1
|
Vlajic K, Bie W, Gilic MB, Tyner AL. Impaired activation of succinate-induced type 2 immunity and secretory cell production in the small intestines of Ptk6-/- male mice. Cell Death Dis 2024; 15:777. [PMID: 39461944 PMCID: PMC11513114 DOI: 10.1038/s41419-024-07149-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 10/08/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024]
Abstract
Protein tyrosine kinase 6 (PTK6) is an intracellular tyrosine kinase that is distantly related to the SRC family of tyrosine kinases. It is expressed in epithelial linings and regulates regeneration and repair of the intestinal epithelium. Analysis of publicly available datasets showed Ptk6 is upregulated in tuft cells upon activation of type 2 immunity. We found that disruption of Ptk6 influences gene expression involved in intestinal immune responses. Administration of succinate, which mimics infection and activates tuft cells, revealed PTK6-dependent activation of innate immune responses in male but not female mice. In contrast to all wild type and Ptk6-/- female mice, Ptk6-/- male mice do not activate innate immunity or upregulate differentiation of the tuft and goblet secretory cell lineages following succinate treatment. Mechanistically, we found that PTK6 regulates Il25 and Irag2, genes that are required for tuft cell effector functions and activation of type 2 innate immunity, in organoids derived from intestines of male but not female mice. In patients with Crohn's disease, PTK6 is upregulated in tuft cells in noninflamed regions of intestine. These data highlight roles for PTK6 in contributing to sex differences in intestinal innate immunity and provide new insights into the regulation of IL-25.
Collapse
Affiliation(s)
- Katarina Vlajic
- Department of Biochemistry and Molecular Genetics, University of Illinois College of Medicine, University of Illinois at Chicago, Chicago, IL, 60607, USA
- University of Washington, Seattle, WA, USA
| | - Wenjun Bie
- Department of Biochemistry and Molecular Genetics, University of Illinois College of Medicine, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Milica B Gilic
- Department of Biochemistry and Molecular Genetics, University of Illinois College of Medicine, University of Illinois at Chicago, Chicago, IL, 60607, USA
- St Jude Children's Hospital, Memphis, TN, USA
| | - Angela L Tyner
- Department of Biochemistry and Molecular Genetics, University of Illinois College of Medicine, University of Illinois at Chicago, Chicago, IL, 60607, USA.
| |
Collapse
|
2
|
Zhang W, Yao W, Meng Y, Luo F, Han M, Mu Q, Jiang L, He W, Fan X, Wang W, Wang B. Effect of Moniezia Benedeni infection on ileal transcriptome profile characteristics of sheep. BMC Genomics 2024; 25:933. [PMID: 39370521 PMCID: PMC11457389 DOI: 10.1186/s12864-024-10853-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 09/30/2024] [Indexed: 10/08/2024] Open
Abstract
BACKGROUND The intestinal mucosal immune system, renowned for its precise and sensitive regulation, can provide comprehensive and effective protection for the body, among which the ileum is a critical induction site for regulating mucosal immune homeostasis. Moniezia benedeni parasitizes the small intestine of sheep and can cause serious pathological damage or even death to the host when the infection is severe. In this study, 5 sheep infected with Moniezia benedeni were selected as the infected group, and 5 uninfected sheep were selected as the control group. The ileal transcriptome profile characteristics of Moniezia benedeni infection were analyzed based on RNA-seq sequencing technology, aiming to lay a foundation for further exploring the perception mechanism of sheep intestines to Moniezia benedeni infection and formulating effective prevention and control strategies. RESULTS The results showed that a total of 3,891 differentially expressed genes (DEGs) were detected in the ileum tissues of sheep between the infected and control groups with 2,429 up-regulated genes and 1,462 down-regulated genes. GO and KEGG pathway enrichment analysis of differential genes, as well as Clue GO analysis showed that differential genes were significantly enriched in immune and metabolic-related biological processes and signaling pathways. Particularly, in immune-related signaling pathways, the B cell receptor signaling pathway was significantly down-regulated, while in metabolic regulation related signaling pathways, Bile secretion, Fat digestion and absorption and Vitamin digestion and absorption were notably up-regulated. On this basis, the differential core genes related to immune metabolism were verified by qRT-PCR method. The results showed that OVAR, CD3E, CD8A, CD4 and CD28 were significantly up-regulated (P < 0.05), while CIITA, BLNK, BCL6 and CD79A were significantly down-regulated (P < 0.05), which were consistent with transcriptome sequencing data. CONCLUSIONS The results demonstrated that Moniezia benedeni infection significantly affected the immune and metabolic processes in sheep ileum, particularly, it significantly inhibited the activation process of host B cells, and also led to an overactive function of bile acid metabolism. This finding provides a solid foundation for further elucidating the response mechanism of Peyer's patches in sheep ileum to Moniezia tapeworm infection.
Collapse
Affiliation(s)
- Wangdong Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Wanling Yao
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Yongcheng Meng
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Fuzhen Luo
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Mengling Han
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Qian Mu
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Lidong Jiang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Wanhong He
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Xiping Fan
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Wenhui Wang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China
| | - Baoshan Wang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, China.
| |
Collapse
|
3
|
Cressler CE, Metz DCG, Chang van Oordt DA, Graham AL. Immunological feedback loops generate parasite persistence thresholds that explain variation in infection duration. Proc Biol Sci 2024; 291:20240934. [PMID: 39317318 PMCID: PMC11421898 DOI: 10.1098/rspb.2024.0934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 07/24/2024] [Accepted: 08/06/2024] [Indexed: 09/26/2024] Open
Abstract
Infection duration affects individual host fitness and between-host transmission. Whether an infection is cleared or becomes chronic depends on the complex interaction between host immune responses and parasite growth. Empirical and theoretical studies have suggested that there are critical thresholds of parasite dose that can determine clearance versus chronicity, driven by the ability of the parasite to manipulate host immunity. However, the mammalian immune response is characterized by strong positive and negative feedback loops that could generate duration thresholds even in the absence of direct immunomodulation. Here, we derive and analyse a simple model for the interaction between T-cell subpopulations and parasite growth. We show that whether an infection is cleared or not is very sensitive to the initial immune state, parasite dose and strength of immunological feedbacks. In particular, chronic infections are possible even when parasites provoke a strong and effective immune response and lack any ability to immunomodulate. Our findings indicate that the initial immune state, which often goes unmeasured in empirical studies, is a critical determinant of infection duration. This work also has implications for epidemiological models, as it implies that infection duration will be highly variable among individuals, and dependent on each individual's infection history.
Collapse
Affiliation(s)
- Clayton E. Cressler
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Daniel C. G. Metz
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA
| | | | - Andrea L. Graham
- Department of Ecology & Evolutionary Biology, Princeton University, Princeton, NJ, USA
| |
Collapse
|
4
|
Desai P, Karl CE, Ying B, Liang CY, Garcia-Salum T, Santana AC, Ten-Caten F, Joseph F Urban, Elbashir SM, Edwards DK, Ribeiro SP, Thackray LB, Sekaly RP, Diamond MS. Intestinal helminth infection impairs vaccine-induced T cell responses and protection against SARS-CoV-2 in mice. Sci Transl Med 2024; 16:eado1941. [PMID: 39167662 DOI: 10.1126/scitranslmed.ado1941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 07/25/2024] [Indexed: 08/23/2024]
Abstract
Although vaccines have reduced the burden of COVID-19, their efficacy in helminth infection-endemic areas is not well characterized. We evaluated the impact of infection by Heligmosomoides polygyrus bakeri (Hpb), a murine intestinal roundworm, on the efficacy of an mRNA vaccine targeting the Wuhan-1 spike protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in mice. Although immunization generated similar B cell responses in Hpb-infected and uninfected mice, polyfunctional CD4+ and CD8+ T cell responses were markedly reduced in Hpb-infected mice. Hpb-infected and mRNA-vaccinated mice were protected against the ancestral SARS-CoV-2 strain WA1/2020, but control of lung infection was diminished against an Omicron variant compared with animals immunized without Hpb infection. Helminth-mediated suppression of spike protein-specific CD8+ T cell responses occurred independently of signal transducer and activator of transcription 6 (STAT6) signaling, whereas blockade of interleukin-10 (IL-10) rescued vaccine-induced CD8+ T cell responses. Together, these data show that, in mice, intestinal helminth infection impaired vaccine-induced T cell responses through an IL-10 pathway, which compromised protection against antigenically drifted SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Pritesh Desai
- Department of Medicine, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| | - Courtney E Karl
- Department of Molecular Microbiology, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| | - Baoling Ying
- Department of Medicine, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| | - Chieh-Yu Liang
- Department of Medicine, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| | - Tamara Garcia-Salum
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30317, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Ana Carolina Santana
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30317, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Felipe Ten-Caten
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30317, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Joseph F Urban
- US Department of Agriculture, Agricultural Research Services, Beltsville Human Nutrition Research Center, Diet, Genomics, and Immunology Laboratory, and Beltsville Agricultural Research Center, Animal Parasitic Diseases Laboratory, Beltsville, MD 20705, USA
| | | | | | - Susan P Ribeiro
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30317, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Larissa B Thackray
- Department of Medicine, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| | - Rafick P Sekaly
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30317, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Michael S Diamond
- Department of Medicine, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
- Department of Molecular Microbiology, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
- Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
5
|
Billipp TE, Fung C, Webeck LM, Sargent DB, Gologorsky MB, Chen Z, McDaniel MM, Kasal DN, McGinty JW, Barrow KA, Rich LM, Barilli A, Sabat M, Debley JS, Wu C, Myers R, Howitt MR, von Moltke J. Tuft cell-derived acetylcholine promotes epithelial chloride secretion and intestinal helminth clearance. Immunity 2024; 57:1243-1259.e8. [PMID: 38744291 PMCID: PMC11168877 DOI: 10.1016/j.immuni.2024.03.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 02/05/2024] [Accepted: 03/29/2024] [Indexed: 05/16/2024]
Abstract
Epithelial cells secrete chloride to regulate water release at mucosal barriers, supporting both homeostatic hydration and the "weep" response that is critical for type 2 immune defense against parasitic worms (helminths). Epithelial tuft cells in the small intestine sense helminths and release cytokines and lipids to activate type 2 immune cells, but whether they regulate epithelial secretion is unknown. Here, we found that tuft cell activation rapidly induced epithelial chloride secretion in the small intestine. This response required tuft cell sensory functions and tuft cell-derived acetylcholine (ACh), which acted directly on neighboring epithelial cells to stimulate chloride secretion, independent of neurons. Maximal tuft cell-induced chloride secretion coincided with immune restriction of helminths, and clearance was delayed in mice lacking tuft cell-derived ACh, despite normal type 2 inflammation. Thus, we have uncovered an epithelium-intrinsic response unit that uses ACh to couple tuft cell sensing to the secretory defenses of neighboring epithelial cells.
Collapse
Affiliation(s)
- Tyler E Billipp
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - Connie Fung
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lily M Webeck
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - Derek B Sargent
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - Matthew B Gologorsky
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Zuojia Chen
- Experimental Immunology Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Margaret M McDaniel
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - Darshan N Kasal
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - John W McGinty
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - Kaitlyn A Barrow
- Center for Respiratory Biology and Therapeutics, Seattle Children's Research Institute, Seattle, WA, USA
| | - Lucille M Rich
- Center for Respiratory Biology and Therapeutics, Seattle Children's Research Institute, Seattle, WA, USA
| | | | - Mark Sabat
- Takeda Pharmaceuticals, San Diego, CA, USA
| | - Jason S Debley
- Center for Respiratory Biology and Therapeutics, Seattle Children's Research Institute, Seattle, WA, USA; Department of Pediatrics, Division of Pulmonary and Sleep Medicine, Seattle Children's Hospital, University of Washington, Seattle, WA, USA
| | - Chuan Wu
- Experimental Immunology Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | | | - Michael R Howitt
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jakob von Moltke
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA.
| |
Collapse
|
6
|
Dean AD, Childs DZ, Corripio‐Miyar Y, Evans M, Hayward A, Kenyon F, McNally L, McNeilly TN, Pakeman RJ, Sweeny AR, Nussey DH, Pedersen AB, Fenton A. Host resources and parasite traits interact to determine the optimal combination of host parasite-mitigation strategies. Ecol Evol 2024; 14:e11310. [PMID: 38903143 PMCID: PMC11187858 DOI: 10.1002/ece3.11310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/29/2024] [Accepted: 04/03/2024] [Indexed: 06/22/2024] Open
Abstract
Organisms have evolved diverse strategies to manage parasite infections. Broadly, hosts may avoid infection by altering behaviour, resist infection by targeting parasites or tolerate infection by repairing associated damage. The effectiveness of a strategy depends on interactions between, for example, resource availability, parasite traits (virulence, life-history) and the host itself (nutritional status, immunopathology). To understand how these factors shape host parasite-mitigation strategies, we developed a mathematical model of within-host, parasite-immune dynamics in the context of helminth infections. The model incorporated host nutrition and resource allocation to different mechanisms of immune response: larval parasite prevention; adult parasite clearance; damage repair (tolerance). We also considered a non-immune strategy: avoidance via anorexia, reducing intake of infective stages. Resources not allocated to immune processes promoted host condition, whereas harm due to parasites and immunopathology diminished it. Maximising condition (a proxy for fitness), we determined optimal host investment for each parasite-mitigation strategy, singly and combined, across different environmental resource levels and parasite trait values. Which strategy was optimal varied with scenario. Tolerance generally performed well, especially with high resources. Success of the different resistance strategies (larval prevention or adult clearance) tracked relative virulence of larval and adult parasites: slowly maturing, highly damaging larvae favoured prevention; rapidly maturing, less harmful larvae favoured clearance. Anorexia was viable only in the short term, due to reduced host nutrition. Combined strategies always outperformed any lone strategy: these were dominated by tolerance, with some investment in resistance. Choice of parasite mitigation strategy has profound consequences for hosts, impacting their condition, survival and reproductive success. We show that the efficacy of different strategies is highly dependent on timescale, parasite traits and resource availability. Models that integrate such factors can inform the collection and interpretation of empirical data, to understand how those drivers interact to shape host immune responses in natural systems.
Collapse
Affiliation(s)
- Andrew D. Dean
- Institute of Infection, Veterinary and Ecological SciencesUniversity of LiverpoolLiverpoolUK
| | | | | | - Mike Evans
- Department for Disease ControlMoredun Research InstitutePenicuikUK
- The University of Edinburgh Royal (Dick) School of Veterinary StudiesRoslinUK
- Institute of Ecology and Evolution, School of Biological SciencesUniversity of EdinburghEdinburghUK
| | - Adam Hayward
- Department for Disease ControlMoredun Research InstitutePenicuikUK
| | - Fiona Kenyon
- Department for Disease ControlMoredun Research InstitutePenicuikUK
| | - Luke McNally
- Institute of Ecology and Evolution, School of Biological SciencesUniversity of EdinburghEdinburghUK
| | - Tom N. McNeilly
- Department for Disease ControlMoredun Research InstitutePenicuikUK
| | | | - Amy R. Sweeny
- School of BiosciencesThe University of SheffieldSheffieldUK
- Institute of Ecology and Evolution, School of Biological SciencesUniversity of EdinburghEdinburghUK
| | - Daniel H. Nussey
- Institute of Ecology and Evolution, School of Biological SciencesUniversity of EdinburghEdinburghUK
| | - Amy B. Pedersen
- Institute of Ecology and Evolution, School of Biological SciencesUniversity of EdinburghEdinburghUK
| | - Andy Fenton
- Institute of Infection, Veterinary and Ecological SciencesUniversity of LiverpoolLiverpoolUK
| |
Collapse
|
7
|
Oser L, Midha A, Schlosser-Brandenburg J, Rausch S, Mugo RM, Kundik A, Elizalde-Velázquez LE, Adjah J, Musimbi ZD, Klopfleisch R, Helm CS, von Samson-Himmelstjerna G, Hartmann S, Ebner F. Ascaris suum infection in juvenile pigs elicits a local Th2 response in a setting of ongoing Th1 expansion. Front Immunol 2024; 15:1396446. [PMID: 38799456 PMCID: PMC11116563 DOI: 10.3389/fimmu.2024.1396446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 04/08/2024] [Indexed: 05/29/2024] Open
Abstract
Ascaris spp. undergo extensive migration within the body before establishing patent infections in the small intestinal tract of humans and pigs. However, whether larval migration is critical for inducing efficient type 2 responses remains poorly understood. Therefore, we investigated systemic versus local adaptive immune responses along the hepato-tracheal migration of Ascaris suum during primary, single infections in conventionally raised pigs. Neither the initial invasion of gut tissue nor migration through the liver resulted in discernable Th2 cell responses. In contrast, lung-stage larvae elicited a Th2-biased pulmonary response, which declined after the larvae had left the lungs. In the small intestine, we observed an accumulation of Th2 cells upon the arrival of fourth-stage larvae (L4) to the small intestinal lumen. In parallel, we noticed robust and increasing Th1 responses in circulation, migration-affected organs, and draining lymph nodes. Phenotypic analysis of CD4+ T cells specifically recognizing A. suum antigens in the circulation and lung tissue of infected pigs confirmed that the majority of Ascaris-specific T cells produced IL-4 (Th2) and, to a much lesser extent, IL-4/IFN-g (Th2/1 hybrids) or IFN-g alone (Th1). These data demonstrate that lung-stage but not the early liver-stage larvae lead to a locally restricted Th2 response. Significant Th2 cell accumulation in the small intestine occurs only when L4 complete the body migration. In addition, Th2 immunity seems to be hampered by the concurrent, nonspecific Th1 bias in growing pigs. Together, the late onset of Th2 immunity at the site of infection and the Th1-biased systemic immunity likely enable the establishment of intestinal infections by sufficiently large L4 stages and pre-adult worms, some of which resist expulsion mechanisms.
Collapse
Affiliation(s)
- Larissa Oser
- Centre for Infection Medicine, Department of Veterinary Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| | - Ankur Midha
- Centre for Infection Medicine, Department of Veterinary Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| | - Josephine Schlosser-Brandenburg
- Centre for Infection Medicine, Department of Veterinary Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| | - Sebastian Rausch
- Centre for Infection Medicine, Department of Veterinary Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| | - Robert M. Mugo
- Centre for Infection Medicine, Department of Veterinary Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| | - Arkadi Kundik
- Centre for Infection Medicine, Department of Veterinary Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| | - Luis E. Elizalde-Velázquez
- Centre for Infection Medicine, Department of Veterinary Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| | - Joshua Adjah
- Centre for Infection Medicine, Department of Veterinary Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| | - Zaneta D. Musimbi
- Centre for Infection Medicine, Department of Veterinary Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| | - Robert Klopfleisch
- Department of Veterinary Medicine, Institute of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Christina S. Helm
- Department of Veterinary Medicine, Institute for Parasitology and Tropical Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| | - Georg von Samson-Himmelstjerna
- Department of Veterinary Medicine, Institute for Parasitology and Tropical Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| | - Susanne Hartmann
- Centre for Infection Medicine, Department of Veterinary Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| | - Friederike Ebner
- Centre for Infection Medicine, Department of Veterinary Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
- Infection Pathogenesis, School of Life Sciences, Technical University of Munich, Freising, Germany
| |
Collapse
|
8
|
Toledo R, Cociancic P, Fiallos E, Esteban JG, Muñoz-Antoli C. Immunology and pathology of echinostomes and other intestinal trematodes. ADVANCES IN PARASITOLOGY 2024; 124:1-55. [PMID: 38754926 DOI: 10.1016/bs.apar.2024.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2024]
Abstract
Intestinal trematodes constitute a major group of helminths that parasitize humans and animals with relevant morbidity and mortality. Despite the importance of the intestinal trematodes in medical and veterinary sciences, immunology and pathology of these helminth infections have been neglected for years. Apart from the work focused on the members of the family Echnistomatidae, there are only very isolated and sporadic studies on the representatives of other families of digeneans, which makes a compilation of all these studies necessary. In the present review, the most salient literature on the immunology and pathology of intestinal trematodes in their definitive hosts in examined. Emphasis will be placed on members of the echinostomatidae family, since it is the group in which the most work has been carried out. However, we also review the information on selected species of the families Brachylaimidae, Diplostomidae, Gymnophallidae, and Heterophyidae. For most of these families, coverage is considered under the following headings: (i) Background; (ii) Pathology of the infection; (iii) Immunology of the infection; and (iv) Human infections.
Collapse
Affiliation(s)
- Rafael Toledo
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, Faculty of Pharmacy, University of Valencia, Valencia, Spain.
| | - Paola Cociancic
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, Faculty of Pharmacy, University of Valencia, Valencia, Spain
| | - Emma Fiallos
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, Faculty of Pharmacy, University of Valencia, Valencia, Spain
| | - J Guillermo Esteban
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, Faculty of Pharmacy, University of Valencia, Valencia, Spain
| | - Carla Muñoz-Antoli
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, Faculty of Pharmacy, University of Valencia, Valencia, Spain
| |
Collapse
|
9
|
Desai P, Karl CE, Ying B, Liang CY, Garcia-Salum T, Santana AC, Caten FT, Urban JF, Elbashir SM, Edwards DK, Ribeiro SP, Thackray LB, Sekaly RP, Diamond MS. Intestinal helminth infection impairs vaccine-induced T cell responses and protection against SARS-CoV-2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.14.575588. [PMID: 38293221 PMCID: PMC10827110 DOI: 10.1101/2024.01.14.575588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Although vaccines have reduced COVID-19 disease burden, their efficacy in helminth infection endemic areas is not well characterized. We evaluated the impact of infection by Heligmosomoides polygyrus bakeri (Hpb), a murine intestinal hookworm, on the efficacy of an mRNA vaccine targeting the Wuhan-1 spike protein of SARS-CoV-2. Although immunization generated similar B cell responses in Hpb-infected and uninfected mice, polyfunctional CD4+ and CD8+ T cell responses were markedly reduced in Hpb-infected mice. Hpb-infected and mRNA vaccinated mice were protected against the ancestral SARS-CoV-2 strain WA1/2020, but control of lung infection was diminished against an Omicron variant compared to animals immunized without Hpb infection. Helminth mediated suppression of spike-specific CD8+ T cell responses occurred independently of STAT6 signaling, whereas blockade of IL-10 rescued vaccine-induced CD8+ T cell responses. In mice, intestinal helminth infection impairs vaccine induced T cell responses via an IL-10 pathway and compromises protection against antigenically shifted SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Pritesh Desai
- Department of Medicine, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
| | - Courtney E. Karl
- Department of Molecular Microbiology, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
| | - Baoling Ying
- Department of Medicine, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
| | - Chieh-Yu Liang
- Department of Medicine, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
| | - Tamara Garcia-Salum
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Ana Carolina Santana
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Felipe Ten Caten
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Joseph F. Urban
- US Department of Agriculture, Agricultural Research Services, Beltsville Human Nutrition Research Center, Diet, Genomics, and Immunology Laboratory, and Beltsville Agricultural Research Center, Animal Parasitic Diseases Laboratory, Beltsville, MD 20705-2350, USA
| | | | | | - Susan P. Ribeiro
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Larissa B. Thackray
- Department of Medicine, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
| | - Rafick P. Sekaly
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Michael S. Diamond
- Department of Medicine, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
- Department of Molecular Microbiology, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
| |
Collapse
|
10
|
Wang J, Zhao X, Li X, Jin X. Akkermansia muciniphila: a deworming partner independent of type 2 immunity. Gut Microbes 2024; 16:2338947. [PMID: 38717824 PMCID: PMC11086001 DOI: 10.1080/19490976.2024.2338947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 04/01/2024] [Indexed: 05/12/2024] Open
Abstract
The gut microbiota has coevolved with the host for hundreds of millions of years, playing a beneficial role in host health. Human parasitic helminths are widespread and pose a pervasive global public health issue. Although Type 2 immunity provides partial resistance to helminth infections, the composition of the gut microbiota can change correspondingly. Therefore, it raises the question of what role the gut microbiota plays during helminth infection. Akkermansia muciniphila has emerged as a notable representative of beneficial microorganisms in the gut microbiota. Recent studies indicate that A. muciniphila is not merely associated with helminth infection but is also causally linked to infection. Here, we provide an overview of the crosstalk between A. muciniphila and enteric helminth infection. Our goal is to enhance our understanding of the interplay among A. muciniphila, helminths, and their hosts while also exploring the potential underlying mechanisms.
Collapse
Affiliation(s)
- Jiaqi Wang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, College of Animal Sciences, Jilin University, Changchun, China
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xiufeng Zhao
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians University of Würzburg, Würzburg, Germany
| | - Xianhe Li
- Department of Microbiology, Immunology and Molecular Genetics, University of California Los Angeles, Los Angeles, USA
| | - Xuemin Jin
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| |
Collapse
|
11
|
Gyu Choi H, Woong Kwon K, Jae Shin S. Importance of adjuvant selection in tuberculosis vaccine development: Exploring basic mechanisms and clinical implications. Vaccine X 2023; 15:100400. [PMID: 37965276 PMCID: PMC10641539 DOI: 10.1016/j.jvacx.2023.100400] [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: 05/02/2023] [Revised: 09/13/2023] [Accepted: 10/18/2023] [Indexed: 11/16/2023] Open
Abstract
The global emergency of unexpected pathogens, exemplified by SARS-CoV-2, has emphasized the importance of vaccines in thwarting infection and curtailing the progression of severe disease. The scourge of tuberculosis (TB), emanating from the Mycobacterium tuberculosis (Mtb) complex, has inflicted a more profound toll in terms of mortality and morbidity than any other infectious agents prior to the SARS-CoV-2 pandemic. Despite the existence of Bacillus Calmette-Guérin (BCG), the only licensed vaccine developed a century ago, its efficacy against TB remains unsatisfactory, particularly in preventing pulmonary Mtb infections in adolescents and adults. However, collaborations between academic and industrial entities have led to a renewed impetus in the development of TB vaccines, with numerous candidates, particularly subunit vaccines with specialized adjuvants, exhibiting promising outcomes in recent clinical studies. Adjuvants are crucial in modulating optimal immunological responses, by endowing immune cells with sufficient antigen and immune signals. As exemplified by the COVID-19 vaccine landscape, the interplay between vaccine efficacy and adverse effects is of paramount importance, particularly for the elderly and individuals with underlying ailments such as diabetes and concurrent infections. In this regard, adjuvants hold the key to optimizing vaccine efficacy and safety. This review accentuates the pivotal roles of adjuvants and their underlying mechanisms in the development of TB vaccines. Furthermore, we expound on the prospects for the development of more efficacious adjuvants and their synergistic combinations for individuals in diverse states, such as aging, HIV co-infection, and diabetes, by examining the immunological alterations that arise with aging and comparing them with those observed in younger cohorts.
Collapse
Affiliation(s)
- Han Gyu Choi
- Department of Microbiology, and Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Kee Woong Kwon
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Sung Jae Shin
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, South Korea
| |
Collapse
|
12
|
Campillo Poveda M, Britton C, Devaney E, McNeilly TN, Gerbe F, Jay P, Maizels RM. Tuft Cells: Detectors, Amplifiers, Effectors and Targets in Parasite Infection. Cells 2023; 12:2477. [PMID: 37887321 PMCID: PMC10605326 DOI: 10.3390/cells12202477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/12/2023] [Accepted: 10/12/2023] [Indexed: 10/28/2023] Open
Abstract
Tuft cells have recently emerged as the focus of intense interest following the discovery of their chemosensory role in the intestinal tract, and their ability to activate Type 2 immune responses to helminth parasites. Moreover, they populate a wide range of mucosal tissues and are intimately connected to immune and neuronal cells, either directly or through the release of pharmacologically active mediators. They are now recognised to fulfil both homeostatic roles, in metabolism and tissue integrity, as well as acting as the first sensors of parasite infection, immunity to which is lost in their absence. In this review we focus primarily on the importance of tuft cells in the intestinal niche, but also link to their more generalised physiological role and discuss their potential as targets for the treatment of gastrointestinal disorders.
Collapse
Affiliation(s)
- Marta Campillo Poveda
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow G12 8TA, UK;
| | - Collette Britton
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow G61 1QH, UK; (C.B.); (E.D.)
| | - Eileen Devaney
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow G61 1QH, UK; (C.B.); (E.D.)
| | - Tom N. McNeilly
- Disease Control Department, Moredun Research Institute, Penicuik EH26 0PZ, UK;
| | - François Gerbe
- Institute of Functional Genomics (IGF), University of Montpellier, CNRS, INSERM, F-34094 Montpellier, France; (F.G.); (P.J.)
| | - Philippe Jay
- Institute of Functional Genomics (IGF), University of Montpellier, CNRS, INSERM, F-34094 Montpellier, France; (F.G.); (P.J.)
| | - Rick M. Maizels
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow G12 8TA, UK;
| |
Collapse
|
13
|
Chen H, Cao Z, Liu M, Diamond MS, Jin X. The impact of helminth-induced immunity on infection with bacteria or viruses. Vet Res 2023; 54:87. [PMID: 37789420 PMCID: PMC10548622 DOI: 10.1186/s13567-023-01216-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 08/21/2023] [Indexed: 10/05/2023] Open
Abstract
Different human and animal pathogens trigger distinct immune responses in their hosts. The infection of bacteria or viruses can trigger type I pro-inflammatory immune responses (e.g., IFN-γ, TNF-α, TH1 cells), whereas infection by helminths typically elicits a type II host resistance and tolerizing immune response (e.g., IL-4, IL-5, IL-13, TH2 cells). In some respects, the type I and II immune responses induced by these different classes of pathogens are antagonistic. Indeed, recent studies indicate that infection by helminths differentially shapes the response and outcome of subsequent infection by viruses and bacteria. In this review, we summarize the current knowledge on how helminth infections influence concurrent or subsequent microbial infections and also discuss the implications for helminth-mediated immunity on the outcome of SARS-CoV-2 disease.
Collapse
Affiliation(s)
- Hong Chen
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Zengguo Cao
- State Key Laboratory of Virology, Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Mingyuan Liu
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, China
| | - Michael S Diamond
- Departments of Medicine, Molecular Microbiology, Pathology, and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Xuemin Jin
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China.
| |
Collapse
|
14
|
Maizels RM, Gause WC. Targeting helminths: The expanding world of type 2 immune effector mechanisms. J Exp Med 2023; 220:e20221381. [PMID: 37638887 PMCID: PMC10460967 DOI: 10.1084/jem.20221381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 06/24/2023] [Accepted: 08/09/2023] [Indexed: 08/29/2023] Open
Abstract
In this new review, Rick Maizels and Bill Gause summarize how type 2 immune responses combat helminth parasites through novel mechanisms, coordinating multiple innate and adaptive cell and molecular players that can eliminate infection and repair-resultant tissue damage.
Collapse
Affiliation(s)
- Rick M. Maizels
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - William C. Gause
- Center for Immunity and Inflammation, Rutgers Biomedical Health Sciences Institute for Infectious and Inflammatory Diseases, New Jersey Medical School, Rutgers Biomedical Health Sciences, Newark, NJ, USA
| |
Collapse
|
15
|
Denning DW, Pfavayi LT. Poorly controlled asthma - Easy wins and future prospects for addressing fungal allergy. Allergol Int 2023; 72:493-506. [PMID: 37544851 DOI: 10.1016/j.alit.2023.07.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 06/25/2023] [Accepted: 06/28/2023] [Indexed: 08/08/2023] Open
Abstract
Poorly controlled asthma is especially common in low resource countries. Aside from lack of access to, or poor technique with, inhaled beta-2 agonists and corticosteroids, the most problematic forms of asthma are frequently associated with both fungal allergy and exposure, especially in adults leading to more asthma exacerbations and worse asthma. The umbrella term 'fungal asthma' describes many disorders linked to fungal exposure and/or allergy to fungi. One fungal asthma endotype, ABPA, is usually marked by a very high IgE and its differential diagnosis is reviewed. Both ABPA and fungal bronchitis in bronchiectasis are marked by thick excess airway mucus production. Dermatophyte skin infection can worsen asthma and eradication of the skin infection improves asthma. Exposure to fungi in the workplace, home and schools, often in damp or water-damaged buildings worsens asthma, and remediation improves symptom control and reduces exacerbations. Antifungal therapy is beneficial for fungal asthma as demonstrated in nine of 13 randomised controlled studies, reducing symptoms, corticosteroid need and exacerbations while improving lung function. Other useful therapies include azithromycin and some biologics approved for the treatment of severe asthma. If all individuals with poorly controlled and severe asthma could be 'relieved' of their fungal allergy and infection through antifungal therapy without systemic corticosteroids, the health benefits would be enormous and relatively inexpensive, improving the long term health of over 20 million adults and many children. Antifungal therapy carries some toxicity, drug interactions and triazole resistance risks, and data are incomplete. Here we summarise what is known and what remains uncertain about this complex topic.
Collapse
Affiliation(s)
- David W Denning
- Manchester Fungal Infection Group, The University of Manchester and Manchester Academic Health Science Centre, Manchester, UK.
| | - Lorraine T Pfavayi
- Institute of Immunology & Infection Research, Ashworth Laboratories, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
16
|
Oehm AW, Leinmueller M, Zablotski Y, Campe A, Hoedemaker M, Springer A, Jordan D, Strube C, Knubben-Schweizer G. Multinomial logistic regression based on neural networks reveals inherent differences among dairy farms depending on the differential exposure to Fasciola hepatica and Ostertagia ostertagi. Int J Parasitol 2023; 53:687-697. [PMID: 37355196 DOI: 10.1016/j.ijpara.2023.05.006] [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: 03/31/2023] [Revised: 05/17/2023] [Accepted: 05/18/2023] [Indexed: 06/26/2023]
Abstract
Fasciola hepatica and Ostertagia ostertagi are cattle parasites with worldwide relevance for economic outcome as well as animal health and welfare. The on-farm exposure of cattle to both parasites is a function of host-associated, intrinsic, as well as environmental and farm-specific, extrinsic, factors. Even though knowledge on the biology of both parasites exists, sophisticated and innovative modelling approaches can help to deepen our understanding of key aspects fostering the exposure of dairy cows to these pathogens. In the present study, multiple multinomial logistic regression models were fitted via neural networks to describe the differences among farms where cattle were not exposed to either F. hepatica or O. ostertagi, to one parasite, or to both, respectively. Farm-specific production and management characteristics were used as covariates to portray these differences. This elucidated inherent farm characteristics associated with parasite exposure. In both studied regions, pasture access for cows, farm-level milk yield, and lameness prevalence were identified as relevant factors. In region 'South', adherence to organic farming principles was a further covariate of importance. In region 'North', the prevalence of cows with a low body condition score, herd size, hock lesion prevalence, farm-level somatic cell count, and study year appeared to be of relevance. The present study broadens our understanding of the complex epidemiological scenarios that could predict differential farm-level parasite status. The analyses have revealed the importance of awareness of dissimilarities between farms in regard to the differential exposure to F. hepatica and O. ostertagi. This provides solid evidence that dynamics and relevant factors differ depending on whether or not cows are exposed to F. hepatica, O. ostertagi, or to both.
Collapse
Affiliation(s)
- Andreas W Oehm
- Institute of Parasitology, Vetsuisse Faculty of Zurich, University of Zurich, Zurich, Switzerland; Clinic for Ruminants with Ambulatory and Herd Health Services, Ludwig-Maximilians-Universität Munich, Oberschleissheim, Germany.
| | - Markus Leinmueller
- Clinic for Ruminants with Ambulatory and Herd Health Services, Ludwig-Maximilians-Universität Munich, Oberschleissheim, Germany
| | - Yury Zablotski
- Clinic for Ruminants with Ambulatory and Herd Health Services, Ludwig-Maximilians-Universität Munich, Oberschleissheim, Germany
| | - Amely Campe
- Department of Biometry, Epidemiology and Information Processing, WHO Collaborating Center for Research and Training for Health at the Human-Animal-Environment Interface, University of Veterinary Medicine, Foundation, Hannover, Germany
| | - Martina Hoedemaker
- Clinic for Cattle, University of Veterinary Medicine Hannover Foundation, Hannover, Germany
| | - Andrea Springer
- Institute for Parasitology, Centre for Infection Medicine, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Daniela Jordan
- Institute for Parasitology, Centre for Infection Medicine, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Christina Strube
- Institute for Parasitology, Centre for Infection Medicine, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Gabriela Knubben-Schweizer
- Clinic for Ruminants with Ambulatory and Herd Health Services, Ludwig-Maximilians-Universität Munich, Oberschleissheim, Germany
| |
Collapse
|
17
|
Wang J, Liu X, Sun R, Mao H, Liu M, Jin X. Akkermansia muciniphila participates in the host protection against helminth-induced cardiac fibrosis via TLR2. PLoS Pathog 2023; 19:e1011683. [PMID: 37788279 PMCID: PMC10547169 DOI: 10.1371/journal.ppat.1011683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 09/12/2023] [Indexed: 10/05/2023] Open
Abstract
Helminth Trichinella spiralis (Ts) is one of the major pathogens of human infective myocarditis that can lead to cardiac fibrosis (CF). The gut microbiota involved in this pathology are of interest. Here, we use mice infected with Ts as a model to examine the interactions between gut microbes and host protection to CF. Infected mice show enhanced CF severity. We find that antibiotics treatment to deplete the microbiota aggravates the disease phenotype. Attempts to restore microbiota using fecal microbiota transplantation ameliorates helminth-induced CF. 16S rRNA gene sequencing and metagenomics sequencing reveal a higher abundance of Akkermansia muciniphila in gut microbiomes of Ts-infected mice. Oral supplementation with alive or pasteurized A. muciniphila improves CF via TLR2. This work represents a substantial advance toward our understanding of causative rather than correlative relationships between the gut microbiota and CF.
Collapse
Affiliation(s)
- Jiaqi Wang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, College of Animal Sciences, Jilin University, Changchun, China
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xiaolei Liu
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Ruohang Sun
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Hanhai Mao
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Mingyuan Liu
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Xuemin Jin
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| |
Collapse
|
18
|
Inaba R, Vujakovic S, Bergstrom K. The gut mucus network: A dynamic liaison between microbes and the immune system. Semin Immunol 2023; 69:101807. [PMID: 37478802 DOI: 10.1016/j.smim.2023.101807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 06/24/2023] [Accepted: 07/08/2023] [Indexed: 07/23/2023]
Abstract
A complex mucus network made up of large polymers of the mucin-family glycoprotein MUC2 exists between the large intestinal microbial mass and epithelial and immune cells. This has long been understood as an innate immune defense barrier against the microbiota and other luminal threats that reinforces the barrier function of the epithelium and limits microbiota contact with the tissues. However, past and recent studies have provided new evidence of how critical the mucus network is to act as a 'liaison' between host and microbe to mediate anti-inflammatory, mutualistic interactions with the microbiota and protection from pathogens. This review summarizes historical and recent insights into the formation of the gut mucus network, how the microbes and immune system influence mucus, and in turn, how the mucus influences immune responses to the microbiota.
Collapse
Affiliation(s)
- Rain Inaba
- Department of Biology, University of British Columbia, Okanagan Campus, 3187 University Way, Kelowna V1V 1V7, British Columbia, Canada
| | - Sara Vujakovic
- Department of Biology, University of British Columbia, Okanagan Campus, 3187 University Way, Kelowna V1V 1V7, British Columbia, Canada
| | - Kirk Bergstrom
- Department of Biology, University of British Columbia, Okanagan Campus, 3187 University Way, Kelowna V1V 1V7, British Columbia, Canada.
| |
Collapse
|
19
|
Smita S, Webb LM, Mooney B, Früh SP, Oyesola OO, Matheson MK, Peng SA, Tait Wojno ED. Basophil responses in susceptible AKR mice upon infection with the intestinal helminth parasite Trichuris muris. Parasite Immunol 2023; 45:e12999. [PMID: 37415265 PMCID: PMC10513073 DOI: 10.1111/pim.12999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 06/01/2023] [Accepted: 06/06/2023] [Indexed: 07/08/2023]
Abstract
Intestinal helminth infection promotes a Type 2 inflammatory response in resistant C57BL/6 mice that is essential for worm clearance. The study of inbred mouse strains has revealed factors that are critical for parasite resistance and delineated the role of Type 1 versus Type 2 immune responses in worm clearance. In C57BL/6 mice, basophils are key innate immune cells that promote Type 2 inflammation and are programmed via the Notch signalling pathway during infection with the helminth Trichuris muris. However, how the host genetic background influences basophil responses and basophil expression of Notch receptors remains unclear. Here we use genetically susceptible inbred AKR/J mice that have a Type 1-skewed immune response during T. muris infection to investigate basophil responses in a susceptible host. Basophil population expansion occurred in AKR/J mice even in the absence of fulminant Type 2 inflammation during T. muris infection. However, basophils in AKR/J mice did not robustly upregulate expression of the Notch2 receptor in response to infection as occurred in C57BL/6 mice. Blockade of the Type 1 cytokine interferon-γ in infected AKR/J mice was not sufficient to elicit infection-induced basophil expression of the Notch2 receptor. These data suggest that the host genetic background, outside of the Type 1 skew, is important in regulating basophil responses during T. muris infection in susceptible AKR/J mice.
Collapse
Affiliation(s)
- Shuchi Smita
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - Lauren M. Webb
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - Bridget Mooney
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - Simon P. Früh
- Baker Institute for Animal Health and Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY, USA
| | - Oyebola O. Oyesola
- Department of Immunology, University of Washington, Seattle, WA, USA
- Baker Institute for Animal Health and Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY, USA
| | - Macy K. Matheson
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - Seth A. Peng
- Baker Institute for Animal Health and Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY, USA
| | | |
Collapse
|
20
|
Chakraborty P, Aravindhan V, Mukherjee S. Helminth-derived biomacromolecules as therapeutic agents for treating inflammatory and infectious diseases: What lessons do we get from recent findings? Int J Biol Macromol 2023; 241:124649. [PMID: 37119907 DOI: 10.1016/j.ijbiomac.2023.124649] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/20/2023] [Accepted: 04/24/2023] [Indexed: 05/01/2023]
Abstract
Despite the tremendous progress in healthcare sectors, a number of life-threatening infectious, inflammatory, and autoimmune diseases are continuously challenging mankind throughout the globe. In this context, recent successes in utilizing helminth parasite-derived bioactive macromolecules viz. glycoproteins, enzymes, polysaccharides, lipids/lipoproteins, nucleic acids/nucleotides, and small organic molecules for treating various disorders primarily resulted from inflammation. Among the several parasites that infect humans, helminths (cestodes, nematodes, and trematodes) are known as efficient immune manipulators owing to their explicit ability to modulate and modify the innate and adaptive immune responses of humans. These molecules selectively bind to immune receptors on innate and adaptive immune cells and trigger multiple signaling pathways to elicit anti-inflammatory cytokines, expansion of alternatively activated macrophages, T-helper 2, and immunoregulatory T regulatory cell types to induce an anti-inflammatory milieu. Reduction of pro-inflammatory responses and repair of tissue damage by these anti-inflammatory mediators have been exploited for treating a number of autoimmune, allergic, and metabolic diseases. Herein, the potential and promises of different helminths/helminth-derived products as therapeutic agents in ameliorating immunopathology of different human diseases and their mechanistic insights of function at cell and molecular level alongside the molecular signaling cross-talks have been reviewed by incorporating up-to-date findings achieved in the field.
Collapse
Affiliation(s)
- Pritha Chakraborty
- Integrative Biochemistry & Immunology Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol 713340, India
| | | | - Suprabhat Mukherjee
- Integrative Biochemistry & Immunology Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol 713340, India.
| |
Collapse
|
21
|
Billipp TE, Fung C, Webeck LM, Sargent DB, Gologorsky MB, McDaniel MM, Kasal DN, McGinty JW, Barrow KA, Rich LM, Barilli A, Sabat M, Debley JS, Myers R, Howitt MR, von Moltke J. Tuft cell-derived acetylcholine regulates epithelial fluid secretion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.17.533208. [PMID: 36993541 PMCID: PMC10055254 DOI: 10.1101/2023.03.17.533208] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Tuft cells are solitary chemosensory epithelial cells that can sense lumenal stimuli at mucosal barriers and secrete effector molecules to regulate the physiology and immune state of their surrounding tissue. In the small intestine, tuft cells detect parasitic worms (helminths) and microbe-derived succinate, and signal to immune cells to trigger a Type 2 immune response that leads to extensive epithelial remodeling spanning several days. Acetylcholine (ACh) from airway tuft cells has been shown to stimulate acute changes in breathing and mucocilliary clearance, but its function in the intestine is unknown. Here we show that tuft cell chemosensing in the intestine leads to release of ACh, but that this does not contribute to immune cell activation or associated tissue remodeling. Instead, tuft cell-derived ACh triggers immediate fluid secretion from neighboring epithelial cells into the intestinal lumen. This tuft cell-regulated fluid secretion is amplified during Type 2 inflammation, and helminth clearance is delayed in mice lacking tuft cell ACh. The coupling of the chemosensory function of tuft cells with fluid secretion creates an epithelium-intrinsic response unit that effects a physiological change within seconds of activation. This response mechanism is shared by tuft cells across tissues, and serves to regulate the epithelial secretion that is both a hallmark of Type 2 immunity and an essential component of homeostatic maintenance at mucosal barriers.
Collapse
Affiliation(s)
- Tyler E. Billipp
- Department of Immunology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Connie Fung
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lily M. Webeck
- Department of Immunology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Derek B. Sargent
- Department of Immunology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Matthew B. Gologorsky
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Margaret M. McDaniel
- Department of Immunology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Darshan N. Kasal
- Department of Immunology, University of Washington School of Medicine, Seattle, Washington, USA
| | - John W. McGinty
- Department of Immunology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Kaitlyn A. Barrow
- Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Lucille M. Rich
- Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, Washington, USA
| | | | - Mark Sabat
- Takeda Pharmaceuticals, San Diego, California, USA
| | - Jason S. Debley
- Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, Washington, USA
- Department of Pediatrics, Division of Pulmonary and Sleep Medicine, Seattle Children’s Hospital, University of Washington, Seattle, WA, USA
| | | | - Michael R. Howitt
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jakob von Moltke
- Department of Immunology, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
22
|
ILCs-Crucial Players in Enteric Infectious Diseases. Int J Mol Sci 2022; 23:ijms232214200. [PMID: 36430676 PMCID: PMC9695539 DOI: 10.3390/ijms232214200] [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: 10/31/2022] [Accepted: 11/12/2022] [Indexed: 11/18/2022] Open
Abstract
Research of the last decade has remarkably increased our understanding of innate lymphoid cells (ILCs). ILCs, in analogy to T helper (Th) cells and their cytokine and transcription factor profile, are categorized into three distinct populations: ILC1s express the transcription factor T-bet and secrete IFNγ, ILC2s depend on the expression of GATA-3 and release IL-5 and IL-13, and ILC3s express RORγt and secrete IL-17 and IL-22. Noteworthy, ILCs maintain a level of plasticity, depending on exposed cytokines and environmental stimuli. Furthermore, ILCs are tissue resident cells primarily localized at common entry points for pathogens such as the gut-associated lymphoid tissue (GALT). They have the unique capacity to initiate rapid responses against pathogens, provoked by changes of the cytokine profile of the respective tissue. Moreover, they regulate tissue inflammation and homeostasis. In case of intracellular pathogens entering the mucosal tissue, ILC1s respond by secreting cytokines (e.g., IFNγ) to limit the pathogen spread. Upon infection with helminths, intestinal epithelial cells produce alarmins (e.g., IL-25) and activate ILC2s to secrete IL-13, which induces differentiation of intestinal stem cells into tuft and goblet cells, important for parasite expulsion. Additionally, during bacterial infection ILC3-derived IL-22 is required for bacterial clearance by regulating antimicrobial gene expression in epithelial cells. Thus, ILCs can limit infectious diseases via secretion of inflammatory mediators and interaction with other cell types. In this review, we will address the role of ILCs during enteric infectious diseases.
Collapse
|
23
|
Immunological Interactions between Intestinal Helminth Infections and Tuberculosis. Diagnostics (Basel) 2022; 12:diagnostics12112676. [PMID: 36359526 PMCID: PMC9689268 DOI: 10.3390/diagnostics12112676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/13/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022] Open
Abstract
Helminth infections are among the neglected tropical diseases affecting billions of people globally, predominantly in developing countries. Helminths’ effects are augmented by coincident tuberculosis disease, which infects a third of the world’s population. The role of helminth infections on the pathogenesis and pathology of active tuberculosis (T.B.) remains controversial. Parasite-induced suppression of the efficacy of Bacille Calmette-Guerin (BCG) has been widely reported in helminth-endemic areas worldwide. T.B. immune response is predominantly proinflammatory T-helper type 1 (Th1)-dependent. On the other hand, helminth infections induce an opposing anti-inflammatory Th2 and Th3 immune-regulatory response. This review summarizes the literature focusing on host immune response profiles during single-helminth, T.B. and dual infections. It also aims to necessitate investigations into the complexity of immunity in helminth/T.B. coinfected patients since the research data are limited and contradictory. Helminths overlap geographically with T.B., particularly in Sub-Saharan Africa. Each disease elicits a response which may skew the immune responses. However, these effects are helminth species-dependent, where some parasites have no impact on the immune responses to concurrent T.B. The implications for the complex immunological interactions that occur during coinfection are highlighted to inform government treatment policies and encourage the development of high-efficacy T.B. vaccines in areas where helminths are prevalent.
Collapse
|
24
|
Wang Y, Song W, Yu S, Liu Y, Chen YG. Intestinal cellular heterogeneity and disease development revealed by single-cell technology. CELL REGENERATION 2022; 11:26. [PMID: 36045190 PMCID: PMC9433512 DOI: 10.1186/s13619-022-00127-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 07/15/2022] [Indexed: 11/10/2022]
Abstract
The intestinal epithelium is responsible for food digestion and nutrient absorption and plays a critical role in hormone secretion, microorganism defense, and immune response. These functions depend on the integral single-layered intestinal epithelium, which shows diversified cell constitution and rapid self-renewal and presents powerful regeneration plasticity after injury. Derailment of homeostasis of the intestine epithelium leads to the development of diseases, most commonly including enteritis and colorectal cancer. Therefore, it is important to understand the cellular characterization of the intestinal epithelium at the molecular level and the mechanisms underlying its homeostatic maintenance. Single-cell technologies allow us to gain molecular insights at the single-cell level. In this review, we summarize the single-cell RNA sequencing applications to understand intestinal cell characteristics, spatiotemporal evolution, and intestinal disease development.
Collapse
|
25
|
Jin X, Liu Y, Vallee I, Karadjian G, Liu M, Liu X. Lentinan -triggered butyrate-producing bacteria drive the expulsion of the intestinal helminth Trichinella spiralis in mice. Front Immunol 2022; 13:926765. [PMID: 35967395 PMCID: PMC9371446 DOI: 10.3389/fimmu.2022.926765] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 07/06/2022] [Indexed: 11/16/2022] Open
Abstract
Trichinellosis caused by Trichinella spiralis is a serious zoonosis with a worldwide distribution. Lentinan (LNT) is known to modulate the intestinal environment with noted health benefits, yet the effect of LNT against intestinal helminth is unknown. In our study, we first observed that LNT could trigger worm expulsion by promoting mucus layer functions through alteration of gut microbiota. LNT restored the abundance of Bacteroidetes and Proteobacteria altered by T. spiralis infection to the control group level. Interestingly, LNT triggered the production of butyrate. Then, we determined the deworming capacity of probiotics (butyrate-producing bacteria) in mice. Collectively, these findings indicated that LNT could modulate intestinal dysbiosis by T. spiralis, drive the expulsion of intestinal helminth and provided an easily implementable strategy to improve the host defence against T. spiralis infection.
Collapse
Affiliation(s)
- Xuemin Jin
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yi Liu
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Isabelle Vallee
- UMR BIPAR, Anses, Ecole Nationale Vétérinaire d’Alfort, INRA, University Paris-Est, Animal Health Laboratory, Maisons-Alfort, France
| | - Gregory Karadjian
- UMR BIPAR, Anses, Ecole Nationale Vétérinaire d’Alfort, INRA, University Paris-Est, Animal Health Laboratory, Maisons-Alfort, France
| | - Mingyuan Liu
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Xiaolei Liu
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
- *Correspondence: Xiaolei Liu,
| |
Collapse
|
26
|
Biller JD, Chagas EC. Mechanisms of resistance and tolerance against parasites in fish: the impairments caused by Neoechinorhynchus buttnerae in Colossoma macropomum. AN ACAD BRAS CIENC 2022; 94:e20210258. [PMID: 35830072 DOI: 10.1590/0001-3765202220210258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 05/10/2021] [Indexed: 08/30/2023] Open
Abstract
Tambaqui is the second native fish most produced species in Brazil. Currently, tambaqui fish farms deals with serious sanitary problems due to the prevalence of the parasite Neoechinorhynchus buttnerae. However, the prevalence of the acanthocephalan parasite infections depends on the resistance and tolerance interactions between the host organisms and parasites. The immune response against parasites is divided between innate and acquired immunity. The innate defense is a result of physical barriers, cellular and humoral compounds. Acquired defense occurs through the production of antibodies (humoral) and is mediated by cells, mainly by type 2 T helper lymphocytes. Most parasites secrete a variety of immunomodulatory compounds that allow coexistence with the host and chronicity of the parasite. The host-parasite relationship is complex and makes prevention and treatment difficult. However, some studies show that the use of immunostimulants may have "systemic" effects. These include improvement of the intestinal mucosa health and also in the production of cellular and humoral compounds in the whole body, thus assisting treatment and control. As such, it is important to understand the mechanisms of resistance and tolerance in the host organisms so that prevention and treatment measures can be effective.
Collapse
Affiliation(s)
- Jaqueline D Biller
- Universidade Estadual Paulista/UNESP, Faculdade de Ciências Agrárias e Tecnológicas, Departamento de Produção Animal, Campus de Dracena, Rodovia Comandante João Ribeiro de Barros, Km 651, Bairro das Antas, 17900-000 Dracena, SP, Brazil
| | - Edsandra C Chagas
- Empresa Brasileira de Pesquisa Agropecuária - Embrapa Amazônia Ocidental, Rodovia AM 010, Km 2, Caixa Postal 319, Zona Rural, 69010970 , AM, Brazil
| |
Collapse
|
27
|
Liu W, McNeilly TN, Mitchell M, Burgess STG, Nisbet AJ, Matthews JB, Babayan SA. Vaccine-induced time- and age-dependent mucosal immunity to gastrointestinal parasite infection. NPJ Vaccines 2022; 7:78. [PMID: 35798788 PMCID: PMC9262902 DOI: 10.1038/s41541-022-00501-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 06/13/2022] [Indexed: 11/10/2022] Open
Abstract
Individuals vary broadly in their response to vaccination and subsequent challenge infection, with poor vaccine responders causing persistence of both infection and transmission in populations. Yet despite having substantial economic and societal impact, the immune mechanisms that underlie such variability, especially in infected tissues, remain poorly understood. Here, to characterise how antihelminthic immunity at the mucosal site of infection developed in vaccinated lambs, we inserted gastric cannulae into the abomasa of three-month- and six-month-old lambs and longitudinally analysed their local immune response during subsequent challenge infection. The vaccine induced broad changes in pre-challenge abomasal immune profiles and reduced parasite burden and egg output post-challenge, regardless of age. However, age affected how vaccinated lambs responded to infection across multiple immune pathways: adaptive immune pathways were typically age-dependent. Identification of age-dependent and age-independent protective immune pathways may help refine the formulation of vaccines, and indicate specificities of pathogen-specific immunity more generally.
Collapse
Affiliation(s)
- Wei Liu
- Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow, G12 8QQ, Scotland, UK
| | - Tom N McNeilly
- The Moredun Research Institute, Pentlands Science Park, Scotland, EH26 0PZ, UK.
| | - Mairi Mitchell
- The Moredun Research Institute, Pentlands Science Park, Scotland, EH26 0PZ, UK
| | - Stewart T G Burgess
- The Moredun Research Institute, Pentlands Science Park, Scotland, EH26 0PZ, UK
| | - Alasdair J Nisbet
- The Moredun Research Institute, Pentlands Science Park, Scotland, EH26 0PZ, UK
| | - Jacqueline B Matthews
- The Moredun Research Institute, Pentlands Science Park, Scotland, EH26 0PZ, UK.,Roslin Technologies Limited, Roslin Innovation Centre, University of Edinburgh, Easter Bush, Scotland, EH25 9RG, UK
| | - Simon A Babayan
- Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow, G12 8QQ, Scotland, UK. .,The Moredun Research Institute, Pentlands Science Park, Scotland, EH26 0PZ, UK.
| |
Collapse
|
28
|
Varyani F, Löser S, Filbey KJ, Harcus Y, Drurey C, Poveda MC, Rasid O, White MPJ, Smyth DJ, Gerbe F, Jay P, Maizels RM. The IL-25-dependent tuft cell circuit driven by intestinal helminths requires macrophage migration inhibitory factor (MIF). Mucosal Immunol 2022; 15:1243-1256. [PMID: 35288645 PMCID: PMC9705247 DOI: 10.1038/s41385-022-00496-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 01/31/2022] [Accepted: 02/10/2022] [Indexed: 02/04/2023]
Abstract
Macrophage migration inhibitory factor (MIF) is a key innate immune mediator with chemokine- and cytokine-like properties in the inflammatory pathway. While its actions on macrophages are well-studied, its effects on other cell types are less understood. Here we report that MIF is required for expansion of intestinal tuft cells during infection with the helminth Nippostrongylus brasiliensis. MIF-deficient mice show defective innate responses following infection, lacking intestinal epithelial tuft cell hyperplasia or upregulation of goblet cell RELMβ, and fail to expand eosinophil, type 2 innate lymphoid cell (ILC2) and macrophage (M2) populations. Similar effects were observed in MIF-sufficient wild-type mice given the MIF inhibitor 4-IPP. MIF had no direct effect on epithelial cells in organoid cultures, and MIF-deficient intestinal stem cells could generate tuft cells in vitro in the presence of type 2 cytokines. In vivo the lack of MIF could be fully compensated by administration of IL-25, restoring tuft cell differentiation and goblet cell expression of RELM-β, demonstrating its requirement upstream of the ILC2-tuft cell circuit. Both ILC2s and macrophages expressed the MIF receptor CXCR4, indicating that MIF may act as an essential co-factor on both cell types to activate responses to IL-25 in helminth infection.
Collapse
Affiliation(s)
- Fumi Varyani
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, UK
| | - Stephan Löser
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Kara J Filbey
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, UK
- Lydia Becker Institute for Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Yvonne Harcus
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, UK
| | - Claire Drurey
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Marta Campillo Poveda
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Orhan Rasid
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Madeleine P J White
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Danielle J Smyth
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
- Division of Cell Signalling and Immunology, University of Dundee, Dundee, UK
| | - François Gerbe
- IGF, University of Montpellier, CNRS, Inserm, Montpellier, France
| | - Philippe Jay
- IGF, University of Montpellier, CNRS, Inserm, Montpellier, France
| | - Rick M Maizels
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK.
| |
Collapse
|
29
|
Gildner TE, Cepon-Robins TJ, Urlacher SS. Cumulative host energetic costs of soil-transmitted helminth infection. Trends Parasitol 2022; 38:629-641. [DOI: 10.1016/j.pt.2022.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/25/2022] [Accepted: 04/26/2022] [Indexed: 11/24/2022]
|
30
|
Oyesola OO, Souza COS, Loke P. The Influence of Genetic and Environmental Factors and Their Interactions on Immune Response to Helminth Infections. Front Immunol 2022; 13:869163. [PMID: 35572520 PMCID: PMC9103684 DOI: 10.3389/fimmu.2022.869163] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 04/04/2022] [Indexed: 12/20/2022] Open
Abstract
Helminth infection currently affect over 2 billion people worldwide, with those with the most pathologies and morbidities, living in regions with unequal and disproportionate access to effective healthcare solutions. Host genetics and environmental factors play critical roles in modulating and regulating immune responses following exposure to various pathogens and insults. However, the interplay of environment and genetic factors in influencing who gets infected and the establishment, persistence, and clearance of helminth parasites remains unclear. Inbred strains of mice have long been used to investigate the role of host genetic factors on pathogenesis and resistance to helminth infection in a laboratory setting. This review will discuss the use of ecological and environmental mouse models to study helminth infections and how this could be used in combination with host genetic variation to explore the relative contribution of these factors in influencing immune response to helminth infections. Improved understanding of interactions between genetics and the environment to helminth immune responses would be important for efforts to identify and develop new prophylactic and therapeutic options for the management of helminth infections and their pathogenesis.
Collapse
Affiliation(s)
- Oyebola O. Oyesola
- Laboratory of Parasitic Disease, National Institute of Allergy and Infectious Disease (NIAID), National Institute of Health, Bethesda, MD, United States
| | | | | |
Collapse
|
31
|
Administration of Hookworm Excretory/Secretory Proteins Improves Glucose Tolerance in a Mouse Model of Type 2 Diabetes. Biomolecules 2022; 12:biom12050637. [PMID: 35625566 PMCID: PMC9138508 DOI: 10.3390/biom12050637] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/19/2022] [Accepted: 04/22/2022] [Indexed: 01/27/2023] Open
Abstract
Diabetes is recognised as the world’s fastest growing chronic condition globally. Helminth infections have been shown to be associated with a lower prevalence of type 2 diabetes (T2D), in part due to their ability to induce a type 2 immune response. Therefore, to understand the molecular mechanisms that underlie the development of T2D-induced insulin resistance, we treated mice fed on normal or diabetes-promoting diets with excretory/secretory products (ES) from the gastrointestinal helminth Nippostrongylus brasiliensis. We demonstrated that treatment with crude ES products from adult worms (AES) or infective third-stage larvae (L3ES) from N. brasiliensis improved glucose tolerance and attenuated body weight gain in mice fed on a high glycaemic index diet. N. brasiliensis ES administration to mice was associated with a type 2 immune response measured by increased eosinophils and IL-5 in peripheral tissues but not IL-4, and with a decrease in the level of IL-6 in adipose tissue and corresponding increase in IL-6 levels in the liver. Moreover, treatment with AES or L3ES was associated with significant changes in the community composition of the gut microbiota at the phylum and order levels. These data highlight a role for N. brasiliensis ES in modulating the immune response associated with T2D, and suggest that N. brasiliensis ES contain molecules with therapeutic potential for treating metabolic syndrome and T2D.
Collapse
|
32
|
Okakpu OK, Dillman AR. Review of the Role of Parasitic Nematode Excretory/Secretory Proteins in Host Immunomodulation. J Parasitol 2022; 108:199-208. [DOI: 10.1645/21-33] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Affiliation(s)
- Ogadinma K. Okakpu
- University of California, Riverside 900 University Avenue, Riverside, California 92521
| | - Adler R. Dillman
- University of California, Riverside 900 University Avenue, Riverside, California 92521
| |
Collapse
|
33
|
Duque-Correa MA, Goulding D, Rodgers FH, Gillis JA, Cormie C, Rawlinson KA, Bancroft AJ, Bennett HM, Lotkowska ME, Reid AJ, Speak AO, Scott P, Redshaw N, Tolley C, McCarthy C, Brandt C, Sharpe C, Ridley C, Moya JG, Carneiro CM, Starborg T, Hayes KS, Holroyd N, Sanders M, Thornton DJ, Grencis RK, Berriman M. Defining the early stages of intestinal colonisation by whipworms. Nat Commun 2022; 13:1725. [PMID: 35365634 PMCID: PMC8976045 DOI: 10.1038/s41467-022-29334-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 03/08/2022] [Indexed: 01/08/2023] Open
Abstract
Whipworms are large metazoan parasites that inhabit multi-intracellular epithelial tunnels in the large intestine of their hosts, causing chronic disease in humans and other mammals. How first-stage larvae invade host epithelia and establish infection remains unclear. Here we investigate early infection events using both Trichuris muris infections of mice and murine caecaloids, the first in-vitro system for whipworm infection and organoid model for live helminths. We show that larvae degrade mucus layers to access epithelial cells. In early syncytial tunnels, larvae are completely intracellular, woven through multiple live dividing cells. Using single-cell RNA sequencing of infected mouse caecum, we reveal that progression of infection results in cell damage and an expansion of enterocytes expressing of Isg15, potentially instigating the host immune response to the whipworm and tissue repair. Our results unravel intestinal epithelium invasion by whipworms and reveal specific host-parasite interactions that allow the whipworm to establish its multi-intracellular niche. Whipworms are large parasites causing chronic disease in humans and other mammals. Here, the authors show how larvae create tunnels inside the gut lining and reveal the early host response to infection via Isg15 in mice and murine caecaloids.
Collapse
Affiliation(s)
- María A Duque-Correa
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK. .,Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, CB2 0AW, UK.
| | - David Goulding
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
| | - Faye H Rodgers
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK.,Mogrify Ltd, 25 Cambridge Science Park, Milton Road, Cambridge, CB4 0FW, UK
| | - J Andrew Gillis
- Department of Zoology, University of Cambridge, Cambridge, CB2 3EJ, UK
| | - Claire Cormie
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK.,Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Kate A Rawlinson
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
| | - Allison J Bancroft
- Lydia Becker Institute of Immunology and Inflammation, Wellcome Trust Centre for Cell Matrix Research and Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK
| | - Hayley M Bennett
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK.,Genentech, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Magda E Lotkowska
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
| | - Adam J Reid
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK.,Wellcome/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, CB2 1QN, UK
| | - Anneliese O Speak
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
| | - Paul Scott
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
| | - Nicholas Redshaw
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
| | - Charlotte Tolley
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
| | - Catherine McCarthy
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
| | - Cordelia Brandt
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
| | - Catherine Sharpe
- Lydia Becker Institute of Immunology and Inflammation, Wellcome Trust Centre for Cell Matrix Research and Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK.,InstilBio, UMIC Bio-Incubator, Manchester, M13 9XX, UK
| | - Caroline Ridley
- Lydia Becker Institute of Immunology and Inflammation, Wellcome Trust Centre for Cell Matrix Research and Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK.,Prime Global Medical Communications, Knutsford, WA16 8GP, UK
| | - Judit Gali Moya
- Faculty of Biology, University of Barcelona, Barcelona, 08028, Spain
| | - Claudia M Carneiro
- Immunopathology Laboratory, NUPEB, Federal University of Ouro Preto, Campus Universitario Morro do Cruzeiro, Ouro Preto, MG, 35400-000, Brazil
| | - Tobias Starborg
- Lydia Becker Institute of Immunology and Inflammation, Wellcome Trust Centre for Cell Matrix Research and Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK.,Rosalind Franklin Institute, Harwell Campus, Didcot, OX11 0FA, UK
| | - Kelly S Hayes
- Lydia Becker Institute of Immunology and Inflammation, Wellcome Trust Centre for Cell Matrix Research and Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK
| | - Nancy Holroyd
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
| | - Mandy Sanders
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
| | - David J Thornton
- Lydia Becker Institute of Immunology and Inflammation, Wellcome Trust Centre for Cell Matrix Research and Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK
| | - Richard K Grencis
- Lydia Becker Institute of Immunology and Inflammation, Wellcome Trust Centre for Cell Matrix Research and Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK
| | - Matthew Berriman
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, UK.
| |
Collapse
|
34
|
Hu RS, Zhang FK, Ma QN, Ehsan M, Zhao Q, Zhu XQ. Transcriptomic landscape of hepatic lymph nodes, peripheral blood lymphocytes and spleen of swamp buffaloes infected with the tropical liver fluke Fasciola gigantica. PLoS Negl Trop Dis 2022; 16:e0010286. [PMID: 35320269 PMCID: PMC8942208 DOI: 10.1371/journal.pntd.0010286] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 02/28/2022] [Indexed: 11/22/2022] Open
Abstract
The tropical liver fluke Fasciola gigantica is a parasitic helminth that has been frequently reported to infect mammals, typically involving water buffaloes. In this study, we characterized the tissue transcriptional landscape of buffaloes following infection by F. gigantica. RNAs were isolated from hepatic lymph nodes (hLNs), peripheral blood lymphocytes (pBLs), and spleen at 3-, 42- and 70-days post-infection (dpi), and all samples were subjected to RNA sequencing analyses. At 3 dpi, 2603, 460, and 162 differentially expressed transcripts (DETs) were detected in hLNs, pBLs, and spleen, respectively. At 42 dpi, 322, 937, and 196 DETs were detected in hLNs, pBLs, and spleen, respectively. At 70 dpi, 376, 334, and 165 DETs were detected in hLNs, pBLs, and spleen, respectively. Functional enrichment analysis identified upregulated immune-related pathways in the infected tissues involved in innate and adaptive immune responses, especially in hLNs at 42 and 70 dpi, and pBLs at 3 and 42 dpi. The upregulated transcripts in spleen were not enriched in any immune-related pathway. Co-expression network analysis further identified transcriptional changes associated with immune response to F. gigantica infection. Receiver operating characteristic (ROC) curve analysis showed that 107 genes in hLNs, 32 genes in pBLs, and 36 genes in spleen correlated with F. gigantica load. These findings provide new insight into molecular mechanisms and signaling pathways associated with F. gigantica infection in buffaloes. Fasciola gigantica is a socioeconomically important tropical liver fluke of mammals, causing fascioliasis–a neglected tropical disease. In the present study, RNA sequencing and bioinformatic approach were employed to explore the global transcriptional changes of hepatic lymph nodes (hLNs), peripheral blood lymphocytes (pBLs), and spleen of water buffaloes during F. gigantica infection at 3-, 42-, and 70-days post-infection (dpi). The results revealed significant transcriptional upregulation of genes associated with innate and adaptive immune responses in infected hLNs (42 and 70 dpi) and pBLs (3 and 42 dpi). However, downregulation of transcripts involved in immune response was detected in pBLs at 70 dpi. The downregulated transcripts were enriched in metabolic pathways, such as drug metabolism-cytochrome P450 in infected hLNs at 3 dpi. These findings provide new insight into the pathogenesis of F. gigantica in its natural mammalian host.
Collapse
Affiliation(s)
- Rui-Si Hu
- College of Life Science, Changchun Sci-Tech University, Shuangyang, Jilin Province, People’s Republic of China
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, People’s Republic of China
| | - Fu-Kai Zhang
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, People’s Republic of China
| | - Qiao-Ni Ma
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, People’s Republic of China
| | - Muhammad Ehsan
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, People’s Republic of China
- Department of Parasitology, Faculty of Veterinary and Animal Sciences, The Islamia University of Bahawalpur, Bahawalpur, Punjab Province, Pakistan
| | - Quan Zhao
- College of Life Science, Changchun Sci-Tech University, Shuangyang, Jilin Province, People’s Republic of China
- * E-mail: (QZ); (X-QZ)
| | - Xing-Quan Zhu
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi Province, People’s Republic of China
- Key Laboratory of Veterinary Public Health of Yunnan Province, College of Veterinary Medicine, Yunnan Agricultural University, Kunming, Yunnan Province, People’s Republic of China
- * E-mail: (QZ); (X-QZ)
| |
Collapse
|
35
|
Corripio-Miyar Y, Hayward A, Lemon H, Sweeny AR, Bal X, Kenyon F, Pilkington JG, Pemberton JM, Nussey DH, McNeilly TN. Functionally distinct T-helper cell phenotypes predict resistance to different types of parasites in a wild mammal. Sci Rep 2022; 12:3197. [PMID: 35210503 PMCID: PMC8873199 DOI: 10.1038/s41598-022-07149-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 02/04/2022] [Indexed: 12/31/2022] Open
Abstract
The adaptive immune system is critical to an effective response to infection in vertebrates, with T-helper (Th) cells pivotal in orchestrating these responses. In natural populations where co-infections are the norm, different Th responses are likely to play an important role in maintaining host health and fitness, a relationship which remains poorly understood in wild animals. In this study, we characterised variation in functionally distinct Th responses in a wild population of Soay sheep by enumerating cells expressing Th-subset specific transcription factors and quantifying Th-associated cytokines. We tested the prediction that raised Th1 and Th2 responses should predict reduced apicomplexan and helminth parasite burdens, respectively. All measures of Th-associated cytokine production increased with age, while Th17- and regulatory Th-associated cytokine production increased more rapidly with age in males than females. Independent of age, sex, and each other, IL-4 and Gata3 negatively predicted gastro-intestinal nematode faecal egg count, while IFN-γ negatively predicted coccidian faecal oocyst count. Our results provide important support from outside the laboratory that Th1 and Th2 responses predict resistance to different kinds of parasites, and illustrate how harnessing specific reagents and tools from laboratory immunology will illuminate our understanding of host-parasite interactions in the wild.
Collapse
Affiliation(s)
- Yolanda Corripio-Miyar
- Moredun Research Institute, Pentlands Science Park, Bush Loan, Midlothian, EH26 0PZ, UK.
| | - Adam Hayward
- Moredun Research Institute, Pentlands Science Park, Bush Loan, Midlothian, EH26 0PZ, UK
| | - Hannah Lemon
- Institute of Evolutionary Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, EH9 3FL, UK
| | - Amy R Sweeny
- Institute of Evolutionary Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, EH9 3FL, UK
| | - Xavier Bal
- Institute of Evolutionary Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, EH9 3FL, UK
| | - Fiona Kenyon
- Moredun Research Institute, Pentlands Science Park, Bush Loan, Midlothian, EH26 0PZ, UK
| | - Jill G Pilkington
- Institute of Evolutionary Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, EH9 3FL, UK
| | - Josephine M Pemberton
- Institute of Evolutionary Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, EH9 3FL, UK
| | - Daniel H Nussey
- Institute of Evolutionary Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, EH9 3FL, UK
| | - Tom N McNeilly
- Moredun Research Institute, Pentlands Science Park, Bush Loan, Midlothian, EH26 0PZ, UK
| |
Collapse
|
36
|
Liu J, Zhou J, Zhao S, Xu X, Li CJ, Li L, Shen T, Hunt PW, Zhang R. Differential responses of abomasal transcriptome to Haemonchus contortus infection between Haemonchus-selected and Trichostrongylus-selected merino sheep. Parasitol Int 2022; 87:102539. [PMID: 35007764 DOI: 10.1016/j.parint.2022.102539] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 12/16/2021] [Accepted: 01/04/2022] [Indexed: 10/19/2022]
Abstract
Haemonchus contortus is the most prevalent and pathogenic gastrointestinal nematode infecting sheep and goats. The two CSIRO sheep resource flocks, the Haemonchus-selected flock (HSF) and Trichostrongylus-selected flock (TSF) were developed for research on host resistance or susceptibility to gastrointestinal nematode infection. A recent study focused on the gene expression differences between resistant and susceptible sheep within each flock, with lymphatic and gastrointestinal tissues. To identify features in the host transcriptome and understand the molecular differences underlying host resistance to H. contortus between flocks with different selective breeding and genetic backgrounds, we compared the abomasal transcriptomic responses of the resistant or susceptible animals between HSF and TSF flocks. A total of 11 and 903 differentially expressed genes were identified in the innate infection treatment in HSF and TSF flocks between resistant and susceptible sheep respectively, while 52 and 485 genes were identified to be differentially expressed in the acquired infection treatment, respectively. Among them, 294 genes had significantly different gene expression levels between HSF and TSF flock animals within the susceptible sheep by both the innate and acquired infections. Moreover, similar expression patterns of the 294 genes were observed, with 273 genes more highly expressed in HSF and 21 more highly expressed in the TSF within the abomasal transcriptome of the susceptible animals. Gene ontology enrichment of the differentially expressed genes identified in this study predicted the likely differing function between the two flock's susceptible lines in response to H. contortus infection. Nineteen pathways were significantly enriched in both the innate and adaptive immune responses in susceptible animals, which indicated that these pathways likely contribute to the host resistance development to H. contortus infection in susceptible sheep. Biological networks built for the set of genes differentially abundant in susceptible animals identified hub genes of PRKG1, PRKACB, PRKACA, and ITGB1 for the innate immune response, and CALM2, MYL1, COL1A1, ITGB1 and ITGB3 for the adaptive immune response, respectively. Our results offered a quantitative snapshot of host transcriptomic changes induced by H. contortus infection between flocks with different selective breeding and genetic backgrounds and provided novel insights into molecular mechanisms of host resistance.
Collapse
Affiliation(s)
- Jing Liu
- College of Life Science, Hubei Key Laboratory of Edible Wild Plants Conservation & Utilization, Huangshi Biomedicine Industry and Technology Research Institute Company Limited, Hubei Normal University, Huangshi, Hubei 435002, China
| | - Jiachang Zhou
- College of Life Science, Hubei Key Laboratory of Edible Wild Plants Conservation & Utilization, Huangshi Biomedicine Industry and Technology Research Institute Company Limited, Hubei Normal University, Huangshi, Hubei 435002, China
| | - Si Zhao
- College of Life Science, Hubei Key Laboratory of Edible Wild Plants Conservation & Utilization, Huangshi Biomedicine Industry and Technology Research Institute Company Limited, Hubei Normal University, Huangshi, Hubei 435002, China; International Medical School, Hebei Foreign Studies University, Shijiazhuang, Hebei 050096, China
| | - Xiangdong Xu
- College of Life Science, Hubei Key Laboratory of Edible Wild Plants Conservation & Utilization, Huangshi Biomedicine Industry and Technology Research Institute Company Limited, Hubei Normal University, Huangshi, Hubei 435002, China
| | - Cong-Jun Li
- United States Department of Agriculture, Agriculture Research Service (USDA-ARS), Animal Genomics and Improvement Laboratory, Beltsville, MD 20705, USA.
| | - Li Li
- College of Life Science, Hubei Key Laboratory of Edible Wild Plants Conservation & Utilization, Huangshi Biomedicine Industry and Technology Research Institute Company Limited, Hubei Normal University, Huangshi, Hubei 435002, China
| | - Tingbo Shen
- College of Life Science, Hubei Key Laboratory of Edible Wild Plants Conservation & Utilization, Huangshi Biomedicine Industry and Technology Research Institute Company Limited, Hubei Normal University, Huangshi, Hubei 435002, China
| | - Peter W Hunt
- CSIRO Agriculture and Food, Armidale, NSW, Australia.
| | - Runfeng Zhang
- College of Life Science, Hubei Key Laboratory of Edible Wild Plants Conservation & Utilization, Huangshi Biomedicine Industry and Technology Research Institute Company Limited, Hubei Normal University, Huangshi, Hubei 435002, China.
| |
Collapse
|
37
|
β-Glucan-triggered Akkermansia muciniphila expansion facilitates the expulsion of intestinal helminth via TLR2 in mice. Carbohydr Polym 2022; 275:118719. [PMID: 34742442 DOI: 10.1016/j.carbpol.2021.118719] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 09/24/2021] [Accepted: 09/28/2021] [Indexed: 12/21/2022]
Abstract
Trichinellosis caused by Trichinella spiralis is a serious zoonosis with a worldwide. β-Glucans (BG) are readily used across the world with noted health benefits, yet the effect and mechanism of BG on host defense against helminth infection remain poorly understood. We observed that BG could trigger worm expulsion via mucus layer independently of type 2 immunity, but was dependent on the gut microbiota in mice. BG restored the abundance of Bacteroidetes and Proteobacteria changed by T. spiralis infection to the control group level and markedly increased the relative abundance of Verrucomicrobia. Akkermansia (belonging to Verrucomicrobia) were significantly expanded in the BG + T. spiralis group. Notably, daily oral supplementation of pasteurized A. muciniphila has a stronger deworming effect than live bacteria and interacted with TLR2. These findings of this study is an easily implementable strategy to facilitate expulsion of gastrointestinal helminth.
Collapse
|
38
|
Lymphatic filariasis and visceral leishmaniasis coinfection: A review on their epidemiology, therapeutic, and immune responses. Acta Trop 2021; 224:106117. [PMID: 34464587 DOI: 10.1016/j.actatropica.2021.106117] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 08/13/2021] [Accepted: 08/18/2021] [Indexed: 12/26/2022]
Abstract
Coinfection is less commonly observed in individuals around the world, yet it is more common than the single infection. Around 800 million people worldwide are infected with helminths as a result of various diseases. Lymphatic filariasis (LF) and visceral leishmaniasis (VL) are chronic, deadly, crippling, and debilitating neglected tropical diseases (NTDs) that are endemic in tropical and subtropical regions of the world. Due to poor hygienic conditions, poverty, and genetic predisposition, those living in endemic areas are more likely to develop both leishmaniasis and filariasis. One of the key challenges in the management of LF/VL coinfection is the development of an effective therapeutic strategy that not only treats the first episode of VL but also prevents LF. However, there is a scarcity of knowledge and data on the relationship between LF and VL coinfection. While reviewing it was apparent that only a few studies relevant to LF/VL coinfections have been reported from southeastern Spain, Sudan, and the Indian subcontinents, highlighting the need for greater research in the most affected areas. We also looked at LF and VL as a single disease and also as a coinfection. Some features of the immune response evolved in mammalian hosts against LF and VL alone or against coinfection are also discussed, including epidemiology, therapeutic regimens, and vaccines. In addition to being potentially useful in clinical research, our findings imply the need for improved diagnostic methodology and therapeutics, which could accelerate the deployment of more specific and effective diagnosis for treatments to lessen the impact of VL/LF coinfections in the population.
Collapse
|
39
|
Differential effects of asymptomatic Ascaris lumbricoides, Schistosoma mansoni or hook worm infection on the frequency and TGF-beta-producing capacity of regulatory T cells during active tuberculosis. Tuberculosis (Edinb) 2021; 131:102126. [PMID: 34601265 DOI: 10.1016/j.tube.2021.102126] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 09/01/2021] [Accepted: 09/09/2021] [Indexed: 02/08/2023]
Abstract
Helminth induced expansion of regulatory T cells (Tregs) may take part in suppressing protective host responses during tuberculosis (TB), although Tregs functionality and link to TB disease severity remains unexplored. We investigated the species-specific effect of helminths on frequency and TGF-β producing capacity of Tregs, and possible connection to TB disease severity. 89 pulmonary TB patients (PTB) and 69 community controls (CCs) from Gondar, Ethiopia, were included. Clinical disease severity was graded by TB score, and flow cytometry used to characterize Treg frequency and functionality measured as their TGF-β-producing capacity. In helminth positive PTB patients (Helminth+PTB+) compared to helminth negative PTB or CCs, TGF-β+ Tregs were significantly increased mainly in hookworm coinfection whereas S. mansoni increased TGF-β+ Tregs in CCs. Treatment of TB and helminths decreased TGF-β+ Tregs in Helminth+PTB+ at 2 months follow-up. There were no overall differences in the frequency of Tregs in CCs or PTB unless stratification on TB disease severity was performed. At inclusion Helminth+PTB+ had increased frequency of Tregs already at low disease severity, and TGF-β+ Tregs correlated to intermediate-to-high disease severity. In conclusion, helminth specific increase of TGF-β+ Tregs in PTB patients was correlated to TB disease severity and was restored following anti-helminth treatment.
Collapse
|
40
|
Rapaka RR, Cross AS, McArthur MA. Using Adjuvants to Drive T Cell Responses for Next-Generation Infectious Disease Vaccines. Vaccines (Basel) 2021; 9:vaccines9080820. [PMID: 34451945 PMCID: PMC8402546 DOI: 10.3390/vaccines9080820] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/16/2021] [Accepted: 07/20/2021] [Indexed: 12/12/2022] Open
Abstract
Using adjuvants to drive features of T cell responses to vaccine antigens is an important technological challenge in the design of new and improved vaccines against infections. Properties such as T helper cell function, T cell memory, and CD8+ T cell cytotoxicity may play critical roles in optimal and long-lived immunity through vaccination. Directly manipulating specific immune activation or antigen delivery pathways with adjuvants may selectively augment desired T cell responses in vaccination and may improve the effectiveness and durability of vaccine responses in humans. In this review we outline recently studied adjuvants in their potential for antigen presenting cell and T cell programming during vaccination, with an emphasis on what has been observed in studies in humans as available.
Collapse
|
41
|
Kushwaha V, Kaur S. Cross-protective efficacy of immuno-stimulatory recombinant Brugia malayi protein HSP60 against the Leishmania donovani in BALB/c mice. Biologicals 2021; 72:18-26. [PMID: 34229924 DOI: 10.1016/j.biologicals.2021.06.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 06/08/2021] [Accepted: 06/10/2021] [Indexed: 11/18/2022] Open
Abstract
Coinfection of Leishmania with bacteria, viruses, protozoans, and nematodes alter the immune system of the host, thereby influencing the disease outcomes. Here, we have determined the immunogenic property and protective efficacy of the cross-reactive molecule HSP60 of filarial parasite B. malayi against the L. donovani in BALB/c mice. Parasitological parameters results showed a significant decrease in the parasite burden (~59%; P < 0.001) and also a substantial increase in the delayed-type hypersensitivity (DTH) response (P < 0.001) in mice immunized with 10 μg of rBmHSP60. Protection against L. donovani in mice immunized with rBmHSP60 resulted from activation of the T cells, which is characterized by higher levels of nitric oxide (NO) production, enhanced cell proliferation, higher levels (expression and release) of IFN- γ, TNF- α, and IL-12, also, higher production of IgG and IgG2a antibodies. This strong Th1 immune response creates an inflammatory domain for L. donovani and protects the host from VL.
Collapse
Affiliation(s)
- Vikas Kushwaha
- Leishmania Research Laboratory, Department of Zoology, Panjab University, Sector-14, Chandigarh, 160014, India.
| | - Sukhbir Kaur
- Leishmania Research Laboratory, Department of Zoology, Panjab University, Sector-14, Chandigarh, 160014, India
| |
Collapse
|
42
|
Bailey C, Strepparava N, Ros A, Wahli T, Schmidt-Posthaus H, Segner H, Tafalla C. It's a hard knock life for some: Heterogeneity in infection life history of salmonids influences parasite disease outcomes. J Anim Ecol 2021; 90:2573-2593. [PMID: 34165799 PMCID: PMC8597015 DOI: 10.1111/1365-2656.13562] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 06/19/2021] [Indexed: 11/27/2022]
Abstract
Heterogeneity in immunity occurs across numerous disease systems with individuals from the same population having diverse disease outcomes. Proliferative kidney disease (PKD) caused by Tetracapsuloides bryosalmonae, is a persistent parasitic disease negatively impacting both wild and farmed salmonids. Little is known of how PKD is spread or maintained within wild susceptible populations. We investigated an aspect of fish disease that has been largely overlooked, that is, the role of the host phenotypic heterogeneity in disease outcome. We examined how host susceptibility to T. bryosalmonae infection, and the disease PKD, varied across different infection life-history stages and how it differs between naïve, re-infected and persistently infected hosts. We investigated the response to parasite exposure in host phenotypes with (a) different ages and (b) heterogeneous infection life histories. Among (a) the age phenotypes were young-of-the-year (YOY) fish and juvenile 1+ fish (fish older than one) and, for (b) juvenile 1+ infection survivors were either re-exposed or not re- exposed to the parasite and response phenotypes were assigned post-hoc dependant on infection status. In fish not re-exposed this included fish that cleared infection (CI) or had a persistent infection (PI). In fish re-exposed these included fish that were re-infected (RI), or re-exposed and uninfected (RCI). We assessed both parasite-centric (infection prevalence, parasite burden, malacospore transmission) and host-centric parameters (growth rates, disease severity, infection tolerance and the immune response). In (a), YOY fish, parasite success and disease severity were greater and differences in the immune response occurred, demonstrating an ontogenetic decline of susceptibility in older fish. In (b), in PI and RI fish, parasite success and disease severity were comparable. However, expression of several adaptive immunity markers was greater in RI fish, indicating concomitant immunity, as re-exposure did not intensify infection. We demonstrate the relevance of heterogeneity in infection life history on disease outcome and describe several distinctive features of immune ontogeny and protective immunity in this model not previously reported. The relevance of such themes on a population level requires greater research in many aquatic disease systems to generate clearer framework for understanding the spread and maintenance of aquatic pathogens.
Collapse
Affiliation(s)
- Christyn Bailey
- Fish Immunology and Pathology Group, Animal Health Research Centre (CISA-INIA), Madrid, Spain
| | - Nicole Strepparava
- Centre for Fish and Wildlife Health, University of Bern, Bern, Switzerland
| | - Albert Ros
- LAZBW, Fischereiforschungsstelle, Langenargen, Germany
| | - Thomas Wahli
- Centre for Fish and Wildlife Health, University of Bern, Bern, Switzerland
| | | | - Helmut Segner
- Centre for Fish and Wildlife Health, University of Bern, Bern, Switzerland
| | - Carolina Tafalla
- Fish Immunology and Pathology Group, Animal Health Research Centre (CISA-INIA), Madrid, Spain
| |
Collapse
|
43
|
Survival of metazoan parasites in fish: Putting into context the protective immune responses of teleost fish. ADVANCES IN PARASITOLOGY 2021; 112:77-132. [PMID: 34024360 DOI: 10.1016/bs.apar.2021.03.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Defence mechanisms of fish can be divided into specific and non-specific that act in concert and are often interdependent. Most fish in both wild and cultured populations are vulnerable to metazoan parasites. Endoparasitic helminths include several species of digeneans, cestodes, nematodes, and acanthocephalans. Although they may occur in large numbers, helminth infections rarely result in fish mortality. Conversely, some ectoparasites cause mass mortality in farmed fish. Given the importance of fish innate immunity, this review addresses non-specific defence mechanisms of fish against metazoan parasites, with emphasis on granulocyte responses involving mast cells, neutrophils, macrophages, rodlet cells, and mucous cells. Metazoan parasites are important disease agents that affect wild and farmed fish and can induce high economic loss and, as pathogen organisms, deserve considerable attention. The paper will provide our light and transmission electron microscopy data on metazoan parasites-fish innate immune and neuroendocrine systems. Insights about the structure and functions of the cell types listed above and a brief account of the effects and harms of each metazoan taxon to specific fish apparati/organs will be presented.
Collapse
|
44
|
Long SR, Liu RD, Kumar DV, Wang ZQ, Su CW. Immune Protection of a Helminth Protein in the DSS-Induced Colitis Model in Mice. Front Immunol 2021; 12:664998. [PMID: 33995396 PMCID: PMC8117093 DOI: 10.3389/fimmu.2021.664998] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 04/08/2021] [Indexed: 12/23/2022] Open
Abstract
Inflammatory bowel disease (IBD) increases the risk of colorectal cancer, and it has the potential to diminish the quality of life. Recent clinical and experimental evidence demonstrate protective aspects of parasitic helminth infection against IBD. Reports have highlighted the potential use of helminths and their byproducts as potential treatment for IBD. In the current study, we studied the effect of a newborn larvae-specific serine protease from Trichinella spiralis (TsSp) on the host immune and inflammatory responses. A 49-kDa recombinant TsSp (rTsSp) was expressed in Escherichia coli BL21 (DE3) and purified. The cytotoxicity of rTsSp was analyzed. The immune protective effect of rTsSp was studied by using dextran sodium sulfate (DSS)-induced mouse colitis model. The result illustrated that rTsSp has no toxic effects on cells. We further demonstrated that administration of the rTsSp without the additional adjuvant before the induction of DSS-induced colitis reduced the severity of intestinal inflammation and the disease index; it suppressed macrophage infiltration, reduced TNF-α secretion, and induced IL-10 expression. Our findings suggest therapeutic potential of rTsSp on colitis by altering the effect of macrophages. Data also suggest immunotherapy with rTsSp holds promise for use as an additional strategy to positively modulate inflammatory processes involved in IBD.
Collapse
Affiliation(s)
- Shao Rong Long
- Department of Parasitology, Medical College of Zhengzhou University, Zhengzhou, China.,Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| | - Ruo Dan Liu
- Department of Parasitology, Medical College of Zhengzhou University, Zhengzhou, China
| | - Deepak Vijaya Kumar
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| | - Zhong Quan Wang
- Department of Parasitology, Medical College of Zhengzhou University, Zhengzhou, China
| | - Chien-Wen Su
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| |
Collapse
|
45
|
Kumar V. Innate Lymphoid Cells and Adaptive Immune Cells Cross-Talk: A Secret Talk Revealed in Immune Homeostasis and Different Inflammatory Conditions. Int Rev Immunol 2021; 40:217-251. [PMID: 33733998 DOI: 10.1080/08830185.2021.1895145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The inflammatory immune response has evolved to protect the host from different pathogens, allergens, and endogenous death or damage-associated molecular patterns. Both innate and adaptive immune components are crucial in inducing an inflammatory immune response depending on the stimulus type and its duration of exposure or the activation of the primary innate immune response. As the source of inflammation is removed, the aggravated immune response comes to its homeostatic level. However, the failure of the inflammatory immune response to subside to its normal level generates chronic inflammatory conditions, including autoimmune diseases and cancer. Innate lymphoid cells (ILCs) are newly discovered innate immune cells, which are present in abundance at mucosal surfaces, including lungs, gastrointestinal tract, and reproductive tract. Also, they are present in peripheral blood circulation, skin, and lymph nodes. They play a crucial role in generating the pro-inflammatory immune response during diverse conditions. On the other hand, adaptive immune cells, including different types of T and B cells are major players in the pathogenesis of autoimmune diseases (type 1 diabetes mellitus, rheumatoid arthritis, psoriasis, and systemic lupus erythematosus, etc.) and cancers. Thus the article is designed to discuss the immunological role of different ILCs and their interaction with adaptive immune cells in maintaining the immune homeostasis, and during inflammatory autoimmune diseases along with other inflammatory conditions (excluding pathogen-induced inflammation), including cancer, graft-versus-host diseases, and human pregnancy.
Collapse
Affiliation(s)
- Vijay Kumar
- Children's Health Queensland Clinical Unit, School of Clinical Medicine, Faculty of Medicine, Mater Research, University of Queensland, St Lucia, Brisbane, Queensland, Australia.,School of Biomedical Sciences, Faculty of Medicine, University of Queensland, St Lucia, Brisbane, Queensland, Australia
| |
Collapse
|
46
|
Yang X, Ding W, Qian X, Jiang P, Chen Q, Zhang X, Lu Y, Wu J, Sun F, Pan Z, Li X, Pan W. Schistosoma japonicum Infection Leads to the Reprogramming of Glucose and Lipid Metabolism in the Colon of Mice. Front Vet Sci 2021; 8:645807. [PMID: 33791356 PMCID: PMC8006365 DOI: 10.3389/fvets.2021.645807] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 02/17/2021] [Indexed: 01/21/2023] Open
Abstract
The deposition of Schistosoma japonicum (S. japonicum) eggs commonly induces inflammation, fibrosis, hyperplasia, ulceration, and polyposis in the colon, which poses a serious threat to human health. However, the underlying mechanism is largely neglected. Recently, the disorder of glucose and lipid metabolism was reported to participate in the liver fibrosis induced by the parasite, which provides a novel clue for studying the underlying mechanism of the intestinal pathology of the disease. This study focused on the metabolic reprogramming profiles of glucose and lipid in the colon of mice infected by S. japonicum. We found that S. japonicum infection shortened the colonic length, impaired intestinal integrity, induced egg-granuloma formation, and increased colonic inflammation. The expression of key enzymes involved in the pathways regulating glucose and lipid metabolism was upregulated in the colon of infected mice. Conversely, phosphatase and tensin homolog deleted on chromosome ten (PTEN) and its downstream signaling targets were significantly inhibited after infection. In line with these results, in vitro stimulation with soluble egg antigens (SEA) downregulated the expression of PTEN in CT-26 cells and induced metabolic alterations similar to that observed under in vivo results. Moreover, PTEN over-expression prevented the reprogramming of glucose and lipid metabolism induced by SEA in CT-26 cells. Overall, the present study showed that S. japonicum infection induces the reprogramming of glucose and lipid metabolism in the colon of mice, and PTEN may play a vital role in mediating this metabolic reprogramming. These findings provide a novel insight into the pathogenicity of S. japonicum in hosts.
Collapse
Affiliation(s)
- Xiaoying Yang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China.,National Experimental Teaching Demonstration Center of Basic Medicine, Xuzhou Medical University, Xuzhou, China
| | - Weimin Ding
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China.,National Experimental Teaching Demonstration Center of Basic Medicine, Xuzhou Medical University, Xuzhou, China.,School of Life Sciences, Xuzhou Medical University, Xuzhou, China
| | - Xinyu Qian
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China.,National Experimental Teaching Demonstration Center of Basic Medicine, Xuzhou Medical University, Xuzhou, China.,The First Clinical Medical College, Xuzhou Medical University, Xuzhou, China
| | - Pengfei Jiang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China.,National Experimental Teaching Demonstration Center of Basic Medicine, Xuzhou Medical University, Xuzhou, China
| | - Qingqing Chen
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China.,National Experimental Teaching Demonstration Center of Basic Medicine, Xuzhou Medical University, Xuzhou, China.,The First Clinical Medical College, Xuzhou Medical University, Xuzhou, China
| | - Xin Zhang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China.,National Experimental Teaching Demonstration Center of Basic Medicine, Xuzhou Medical University, Xuzhou, China.,The First Clinical Medical College, Xuzhou Medical University, Xuzhou, China
| | - Yang Lu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China.,National Experimental Teaching Demonstration Center of Basic Medicine, Xuzhou Medical University, Xuzhou, China.,The First Clinical Medical College, Xuzhou Medical University, Xuzhou, China
| | - Jiacheng Wu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China.,National Experimental Teaching Demonstration Center of Basic Medicine, Xuzhou Medical University, Xuzhou, China.,The Second Clinical Medical College, Xuzhou Medical University, Xuzhou, China
| | - Fenfen Sun
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China.,National Experimental Teaching Demonstration Center of Basic Medicine, Xuzhou Medical University, Xuzhou, China
| | - Zhihua Pan
- National Experimental Teaching Demonstration Center of Basic Medicine, Xuzhou Medical University, Xuzhou, China
| | - Xiangyang Li
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China.,National Experimental Teaching Demonstration Center of Basic Medicine, Xuzhou Medical University, Xuzhou, China
| | - Wei Pan
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China.,National Experimental Teaching Demonstration Center of Basic Medicine, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
47
|
Sweeny AR, Clerc M, Pontifes PA, Venkatesan S, Babayan SA, Pedersen AB. Supplemented nutrition decreases helminth burden and increases drug efficacy in a natural host-helminth system. Proc Biol Sci 2021; 288:20202722. [PMID: 33468010 PMCID: PMC7893286 DOI: 10.1098/rspb.2020.2722] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/15/2020] [Indexed: 12/17/2022] Open
Abstract
Gastrointestinal (GI) helminths are common parasites of humans, wildlife, and livestock, causing chronic infections. In humans and wildlife, poor nutrition or limited resources can compromise an individual's immune response, predisposing them to higher helminth burdens. This relationship has been tested in laboratory models by investigating infection outcomes following reductions of specific nutrients. However, much less is known about how diet supplementation can impact susceptibility to infection, acquisition of immunity, and drug efficacy in natural host-helminth systems. We experimentally supplemented the diet of wood mice (Apodemus sylvaticus) with high-quality nutrition and measured resistance to the common GI nematode Heligmosomoides polygyrus. To test whether diet can enhance immunity to reinfection, we also administered anthelmintic treatment in both natural and captive populations. Supplemented wood mice were more resistant to H. polygyrus infection, cleared worms more efficiently after treatment, avoided a post-treatment infection rebound, produced stronger general and parasite-specific antibody responses, and maintained better body condition. In addition, when applied in conjunction with anthelmintic treatment, supplemented nutrition significantly reduced H. polygyrus transmission potential. These results show the rapid and extensive benefits of a well-balanced diet and have important implications for both disease control and wildlife health under changing environmental conditions.
Collapse
Affiliation(s)
- Amy R. Sweeny
- Institute of Evolutionary Biology and Centre for Immunity, Infection and Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FL, UK
| | - Melanie Clerc
- MRC Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Paulina A. Pontifes
- Departamento de Etología, Fauna Silvestre y Animales de Laboratorio, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Avenida Ciudad Universitaria 3000, CP 04510 Coyoacán, Ciudad de México, México
| | - Saudamini Venkatesan
- Institute of Evolutionary Biology and Centre for Immunity, Infection and Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FL, UK
| | - Simon A. Babayan
- Institute of Biodiversity Animal Health and Comparative Medicine, University of Glasgow, Glasgow G12 8QQ, UK
| | - Amy B. Pedersen
- Institute of Evolutionary Biology and Centre for Immunity, Infection and Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FL, UK
| |
Collapse
|
48
|
Jirků M, Lhotská Z, Frgelecová L, Kadlecová O, Petrželková KJ, Morien E, Jirků-Pomajbíková K. Helminth Interactions with Bacteria in the Host Gut Are Essential for Its Immunomodulatory Effect. Microorganisms 2021; 9:microorganisms9020226. [PMID: 33499240 PMCID: PMC7910914 DOI: 10.3390/microorganisms9020226] [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: 12/31/2020] [Revised: 01/20/2021] [Accepted: 01/21/2021] [Indexed: 12/14/2022] Open
Abstract
Colonization by the benign tapeworm, Hymenolepis diminuta, has been associated with a reduction in intestinal inflammation and changes in bacterial microbiota. However, the role of microbiota in the tapeworm anti-inflammatory effect is not yet clear, and the aim of this study was to determine whether disruption of the microflora during worm colonization can affect the course of intestinal inflammation. We added a phase for disrupting the intestinal microbiota using antibiotics to the experimental design for which we previously demonstrated the protective effect of H. diminuta. We monitored the immunological markers, clinical parameters, bacterial microbiota, and histological changes in the colon of rats. After a combination of colonization, antibiotics, and colitis induction, we had four differently affected experimental groups. We observed a different course of the immune response in each group, but no protective effect was found. Rats treated with colonization and antibiotics showed a strong induction of the Th2 response as well as a significant change in microbial diversity. The microbial results also revealed differences in the richness and abundance of some bacterial taxa, influenced by various factors. Our data suggest that interactions between the tapeworm and bacteria may have a major impact on its protective effect.
Collapse
Affiliation(s)
- Milan Jirků
- Biology Centre, Czech Academy of Sciences, Institute of Parasitology, Branišovská 31, 370 05 České Budějovice, Czech Republic; (Z.L.); (O.K.); (K.J.P.)
- Correspondence: (M.J.); (K.J.-P.); Tel.: +420-38-777-5470 (M.J.); +420-38-777-5470 (K.J.P.)
| | - Zuzana Lhotská
- Biology Centre, Czech Academy of Sciences, Institute of Parasitology, Branišovská 31, 370 05 České Budějovice, Czech Republic; (Z.L.); (O.K.); (K.J.P.)
- Department of Medical Biology, Faculty of Science, University of South Bohemia, Branišovská 31, 370 05 České Budějovice, Czech Republic
| | - Lucia Frgelecová
- Department of Pathology and Parasitology, University of Veterinary and Pharmaceutical Sciences Brno, Palackého tř. 1/3, 612 42 Brno, Czech Republic;
| | - Oldřiška Kadlecová
- Biology Centre, Czech Academy of Sciences, Institute of Parasitology, Branišovská 31, 370 05 České Budějovice, Czech Republic; (Z.L.); (O.K.); (K.J.P.)
| | - Klára Judita Petrželková
- Biology Centre, Czech Academy of Sciences, Institute of Parasitology, Branišovská 31, 370 05 České Budějovice, Czech Republic; (Z.L.); (O.K.); (K.J.P.)
- Institute of Vertebrate Biology, Czech Academy of Sciences, Květná, 8603 65 Brno, Czech Republic
| | - Evan Morien
- Department of Botany, University of British Columbia, 3156-6270 University Blvd., Vancouver, BC V6T 1Z4, Canada;
| | - Kateřina Jirků-Pomajbíková
- Biology Centre, Czech Academy of Sciences, Institute of Parasitology, Branišovská 31, 370 05 České Budějovice, Czech Republic; (Z.L.); (O.K.); (K.J.P.)
- Department of Medical Biology, Faculty of Science, University of South Bohemia, Branišovská 31, 370 05 České Budějovice, Czech Republic
- Correspondence: (M.J.); (K.J.-P.); Tel.: +420-38-777-5470 (M.J.); +420-38-777-5470 (K.J.P.)
| |
Collapse
|
49
|
Agache I, Akdis CA, Akdis M, Canonica GW, Casale T, Chivato T, Corren J, Chu DK, Del Giacco S, Eiwegger T, Flood B, Firinu D, Gern JE, Hamelmann E, Hanania N, Hernández‐Martín I, Knibb R, Mäkelä M, Nair P, O’Mahony L, Papadopoulos NG, Papi A, Park H, Pérez de Llano L, Pfaar O, Quirce S, Sastre J, Shamji M, Schwarze J, Palomares O, Jutel M. EAACI Biologicals Guidelines-Recommendations for severe asthma. Allergy 2021; 76:14-44. [PMID: 32484954 DOI: 10.1111/all.14425] [Citation(s) in RCA: 130] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 05/19/2020] [Indexed: 12/20/2022]
Abstract
Severe asthma imposes a significant burden on patients, families and healthcare systems. Management is difficult, due to disease heterogeneity, co-morbidities, complexity in care pathways and differences between national or regional healthcare systems. Better understanding of the mechanisms has enabled a stratified approach to the management of severe asthma, supporting the use of targeted treatments with biologicals. However, there are still many issues that require further clarification. These include selection of a certain biological (as they all target overlapping disease phenotypes), the definition of response, strategies to enhance the responder rate, the duration of treatment and its regimen (in the clinic or home-based) and its cost-effectiveness. The EAACI Guidelines on the use of biologicals in severe asthma follow the GRADE approach in formulating recommendations for each biological and each outcome. In addition, a management algorithm for the use of biologicals in the clinic is proposed, together with future approaches and research priorities.
Collapse
Affiliation(s)
- Ioana Agache
- Faculty of Medicine Transylvania University Brasov Romania
| | - Cezmi A. Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
- Christine‐Kühne‐Center for Allergy Research and Education (CK‐CARE) Davos Switzerland
| | - Mubeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
| | - Giorgio Walter Canonica
- Personalized Medicine, Asthma and Allergy Humanitas Clinical and Research Center IRCCS Rozzano Italy
| | - Thomas Casale
- Division of Allergy and Immunology University of South Florida Morsani College of Medicine Tampa FL USA
| | - Tomas Chivato
- School of Medicine University CEU San Pablo Madrid Spain
| | | | - Derek K. Chu
- Department of Health Research Methods, Evidence and Impact Division of Immunology and Allergy, and Department of Medicine McMaster University Hamilton ON Canada
| | - Stefano Del Giacco
- Department of Medical Sciences and Public Health University of Cagliari Cagliari Italy
| | - Thomas Eiwegger
- Translational Medicine Program, Research Institute Hospital for Sick Children Toronto ON Canada
- Department of Immunology University of Toronto Toronto ON Canada
- Division of Immunology and Allergy Food Allergy and Anaphylaxis Program The Hospital for Sick Children Departments of Paediatrics and Immunology University of Toronto Toronto ON Canada
| | - Breda Flood
- European Federation of Allergy and Airway Diseases Brussels Belgium
| | - Davide Firinu
- Department of Medical Sciences and Public Health University of Cagliari Cagliari Italy
| | - James E. Gern
- Department of Pediatrics School of Medicine and Public Health University of Wisconsin Madison WI USA
| | - Eckard Hamelmann
- Children’s Center Bethel Evangelical Hospital Bethel University of Bielefeld Bielefeld Germany
| | - Nicola Hanania
- Section of Pulmonary, Critical Care and Sleep Medicine Baylor College of Medicine Houston TX USA
| | | | - Rebeca Knibb
- Department of Psychology School of Life and Health Sciences Aston University Birmingham UK
| | - Mika Mäkelä
- Skin and Allergy Hospital Helsinki University Hospital and University of Helsinki Helsinki Finland
| | - Parameswaran Nair
- Division of Respirology Department of Medicine McMaster University Hamilton ON Canada
- Firestone Institute for Respiratory Health St Joseph's Healthcare Hamilton ON Canada
| | - Liam O’Mahony
- Departments of Medicine and Microbiology APC Microbiome Ireland University College Cork Cork Ireland
| | - Nikolaos G. Papadopoulos
- Division of Infection, Immunity and Respiratory Medicine University of Manchester Manchester UK
- Allergy Department 2nd Pediatric Clinic National Kapodistrian University of Athens Athens Greece
| | - Alberto Papi
- Research Center on Asthma and COPD Department of Medical Sciences University of Ferrara Ferrara Italy
| | - Hae‐Sim Park
- Department of Allergy and Clinical Immunology Ajou University Ajou Korea
| | | | - Oliver Pfaar
- Department of Otorhinolaryngology, Head and Neck Surgery Section of Rhinology and Allergy University Hospital Marburg Philipps‐Universität Marburg Marburg Germany
| | - Santiago Quirce
- Department of Allergy La Paz University Hospital IdiPAZ CIBER of Respiratory Diseases (CIBERES) Universidad Autónoma de Madrid Madrid Spain
| | - Joaquin Sastre
- Facultad de Medicina Universidad Autónoma de Madrid Madrid Spain
| | - Mohamed Shamji
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Inflammation, Repair, Development National Heart and Lung Institute London UK
- Imperial College NIHR Biomedical Research Centre Asthma UK Centre in Allergic Mechanisms of Asthma London UK
| | - Jurgen Schwarze
- Centre for Inflammation Research, Child Life and Health The University of Edinburgh Edinburgh UK
| | - Oscar Palomares
- Department of Biochemistry and Molecular Biology Chemistry School Complutense University of Madrid Madrid Spain
| | - Marek Jutel
- Department of Clinical Immunology Wroclaw Medical University Wroclaw Poland
- All‐MED Medical Research Institute Wroclaw Poland
| |
Collapse
|
50
|
Cristina Borges Araujo E, Cariaco Y, Paulo Oliveira Almeida M, Patricia Pallete Briceño M, Neto de Sousa JE, Rezende Lima W, Maria Costa-Cruz J, Maria Silva N. Beneficial effects of Strongyloides venezuelensis antigen extract in acute experimental toxoplasmosis. Parasite Immunol 2020; 43:e12811. [PMID: 33247953 DOI: 10.1111/pim.12811] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 11/21/2020] [Accepted: 11/23/2020] [Indexed: 01/13/2023]
Abstract
BACKGROUND Toxoplasma gondii is a protozoan with worldwide distribution and triggers a strong Th1 immune response in infected susceptible hosts. On the contrary, most helminth infections are characterized by Th2 immune response and the use of helminth-derived antigens to regulate immune response in inflammatory disorders has been broadly investigated. OBJECTIVES The aim of this study was to investigate whether treatment with Strongyloides venezuelensis antigen extract (SvAg) would alter immune response against T gondii. METHODS C57BL/6 mice were orally infected with T gondii and treated with SvAg, and parasitological, histological and immunological parameters were investigated. RESULTS It was observed that SvAg treatment improved survival rates of T gondii-infected mice. At day 7 post-infection, the parasite load was lower in the lung and small intestine of infected SvAg-treated mice than untreated infected mice. Remarkably, SvAg-treated mice infected with T gondii presented reduced inflammatory lesions in the small intestine than infected untreated mice and decreased intestinal and systemic levels of IFN-γ, TNF-α and IL-6. In contrast, SvAg treatment increased T gondii-specific IgA serum levels in infected mice. CONCLUSIONS S venezuelensis antigen extract has anti-parasitic and anti-inflammatory properties during T gondii infection suggesting as a possible alternative to parasite and inflammation control.
Collapse
Affiliation(s)
- Ester Cristina Borges Araujo
- Laboratório de Imunopatologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brasil
| | - Yusmaris Cariaco
- Laboratório de Imunopatologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brasil
| | - Marcos Paulo Oliveira Almeida
- Laboratório de Imunopatologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brasil
| | | | - José Eduardo Neto de Sousa
- Laboratório de Diagnóstico de Parasitoses, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brasil
| | - Wânia Rezende Lima
- Instituto de Biotecnologia, Universidade Federal de Catalão, Rua Terezinha Margon Vaz, s/n Residencial Barka II, Catalão, Brasil
| | - Julia Maria Costa-Cruz
- Laboratório de Diagnóstico de Parasitoses, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brasil
| | - Neide Maria Silva
- Laboratório de Imunopatologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, Brasil
| |
Collapse
|