2022.65796.14471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 09/26/2022] [Indexed: 12/24/2022]
Abstract
Objectives Adaptive immunity is crucial in controlling Giardia lamblia infection in the intestinal mucosa, and some dietary lipids may improve mucosal immune function. The aim of this study was to evaluate conjugated linoleic acid (CLA) on the Th17/Treg response and secretory IgA production in a model of giardiasis infection. Materials and Methods C3H/HeN male mice were infected with 5×106 G. lamblia trophozoites (GS/M-83-H7, ATCC collection). Mice were assigned randomly to experimental and control groups. CLA was administered to the experimental group and phosphate-buffered saline (PBS) was given to the control group. Parasite load kinetics was determined. Enzyme-linked immunosorbent assay (ELISA) was performed to evaluate IgA and cytokines. Nuclear transcription factors and cytokines were measured by RT-qPCR, and histology of small bowel cells was evaluated. Results CLA administration reduced the parasite load (P<0.05) and increased early Giardia-specific secretory IgA production. CLA also increased the expression of interleukin-10, transforming growth factor (TGF)-β, and inducible nitric oxide synthase (iNOS) (P<0.05), while infection elevated the expression of Foxp3, with a peak at 40 days post-infection (P<0.05). There were no pathological changes in the colonic mucosa due to infection or treatment. Thus, CLA stimulated mucosal immunity and enhanced the humoral response against G. lamblia, not only for early infection control but also to promote regulatory cytokine production at 40 dpi, restoring the intestinal balance after parasite elimination. Conclusion Our findings reveal novel anti-parasitic effects through the immune-modulatory activity of CLA against the intestinal parasite G. lamblia.
Collapse
Affiliation(s)
- Itzel Reyes-Duarte
- Departamento de Nutrición y Metabolismo, Centro de Investigación en Alimentación y Desarrollo, A.C, Hermosillo, México
| | | | - Lorena Bustamante-Córdova
- Departamento de Nutrición y Metabolismo, Centro de Investigación en Alimentación y Desarrollo, A.C, Hermosillo, México
| | - Silvia Y. Moya-Camarena
- Departamento de Nutrición y Metabolismo, Centro de Investigación en Alimentación y Desarrollo, A.C, Hermosillo, México
| | - Héctor Parra-Sánchez
- Departamento de Nutrición y Metabolismo, Centro de Investigación en Alimentación y Desarrollo, A.C, Hermosillo, México
| | - Osiris Álvarez-Bajo
- CONACYT-Departamento de Investigación en Física, Universidad de Sonora, Hermosillo, México
| | | | - Maricela Montalvo-Corral
- Departamento de Nutrición y Metabolismo, Centro de Investigación en Alimentación y Desarrollo, A.C, Hermosillo, México,Corresponding author: Maricela Montalvo-Corral. Departamento de Nutrición y Metabolismo. Centro de Investigación en Alimentación y Desarrollo, A.C, Hermosillo, México. Tel/Fax: +52-6622892400;
| |
Collapse
|
7
|
Porcine Interleukin-17 and 22 Co-Expressed by Yarrowia lipolytica Enhance Immunity and Increase Protection against Bacterial Challenge in Mice and Piglets. BIOLOGY 2022; 11:biology11121747. [PMID: 36552257 PMCID: PMC9774966 DOI: 10.3390/biology11121747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/24/2022] [Accepted: 11/24/2022] [Indexed: 12/03/2022]
Abstract
Drug resistance in economic animals to pathogens is a matter of widespread concern due to abuse of antibiotics. In order to develop a safe and economical immunopotentiator to raise the immunity and antibacterial response as a replacement for antibiotics, a recombinant yeast co-expressing pig interleukin-17 (IL-17) and IL-22 was constructed and designated as Po1h-pINA1297-IL-17/22. To evaluate the immunoregulator activities of Po1h-pINA1297-IL-17/22, two experiment groups (oral inoculation with Po1h-pINA1297 or Po1h-pINA1297-IL-17/22) and a negative control group (PBS) were set up using 4-week-old female BALB/c mice (10/group). The level of cytokines, including IL-2, IL-4, IL-10, and IFN-γ, were detected by ELISA, and the circulating CD4+ and CD8+ lymphocytes were quantified by flow cytometry. The IgG and secretory IgA (SIgA) levels in both small intestine and fecal matter were also measured by ELISA. The results indicated that the IgG antibody titer and SIgA concentration increased significantly in the Po1h-pINA1297-IL17/22 group in comparison with the controls (p < 0.05) and so did the cytokine levels in the serum (IL-2, IL-4, IL-10, and IFN-γ). In addition, CD4+ and CD8+ T cells were also obviously elevated in the Po1h-pINA1297-IL17/22 group on 35th day (p < 0.05). After challenge with pathogenic Salmonella typhimurium, the Po1h-pINA1297-IL17/22 group showed a relatively higher survival rate without obvious infectious symptoms. On the contrary, the mortality of control group reached 80% due to bacterial infection. As for the piglet experiment, 30 healthy 7-day piglets were similarly attributed into three groups. The oral inoculation of piglets with Po1h-pINA1297-IL17/22 also markedly improved the growth performance and systemic immunity (up-regulations of IL-4, IL-6, IL-15, IL-17, IL-22, and IL-23). Overall, the results indicated that Po1h-pINA1297-IL17/22 effectively promoted the humoral and cellular immunity against bacterial infection. These proved the promising potential of Po1h-pINA1297-IL-17/22 to be a potent immunopotentiator for the prevention of microbial pathogen infections.
Collapse
|
8
|
Garzon T, Ortega-Tirado D, Lopez-Romero G, Alday E, Robles-Zepeda RE, Garibay-Escobar A, Velazquez C. "Immunoinformatic Identification of T-Cell and B-Cell Epitopes From Giardia lamblia Immunogenic Proteins as Candidates to Develop Peptide-Based Vaccines Against Giardiasis". Front Cell Infect Microbiol 2021; 11:769446. [PMID: 34778111 PMCID: PMC8579046 DOI: 10.3389/fcimb.2021.769446] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 10/08/2021] [Indexed: 11/19/2022] Open
Abstract
Giardiasis is one of the most common gastrointestinal infections worldwide, mainly in developing countries. The etiological agent is the Giardia lamblia parasite. Giardiasis mainly affects children and immunocompromised people, causing symptoms such as diarrhea, dehydration, abdominal cramps, nausea, and malnutrition. In order to develop an effective vaccine against giardiasis, it is necessary to understand the host-Giardia interactions, the immunological mechanisms involved in protection against infection, and to characterize the parasite antigens that activate the host immune system. In this study, we identify and characterize potential T-cell and B-cell epitopes of Giardia immunogenic proteins by immunoinformatic approaches, and we discuss the potential role of those epitopes to stimulate the host´s immune system. We selected the main immunogenic and protective proteins of Giardia experimentally investigated. We predicted T-cell and B-cell epitopes using immunoinformatic tools (NetMHCII and BCPREDS). Variable surface proteins (VSPs), structural (giardins), metabolic, and cyst wall proteins were identified as the more relevant immunogens of G. lamblia. We described the protein sequences with the highest affinity to bind MHC class II molecules from mouse (I-Ak and I-Ad) and human (DRB1*03:01 and DRB1*13:01) alleles, as well as we selected promiscuous epitopes, which bind to the most common range of MHC class II molecules in human population. In addition, we identified the presence of conserved epitopes within the main protein families (giardins, VSP, CWP) of Giardia. To our knowledge, this is the first in silico study that analyze immunogenic proteins of G. lamblia by combining bioinformatics strategies to identify potential T-cell and B-cell epitopes, which can be potential candidates in the development of peptide-based vaccines. The bioinformatics analysis demonstrated in this study provides a deeper understanding of the Giardia immunogens that bind to critical molecules of the host immune system, such as MHC class II and antibodies, as well as strategies to rational design of peptide-based vaccine against giardiasis.
Collapse
Affiliation(s)
- Thania Garzon
- Department of Chemistry-Biology, University of Sonora, Hermosillo, Mexico
| | | | | | - Efrain Alday
- Department of Chemistry-Biology, University of Sonora, Hermosillo, Mexico
| | | | | | - Carlos Velazquez
- Department of Chemistry-Biology, University of Sonora, Hermosillo, Mexico
| |
Collapse
|
9
|
Abstract
Giardia duodenalis captured the attention of Leeuwenhoek in 1681 while he was examining his own diarrheal stool, but, ironically, it did not really gain attention as a human pathogen until the 1960s, when outbreaks were reported. Key technological advances, including in vitro cultivation, genomic and proteomic databases, and advances in microscopic and molecular approaches, have led to an understanding that this is a eukaryotic organism with a reduced genome rather than a truly premitochondriate eukaryote. This has included the discovery of mitosomes (vestiges of mitochondria), a transport system with many of the features of the Golgi apparatus, and even evidence for a sexual or parasexual cycle. Cell biology approaches have led to a better understanding of how Giardia survives with two nuclei and how it goes through its life cycle as a noninvasive organism in the hostile environment of the lumen of the host intestine. Studies of its immunology and pathogenesis have moved past the general understanding of the importance of the antibody response in controlling infection to determining the key role of the Th17 response. This work has led to understanding of the requirement for a balanced host immune response that avoids the extremes of an excessive response with collateral damage or one that is unable to clear the organism. This understanding is especially important in view of the remarkable ranges of early manifestations, which range from asymptomatic to persistent diarrhea and weight loss, and longer-term sequelae that include growth stunting in children who had no obvious symptoms and a high frequency of postinfectious irritable bowel syndrome (IBS).
Collapse
|
10
|
Dashti N, Zarebavani M. Probiotics in the management of Giardia duodenalis: an update on potential mechanisms and outcomes. Naunyn Schmiedebergs Arch Pharmacol 2021; 394:1869-1878. [PMID: 34324017 DOI: 10.1007/s00210-021-02124-z] [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: 04/27/2021] [Accepted: 07/07/2021] [Indexed: 10/20/2022]
Abstract
Giardia duodenalis is a common cause of infection in children and travelers. The most frequent symptom is diarrhea in these patients. G. duodenalis trophozoites use a highly specialized adhesive disc to attach the host intestinal epithelium to induce intestinal damages. Pathological features of the small intestine following giardiasis include villous atrophy; infiltration of granulocytes, lymphocytes, and plasma cells into the lamina propria; and nodular lymphoid hyperplasia. The disturbed intestinal microbiota has been observed in patients with giardiasis. Therefore, a growing body of evidence has emphasized restoring the gut microbiome by probiotics in giardiasis. This study aimed to review the literature to find the pathologic features of giardiasis and its relationship with imbalanced microbiota. Then, benefits of probiotics in giardiasis and their potential molecular mechanisms were discussed. It has been illustrated that using probiotics (e.g., Lactobacillus and Saccharomyces) can reduce the time of gastrointestinal symptoms and repair the damages, particularly in giardiasis. Probiotics' capability in restoring the composition of commensal microbiota may lead to therapeutic outcomes. According to preclinical and clinical studies, probiotics can protect against parasite-induced mucosal damages via increasing the antioxidant capacity, suppressing oxidative products, and regulating the systemic and mucosal immune responses. In addition, they can reduce the proportion of G. duodenalis load by directly targeting the parasite. They can destroy the cellular architecture of parasites and suppress the proliferation and growth of trophozoites via the production of some factors with anti-giardial features. Further researches are required to find suitable probiotics for the prevention and treatment of giardiasis.
Collapse
Affiliation(s)
- Nasrin Dashti
- Department of Clinical Laboratory Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Mitra Zarebavani
- Department of Clinical Laboratory Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
11
|
Li Z, Peirasmaki D, Svärd S, Åbrink M. Serglycin-Deficiency Causes Reduced Weight Gain and Changed Intestinal Cytokine Responses in Mice Infected With Giardia intestinalis. Front Immunol 2021; 12:677722. [PMID: 34335577 PMCID: PMC8316049 DOI: 10.3389/fimmu.2021.677722] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 06/14/2021] [Indexed: 11/13/2022] Open
Abstract
The proteoglycan serglycin (SG) is expressed by different innate and adaptive immune cells, e.g. mast cells, macrophages, neutrophils, and cytotoxic T lymphocytes, where SG contributes to correct granule storage and extracellular activity of inflammatory mediators. Here the serglycin-deficient (SG-/-) mouse strain was used to investigate the impact of SG on intestinal immune responses during infection with the non-invasive protozoan parasite Giardia intestinalis. Young (≈11 weeks old) oral gavage-infected congenic SG-/- mice showed reduced weight gain as compared with the infected SG+/+ littermate mice and the PBS-challenged SG-/- and SG+/+ littermate mice. The infection caused no major morphological changes in the small intestine. However, a SG-independent increased goblet cell and granulocyte cell count was observed, which did not correlate with an increased myeloperoxidase or neutrophil elastase activity. Furthermore, infected mice showed increased serum IL-6 levels, with significantly reduced serum IL-6 levels in infected SG-deficient mice and decreased intestinal expression levels of IL-6 in the infected SG-deficient mice. In infected mice the qPCR analysis of alarmins, chemokines, cytokines, and nitric oxide synthases (NOS), showed that the SG-deficiency caused reduced intestinal expression levels of TNF-α and CXCL2, and increased IFN-γ, CXCL1, and NOS1 levels as compared with SG-competent mice. This study shows that SG plays a regulatory role in intestinal immune responses, reflected by changes in chemokine and cytokine expression levels and a delayed weight gain in young SG-/- mice infected with G. intestinalis.
Collapse
Affiliation(s)
- Zhiqiang Li
- The Key and Characteristic Laboratory of Modern Pathogen Biology, College of Basic Medicine, Guizhou Medical University, Guiyang, China.,Department of Biomedical Sciences and Veterinary Public Health, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Dimitra Peirasmaki
- SciLifeLab, Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| | - Staffan Svärd
- SciLifeLab, Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| | - Magnus Åbrink
- Department of Biomedical Sciences and Veterinary Public Health, Swedish University of Agricultural Sciences, Uppsala, Sweden
| |
Collapse
|
12
|
Fahmy A, Abuelenain GL, Rasheed N, Abdou A. 'de Novo' repurposing of Daflon as anti-intestinal parasitic drug in experimental giardiasis. Exp Parasitol 2021; 226-227:108124. [PMID: 34139241 DOI: 10.1016/j.exppara.2021.108124] [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: 09/13/2020] [Revised: 04/19/2021] [Accepted: 06/07/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND There is a necessity to develop or discover an alternative drug to combat the drug resistance by Giardia duodenalis and minimize the multiple doses and frequency of the conventional drug administration. Progressive repositioning or 'repurposing' of drugs has become widespread due to economic circumstances and medical emergency needs. Daflon 500 mg (DFL) is a natural product used safely as a nutrient supplement and an antidiabetic drug in many European countries and the US. OBJECTIVE This study aimed at investigating the efficiency of DFL, in vivo, in a murine model as a safe alternative or co-drug for giardiasis. MATERIALS AND METHODS Swiss Albino mice (n = 32) were inoculated with 1X104Giardia cysts and assigned to four groups: One group was the infected non-treated control mice and three experimental groups that were treated differently, either with Metronidazole (MTZ), DFL, or combined therapy of DFL/MTZ. Also, eight normal mice served as a control group. All mice were sacrificed 13 days post-infection for the parasitic, histopathological, and oxidative stress analysis. RESULTS MTZ, DFL, and the combined therapy significantly reduced the number of trophozoites and cysts compared to their counterparts of the infected mice. The histopathological analysis of the small intestines of the mice treated with the combined therapy retained typical intestinal architecture and normal levels of malondialdehyde (MDA), superoxide dismutase (SOD), and glutathione. CONCLUSION This study indicated promising actions of Daflon 500 as an anti-giardial drug, and the results demonstrated its potential effect in improving the intestinal epithelial tissue and disturbing the Giardia stages when it was taken collectively with Metronidazole.
Collapse
Affiliation(s)
- Azza Fahmy
- Parasitology Lab, Department of Immunology and Drug Evaluation, Theodor Bilharz Research Institute, Imbaba, Egypt
| | - Gehan Labib Abuelenain
- Parasitology Lab, Department of Immunology and Drug Evaluation, Theodor Bilharz Research Institute, Imbaba, Egypt.
| | | | - Amr Abdou
- Microbiology and Immunology Department, NRC, Giza, Egypt
| |
Collapse
|
13
|
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
|
14
|
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
|
15
|
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
|
16
|
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
|
17
|
Amezcua Vesely MC, Rodríguez C, Gruppi A, Acosta Rodríguez EV. Interleukin-17 mediated immunity during infections with Trypanosoma cruzi and other protozoans. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165706. [PMID: 31987839 PMCID: PMC7071987 DOI: 10.1016/j.bbadis.2020.165706] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 12/05/2019] [Accepted: 01/22/2020] [Indexed: 12/31/2022]
Abstract
Host resistance during infection with Trypanosoma cruzi, and other protozoans, is dependent on a balanced immune response. Robust immunity against these pathogens requires of the concerted action of many innate and adaptive cell populations including macrophages, neutrophils, dendritic cells, CD4+, and CD8+ T cells and B cells among others. Indeed, during most protozoan infections only a balanced production of inflammatory (TH1) and anti-inflammatory (TH2/regulatory) cytokines will allow the control of parasite spreading without compromising host tissue integrity. The description of TH17 cells, a novel effector helper T cell lineage that produced IL-17 as signature cytokine, prompted the revision of our knowledge about the mechanisms that mediate protection and immunopathology during protozoan infections. In this manuscript we discuss the general features of IL-17 mediated immune responses as well as the cellular sources, effector mechanisms and overall role of IL-17 in the immune response to T. cruzi and other protozoan infections.
Collapse
Affiliation(s)
- María Carolina Amezcua Vesely
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI)-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Argentina
| | - Constanza Rodríguez
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI)-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Argentina
| | - Adriana Gruppi
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI)-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Argentina
| | - Eva Virginia Acosta Rodríguez
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI)-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Argentina.
| |
Collapse
|
18
|
Faria CP, Neves BM, Lourenço Á, Cruz MT, Martins JD, Silva A, Pereira S, Sousa MDC. Giardia lamblia Decreases NF-κB p65 RelA Protein Levels and Modulates LPS-Induced Pro-Inflammatory Response in Macrophages. Sci Rep 2020; 10:6234. [PMID: 32277133 PMCID: PMC7148380 DOI: 10.1038/s41598-020-63231-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 03/20/2020] [Indexed: 11/09/2022] Open
Abstract
The protozoan Giardia lamblia is the most common cause of parasitic gastrointestinal infection worldwide. The parasite developed sophisticated, yet not completely disclosed, mechanisms to escape immune system and growth in the intestine. To further understand the interaction of G. lamblia with host immune cells, we investigated the ability of parasites to modulate the canonical activation of mouse macrophages (Raw 264.7 cell line) and human monocyte-derived macrophages triggered by the TLR4 agonist, lipopolysaccharide (LPS). We observed that G. lamblia impairs LPS-evoked pro-inflammatory status in these macrophage-like cells through inhibition of cyclooxygenase-2 and inducible nitric oxide synthase expression and subsequent NO production. This effect was in part due to the activity of three G. lamblia proteases, a 135 kDa metalloprotease and two cysteine proteases with 75 and 63 kDa, that cleave the p65RelA subunit of the nuclear factor-kappa B (NF-κB). Moreover, Tnf and Ccl4 transcription was increased in the presence of the parasite. Overall, our data indicates that G. lamblia modulates macrophages inflammatory response through impairment of the NF-κB, thus silencing a crucial signaling pathway of the host innate immune response.
Collapse
Affiliation(s)
- Clarissa Perez Faria
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.,Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Bruno Miguel Neves
- Department of Medical Sciences and Institute of Biomedicine - iBiMED, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Ágata Lourenço
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.,Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Maria Teresa Cruz
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.,Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - João D Martins
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Ana Silva
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Sónia Pereira
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.,Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Maria do Céu Sousa
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal. .,Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
19
|
The Chymase Mouse Mast Cell Protease-4 Regulates Intestinal Cytokine Expression in Mature Adult Mice Infected with Giardia intestinalis. Cells 2020; 9:cells9040925. [PMID: 32283818 PMCID: PMC7226739 DOI: 10.3390/cells9040925] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 04/06/2020] [Accepted: 04/07/2020] [Indexed: 12/16/2022] Open
Abstract
Mast cells have been shown to affect the control of infections with the protozoan parasite Giardia intestinalis. Recently, we demonstrated that Giardia excretory-secretory proteins inhibited the activity of the connective tissue mast cell-specific protease chymase. To study the potential role of the chymase mouse mast cell protease (mMCP)-4 during infections with Giardia, mMCP-4+/+ and mMCP-4−/− littermate mice were gavage-infected with G. intestinalis trophozoites of the human assemblage B isolate GS. No significant changes in weight gain was observed in infected young (≈10 weeks old) mMCP-4−/− and mMCP-4+/+ littermate mice. In contrast, infections of mature adult mice (>18 weeks old) caused significant weight loss as compared to uninfected control mice. We detected a more rapid weight loss in mMCP-4−/− mice as compared to littermate mMCP-4+/+ mice. Submucosal mast cell and granulocyte counts in jejunum increased in the infected adult mMCP-4−/− and mMCP-4+/+ mice. This increase was correlated with an augmented intestinal trypsin-like and chymotrypsin-like activity, but the myeloperoxidase activity was constant. Infected mice showed a significantly lower intestinal neutrophil elastase (NE) activity, and in vitro, soluble Giardia proteins inhibited human recombinant NE. Serum levels of IL-6 were significantly increased eight and 13 days post infection (dpi), while intestinal IL-6 levels showed a trend to significant increase 8 dpi. Strikingly, the lack of mMCP-4 resulted in significantly less intestinal transcriptional upregulation of IL-6, TNF-α, IL-25, CXCL2, IL-2, IL-4, IL-5, and IL-10 in the Giardia-infected mature adult mice, suggesting that chymase may play a regulatory role in intestinal cytokine responses.
Collapse
|
20
|
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
|
21
|
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
|
22
|
Fink MY, Maloney J, Keselman A, Li E, Menegas S, Staniorski C, Singer SM. Proliferation of Resident Macrophages Is Dispensable for Protection during Giardia duodenalis Infections. Immunohorizons 2019; 3:412-421. [PMID: 31455692 PMCID: PMC7033283 DOI: 10.4049/immunohorizons.1900041] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 08/06/2019] [Indexed: 12/28/2022] Open
Abstract
Infection with the intestinal parasite Giardia duodenalis is one of the most common causes of diarrheal disease in the world. Previous work has demonstrated that the cells and mechanisms of the adaptive immune system are critical for clearance of this parasite. However, the innate system has not been as well studied in the context of Giardia infection. We have previously demonstrated that Giardia infection leads to the accumulation of a population of CD11b+, F4/80+, ARG1+, and NOS2+ macrophages in the small intestinal lamina propria. In this report, we sought to identify the accumulation mechanism of duodenal macrophages during Giardia infection and to determine if these cells were essential to the induction of protective Giardia immunity. We show that F4/80+, CD11b+, CD11cint, CX3CR1+, MHC class II+, Ly6C−, ARG1+, and NOS2+ macrophages accumulate in the small intestine during infections in mice. Consistent with this resident macrophage phenotype, macrophage accumulation does not require CCR2, and the macrophages incorporate EdU, indicating in situ proliferation rather than the recruitment of monocytes. Depletion of macrophages using anti-CSF1R did not impact parasite clearance nor development of regulatory T cell or Th17 cellular responses, suggesting that these macrophages are dispensable for protective Giardia immunity.
Collapse
Affiliation(s)
- Marc Y Fink
- Department of Biology, Georgetown University, Washington, DC 20057
| | - Jenny Maloney
- Department of Biology, Georgetown University, Washington, DC 20057
| | | | - Erqiu Li
- Department of Biology, Georgetown University, Washington, DC 20057
| | - Samantha Menegas
- Department of Biology, Georgetown University, Washington, DC 20057
| | | | - Steven M Singer
- Department of Biology, Georgetown University, Washington, DC 20057
| |
Collapse
|
23
|
Singer SM, Fink MY, Angelova VV. Recent insights into innate and adaptive immune responses to Giardia. ADVANCES IN PARASITOLOGY 2019; 106:171-208. [PMID: 31630758 DOI: 10.1016/bs.apar.2019.07.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Infection with Giardia produces a wide range of clinical outcomes. Acutely infected patients may have no overt symptoms or suffer from severe cramps, diarrhea, nausea and even urticaria. Recently, post-infectious irritable bowel syndrome and chronic fatigue syndrome have been identified as long-term sequelae of giardiasis. Frequently, recurrent and chronic Giardia infection is considered a major contributor to stunting in children from low and middle income countries. Perhaps the most unusual outcome of infection with Giardia is the apparent reduced risk of developing moderate-to-severe diarrhea due to other enteric infections which has been noted in several recent studies. The goal of understanding immune responses against Giardia is therefore to identify protective mechanisms which could become targets for vaccine development, but also to identify mechanisms whereby infections lead to these other diverse outcomes. Giardia induces a robust adaptive immune response in both humans and animals. It has been known for many years that there is production of large amounts of parasite-specific IgA following infection and that CD4+ T cell responses contribute to this IgA production and control of the infection. In the past decade, there have been advances in our understanding of the non-antibody effector mechanisms used by the host to fight Giardia infections, in particular the importance of the cytokine interleukin (IL)-17 in orchestrating these responses. There have also been major advances in understanding how the innate response to Giardia infection is initiated and how it contributes to the development of adaptive immunity. Finally, there here have been significant increases in our knowledge of how the resident microbial community influences the immune response and how these responses contribute to the development of some of the symptoms of giardiasis. In this article, we will focus on data generated in the last 10 years and how it has advanced our knowledge about this important parasitic disease.
Collapse
Affiliation(s)
- Steven M Singer
- Department of Biology, Georgetown University, Washington, DC, United States.
| | - Marc Y Fink
- Department of Biology, Georgetown University, Washington, DC, United States
| | - Vanessa V Angelova
- Department of Biology, Georgetown University, Washington, DC, United States
| |
Collapse
|
24
|
Lee HY, Park EA, Lee KJ, Lee KH, Park SJ. Increased Innate Lymphoid Cell 3 and IL-17 Production in Mouse Lamina Propria Stimulated with Giardia lamblia. THE KOREAN JOURNAL OF PARASITOLOGY 2019; 57:225-232. [PMID: 31284344 PMCID: PMC6616171 DOI: 10.3347/kjp.2019.57.3.225] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 05/23/2019] [Indexed: 12/14/2022]
Abstract
Innate lymphoid cells (ILCs) are key players during an immune response at the mucosal surfaces, such as lung, skin, and gastrointestinal tract. Giardia lamblia is an extracellular protozoan pathogen that inhabits the human small intestine. In this study, ILCs prepared from the lamina propria of mouse small intestine were incubated with G. lamblia trophozoites. Transcriptional changes in G. lamblia-exposed ILCs resulted in identification of activation of several immune pathways. Secretion of interleukin (IL)-17A, IL-17F, IL-1β, and interferon-γ was increased, whereas levels of IL-13, IL-5, and IL22, was maintained or reduced upon exposure to G. lamblia. Goup 3 ILC (ILC3) was found to be dominant amongst the ILCs, and increased significantly upon co-cultivation with G. lamblia trophozoites. Oral inoculation of G. lamblia trophozoites into mice resulted in their presence in the small intestine, of which, the highest number of parasites was detected at the 5 days-post infection. Increased ILC3 was observed amongst the ILC population at the 5 days-post infection. These findings indicate that ILC3 from the lamina propria secretes IL-17 in response to G. lamblia, leading to the intestinal pathology observed in giardiasis.
Collapse
Affiliation(s)
- Hye-Yeon Lee
- Department of Environmental Medical Biology and Institute of Tropical Medicine, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Eun-Ah Park
- Department of Environmental Medical Biology and Institute of Tropical Medicine, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Kyung-Jo Lee
- Department of Life Science, Sogang University, Seoul 04107, Korea
| | - Kyu-Ho Lee
- Department of Life Science, Sogang University, Seoul 04107, Korea
| | - Soon-Jung Park
- Department of Environmental Medical Biology and Institute of Tropical Medicine, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
| |
Collapse
|
25
|
Paerewijck O, Maertens B, Gagnaire A, De Bosscher K, Geldhof P. Delayed development of the protective IL-17A response following a Giardia muris infection in neonatal mice. Sci Rep 2019; 9:8959. [PMID: 31222079 PMCID: PMC6586865 DOI: 10.1038/s41598-019-45544-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 06/04/2019] [Indexed: 11/09/2022] Open
Abstract
Giardia is an intestinal protozoan parasite that has the ability to infect a wide range of hosts, which can result in the clinical condition 'giardiasis'. Over the years, experimental research has shown the crucial involvement of IL-17A to steer the protective immune response against Giardia. The development of the protective response, as reflected by a significant drop in cyst secretion, typically takes around 3 to 4 weeks. However, early-life infections often have a more chronic character lasting for several weeks or months. Therefore, the aim of the current study was to investigate the dynamics of a Giardia muris infection and the subsequent host immune response in neonatal mice infected 4 days after birth. The outcome of the study showed that a G. muris infection in pre-weaned mice failed to trigger a protective IL-17A response, which could explain the prolonged course of infection in comparison to older mice. Only after weaning, a protective intestinal immune response started to develop, characterized by an upregulation of IL-17A and Mbl2 and the secretion of parasite-specific IgA.
Collapse
Affiliation(s)
- Oonagh Paerewijck
- Department of Virology, Parasitology and Immunology, Laboratory of Parasitology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Brecht Maertens
- Department of Virology, Parasitology and Immunology, Laboratory of Parasitology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Aurélie Gagnaire
- Department of Virology, Parasitology and Immunology, Laboratory of Parasitology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Karolien De Bosscher
- VIB Department of Medical Protein Research, Receptor Research laboratories, Nuclear Receptor Lab, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Peter Geldhof
- Department of Virology, Parasitology and Immunology, Laboratory of Parasitology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium.
| |
Collapse
|
26
|
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]
|
27
|
Bougarne N, Weyers B, Desmet SJ, Deckers J, Ray DW, Staels B, De Bosscher K. Molecular Actions of PPARα in Lipid Metabolism and Inflammation. Endocr Rev 2018; 39:760-802. [PMID: 30020428 DOI: 10.1210/er.2018-00064] [Citation(s) in RCA: 482] [Impact Index Per Article: 68.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Accepted: 07/10/2018] [Indexed: 12/13/2022]
Abstract
Peroxisome proliferator-activated receptor α (PPARα) is a nuclear receptor of clinical interest as a drug target in various metabolic disorders. PPARα also exhibits marked anti-inflammatory capacities. The first-generation PPARα agonists, the fibrates, have however been hampered by drug-drug interaction issues, statin drop-in, and ill-designed cardiovascular intervention trials. Notwithstanding, understanding the molecular mechanisms by which PPARα works will enable control of its activities as a drug target for metabolic diseases with an underlying inflammatory component. Given its role in reshaping the immune system, the full potential of this nuclear receptor subtype as a versatile drug target with high plasticity becomes increasingly clear, and a novel generation of agonists may pave the way for novel fields of applications.
Collapse
Affiliation(s)
- Nadia Bougarne
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Receptor Research Laboratories, Nuclear Receptor Laboratory, VIB Center for Medical Biotechnology, Ghent, Belgium
| | - Basiel Weyers
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Receptor Research Laboratories, Nuclear Receptor Laboratory, VIB Center for Medical Biotechnology, Ghent, Belgium
| | - Sofie J Desmet
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Receptor Research Laboratories, Nuclear Receptor Laboratory, VIB Center for Medical Biotechnology, Ghent, Belgium
| | - Julie Deckers
- Department of Internal Medicine, Ghent University, Ghent, Belgium
- Laboratory of Immunoregulation, VIB Center for Inflammation Research, Ghent (Zwijnaarde), Belgium
| | - David W Ray
- Division of Metabolism and Endocrinology, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, United Kingdom
| | - Bart Staels
- Université de Lille, U1011-European Genomic Institute for Diabetes, Lille, France
- INSERM, U1011, Lille, France
- Centre Hospitalier Universitaire de Lille, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Karolien De Bosscher
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Receptor Research Laboratories, Nuclear Receptor Laboratory, VIB Center for Medical Biotechnology, Ghent, Belgium
| |
Collapse
|
28
|
Dann SM, Le CHY, Hanson EM, Ross MC, Eckmann L. Giardia Infection of the Small Intestine Induces Chronic Colitis in Genetically Susceptible Hosts. THE JOURNAL OF IMMUNOLOGY 2018; 201:548-559. [PMID: 29898958 DOI: 10.4049/jimmunol.1700824] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 05/07/2018] [Indexed: 01/01/2023]
Abstract
The lumen-dwelling protozoan Giardia is an important parasitic cause of diarrheal disease worldwide. Infection can persist over extended periods with minimal intestinal inflammation, suggesting that Giardia may attenuate host responses to ensure its survival, although clearance eventually occurs in most cases. IL-10 is an anti-inflammatory regulator critical for intestinal homeostasis and controlling host responses to bacterial exposure, yet its potential role in coordinating antiprotozoal host defense in the intestine is not known. In this study, we found that murine infection with the natural enteric pathogen Giardia muris induced a transient IL-10 response after 2-4 wk at the primary site of infection in the upper small intestine, but parasite colonization and eradication were not affected by the absence of the cytokine in gene-targeted mice. However, IL-10 was critical for controlling infection-associated immunological sequelae in the colon because severe and persistent diarrhea and colitis were observed in IL-10-deficient mice within 1-2 wk postinfection but not in uninfected littermate controls. Inflammation was characterized by epithelial hyperplasia, neutrophil and macrophage expansion, and Th1 induction and could be prevented by blockade of IL-12/IL-23 p40 but not depletion of CD11c+ dendritic cells. Furthermore, the intestinal microbiota underwent characteristic shifts in composition and was required for disease because antibiotics and loss of TLR signaling in MyD88-deficient mice protected against colitis. Together, our data suggest that transient infection by a luminal and seemingly noninflammatory pathogen can trigger sustained colitis in genetically susceptible hosts, which has broader implications for understanding postinfectious syndromes and other chronic intestinal inflammatory conditions.
Collapse
Affiliation(s)
- Sara M Dann
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555.,Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555
| | - Christine H Y Le
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093; and
| | - Elaine M Hanson
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093; and
| | - Matthew C Ross
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030
| | - Lars Eckmann
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093; and
| |
Collapse
|
29
|
Muñoz-Cruz S, Gomez-García A, Matadamas-Martínez F, Alvarado-Torres JA, Meza-Cervantez P, Arriaga-Pizano L, Yépez-Mulia L. Giardia lamblia: identification of molecules that contribute to direct mast cell activation. Parasitol Res 2018; 117:2555-2567. [DOI: 10.1007/s00436-018-5944-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 05/24/2018] [Indexed: 12/01/2022]
|
30
|
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
|
31
|
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: 129] [Impact Index Per Article: 16.1] [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
|
32
|
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: 73] [Impact Index Per Article: 9.1] [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
|
33
|
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
|
34
|
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]
|
35
|
Synergistic effects of fenbendazole and metronidazole against Giardia muris in Swiss mice naturally infected. Parasitol Res 2016; 116:939-944. [DOI: 10.1007/s00436-016-5367-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 12/25/2016] [Indexed: 10/20/2022]
|
36
|
Mercer F, Diala FGI, Chen YP, Molgora BM, Ng SH, Johnson PJ. Leukocyte Lysis and Cytokine Induction by the Human Sexually Transmitted Parasite Trichomonas vaginalis. PLoS Negl Trop Dis 2016; 10:e0004913. [PMID: 27529696 PMCID: PMC4986988 DOI: 10.1371/journal.pntd.0004913] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 07/19/2016] [Indexed: 11/18/2022] Open
Abstract
Trichomonas vaginalis (Tv) is an extracellular protozoan parasite that causes the most common non-viral sexually transmitted infection: trichomoniasis. While acute symptoms in women may include vaginitis, infections are often asymptomatic, but can persist and are associated with medical complications including increased HIV susceptibility, infertility, pre-term labor, and higher incidence of cervical cancer. Heightened inflammation resulting from Tv infection could account for these complications. Effective cellular immune responses to Tv have not been characterized, and re-infection is common, suggesting a dysfunctional adaptive immune response. Using primary human leukocyte components, we have established an in vitro co-culture system to assess the interaction between Tv and the cells of the human immune system. We determined that in vitro, Tv is able to lyse T-cells and B-cells, showing a preference for B-cells. We also found that Tv lysis of lymphocytes was mediated by contact-dependent and soluble factors. Tv lysis of monocytes is far less efficient, and almost entirely contact-dependent. Interestingly, a common symbiont of Tv, Mycoplasma hominis, did not affect cytolytic activity of the parasite, but had a major impact on cytokine responses. M. hominis enabled more diverse inflammatory cytokine secretion in response to Tv and, of the cytokines tested, Tv strains cleared of M. hominis induced only IL-8 secretion from monocytes. The quality of the adaptive immune response to Tv is therefore likely influenced by Tv symbionts, commensals, and concomitant infections, and may be further complicated by direct parasite lysis of effector immune cells.
Collapse
Affiliation(s)
- Frances Mercer
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Fitz Gerald I. Diala
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Yi-Pei Chen
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Brenda M. Molgora
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Shek Hang Ng
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Patricia J. Johnson
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
37
|
Einarsson E, Ma'ayeh S, Svärd SG. An up-date on Giardia and giardiasis. Curr Opin Microbiol 2016; 34:47-52. [PMID: 27501461 DOI: 10.1016/j.mib.2016.07.019] [Citation(s) in RCA: 218] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 07/27/2016] [Accepted: 07/27/2016] [Indexed: 12/11/2022]
Abstract
Giardia intestinalis is a non-invasive protozoan parasite infecting the upper small intestine causing acute, watery diarrhea or giardiasis in 280 million people annually. Asymptomatic infections are equally common and recent data have suggested that infections even can be protective against other diarrheal diseases. Most symptomatic infections resolve spontaneously but infections can lead to chronic disease and treatment failures are becoming more common world-wide. Giardia infections can also result in irritable bowel syndrome (IBS) and food allergies after resolution. Until recently not much was known about the mechanism of giardiasis or the cause of post-giardiasis syndromes and treatment failures, but here we will describe the recent progress in these areas.
Collapse
Affiliation(s)
- Elin Einarsson
- Department of Cell and Molecular Biology, BMC, Box 596, Uppsala University, SE-751 24 Uppsala, Sweden
| | - Showgy Ma'ayeh
- Department of Cell and Molecular Biology, BMC, Box 596, Uppsala University, SE-751 24 Uppsala, Sweden
| | - Staffan G Svärd
- Department of Cell and Molecular Biology, BMC, Box 596, Uppsala University, SE-751 24 Uppsala, Sweden.
| |
Collapse
|
38
|
Li E, Tako EA, Singer SM. Complement Activation by Giardia duodenalis Parasites through the Lectin Pathway Contributes to Mast Cell Responses and Parasite Control. Infect Immun 2016; 84:1092-1099. [PMID: 26831470 PMCID: PMC4807472 DOI: 10.1128/iai.00074-16] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 01/26/2016] [Indexed: 11/20/2022] Open
Abstract
Infection with Giardia duodenalis is one of the most common causes of diarrheal disease in the world. While numerous studies have identified important contributions of adaptive immune responses to parasite control, much less work has examined innate immunity and its connections to the adaptive response during this infection. We explored the role of complement in immunity to Giardia using mice deficient in mannose-binding lectin (Mbl2) or complement factor 3a receptor (C3aR). Both strains exhibited delayed clearance of parasites and a reduced ability to recruit mast cells in the intestinal submucosa. C3aR-deficient mice had normal production of antiparasite IgA, butex vivo T cell recall responses were impaired. These data suggest that complement is a key factor in the innate recognition of Giardia and that recruitment of mast cells and activation of T cell immunity through C3a are important for parasite control.
Collapse
Affiliation(s)
- Erqiu Li
- Department of Biology, Georgetown University, Washington, DC, USA
| | - Ernest A Tako
- Department of Biology, Georgetown University, Washington, DC, USA
| | - Steven M Singer
- Department of Biology, Georgetown University, Washington, DC, USA
| |
Collapse
|
39
|
Di Genova BM, Tonelli RR. Infection Strategies of Intestinal Parasite Pathogens and Host Cell Responses. Front Microbiol 2016; 7:256. [PMID: 26973630 PMCID: PMC4776161 DOI: 10.3389/fmicb.2016.00256] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 02/16/2016] [Indexed: 12/24/2022] Open
Abstract
Giardia lamblia, Cryptosporidium sp., and Entamoeba histolytica are important pathogenic intestinal parasites and are amongst the leading causes worldwide of diarrheal illness in humans. Diseases caused by these organisms, giardiasis, cryptosporidiosis, and amoebiasis, respectively, are characterized by self-limited diarrhea but can evolve to long-term complications. The cellular and molecular mechanisms underlying the pathogenesis of diarrhea associated with these three pathogens are being unraveled, with knowledge of both the strategies explored by the parasites to establish infection and the methods evolved by hosts to avoid it. Special attention is being given to molecules participating in parasite–host interaction and in the mechanisms implicated in the diseases’ pathophysiologic processes. This review focuses on cell mechanisms that are modulated during infection, including gene transcription, cytoskeleton rearrangements, signal transduction pathways, and cell death.
Collapse
Affiliation(s)
- Bruno M Di Genova
- Departamento de Microbiologia e Imunologia, Universidade Federal de São Paulo São Paulo, Brazil
| | - Renata R Tonelli
- Departamento de Microbiologia e Imunologia, Universidade Federal de São PauloSão Paulo, Brazil; Instituto de Ciências Ambientais, Químicas e Farmacêuticas, Departamento de Ciências Biológicas, Universidade Federal de São PauloDiadema, Brazil
| |
Collapse
|
40
|
Sarre C, González-Hernández A, Van Coppernolle S, Grit R, Grauwet K, Van Meulder F, Chiers K, Van den Broeck W, Geldhof P, Claerebout E. Comparative immune responses against Psoroptes ovis in two cattle breeds with different susceptibility to mange. Vet Res 2015; 46:131. [PMID: 26582546 PMCID: PMC4652412 DOI: 10.1186/s13567-015-0277-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 10/19/2015] [Indexed: 12/22/2022] Open
Abstract
The sheep scab mite, Psoroptes ovis, is a major problem in the beef cattle industry, especially in Belgian Blue (BB) cattle. This breed is naturally more predisposed to psoroptic mange but reasons for this high susceptibility remain unknown. Different immune responses could be a potential cause; thus in this study, the cutaneous immune response and in vitro cellular immune response after antigen re-stimulation were examined in naturally infested BB. Cytokine production in the skin and in circulating re-stimulated peripheral blood mononuclear cells (PBMC) demonstrated a mixed pro-inflammatory Th2/Th17 profile, with transcription of IL-4, IL-13, IL-6 and IL-17. Strong IL-17 up-regulation in the skin of BB was associated with an influx of eosinophils and other immune cells, potentially leading towards more severe symptoms. Virtually no changes in cutaneous IFN-γ transcription were detected, while there was substantial IFN-γ up-regulation in re-stimulated PBMC from infested and uninfested animals, potentially indicating a role of this pro-inflammatory cytokine in the innate immune response. In Holstein–Friesian (HF) cattle, generally more resistant to P. ovis infection, a largely similar immunologic response was observed. Differences between HF and BB were the lack of cutaneous IL-17 response in infested HF and low transcription levels of IFN-γ and high IL-10 transcription in re-stimulated PBMC from both infested and uninfested animals. Further research is needed to identify potential cell sources and biological functions for these cytokines and to fully unravel the basis of this different breed susceptibility to P. ovis.
Collapse
Affiliation(s)
- Charlotte Sarre
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium.
| | - Ana González-Hernández
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium.
| | - Stefanie Van Coppernolle
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium.
| | - Rika Grit
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium.
| | - Korneel Grauwet
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium.
| | - Frederik Van Meulder
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium.
| | - Koen Chiers
- Department of Pathology, Bacteriology and Poultry Diseases, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium.
| | - Wim Van den Broeck
- Department of Morphology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium.
| | - Peter Geldhof
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium.
| | - Edwin Claerebout
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium.
| |
Collapse
|
41
|
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
|
42
|
Control of Giardiasis by Interleukin-17 in Humans and Mice--Are the Questions All Answered? CLINICAL AND VACCINE IMMUNOLOGY : CVI 2015; 23:2-5. [PMID: 26581888 DOI: 10.1128/cvi.00648-15] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
For years, studies of the immune response to Giardia lamblia infection focused on the production of IgA by infected hosts and antigenic variation by the parasite to escape destruction by this IgA. A new study by Hanevik and colleagues (C. S. Saghaug, S. Sørnes, D. Peirasmaki, S. Svärd, N. Langeland, and K. Hanevik, Clin Vaccine Immunol 23:11-18, 2016, http://dx.doi.org/10.1128/CVI.00419-15) highlights the emerging role of interleukin-17 (IL-17) in immunity to this parasite. Along with recent studies of Giardia infections of animals, this work shows that IL-17 appears to be essential for the control of these infections and to be a key factor linking cellular and humoral immune responses.
Collapse
|
43
|
Luo Y, Van Nguyen U, de la Fe Rodriguez PY, Devriendt B, Cox E. F4+ ETEC infection and oral immunization with F4 fimbriae elicits an IL-17-dominated immune response. Vet Res 2015; 46:121. [PMID: 26490738 PMCID: PMC4618862 DOI: 10.1186/s13567-015-0264-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 10/02/2015] [Indexed: 02/06/2023] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) are an important cause of post-weaning diarrhea (PWD) in piglets. Porcine-specific ETEC strains possess different fimbrial subtypes of which F4 fimbriae are the most frequently associated with ETEC-induced diarrhea in piglets. These F4 fimbriae are potent oral immunogens that induce protective F4-specific IgA antibody secreting cells at intestinal tissues. Recently, T-helper 17 (Th17) cells have been implicated in the protection of the host against extracellular pathogens. However, it remains unknown if Th17 effector responses are needed to clear ETEC infections. In the present study, we aimed to elucidate if ETEC elicits a Th17 response in piglets and if F4 fimbriae trigger a similar response. F4+ ETEC infection upregulated IL-17A, IL-17F, IL-21 and IL-23p19, but not IL-12 and IFN-γ mRNA expression in the systemic and mucosal immune system. Similarly, oral immunization with F4 fimbriae triggered a Th17 signature evidenced by an upregulated mRNA expression of IL-17F, RORγt, IL-23p19 and IL-21 in the peripheral blood mononuclear cells (PBMCs). Intriguingly, IL-17A mRNA levels were unaltered. To further evaluate this difference between systemic and mucosal immune responses, we assayed the cytokine mRNA profile of F4 fimbriae stimulated PBMCs. F4 fimbriae induced IL-17A, IL-17F, IL-22 and IL-23p19, but downregulated IL-17B mRNA expression. Altogether, these data indicate a Th17 dominated response upon oral immunization with F4 fimbriae and F4+ ETEC infection. Our work also highlights that IL-17B and IL-17F participate in the immune response to protect the host against F4+ ETEC infection and could aid in the design of future ETEC vaccines.
Collapse
Affiliation(s)
- Yu Luo
- Laboratory of Veterinary Immunology, Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium.
| | - Ut Van Nguyen
- Laboratory of Veterinary Immunology, Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium.
| | - Pedro Y de la Fe Rodriguez
- Department of Veterinary Medicine and Zootechnics, Universidad Central "Marta Abreu" de Las Villas, Carretera a Camajuani km 5½, 54830, Santa Clara, Villa Clara, Cuba.
| | - Bert Devriendt
- Laboratory of Veterinary Immunology, Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium.
| | - Eric Cox
- Laboratory of Veterinary Immunology, Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium.
| |
Collapse
|
44
|
Human Memory CD4+ T Cell Immune Responses against Giardia lamblia. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2015; 23:11-8. [PMID: 26376930 DOI: 10.1128/cvi.00419-15] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 09/10/2015] [Indexed: 01/17/2023]
Abstract
The intestinal protozoan parasite Giardia lamblia may cause severe prolonged diarrheal disease or pass unnoticed as an asymptomatic infection. T cells seem to play an important role in the immune response to Giardia infection, and memory responses may last years. Recently, TH17 responses have been found in three animal studies of Giardia infection. The aim of this study was to characterize the human CD4(+) T cell responses to Giardia. Peripheral blood mononuclear cells (PBMCs) were obtained from 21 returning travelers with recent or ongoing giardiasis and 12 low-risk healthy controls and stimulated in vitro with Giardia lamblia proteins. Production of tumor necrosis factor alpha (TNF-α), gamma interferon, interleukin-17A (IL-17A), IL-10, and IL-4 was measured in CD4(+) effector memory (EM) T cells after 24 h by flow cytometry. After 6 days of culture, activation and proliferation were measured by flow cytometry, while an array of inflammatory cytokine levels in supernatants were measured with multiplex assays. We found the number of IL-17A-producing CD4(+) EM T cells, as well as that of cells simultaneously producing both IL-17A and TNF-α, to be significantly elevated in the Giardia-exposed individuals after 24 h of antigen stimulation. In supernatants of PBMCs stimulated with Giardia antigens for 6 days, we found inflammation-associated cytokines, including 1L-17A, as well as CD4(+) T cell activation and proliferation, to be significantly elevated in the Giardia-exposed individuals. We conclude that symptomatic Giardia infection in humans induces a CD4(+) EM T cell response of which IL-17A production seems to be an important component.
Collapse
|
45
|
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
|
46
|
Gupta S, Smith C, Auclair S, Delgadillo ADJ, Garg NJ. Therapeutic Efficacy of a Subunit Vaccine in Controlling Chronic Trypanosoma cruzi Infection and Chagas Disease Is Enhanced by Glutathione Peroxidase Over-Expression. PLoS One 2015; 10:e0130562. [PMID: 26075398 PMCID: PMC4468200 DOI: 10.1371/journal.pone.0130562] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 05/22/2015] [Indexed: 12/15/2022] Open
Abstract
Trypanosoma cruzi-induced oxidative and inflammatory responses are implicated in chagasic cardiomyopathy. In this study, we examined the therapeutic utility of a subunit vaccine against T. cruzi and determined if glutathione peroxidase (GPx1, antioxidant) protects the heart from chagasic pathogenesis. C57BL/6 mice (wild-type (WT) and GPx1 transgenic (GPxtg) were infected with T. cruzi and at 45 days post-infection (dpi), immunized with TcG2/TcG4 vaccine delivered by a DNA-prime/Protein-boost (D/P) approach. The plasma and tissue-sections were analyzed on 150 dpi for parasite burden, inflammatory and oxidative stress markers, inflammatory infiltrate and fibrosis. WT mice infected with T. cruzi had significantly more blood and tissue parasite burden compared with infected/GPxtg mice (n = 5-8, p<0.01). Therapeutic vaccination provided >15-fold reduction in blood and tissue parasites in both WT and GPxtg mice. The increase in plasma levels of myeloperoxidase (MPO, 24.7%) and nitrite (iNOS activity, 45%) was associated with myocardial increase in oxidant levels (3-4-fold) and non-responsive antioxidant status in chagasic/WT mice; and these responses were not controlled after vaccination (n = 5-7). The GPxtg mice were better equipped than the WT mice in controlling T. cruzi-induced inflammatory and oxidative stress markers. Extensive myocardial and skeletal tissue inflammation noted in chagasic/WT mice, was significantly more compared with chagasic/GPxtg mice (n = 4-6, p<0.05). Vaccination was equally effective in reducing the chronic inflammatory infiltrate in the heart and skeletal tissue of infected WT and GPxtg mice (n = 6, p<0.05). Hypertrophy (increased BNP and ANP mRNA) and fibrosis (increased collagen) of the heart were extensively present in chronically-infected WT and GPxtg mice and notably decreased after therapeutic vaccination. We conclude the therapeutic delivery of D/P vaccine was effective in arresting the chronic parasite persistence and chagasic pathology; and GPx1 over-expression provided additive benefits in reducing the parasite burden, inflammatory/oxidative stress and cardiac remodeling in Chagas disease.
Collapse
Affiliation(s)
- Shivali Gupta
- Department of Microbiology and Immunology, School of Medicine, University of Texas Medical Branch, Galveston, Texas, United States of America
- * E-mail: (SG); (NG)
| | - Charity Smith
- Department of Microbiology and Immunology, School of Medicine, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Sarah Auclair
- Department of Microbiology and Immunology, School of Medicine, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Anahi De Jesus Delgadillo
- Department of Microbiology and Immunology, School of Medicine, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Nisha Jain Garg
- Department of Microbiology and Immunology, School of Medicine, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Institute for Human Infections and Immunity and the Sealy Center for Vaccine Development, University of Texas Medical Branch, Galveston, Galveston, Texas, United States of America
- * E-mail: (SG); (NG)
| |
Collapse
|
47
|
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
|
48
|
Grit G, Van Coppernolle S, Devriendt B, Geurden T, Dreesen L, Hope J, Vercruysse J, Cox E, Geldhof P, Claerebout E. Evaluation of cellular and humoral systemic immune response against Giardia duodenalis infection in cattle. Vet Parasitol 2014; 202:145-55. [DOI: 10.1016/j.vetpar.2014.03.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Revised: 03/06/2014] [Accepted: 03/08/2014] [Indexed: 11/15/2022]
|