1
|
Whitehead B, Sørensen Rossen L, Zippor M, Boysen AT, Indira Chandran V, Skallerup P, Thamsborg SM, Nejsum P. Micro RNA profiles of host extracellular vesicles are modulated by Ascaris suum infection but parasite extracellular vesicle miRNAs are systemically undetectable using in-depth miRNA sequencing. Int J Parasitol 2024; 54:691-696. [PMID: 39116918 DOI: 10.1016/j.ijpara.2024.08.001] [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: 02/26/2024] [Revised: 06/05/2024] [Accepted: 08/02/2024] [Indexed: 08/10/2024]
Abstract
The intestinal helminth Ascaris lumbricoides infects over 800 million people. Infections are often chronic and immunity is not sterilizing due to host-immune modulation, therefore reinfection is common after antihelmintic treatment. We have previously demonstrated a role for Ascaris spp. extracellular vesicles (EVs) in host immune modulation but whether EVs are recognized by the adaptive immune system and are present systemically in the host remains unknown. Therefore, we employed a well-established trickle infection model in pigs to mimic natural Ascaris infection in humans. EVs were isolated from adult Ascaris suum followed by immunoblotting of EV and EV-depleted secretory fractions using plasma from infected and uninfected pigs. Next, EVs were isolated from pig plasma at day 56 post first infection and subjected to deep small RNAseq analysis. RNAs were aligned to A. suum and Sus scrofa miRNA complements to detect A. suum EVs and elucidate the host EV micro RNA (miRNA) response to infection, respectively. Infection generates robust antibody responses against A. suum EVs that is distinct from EV-depleted fractions. However, A. suum miRNAs were not detectable in EVs from the peripheral blood. Notably, host plasma-derived EV miRNA profiles showed significant changes between infected and uninfected pigs, indicating that Ascaris infection drives systemic changes in host EV composition.
Collapse
Affiliation(s)
- Bradley Whitehead
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark.
| | - Litten Sørensen Rossen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Mads Zippor
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Anders T Boysen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Vineesh Indira Chandran
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | | | - Stig M Thamsborg
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Peter Nejsum
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark.
| |
Collapse
|
2
|
Partida-Rodríguez O, Brown EM, Woodward SE, Cirstea M, Reynolds LA, Petersen C, Vogt SL, Peña-Díaz J, Thorson L, Arrieta MC, Hernández EG, Rojas-Velázquez L, Moran P, González Rivas E, Serrano-Vázquez A, Pérez-Juárez H, Torres J, Ximénez C, Finlay BB. Fecal microbiota transplantation from protozoa-exposed donors downregulates immune response in a germ-free mouse model, its role in immune response and physiology of the intestine. PLoS One 2024; 19:e0312775. [PMID: 39466773 PMCID: PMC11515975 DOI: 10.1371/journal.pone.0312775] [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: 03/26/2024] [Accepted: 10/12/2024] [Indexed: 10/30/2024] Open
Abstract
Intestinal parasites are part of the intestinal ecosystem and have been shown to establish close interactions with the intestinal microbiota. However, little is known about the influence of intestinal protozoa on the regulation of the immune response. In this study, we analyzed the regulation of the immune response of germ-free mice transplanted with fecal microbiota (FMT) from individuals with multiple parasitic protozoans (P) and non-parasitized individuals (NP). We determined the production of intestinal cytokines, the lymphocyte populations in both the colon and the spleen, and the genetic expression of markers of intestinal epithelial integrity. We observed a general downregulation of the intestinal immune response in mice receiving FMT-P. We found significantly lower intestinal production of the cytokines IL-6, TNF, IFN-γ, MCP-1, IL-10, and IL-12 in the FMT-P. Furthermore, a significant decrease in the proportion of CD3+, CD4+, and Foxp3+ T regulatory cells (Treg) was observed in both, the colon and spleen with FMT-P in contrast to FMT-NP. We also found that in FMT-P mice there was a significant decrease in tjp1 expression in all three regions of the small intestine; ocln in the ileum; reg3γ in the duodenum and relmβ in both the duodenum and ileum. We also found an increase in colonic mucus layer thickness in mice colonized with FMT-P in contrast with FMT-NP. Finally, our results suggest that gut protozoa, such as Blastocystis hominis, Entamoeba coli, Endolimax nana, Entamoeba histolytica/E. dispar, Iodamoeba bütschlii, and Chilomastix mesnili consortia affect the immunoinflammatory state and induce functional changes in the intestine via the gut microbiota. Likewise, it allows us to establish an FMT model in germ-free mice as a viable alternative to explore the effects that exposure to intestinal parasites could have on the immune response in humans.
Collapse
Affiliation(s)
- Oswaldo Partida-Rodríguez
- Unidad de Investigación en Medicina Experimental, Hospital General de Mexico, Universidad Nacional Autónoma de México, Mexico, Mexico
- Michael Smith Laboratories, Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
| | - Eric M. Brown
- Michael Smith Laboratories, Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
| | - Sarah E. Woodward
- Michael Smith Laboratories, Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
| | - Mihai Cirstea
- Michael Smith Laboratories, Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
| | - Lisa A. Reynolds
- Michael Smith Laboratories, Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
- Department of Biochemistry and Microbiology, Faculty of Science, University of Victoria, Victoria, Canada
| | - Charisse Petersen
- Michael Smith Laboratories, Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
| | - Stefanie L. Vogt
- Michael Smith Laboratories, Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
| | - Jorge Peña-Díaz
- Michael Smith Laboratories, Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
| | - Lisa Thorson
- Michael Smith Laboratories, Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
| | - Marie-Claire Arrieta
- Michael Smith Laboratories, Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Eric G. Hernández
- Unidad de Investigación en Medicina Experimental, Hospital General de Mexico, Universidad Nacional Autónoma de México, Mexico, Mexico
| | - Liliana Rojas-Velázquez
- Unidad de Investigación en Medicina Experimental, Hospital General de Mexico, Universidad Nacional Autónoma de México, Mexico, Mexico
| | - Patricia Moran
- Unidad de Investigación en Medicina Experimental, Hospital General de Mexico, Universidad Nacional Autónoma de México, Mexico, Mexico
| | - Enrique González Rivas
- Unidad de Investigación en Medicina Experimental, Hospital General de Mexico, Universidad Nacional Autónoma de México, Mexico, Mexico
| | - Angélica Serrano-Vázquez
- Unidad de Investigación en Medicina Experimental, Hospital General de Mexico, Universidad Nacional Autónoma de México, Mexico, Mexico
| | - Horacio Pérez-Juárez
- Unidad de Investigación en Medicina Experimental, Hospital General de Mexico, Universidad Nacional Autónoma de México, Mexico, Mexico
| | - Javier Torres
- Unidad de Investigación en Enfermedades Infecciosas y Parasitarias, Instituto Mexicano del Seguro Social (IMSS), Mexico, Mexico
| | - Cecilia Ximénez
- Unidad de Investigación en Medicina Experimental, Hospital General de Mexico, Universidad Nacional Autónoma de México, Mexico, Mexico
| | - B. B. Finlay
- Michael Smith Laboratories, Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
- Department of Microbiology and Immunology, Faculty of Science, University of British Columbia, Vancouver, Canada
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| |
Collapse
|
3
|
Shen C, Zhu X, Chang H, Li C, Hou M, Chen L, Lu Chen, Zhou Z, Ji M, Xu Z. The rebalancing of the immune system at the maternal-fetal interface ameliorates autism-like behavior in adult offspring. Cell Rep 2024; 43:114787. [PMID: 39321022 DOI: 10.1016/j.celrep.2024.114787] [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: 04/08/2024] [Revised: 07/04/2024] [Accepted: 09/06/2024] [Indexed: 09/27/2024] Open
Abstract
Maternal immune activation (MIA) is critical for imparting neuropathology and altered behaviors in offspring; however, maternal-fetal immune cell populations have not been thoroughly investigated in MIA-induced autism spectrum disorders (ASDs). Here, we report the single-cell transcriptional landscape of placental cells in both PBS- and poly(I:C)-induced MIA dams. We observed a decrease in regulatory T (Treg) cells but an increase in the M1 macrophage population at the maternal-fetal interface in MIA dams. Based on the Treg-targeting approach, we investigate an immunoregulatory protein, the helminth-derived heat shock protein 90α (Sjp90α), that induces maternal Treg cells and subsequently rescues the autism-like behaviors in adult offspring. Furthermore, in vivo depletion of maternal macrophages attenuates placental inflammatory reaction and reverses behavioral abnormalities in adult offspring. Notably, Sjp90α induces CD4+ T cell differentiation via scavenger receptor A (SR-A) on the macrophage in vitro. Our findings suggest a maternal Treg-targeted approach to alleviate MIA-induced autism-like behavior in adult offspring.
Collapse
Affiliation(s)
- Chunxiang Shen
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Xinyi Zhu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Hao Chang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Chen Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Min Hou
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China; Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing, Jiangsu 211166, P.R. China
| | - Lin Chen
- Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing, Jiangsu 211166, P.R. China
| | - Lu Chen
- Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing, Jiangsu 211166, P.R. China
| | - Zikai Zhou
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong 528400, P.R. China.
| | - Minjun Ji
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China; Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing, Jiangsu 211166, P.R. China; NHC Key Laboratory of Antibody Technique, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China.
| | - Zhipeng Xu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China; Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing, Jiangsu 211166, P.R. China; NHC Key Laboratory of Antibody Technique, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China.
| |
Collapse
|
4
|
Hussain A, Khan H, Rasool A, Rafiq N, Badshah F, Tariq M, Khan MS, Ibáñez-Arancibia E, De Los Ríos-Escalante PR, Badshah S, Ben Said M. Diversity of aquatic parasites in pristine spring waters in Tehsil Babuzai, Swat, Pakistan. BRAZ J BIOL 2024; 84:e282008. [PMID: 39383363 DOI: 10.1590/1519-6984.282008] [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: 01/05/2024] [Accepted: 06/03/2024] [Indexed: 10/11/2024] Open
Abstract
Global access to clean and safe drinking water remains a formidable challenge, contributing to a myriad of health issues. This research exposes the existence of waterborne parasites in seemingly pristine spring waters, indicating potential contamination. Daily extensive sampling of Seventeen water sources was conducted in the untarnished freshwater streams of Tehsil Babuzai, District Swat, Khyber Pakhtunkhwa, Pakistan, from February to September 2021. Employing a stringent filtration process, the collected samples were effectively concentrated to detect any waterborne parasites. Subsequent application of the wet mount technique, combined with the capabilities of a compound microscope, revealed a disconcerting reality: all examined samples tested positive for various parasites. Identified parasites included Schistosoma species, Ascaris lumbricoides, Trichiuria trichiuria, Taenia saginata, Entamoeba histolytica, Amoeba, Lacrymana olor, Tintinnids, Paramecium, Dileptus, Euglena, Loxodes striatus, Acanthocyclops lynceus, Spondylosium, Oscillatoria, Cyanobacteria, Cilindros, Cilindros cerro, Commensal amoeba mature cysts,, Filliform larva of Strongyloides, Cercaria larva, Larva of Taenia solium, Egg of Enterobius vermiculais, Egg of Isospora belli, Egg of Tapeworm, Egg of Schistosoma species, Egg of Toxocara, and Egg of Diphyllobothrium latum. These findings clearly demonstrate the presence of a diverse array of parasites in the freshwater springs of Tehsil Babuzai, Swat, Pakistan. Implementing robust water treatment protocols, conducting regular monitoring and testing, and raising awareness about the risks of waterborne parasites are crucial steps to safeguard public health in the region.
Collapse
Affiliation(s)
- A Hussain
- University of Swat, Department of Zoology, Swat, Pakistan
| | - H Khan
- University of Swat, Department of Zoology, Swat, Pakistan
| | - A Rasool
- University of Swat, Department of Zoology, Swat, Pakistan
| | - N Rafiq
- Abdul Wali Khan University Mardan, Department of Zoology, Mardan, Pakistan
| | - F Badshah
- Abdul Wali Khan University Mardan, Department of Zoology, Mardan, Pakistan
- Chinese Academy of Agricultural Sciences - GSCAAS, Institute of Animal Science, State Key Laboratory of Animal Biotech Breeding, Beijing, China
- Chinese Academy of Agricultural Sciences - GSCAAS, Agricultural Genomics Institute, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Guangdong Laboratory of Lingnan Modern Agriculture, Shenzhen, China
| | - M Tariq
- Nanjing Agricultural University, College of Animal Science and Technology, Nanjing, Jiangsu, PR China
| | - M S Khan
- Abdul Wali Khan University Mardan, Department of Zoology, Mardan, Pakistan
| | - E Ibáñez-Arancibia
- Universidad de La Frontera, Programa de Doctorado en Ciencias Mención Biología Celular y Molecular Aplicada, Temuco, Chile
- Universidad de La Frontera, Facultad de Ingeniería y Ciencias, Departamento de Ingeniería Química, Laboratorio de Ingeniería, Biotecnología y Bioquímica Aplicada - LIBBA, Temuco, Chile
- Universidad Católica de Temuco, Facultad de Recursos Naturales, Departamento de Ciencias Biológicas y Químicas, Temuco, Chile
| | - P R De Los Ríos-Escalante
- Universidad Católica de Temuco, Facultad de Recursos Naturales, Departamento de Ciencias Biológicas y Químicas, Temuco, Chile
- Universidad Católica de Temuco, Facultad de Recursos Naturales, Nucleo de Estudios Ambientales, Temuco, Chile
| | - S Badshah
- Kohat University of Science and Technology, Department of Botany, Kohat, Pakistan
| | - M Ben Said
- University of Manouba, Higher Institute of Biotechnology of Sidi Thabet, Department of Basic Sciences, Manouba, Tunisia
- University of Manouba, National School of Veterinary Medicine of Sidi Thabet, Laboratory of Microbiology, Manouba, Tunisia
| |
Collapse
|
5
|
Sanku G, Ricciardi A, Redekar NR, Schaughency P, Lack J, Gazzinelli-Guimaraes PH, Nutman TB. Brugia malayi filarial helminth-derived extracellular vesicles suppress antigen presenting cell function and antigen-specific CD4+ T cell responses. Front Immunol 2024; 15:1436818. [PMID: 39434874 PMCID: PMC11491353 DOI: 10.3389/fimmu.2024.1436818] [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: 05/22/2024] [Accepted: 08/08/2024] [Indexed: 10/23/2024] Open
Abstract
Introduction Live microfilariae (mf) and mf-derived extracellular vesicles (EVs) have been shown to modulate human antigen presenting cell (APC) function, most notably by suppressing the induction of IL-12 (and other pro-inflammatory cytokines) following activation with LPS and interferon-y. Methods To explore further how EVs alter human APC function, we studied the effect of mf and EVs on human elutriated monocyte-derived dendritic cells (DC) following exposure to Mf, mf-derived excretory/secretory (E/S) products, E/S depleted of EVs through ultracentrifugation and purified EVs. After demonstrating that the measurable responses induced by live mf could be recapitulated by EVs and EV-containing E/S, we next performed RNAseq analysis of human DC following exposure to live mf, EVs, E/S, or EV-depleted E/S. Results In our analyses of the data for the DC, using a false discovery rate (FDR)<0.05, EV-exposed DC had induced the expression of 212 differentially expressed genes (DEGs) when compared to unexposed DC and 157 when compared to E/S-depleted EVs. These genes were enriched in GO biological processes associated with neutrophil degranulation and 15 DEGs associated with KEGG Lysosome pathways. IPA analysis point to immune dysregulation. We next aimed to understand the intracellular processes altered by EVs and the effect these have on effector T cells. When SARS CoV-2 Membrane-specific CD4+ TCLs were assessed following EV conditioning of autologous DC and activation with the SARS CoV-2-Membrane peptide pool, we found conditioning reduced the frequency of SARS CoV-2 Membrane-specific CD3+ CD4+ CD154+ cells (p=.015). Similarly, EV-conditioning of SARS CoV-2 Membrane-specific CD3+ CD4+ cells induced fewer cell capable of producing IFN-γ (p=.045). Discussion Taken together, our data suggest a modulatory role of EVs on APC function that likely leads to defects in T cell effector function.
Collapse
Affiliation(s)
- Gayatri Sanku
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Alessandra Ricciardi
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Neelam R. Redekar
- Integrated Data Science Section (IDSS), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Paul Schaughency
- Integrated Data Science Section (IDSS), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Justin Lack
- Integrated Data Science Section (IDSS), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Pedro H. Gazzinelli-Guimaraes
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Thomas B. Nutman
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda, MD, United States
| |
Collapse
|
6
|
Yang Y, Azzuolo A, Fodil N, Gros P. Gene: environment interactions in immune and inflammatory responses to severe acute respiratory syndrome coronavirus 2 infection. Curr Opin Immunol 2024; 90:102459. [PMID: 39243725 DOI: 10.1016/j.coi.2024.102459] [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: 01/23/2024] [Revised: 08/07/2024] [Accepted: 08/19/2024] [Indexed: 09/09/2024]
Abstract
Despite its devastating human cost, the rapid spread and global establishment of the severe acute respiratory syndrome coronavirus (SARS-CoV-2) pandemic had the benefit of providing unique insights into the intricate interplay between genetic, environmental, and socioeconomic factors, which collectively impact susceptibility to infection with SARS-CoV-2. Preceding the implementation of broad vaccination programs and assuming the absence of significant acquired immunity, examining the innate vulnerability to the virus becomes essential. There is indeed considerable heterogeneity observed at both the population and individual levels for various SARS-CoV-2 infection phenotypes, including emergence, progression, and survival from the coronavirus disease 2019 (COVID-19) syndrome. Particularly intriguing is the seemingly milder course of COVID-19 disease reported for the African continent early during the pandemic. This was characterized by significantly lower mortality rates in SARS-CoV-2 patients compared with the European and American continents and globally. We will discuss some of the demographic and socioeconomic factors that may have contributed to these observations. We review the mapped COVID-19 genetic architecture, including the remarkable association of type I interferon as a single protective mechanism and a major determinant of susceptibility. Furthermore, we speculate on potential 'environmental' modulators of penetrance and expressivity of intrinsic vulnerability factors, with a focus on the microbiome and associated metabolomes. Additionally, this review explores the potential immunomodulatory contribution of helminth parasites to the human host immune and inflammatory responses to respiratory viral infections.
Collapse
Affiliation(s)
- Yunxiang Yang
- Department of Biochemistry, McGill Research Center of Complex Traits, and Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, QC, Canada
| | - Alessia Azzuolo
- Department of Biochemistry, McGill Research Center of Complex Traits, and Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, QC, Canada
| | - Nassima Fodil
- Department of Biochemistry, McGill Research Center of Complex Traits, and Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, QC, Canada
| | - Philippe Gros
- Department of Biochemistry, McGill Research Center of Complex Traits, and Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, QC, Canada.
| |
Collapse
|
7
|
Midha A, Oser L, Schlosser-Brandenburg J, Laubschat A, Mugo RM, Musimbi ZD, Höfler P, Kundik A, Hayani R, Adjah J, Groenhagen S, Tieke M, Elizalde-Velázquez LE, Kühl AA, Klopfleisch R, Tedin K, Rausch S, Hartmann S. Concurrent Ascaris infection modulates host immunity resulting in impaired control of Salmonella infection in pigs. mSphere 2024; 9:e0047824. [PMID: 39140728 PMCID: PMC11423588 DOI: 10.1128/msphere.00478-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 07/09/2024] [Indexed: 08/15/2024] Open
Abstract
Ascaris is one of the most widespread helminth infections, leading to chronic morbidity in humans and considerable economic losses in pig farming. In addition, pigs are an important reservoir for the zoonotic salmonellosis, where pigs can serve as asymptomatic carriers. Here, we investigated the impact of an ongoing Ascaris infection on the immune response to Salmonella in pigs. We observed higher bacterial burdens in experimentally coinfected pigs compared to pigs infected with Salmonella alone. The impaired control of Salmonella in the coinfected pigs was associated with repressed interferon gamma responses in the small intestine and with the alternative activation of gut macrophages evident in elevated CD206 expression. Ascaris single and coinfection were associated with a rise of CD4-CD8α+FoxP3+ Treg in the lymph nodes draining the small intestine and liver. In addition, macrophages from coinfected pigs showed enhanced susceptibility to Salmonella infection in vitro and the Salmonella-induced monocytosis and tumor necrosis factor alpha production by myeloid cells was repressed in pigs coinfected with Ascaris. Hence, our data indicate that acute Ascaris infection modulates different immune effector functions with important consequences for the control of tissue-invasive coinfecting pathogens.IMPORTANCEIn experimentally infected pigs, we show that an ongoing infection with the parasitic worm Ascaris suum modulates host immunity, and coinfected pigs have higher Salmonella burdens compared to pigs infected with Salmonella alone. Both infections are widespread in pig production and the prevalence of Salmonella is high in endemic regions of human Ascariasis, indicating that this is a clinically meaningful coinfection. We observed the type 2/regulatory immune response to be induced during an Ascaris infection correlates with increased susceptibility of pigs to the concurrent bacterial infection.
Collapse
Affiliation(s)
- Ankur Midha
- Department of Veterinary Medicine, Institute of Immunology, Centre for Infection Medicine, Freie Universität Berlin, Berlin, Germany
| | - Larissa Oser
- Department of Veterinary Medicine, Institute of Immunology, Centre for Infection Medicine, Freie Universität Berlin, Berlin, Germany
| | - Josephine Schlosser-Brandenburg
- Department of Veterinary Medicine, Institute of Immunology, Centre for Infection Medicine, Freie Universität Berlin, Berlin, Germany
| | - Alexandra Laubschat
- Department of Veterinary Medicine, Institute of Immunology, Centre for Infection Medicine, Freie Universität Berlin, Berlin, Germany
| | - Robert M Mugo
- Department of Veterinary Medicine, Institute of Immunology, Centre for Infection Medicine, Freie Universität Berlin, Berlin, Germany
| | - Zaneta D Musimbi
- Department of Veterinary Medicine, Institute of Immunology, Centre for Infection Medicine, Freie Universität Berlin, Berlin, Germany
| | - Philipp Höfler
- Department of Veterinary Medicine, Institute of Immunology, Centre for Infection Medicine, Freie Universität Berlin, Berlin, Germany
| | - Arkadi Kundik
- Department of Veterinary Medicine, Institute of Immunology, Centre for Infection Medicine, Freie Universität Berlin, Berlin, Germany
| | - Rima Hayani
- Department of Veterinary Medicine, Institute of Immunology, Centre for Infection Medicine, Freie Universität Berlin, Berlin, Germany
| | - Joshua Adjah
- Department of Veterinary Medicine, Institute of Immunology, Centre for Infection Medicine, Freie Universität Berlin, Berlin, Germany
| | - Saskia Groenhagen
- Department of Veterinary Medicine, Institute of Immunology, Centre for Infection Medicine, Freie Universität Berlin, Berlin, Germany
| | - Malte Tieke
- Department of Veterinary Medicine, Institute of Immunology, Centre for Infection Medicine, Freie Universität Berlin, Berlin, Germany
| | - Luis E Elizalde-Velázquez
- Department of Veterinary Medicine, Institute of Immunology, Centre for Infection Medicine, Freie Universität Berlin, Berlin, Germany
| | - Anja A Kühl
- Charité Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, iPATH.Berlin, Core unit of Charité, Campus Benjamin Franklin, Berlin, Germany
| | - Robert Klopfleisch
- Department of Veterinary Medicine, Institute of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Karsten Tedin
- Department of Veterinary Medicine, Institute of Microbiology and Epizootics, Centre for Infection Medicine, Freie Universität Berlin, Berlin, Germany
| | - Sebastian Rausch
- Department of Veterinary Medicine, Institute of Immunology, Centre for Infection Medicine, Freie Universität Berlin, Berlin, Germany
| | - Susanne Hartmann
- Department of Veterinary Medicine, Institute of Immunology, Centre for Infection Medicine, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
8
|
Lins JGG, Albuquerque ACA, Louvandini H, Amarante AFT. Immunohistochemistry analyses of the abomasal mucosa show differences in cellular-mediated immune responses to Haemonchus contortus infection in resistant and susceptible young lambs. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2024; 161:105259. [PMID: 39216779 DOI: 10.1016/j.dci.2024.105259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Haemonchus contortus is known for its high pathogenicity in sheep, and the uncontrolled use of anthelmintics resulted in the emergence of multiple drug-resistant populations. Breeding sheep for gastrointestinal nematode resistance is a sustainable alternative to reduce dependence of anthelmintic drugs, and differences in the degree of resistance between breeds have been reported. Here we compare two sheep breeds (Santa Ines and Ile de France), concerning the differences in innate and adaptive immune response involved in the resistance against H. contortus infection. Immunohistochemical analyses of the abomasum were conducted in naïve Santa Ines (n = 14) and Ile de France (n = 12) lambs randomized into four groups: infected Santa Ines (n = 8), non-infected control Santa Ines (n = 6), infected Ile de France (n = 8), and non-infected control Ile de France (n = 4). The infected lambs were initially infected with H. contortus infective larvae at 14 days of age, and multiple infections were conducted every second day until they reached 66 days of age. There was a significant effect (P < 0.001) of the infection with increase in numbers of CD3+ T; CD79α+ B; GATA3+ Th2/ILC2; POU2F3+ tuft cells; FOXP3+ T reg; and IgE + cells in the fundus of the abomasal mucosa in both Santa Ines and Ile de France lambs. Nevertheless, the infected Santa Ines lambs presented the highest averages for CD79α+ B; GATA3+ Th2/ILC; IgE + cells; and POU2F3+ tuft cells and there was a significant association of the breed and infection status with regards to POU2F3+ tuft cells, with the highest mean in the infected Santa Ines group. The infected Santa Ines group had three lambs with high degree of resistance and five lambs that showed a moderate infection. Our results suggest a mechanism of synergistic coordination between different immune-cell types in promoting resistance of suckling lambs under H. contortus infection.
Collapse
Affiliation(s)
- José Gabriel Gonçalves Lins
- São Paulo State University (UNESP), School of Veterinary Medicine and Animal Science, Botucatu, SP, Brazil; São Paulo State University (UNESP), Department of Biodiversity and Biostatistics of the Bioscience Institute, Botucatu, SP, Brazil.
| | - Ana Cláudia A Albuquerque
- São Paulo State University (UNESP), Department of Biodiversity and Biostatistics of the Bioscience Institute, Botucatu, SP, Brazil
| | - Helder Louvandini
- Center for Nuclear Energy in Agriculture, University of São Paulo, Piracicaba, SP, Brazil
| | - Alessandro F T Amarante
- São Paulo State University (UNESP), Department of Biodiversity and Biostatistics of the Bioscience Institute, Botucatu, SP, Brazil
| |
Collapse
|
9
|
Bamorovat M, Sharifi I, Shafiei Bafti M, Agha Kuchak Afshari S, Aflatoonian MR, Karamoozian A, Jafarzadeh A, Amirzadeh R, Khosravi A, Babaei Z, Safa F, Sharifi F, Heshmatkhah A. Cutaneous Leishmaniasis Hampers COVID-19: A Controlled Cross-Sectional Study in High-Burden Endemic Areas of Iran. J Epidemiol Glob Health 2024; 14:142-153. [PMID: 38190050 PMCID: PMC11043327 DOI: 10.1007/s44197-023-00179-0] [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: 08/28/2023] [Accepted: 12/06/2023] [Indexed: 01/09/2024] Open
Abstract
INTRODUCTION Emerging infectious diseases such as SARS-CoV-2 can cause pandemics and create a critical risk for humans. In a previous pilot study, we reported that the immunological responses induced by cutaneous leishmaniasis (CL) could decrease the incidence and severity of COVID-19. In this large-scale case-control study, we assessed the possible relationship between mortality and morbidity of COVID-19 in healed CL persons suffering scars compared to cases without CL history. METHODS This controlled cross-sectional study was conducted between July 2020 and December 2022 in the endemic and high-burden areas of CL in southeastern Iran. In the study, 1400 previous CL cases with scars and 1,521,329 subjects who had no previous CL were analyzed. We used R 4.0.2 to analyze the data. Firth's bias reduction approach corresponding to the penalization of likelihood logistic regression by Jeffreys was also employed to influence the variables in the dataset. Also, a Bayesian ordinal logistic regression model was performed to explore the COVID-19 severity in both case and referent groups. RESULTS The occurrence and severity rate of COVID-19 in CL scar cases are significantly less than in the non-CL control group, while in the CL scar subjects, patients with critical conditions and mortality were not observed. The morbidity (OR = 0.11, CI 0.06-0.20 and P < 0.001) and severity of COVID-19 in previous cases with CL scars were significantly diminished than that in the control group (credible interval - 2.57, - 1.62). CONCLUSIONS The results represented a durable negative relationship between cured CL and COVID-19 incidence and severity. Additional studies seem necessary and should be designed to further validate the true impact and underlying mechanistic action of CL on COVID-19.
Collapse
Affiliation(s)
- Mehdi Bamorovat
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran.
| | - Iraj Sharifi
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran.
| | - Mehdi Shafiei Bafti
- Institute for Studies in Medicine History, Persian and Complementary Medicine, Iran University of Medical Sciences, Tehran, Iran
- Deputy for Health, Kerman University of Medical Sciences, Kerman, Iran
| | | | | | - Ali Karamoozian
- Research Center for Modeling in Health, Institute for Futures Studies in Health, Kerman University of Medical Sciences, Kerman, Iran
| | - Abdollah Jafarzadeh
- Department of Immunology, Kerman University of Medical Sciences, Kerman, Iran
| | - Raheleh Amirzadeh
- Research Center for Social Determinants of Health, Institute for Futures Studies in Health, Kerman University of Medical Sciences, Kerman, Iran
| | - Ahmad Khosravi
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Zahra Babaei
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Farzane Safa
- Deputy for Health, Kerman University of Medical Sciences, Kerman, Iran
| | - Fatemeh Sharifi
- Research Center for Tropical and Infectious Diseases, Kerman University of Medical Sciences, Kerman, Iran
| | - Amireh Heshmatkhah
- Dadbin Health Clinic, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
10
|
Tong M, Yang X, Qiao Y, Liu G, Ge H, Huang G, Wang Y, Yang Y, Fan W. Serine protease inhibitor from the muscle larval Trichinella spiralis ameliorates non-alcoholic fatty liver disease in mice via anti-inflammatory properties and gut-liver crosstalk. Biomed Pharmacother 2024; 172:116223. [PMID: 38325266 DOI: 10.1016/j.biopha.2024.116223] [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: 10/31/2023] [Revised: 01/22/2024] [Accepted: 01/29/2024] [Indexed: 02/09/2024] Open
Abstract
Trichinella spiralis is recognized for its ability to regulate host immune responses. The serine protease inhibitor of T. spiralis (Ts-SPI) participates in T. spiralis-mediated immunoregulatory effects. Studies have shown that helminth therapy exhibits therapeutic effects on metabolic diseases. In addition, we previously found that T. spiralis-derived crude antigens could alleviate diet-induced obesity. Thus, Ts-SPI was hypothesized to alleviate non-alcoholic fatty liver disease (NAFLD). Herein, recombinant Ts-SPI (rTs-SPI) was prepared from the muscle larvae T. spiralis. The relative molecular mass of rTs-SPI was approximately 35,000 Da, and western blot analysis indicated good immunoreactivity. rTs-SPI ameliorated hepatic steatosis, inflammation, and pyroptosis in NAFLD mice, which validated the hypothesis. rTs-SPI also reduced macrophage infiltration, significantly expanded Foxp3+ Treg population, and inactivated TLR4/NF-κB/NLRP3 signaling in the liver. Furthermore, rTs-SPI treatment significantly shifted the gut microbiome structure, with a remarkable increase in beneficial bacteria and reduction in harmful bacteria to improve gut barrier integrity. Finally, Abx-treated mice and FMT confirmed that gut-liver crosstalk contributed to NAFLD improvement after rTs-SPI treatment. Taken together, Taken together, these findings suggest that rTs-SPI exerts therapeutic effects in NAFLD via anti-inflammatory activity and gut-liver crosstalk.
Collapse
Affiliation(s)
- Mingwei Tong
- School of Basic Medical Sciences, Shanxi Medical University, Jinzhong 030619, China; Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, and Shanxi Key Laboratory of Cellular Physiology, Taiyuan 030001, China.
| | - Xiaodan Yang
- School of Basic Medical Sciences, Shanxi Medical University, Jinzhong 030619, China
| | - Yuyu Qiao
- School of Basic Medical Sciences, Shanxi Medical University, Jinzhong 030619, China
| | - Ge Liu
- School of Basic Medical Sciences, Shanxi Medical University, Jinzhong 030619, China
| | - Huihui Ge
- School of Basic Medical Sciences, Shanxi Medical University, Jinzhong 030619, China
| | - Guangrong Huang
- School of Basic Medical Sciences, Shanxi Medical University, Jinzhong 030619, China
| | - Yanhong Wang
- School of Basic Medical Sciences, Shanxi Medical University, Jinzhong 030619, China; Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, and Shanxi Key Laboratory of Cellular Physiology, Taiyuan 030001, China
| | - Yong Yang
- School of Basic Medical Sciences, Shanxi Medical University, Jinzhong 030619, China; Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, and Shanxi Key Laboratory of Cellular Physiology, Taiyuan 030001, China.
| | - Weiping Fan
- School of Basic Medical Sciences, Shanxi Medical University, Jinzhong 030619, China; Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, and Shanxi Key Laboratory of Cellular Physiology, Taiyuan 030001, China.
| |
Collapse
|
11
|
Tanabe MB, Caravedo MA, Clinton White A, Cabada MM. An Update on the Pathogenesis of Fascioliasis: What Do We Know? Res Rep Trop Med 2024; 15:13-24. [PMID: 38371362 PMCID: PMC10874186 DOI: 10.2147/rrtm.s397138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 01/27/2024] [Indexed: 02/20/2024] Open
Abstract
Fasciola hepatica is a trematode parasite distributed worldwide. It is known to cause disease in mammals, producing significant economic loses to livestock industry and burden to human health. After ingestion, the parasites migrate through the liver and mature in the bile ducts. A better understanding of the parasite's immunopathogenesis would help to develop efficacious therapeutics and vaccines. Currently, much of our knowledge comes from in vitro and in vivo studies in animal models. Relatively little is known about the host-parasite interactions in humans. Here, we provide a narrative review of what is currently know about the pathogenesis and host immune responses to F. hepatica summarizing the evidence available from the multiple hosts that this parasite infects.
Collapse
Affiliation(s)
- Melinda B Tanabe
- Division of Infectious Disease, Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, USA
| | - Maria A Caravedo
- Division of Infectious Disease, Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, USA
| | - A Clinton White
- Division of Infectious Disease, Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, USA
- Cusco Branch – Alexander von Humboldt Tropical Medicine Institute, Universidad Peruana Cayetano Heredia, Cusco, Peru
| | - Miguel M Cabada
- Division of Infectious Disease, Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, USA
- Cusco Branch – Alexander von Humboldt Tropical Medicine Institute, Universidad Peruana Cayetano Heredia, Cusco, Peru
| |
Collapse
|
12
|
Eckmair B, Gao C, Mehta AY, Dutkiewicz Z, Vanbeselaere J, Cummings RD, Paschinger K, Wilson IBH. Recognition of Highly Branched N-Glycans of the Porcine Whipworm by the Immune System. Mol Cell Proteomics 2024; 23:100711. [PMID: 38182041 PMCID: PMC10850124 DOI: 10.1016/j.mcpro.2024.100711] [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: 10/04/2023] [Revised: 12/14/2023] [Accepted: 01/02/2024] [Indexed: 01/07/2024] Open
Abstract
Glycans are key to host-pathogen interactions, whereby recognition by the host and immunomodulation by the pathogen can be mediated by carbohydrate binding proteins, such as lectins of the innate immune system, and their glycoconjugate ligands. Previous studies have shown that excretory-secretory products of the porcine nematode parasite Trichuris suis exert immunomodulatory effects in a glycan-dependent manner. To better understand the mechanisms of these interactions, we prepared N-glycans from T. suis and both analyzed their structures and used them to generate a natural glycan microarray. With this array, we explored the interactions of glycans with C-type lectins, C-reactive protein, and sera from T. suis-infected pigs. Glycans containing LacdiNAc and phosphorylcholine-modified glycans were associated with the highest binding by most of these proteins. In-depth analysis revealed not only fucosylated LacdiNAc motifs with and without phosphorylcholine moieties but phosphorylcholine-modified mannose and N-acetylhexosamine-substituted fucose residues, in the context of maximally tetraantennary N-glycan scaffolds. Furthermore, O-glycans also contained fucosylated motifs. In summary, the glycans of T. suis are recognized by both the innate and adaptive immune systems and also exhibit species-specific features distinguishing its glycome from those of other nematodes.
Collapse
Affiliation(s)
- Barbara Eckmair
- Department für Chemie, Institut für Biochemie, Universität für Bodenkultur, Wien, Austria
| | - Chao Gao
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Akul Y Mehta
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Zuzanna Dutkiewicz
- Department für Chemie, Institut für Biochemie, Universität für Bodenkultur, Wien, Austria
| | - Jorick Vanbeselaere
- Department für Chemie, Institut für Biochemie, Universität für Bodenkultur, Wien, Austria
| | - Richard D Cummings
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Katharina Paschinger
- Department für Chemie, Institut für Biochemie, Universität für Bodenkultur, Wien, Austria
| | - Iain B H Wilson
- Department für Chemie, Institut für Biochemie, Universität für Bodenkultur, Wien, Austria.
| |
Collapse
|
13
|
Toru M, Atnaf A, Mengist HM, Reta A. The COVID-19 Severity and Its Association with Intestinal Parasite Coinfection and Urine Biochemical Parameters among COVID-19-Confirmed Patients Admitted to Debre Markos University COVID-19 Center, Northwest Ethiopia. BIOMED RESEARCH INTERNATIONAL 2024; 2024:3064374. [PMID: 38249633 PMCID: PMC10799708 DOI: 10.1155/2024/3064374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 12/17/2023] [Accepted: 01/02/2024] [Indexed: 01/23/2024]
Abstract
Background Though most people with COVID-19 disease show asymptomatic to mild illness, a substantial number of patients are at high risk of developing severe disease and adverse outcomes with long COVID-19 and death. Even though some studies showed that previously existing infections with parasites amend the host's body defenses to increase resistance to infection with SARS-CoV-2, there is limited data in Ethiopia. Objectives This study is aimed at determining the COVID-19 disease severity and its association with intestinal parasite coinfection and urine biochemical parameters among COVID-19-confirmed patients admitted at Debre Markos University COVID-19 Center, 2021. Methods A prospective cohort study was conducted on 136 RT-qPCR-confirmed COVID-19 patients admitted at Debre Markos University COVID-19 Center from January 1 to March 30, 2021. Sociodemographic and clinical data were collected by using standardized data collection forms. A urine biochemical test was performed using a dry urine dipstick kit and stool examination using direct wet mount microscopic examination and formalin-ether concentration method. The chi-square test, Fisher exact test, and ordinal logistic regression analysis were computed to assess association with outcome variables using Statistical Package for Social Science software (version 24). Result A total of 136 COVID-19-confirmed patients participated in this study. The median age of the participants was 48 years. The majority (86 (62.5%)) of them were male in sex. Of the 136 cases, 39 (28.7%) had died. Among the 136 patients, 22 (16.2%) were coinfected with intestinal parasites. COVID-19 patients who have intestinal parasite coinfection had lower odds of developing clinically severe COVID-19 compared to noninfected (AOR = 0.37; 95% CI = 0.147-0.944; P = 0.037). The majority (104 (76.5%)) of them have abnormal urine biochemical results. From the abnormal urine biochemical tests observed, the urine blood, glucose, and ketone tests were positive for 54 (39.7%), 36 (26.5%), and 30 (21.1%) patients, respectively. Among the 31 critical COVID-19 patients, 25 (80.6%) showed abnormal urine biochemical parameters. Age and comorbidity were significantly associated with COVID-19 severity (P < 0.05). Conclusion Patients with old age and comorbidity had an increased risk of developing severe COVID-19 disease. Patients having SARS-CoV-2 and intestinal parasitic coinfections demonstrated mild COVID-19 disease severity. Abnormal urine biochemical results were common among critical COVID-19 patients. Thus, advanced study on the effect of the interaction among intestinal parasites on COVID-19 clinical severity and its mechanisms is essential.
Collapse
Affiliation(s)
- Milkiyas Toru
- Department of Medical Laboratory Science, College of Health Science, Debre Markos University, Debre Markos, Ethiopia
| | - Aytenew Atnaf
- Department of Medical Laboratory Science, College of Health Science, Debre Markos University, Debre Markos, Ethiopia
| | - Hylemariam Mihiretie Mengist
- Department of Medical Laboratory Science, College of Health Science, Debre Markos University, Debre Markos, Ethiopia
| | - Alemayehu Reta
- Department of Medical Laboratory Science, College of Health Science, Debre Markos University, Debre Markos, Ethiopia
| |
Collapse
|
14
|
Lekki-Jóźwiak J, Bąska P. The Roles of Various Immune Cell Populations in Immune Response against Helminths. Int J Mol Sci 2023; 25:420. [PMID: 38203591 PMCID: PMC10778651 DOI: 10.3390/ijms25010420] [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: 11/17/2023] [Revised: 12/15/2023] [Accepted: 12/22/2023] [Indexed: 01/12/2024] Open
Abstract
Helminths are multicellular parasites that are a substantial problem for both human and veterinary medicine. According to estimates, 1.5 billion people suffer from their infection, resulting in decreased life quality and burdens for healthcare systems. On the other hand, these infections may alleviate autoimmune diseases and allergy symptoms. The immune system is programmed to combat infections; nevertheless, its effector mechanisms may result in immunopathologies and exacerbate clinical symptoms. This review summarizes the role of the immune response against worms, with an emphasis on the Th2 response, which is a hallmark of helminth infections. We characterize non-immune cells (enteric tuft cells-ETCs) responsible for detecting parasites, as well as the role of hematopoietic-derived cells (macrophages, basophils, eosinophils, neutrophils, innate lymphoid cells group 2-ILC2s, mast cells, T cells, and B cells) in initiating and sustaining the immune response, as well as the functions they play in granulomas. The aim of this paper is to review the existing knowledge regarding the immune response against helminths, to attempt to decipher the interactions between cells engaged in the response, and to indicate the gaps in the current knowledge.
Collapse
Affiliation(s)
- Janina Lekki-Jóźwiak
- Division of Parasitology and Parasitic Diseases, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-786 Warsaw, Poland;
| | - Piotr Bąska
- Division of Pharmacology and Toxicology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-786 Warsaw, Poland
| |
Collapse
|
15
|
Kumar M, Mazumder P, Silori R, Manna S, Panday DP, Das N, Sethy SK, Kuroda K, Mahapatra DM, Mahlknecht J, Tyagi VK, Singh R, Zang J, Barceló D. Prevalence of pharmaceuticals and personal care products, microplastics and co-infecting microbes in the post-COVID-19 era and its implications on antimicrobial resistance and potential endocrine disruptive effects. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 904:166419. [PMID: 37625721 DOI: 10.1016/j.scitotenv.2023.166419] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 08/15/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023]
Abstract
The COVID-19 (coronavirus disease 2019) pandemic's steady condition coupled with predominance of emerging contaminants in the environment and its synergistic implications in recent times has stoked interest in combating medical emergencies in this dynamic environment. In this context, high concentrations of pharmaceutical and personal care products (PPCPs), microplastics (MPs), antimicrobial resistance (AMR), and soaring coinfecting microbes, tied with potential endocrine disruptive (ED) are critical environmental concerns that requires a detailed documentation and analysis. During the pandemic, the identification, enumeration, and assessment of potential hazards of PPCPs and MPs and (used as anti-COVID-19 agents/applications) in aquatic habitats have been attempted globally. Albeit receding threats in the magnitude of COVID-19 infections, both these pollutants have still posed serious consequences to aquatic ecosystems and the very health and hygiene of the population in the vicinity. The surge in the contaminants post-COVID also renders them to be potent vectors to harbor and amplify AMR. Pertinently, the present work attempts to critically review such instances to understand the underlying mechanism, interactions swaying the current health of our environment during this post-COVID-19 era. During this juncture, although prevention of diseases, patient care, and self-hygiene have taken precedence, nevertheless antimicrobial stewardship (AMS) efforts have been overlooked. Unnecessary usage of PPCPs and plastics during the pandemic has resulted in increased emerging contaminants (i.e., active pharmaceutical ingredients and MPs) in various environmental matrices. It was also noticed that among COVID-19 patients, while the bacterial co-infection prevalence was 0.2-51%, the fungi, viral, protozoan and helminth were 0.3-49, 1-22, 2-15, 0.4-15% respectively, rendering them resistant to residual PPCPs. There are inevitable chances of ED effects from PPCPs and MPs applied previously, that could pose far-reaching health concerns. Furthermore, clinical and other experimental evidence for many newer compounds is very scarce and demands further research. Pro-active measures targeting effective waste management, evolved environmental policies aiding strict regulatory measures, and scientific research would be crucial in minimizing the impact and creating better preparedness towards such events among the masses fostering sustainability.
Collapse
Affiliation(s)
- Manish Kumar
- Sustainability Cluster, School of Advanced Engineering, UPES, Dehradun, Uttarakhand 248007, India; Escuela de Ingenieria y Ciencias, Tecnologico de Monterrey, Campus Monterrey, Monterrey 64849, Nuevo Leon, Mexico.
| | - Payal Mazumder
- Sustainability Cluster, School of Advanced Engineering, UPES, Dehradun, Uttarakhand 248007, India
| | - Rahul Silori
- Sustainability Cluster, School of Advanced Engineering, UPES, Dehradun, Uttarakhand 248007, India
| | - Suvendu Manna
- Sustainability Cluster, School of Advanced Engineering, UPES, Dehradun, Uttarakhand 248007, India
| | - Durga Prasad Panday
- Sustainability Cluster, School of Advanced Engineering, UPES, Dehradun, Uttarakhand 248007, India
| | - Nilotpal Das
- ENCORE Insoltech Pvt. Ltd, Randesan, Gandhinagar, Gujarat 382421, India
| | - Susanta Kumar Sethy
- Sustainability Cluster, School of Advanced Engineering, UPES, Dehradun, Uttarakhand 248007, India
| | - Keisuke Kuroda
- Department of Environmental and Civil Engineering, Toyama Prefectural University, Imizu 939 0398, Japan
| | - Durga Madhab Mahapatra
- Department of Chemical and Petroleum Engineering, School of Advanced Engineering, UPES, Dehradun, Uttarakhand 248007, India; Department of Biological and Ecological Engineering, Oregon State University, Corvallis, OR 97331, USA
| | - Jürgen Mahlknecht
- Escuela de Ingenieria y Ciencias, Tecnologico de Monterrey, Campus Monterrey, Monterrey 64849, Nuevo Leon, Mexico
| | - Vinay Kumar Tyagi
- Wastewater Division, National Institute of Hydrology Roorkee, Roorkee, Uttranchal, India
| | - Rajesh Singh
- Wastewater Division, National Institute of Hydrology Roorkee, Roorkee, Uttranchal, India
| | - Jian Zang
- Department of Civil Engineering, Chongqing University, China
| | - Damià Barceló
- Sustainability Cluster, School of Advanced Engineering, UPES, Dehradun, Uttarakhand 248007, India; Water and Soil Quality Research Group, Department of Environmental Chemistry, Institute of Environmental Assessment and Water Research (IDAEA-CSIC), Jordi Girona 1826, Barcelona 08034, Spain
| |
Collapse
|
16
|
Singh SP, Smyth DJ, Cunningham K, Mukundan A, Byeon CH, Hinck CS, White MPJ, Ciancia C, Wosowska N, Sanders A, Jin R, Lilla S, Zanivan S, Schoenherr C, Inman G, van Dinther M, ten Dijke P, Hinck AP, Maizels RM. The helminth TGF-β mimic TGM4 is a modular ligand that binds CD44, CD49d and TGF-β receptors to preferentially target myeloid cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.13.566701. [PMID: 38014296 PMCID: PMC10680678 DOI: 10.1101/2023.11.13.566701] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
The murine helminth parasite Heligmosomoides polygyrus expresses a family of modular proteins which, replicating the functional activity of the immunomodulatory cytokine TGF-β, have been named TGM (TGF-β Μimic). Multiple domains bind to different receptors, including TGF-β receptors TβRI (ALK5) and TβRII through domains 1-3, and prototypic family member TGM1 binds the cell surface co-receptor CD44 through domains 4-5. This allows TGM1 to induce T lymphocyte Foxp3 expression, characteristic of regulatory (Treg) cells, and to activate a range of TGF-β-responsive cell types. In contrast, a related protein, TGM4, targets a much more restricted cell repertoire, primarily acting on myeloid cells, with less potent effects on T cells and lacking activity on other TGF-β-responsive cell types. TGM4 binds avidly to myeloid cells by flow cytometry, and can outcompete TGM1 for cell binding. Analysis of receptor binding in comparison to TGM1 reveals a 10-fold higher affinity than TGM1 for TGFβR-I (TβRI), but a 100-fold lower affinity for TβRII through Domain 3. Consequently, TGM4 is more dependent on co-receptor binding; in addition to CD44, TGM4 also engages CD49d (Itga4) through Domains 1-3, as well as CD206 and Neuropilin-1 through Domains 4 and 5. TGM4 was found to effectively modulate macrophage populations, inhibiting lipopolysaccharide-driven inflammatory cytokine production and boosting interleukin (IL)-4-stimulated responses such as Arginase-1 in vitro and in vivo. These results reveal that the modular nature of TGMs has allowed the fine tuning of the binding affinities of the TβR- and co-receptor binding domains to establish cell specificity for TGF-β signalling in a manner that cannot be attained by the mammalian cytokine.
Collapse
Affiliation(s)
- Shashi P. Singh
- Wellcome Centre of Integrative Parasitology, School of Infection and Immunity, University of Glasgow, G12 8TA, UK
| | - Danielle J. Smyth
- Wellcome Centre of Integrative Parasitology, School of Infection and Immunity, University of Glasgow, G12 8TA, UK
| | - Kyle Cunningham
- Wellcome Centre of Integrative Parasitology, School of Infection and Immunity, University of Glasgow, G12 8TA, UK
| | - Ananya Mukundan
- Department of Structural Biology, University of Pittsburgh, USA
| | | | | | - Madeleine P. J. White
- Wellcome Centre of Integrative Parasitology, School of Infection and Immunity, University of Glasgow, G12 8TA, UK
| | - Claire Ciancia
- Wellcome Centre of Integrative Parasitology, School of Infection and Immunity, University of Glasgow, G12 8TA, UK
| | - Nątalia Wosowska
- Wellcome Centre of Integrative Parasitology, School of Infection and Immunity, University of Glasgow, G12 8TA, UK
| | - Anna Sanders
- Wellcome Centre of Integrative Parasitology, School of Infection and Immunity, University of Glasgow, G12 8TA, UK
| | - Regina Jin
- Wellcome Centre of Integrative Parasitology, School of Infection and Immunity, University of Glasgow, G12 8TA, UK
| | - Sergio Lilla
- Cancer Research UK Scotland Institute, Glasgow, G61 1BD, UK
| | - Sara Zanivan
- Cancer Research UK Scotland Institute, Glasgow, G61 1BD, UK
| | | | - Gareth Inman
- Cancer Research UK Scotland Institute, Glasgow, G61 1BD, UK
| | - Maarten van Dinther
- Oncode Institute and Department of Cell and Chemical Biology, University of Leiden, The Netherlands
| | - Peter ten Dijke
- Oncode Institute and Department of Cell and Chemical Biology, University of Leiden, The Netherlands
| | - Andrew P. Hinck
- Department of Structural Biology, University of Pittsburgh, USA
| | - Rick M. Maizels
- Wellcome Centre of Integrative Parasitology, School of Infection and Immunity, University of Glasgow, G12 8TA, UK
| |
Collapse
|
17
|
Conte M, Varraso R, Fournier A, Rothwell JA, Baglietto L, Fornili M, Sbidian E, Severi G. A prospective study of the association between living in a rural environment during childhood and risk of psoriasis. ENVIRONMENTAL RESEARCH 2023; 237:117062. [PMID: 37660877 DOI: 10.1016/j.envres.2023.117062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 08/04/2023] [Accepted: 08/31/2023] [Indexed: 09/05/2023]
Abstract
Psoriasis is one of the most common immune-mediated inflammatory diseases (IMIDs). Living in a rural environment during childhood is associated with a decreased risk of certain IMIDs, like asthma, in adulthood. However, its role in other IMIDs, such as psoriasis is still unclear. To evaluate the relationships between different factors related to the environment during childhood and the risk of psoriasis in adulthood we conducted a study in E3N, a French prospective cohort composed of 98 995 women. During the 1990-2018 follow-up of 72 154 study participants, we identified 1 967 incident cases of psoriasis from self-reports in self-administered structured questionnaires. During the 2004-2018 follow-up of 67 917 study participants, 188 moderate-to-severe cases of psoriasis were identified through self-reports and from data from a drug reimbursement database. We fitted Cox proportional hazards regression models with age as the time scale from which we estimated hazard ratios adjusted for putative confounders (aHRs). We found inverse associations with risk of psoriasis for rural birthplace [aHR: 0.89 (95%CI: 0.79-0.96)] and for having farming parents [aHR: 0.84 (95%CI: 0.72-0.97)]. For moderate-to-severe psoriasis we found a nominally similar inverse association with rural birthplace but not with having farming parents. Our results suggest that an exposure to a rural environment during childhood may be associated with a reduced risk of psoriasis. These findings may help to improve our understanding of the pathogenesis of psoriasis.
Collapse
Affiliation(s)
- Marco Conte
- Université Paris-Saclay, UVSQ, Inserm, Gustave Roussy, Exposome and Heredity team, CESP, Villejuif, 94805, France
| | - Raphaëlle Varraso
- Université Paris-Saclay, UVSQ, Inserm, Integrative Respiratory Epidemiology Team, CESP, Villejuif, 94807, France
| | - Agnès Fournier
- Université Paris-Saclay, UVSQ, Inserm, Gustave Roussy, Exposome and Heredity team, CESP, Villejuif, 94805, France
| | - Joseph A Rothwell
- Université Paris-Saclay, UVSQ, Inserm, Gustave Roussy, Exposome and Heredity team, CESP, Villejuif, 94805, France
| | - Laura Baglietto
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, 56126, Italy
| | - Marco Fornili
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, 56126, Italy
| | - Emilie Sbidian
- Hôpital Henri Mondor, Department of Dermatology, Créteil, 94010, France; Université Paris Est Créteil (UPEC), Epidemiology in Dermatology and Evaluation of Therapeutics (Epi-DermE), Créteil, 94010, France
| | - Gianluca Severi
- Université Paris-Saclay, UVSQ, Inserm, Gustave Roussy, Exposome and Heredity team, CESP, Villejuif, 94805, France; Department of Statistics, Computer Science and Applications « G. Parenti », University of Florence, Florence, 50134, Italy.
| |
Collapse
|
18
|
Mules TC, Inns S, Le Gros G. Helminths' therapeutic potential to treat intestinal barrier dysfunction. Allergy 2023; 78:2892-2905. [PMID: 37449458 DOI: 10.1111/all.15812] [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: 05/16/2023] [Revised: 06/20/2023] [Accepted: 07/02/2023] [Indexed: 07/18/2023]
Abstract
The intestinal barrier is a dynamic multi-layered structure which can adapt to environmental changes within the intestinal lumen. It has the complex task of allowing nutrient absorption while limiting entry of harmful microbes and microbial antigens present in the intestinal lumen. Excessive entry of microbial antigens via microbial translocation due to 'intestinal barrier dysfunction' is hypothesised to contribute to the increasing incidence of allergic, autoimmune and metabolic diseases, a concept referred to as the 'epithelial barrier theory'. Helminths reside in the intestinal tract are in intimate contact with the mucosal surfaces and induce a range of local immunological changes which affect the layers of the intestinal barrier. Helminths are proposed to prevent, or even treat, many of the diseases implicated in the epithelial barrier theory. This review will focus on the effect of helminths on intestinal barrier function and explore whether this could explain the proposed health benefits delivered by helminths.
Collapse
Affiliation(s)
- Thomas C Mules
- Malaghan Institute of Medical Research, Wellington, New Zealand
- University of Otago, Wellington, New Zealand
| | | | - Graham Le Gros
- Malaghan Institute of Medical Research, Wellington, New Zealand
| |
Collapse
|
19
|
Singh AK, de Gooyer T, Singh OP, Pandey S, Neyaz A, Cloots K, Kansal S, Malaviya P, Rai M, Nylén S, Chakravarty J, Hasker E, Sundar S. Wuchereria bancrofti infection is associated with progression to clinical visceral leishmaniasis in VL- endemic areas in Muzaffarpur, Bihar, India. PLoS Negl Trop Dis 2023; 17:e0011729. [PMID: 37903175 PMCID: PMC10635566 DOI: 10.1371/journal.pntd.0011729] [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: 02/08/2023] [Revised: 11/09/2023] [Accepted: 10/16/2023] [Indexed: 11/01/2023] Open
Abstract
BACKGROUND Co-endemicity of neglected tropical diseases (NTDs) necessitates that these diseases should be considered concomitantly to understand the relationship between pathology and to support disease management and control programs. The aims of the study were to assess the prevalence of filarial infection in asymptomatic Leishmania donovani infected individuals and the correlation of Wuchereria bancrofti infection with progression to clinical visceral leishmaniasis (VL) in Bihar, India. METHODOLOGY/PRINCIPAL FINDINGS Within the Muzaffarpur-TMRC Health and Demographic Surveillance System (HDSS) area, a cohort of Leishmania seropositive (n = 476) or seronegative individuals (n = 1130) were sampled annually for three years for filarial infection and followed for progression to clinical VL. To corroborate the results from the cohort study, we also used a retrospective case-control study of 36 VL cases and 71 controls selected from a subset of the HDSS population to investigate the relationship between progression to clinical VL and the prevalence of filarial infection at baseline. Our findings suggest a higher probability of progression to clinical VL in individuals with a history of filarial infection: in both the cohort and case-control studies, progression to clinical VL was higher among filaria infected individuals (RR = 2.57, p = 0.056, and OR = 2.52, p = 0.046 respectively). CONCLUSION This study describes that progression to clinical VL disease is associated with serological evidence of prior infection with W. bancrofti. The integration of disease programs for Leishmania and lymphatic filariasis extend beyond the relationship of sequential or co-infection with disease burden. To ensure elimination targets can be reached and sustained, we suggest areas of co-endemicity would benefit from overlapping vector control activities, health system networks and surveillance infrastructure.
Collapse
Affiliation(s)
- Abhishek Kumar Singh
- Infectious Disease Research Laboratory, Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Tanyth de Gooyer
- Department of Public Health, Institute of Tropical Medicine, Antwerp, Belgium
| | - Om Prakash Singh
- Infectious Disease Research Laboratory, Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
- Department of Biochemistry, Institute of Sciences, Banaras Hindu University, Varanasi, India
| | - Sundaram Pandey
- Infectious Disease Research Laboratory, Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Aziza Neyaz
- Infectious Disease Research Laboratory, Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Kristien Cloots
- Department of Public Health, Institute of Tropical Medicine, Antwerp, Belgium
| | - Sangeeta Kansal
- Department of Community Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Paritosh Malaviya
- Infectious Disease Research Laboratory, Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Madhukar Rai
- Infectious Disease Research Laboratory, Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Susanne Nylén
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solna, Sweden
| | - Jaya Chakravarty
- Infectious Disease Research Laboratory, Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Epco Hasker
- Department of Public Health, Institute of Tropical Medicine, Antwerp, Belgium
| | - Shyam Sundar
- Infectious Disease Research Laboratory, Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| |
Collapse
|
20
|
Eckmair B, Gao C, Mehta AY, Dutkiewicz Z, Vanbeselaere J, Cummings RD, Paschinger K, Wilson IBH. Recognition of highly branched N-glycans of the porcine whipworm by the immune system. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.21.557549. [PMID: 37790353 PMCID: PMC10542551 DOI: 10.1101/2023.09.21.557549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Glycans are key to host-pathogen interactions, whereby recognition by the host and immunomodulation by the pathogen can be mediated by carbohydrate binding proteins, such as lectins of the innate immune system, and their glycoconjugate ligands. Previous studies have shown that excretory-secretory products of the porcine nematode parasite Trichuris suis exert immunomodulatory effects in a glycan-dependent manner. To better understand the mechanisms of these interactions, we prepared N-glycans from T. suis and both analyzed their structures and used them to generate a natural glycan microarray. With this array we explored the interactions of glycans with C-type lectins, C-reactive protein and sera from T. suis infected pigs. Glycans containing LacdiNAc and phosphorylcholine-modified glycans were associated with the highest binding by most of these proteins. In-depth analysis revealed not only fucosylated LacdiNAc motifs with and without phosphorylcholine moieties, but phosphorylcholine-modified mannose and N-acetylhexosamine-substituted fucose residues, in the context of maximally tetraantennary N-glycan scaffolds. Furthermore, O-glycans also contained fucosylated motifs. In summary, the glycans of T. suis are recognized by both the innate and adaptive immune systems, and also exhibit species-specific features distinguishing its glycome from those of other nematodes.
Collapse
Affiliation(s)
- Barbara Eckmair
- Institut für Biochemie, Department für Chemie, Universität für Bodenkultur, Muthgasse 18, 1190 Wien, Austria
| | - Chao Gao
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, 3 Blackfan Circle, Boston, MA 02115, USA
| | - Akul Y Mehta
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, 3 Blackfan Circle, Boston, MA 02115, USA
| | - Zuzanna Dutkiewicz
- Institut für Biochemie, Department für Chemie, Universität für Bodenkultur, Muthgasse 18, 1190 Wien, Austria
| | - Jorick Vanbeselaere
- Institut für Biochemie, Department für Chemie, Universität für Bodenkultur, Muthgasse 18, 1190 Wien, Austria
| | - Richard D Cummings
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, 3 Blackfan Circle, Boston, MA 02115, USA
| | - Katharina Paschinger
- Institut für Biochemie, Department für Chemie, Universität für Bodenkultur, Muthgasse 18, 1190 Wien, Austria
| | - Iain B H Wilson
- Institut für Biochemie, Department für Chemie, Universität für Bodenkultur, Muthgasse 18, 1190 Wien, Austria
| |
Collapse
|
21
|
Hubbard IC, Thompson JS, Else KJ, Shears RK. Another decade of Trichuris muris research: An update and application of key discoveries. ADVANCES IN PARASITOLOGY 2023; 121:1-63. [PMID: 37474238 DOI: 10.1016/bs.apar.2023.05.002] [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: 07/22/2023]
Abstract
The mouse whipworm, Trichuris muris, has been used for over 60 years as a tractable model for human trichuriasis, caused by the related whipworm species, T. trichiura. The history of T. muris research, from the discovery of the parasite in 1761 to understanding the lifecycle and outcome of infection with different doses (high versus low dose infection), as well as the immune mechanisms associated with parasite expulsion and chronic infection have been detailed in an earlier review published in 2013. Here, we review recent advances in our understanding of whipworm biology, host-parasite interactions and basic immunology brought about using the T. muris mouse model, focussing on developments from the last decade. In addition to the traditional high/low dose infection models that have formed the mainstay of T. muris research to date, novel models involving trickle (repeated low dose) infection in laboratory mice or infection in wild or semi-wild mice have led to important insights into how immunity develops in situ in a multivariate environment, while the use of novel techniques such as the development of caecal organoids (enabling the study of larval development ex vivo) promise to deliver important insights into host-parasite interactions. In addition, the genome and transcriptome analyses of T. muris and T. trichiura have proven to be invaluable tools, particularly in the context of vaccine development and identification of secreted products including proteins, extracellular vesicles and micro-RNAs, shedding further light on how these parasites communicate with their host and modulate the immune response to promote their own survival.
Collapse
Affiliation(s)
- Isabella C Hubbard
- Centre for Bioscience, Manchester Metropolitan University, Manchester, United Kingdom; Department of Life Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, United Kingdom
| | - Jacob S Thompson
- Lydia Becker Institute for Immunology and Inflammation, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Kathryn J Else
- Lydia Becker Institute for Immunology and Inflammation, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Rebecca K Shears
- Centre for Bioscience, Manchester Metropolitan University, Manchester, United Kingdom; Department of Life Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, United Kingdom.
| |
Collapse
|
22
|
McManus CM, Maizels RM. Regulatory T cells in parasite infections: susceptibility, specificity and specialisation. Trends Parasitol 2023; 39:547-562. [PMID: 37225557 DOI: 10.1016/j.pt.2023.04.002] [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: 02/28/2023] [Revised: 04/06/2023] [Accepted: 04/06/2023] [Indexed: 05/26/2023]
Abstract
Regulatory T cells (Tregs) are essential to control immune system responses to innocuous self-specificities, intestinal and environmental antigens. However, they may also interfere with immunity to parasites, particularly in chronic infection. Susceptibility to many parasite infections is, to a greater or lesser extent, controlled by Tregs, but often they play a more prominent role in moderating the immunopathological consequences of parasitism, and dampening bystander reactions in an antigen-nonspecific manner. More recently, Treg subtypes have been defined which may preferentially act in different contexts; we also discuss the degree to which this specialisation is now being mapped onto how Tregs maintain the delicate balance between tolerance, immunity, and pathology in infection.
Collapse
Affiliation(s)
- Caitlin M McManus
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, 120 University Place, Glasgow G12 8TA, UK
| | - Rick M Maizels
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, 120 University Place, Glasgow G12 8TA, UK.
| |
Collapse
|
23
|
Wang Y, Guo A, Zou Y, Mu W, Zhang S, Shi Z, Liu Z, Cai X, Zhu XQ, Wang S. Interaction between tissue-dwelling helminth and the gut microbiota drives mucosal immunoregulation. NPJ Biofilms Microbiomes 2023; 9:43. [PMID: 37355675 DOI: 10.1038/s41522-023-00410-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 06/13/2023] [Indexed: 06/26/2023] Open
Abstract
Tissue-dwelling helminths affect billions of people around the world. They are potent manipulators of the host immune system, prominently by promoting regulatory T cells (Tregs) and are generally associated with a modified host gut microbiome. However, the role of the gut microbiota in the immunomodulatory processes for these non-intestinal parasites is still unclear. In the present study, we used an extra-intestinal cestode helminth model-larval Echinococcus multilocularis to explore the tripartite partnership (host-helminth-bacteria) in the context of regulating colonic Tregs in Balb/c mice. We showed that larval E. multilocularis infection in the peritoneal cavity attenuated colitis in Balb/c mice and induced a significant expansion of colonic Foxp3+ Treg populations. Fecal microbiota depletion and transplantation experiments showed that the gut microbiota contributed to increasing Tregs after the helminth infection. Shotgun metagenomic and metabolic analyses revealed that the gut microbiome structure after infection was significantly shifted with a remarkable increase of Lactobacillus reuteri and that the microbial metabolic capability was reprogrammed to produce more Treg cell regulator-short-chain fatty acids in feces. Furthermore, we also prove that the L. reuteri strain elevated in infected mice was sufficient to promote the colonic Treg frequency and its growth was potentially associated with T cell-dependent immunity in larval E. multilocularis infection. Collectively, these findings indicate that the extraintestinal helminth drives expansions of host colonic Tregs through the gut microbes. This study suggests that the gut microbiome serves as a critical component of anti-inflammation effects even for a therapy based on an extraintestinal helminth.
Collapse
Affiliation(s)
- Yugui Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi Province, 030801, China
- State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, 730000, China
| | - Aijiang Guo
- State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, 730000, China
- Key Laboratory of Veterinary Parasitology of Gansu Province, Gansu, 730046, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, 225009, China
| | - Yang Zou
- State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, 730000, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, 225009, China
| | - Wenjie Mu
- State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, 730000, China
| | - Shengying Zhang
- State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, 730000, China
| | - Zhiqi Shi
- State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, 730000, China
| | - Zhongli Liu
- State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, 730000, China
| | - Xuepeng Cai
- State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, 730000, China
| | - Xing-Quan Zhu
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi Province, 030801, China.
| | - Shuai Wang
- State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, 730000, China.
- Key Laboratory of Veterinary Parasitology of Gansu Province, Gansu, 730046, China.
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, 225009, China.
| |
Collapse
|
24
|
Sun L, Su Y, Jiao A, Wang X, Zhang B. T cells in health and disease. Signal Transduct Target Ther 2023; 8:235. [PMID: 37332039 PMCID: PMC10277291 DOI: 10.1038/s41392-023-01471-y] [Citation(s) in RCA: 133] [Impact Index Per Article: 133.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 04/21/2023] [Accepted: 04/24/2023] [Indexed: 06/20/2023] Open
Abstract
T cells are crucial for immune functions to maintain health and prevent disease. T cell development occurs in a stepwise process in the thymus and mainly generates CD4+ and CD8+ T cell subsets. Upon antigen stimulation, naïve T cells differentiate into CD4+ helper and CD8+ cytotoxic effector and memory cells, mediating direct killing, diverse immune regulatory function, and long-term protection. In response to acute and chronic infections and tumors, T cells adopt distinct differentiation trajectories and develop into a range of heterogeneous populations with various phenotype, differentiation potential, and functionality under precise and elaborate regulations of transcriptional and epigenetic programs. Abnormal T-cell immunity can initiate and promote the pathogenesis of autoimmune diseases. In this review, we summarize the current understanding of T cell development, CD4+ and CD8+ T cell classification, and differentiation in physiological settings. We further elaborate the heterogeneity, differentiation, functionality, and regulation network of CD4+ and CD8+ T cells in infectious disease, chronic infection and tumor, and autoimmune disease, highlighting the exhausted CD8+ T cell differentiation trajectory, CD4+ T cell helper function, T cell contributions to immunotherapy and autoimmune pathogenesis. We also discuss the development and function of γδ T cells in tissue surveillance, infection, and tumor immunity. Finally, we summarized current T-cell-based immunotherapies in both cancer and autoimmune diseases, with an emphasis on their clinical applications. A better understanding of T cell immunity provides insight into developing novel prophylactic and therapeutic strategies in human diseases.
Collapse
Affiliation(s)
- Lina Sun
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China
| | - Yanhong Su
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China
| | - Anjun Jiao
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China
| | - Xin Wang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China
| | - Baojun Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China.
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China.
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China.
| |
Collapse
|
25
|
Marascio N, Loria MT, Pavia G, Peronace C, Adams NJ, Campolo M, Divenuto F, Lamberti AG, Giancotti A, Barreca GS, Mazzitelli M, Trecarichi EM, Torti C, Perandin F, Bisoffi Z, Quirino A, Matera G. Evaluation of IL-35, as a Possible Biomarker for Follow-Up after Therapy, in Chronic Human Schistosoma Infection. Vaccines (Basel) 2023; 11:vaccines11050995. [PMID: 37243099 DOI: 10.3390/vaccines11050995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/10/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
The host response to helminth infections is characterized by systemic and tissue-related immune responses that play a crucial role in pathological diseases. Recently, experimental studies have highlighted the role of regulatory T (Tregs) and B (Bregs) cells with secreted cytokines as important markers in anti-schistosomiasis immunity. We investigated the serical levels of five cytokines (TNFα, IFN-γ, IL-4, IL-10 and IL-35) in pre- and post-treatment samples from chronic Schistosoma infected patients to identify potential serological markers during follow-up therapy. Interestingly, we highlighted an increased serum level of IL-35 in the pre-therapy samples (median 439 pg/mL for Schistosoma haematobium and 100.5 pg/mL for Schistsoma mansoni infected patients) compared to a control group (median 62 pg/mL and 58 pg/mL, respectively, p ≤ 0.05), and a significantly lower concentration in post-therapy samples (181 pg/mL for S. haematobium and 49.5 pg/mL for S. mansoni infected patients, p ≤ 0.05). The present study suggests the possible role of IL-35 as a novel serological biomarker in the evaluation of Schistosoma therapy follow-up.
Collapse
Affiliation(s)
- Nadia Marascio
- Clinical Microbiology Unit, Department of Health Sciences, "Magna Græcia" University of Catanzaro-"Mater Domini" Teaching Hospital, 88100 Catanzaro, Italy
| | - Maria Teresa Loria
- Clinical Microbiology Unit, Department of Health Sciences, "Magna Græcia" University of Catanzaro-"Mater Domini" Teaching Hospital, 88100 Catanzaro, Italy
| | - Grazia Pavia
- Clinical Microbiology Unit, Department of Health Sciences, "Magna Græcia" University of Catanzaro-"Mater Domini" Teaching Hospital, 88100 Catanzaro, Italy
| | - Cinzia Peronace
- Clinical Microbiology Unit, Department of Health Sciences, "Magna Græcia" University of Catanzaro-"Mater Domini" Teaching Hospital, 88100 Catanzaro, Italy
| | - Neill James Adams
- Clinical Microbiology Unit, Department of Health Sciences, "Magna Græcia" University of Catanzaro-"Mater Domini" Teaching Hospital, 88100 Catanzaro, Italy
| | - Morena Campolo
- Clinical Microbiology Unit, Department of Health Sciences, "Magna Græcia" University of Catanzaro-"Mater Domini" Teaching Hospital, 88100 Catanzaro, Italy
| | - Francesca Divenuto
- Clinical Microbiology Unit, Department of Health Sciences, "Magna Græcia" University of Catanzaro-"Mater Domini" Teaching Hospital, 88100 Catanzaro, Italy
| | - Angelo Giuseppe Lamberti
- Clinical Microbiology Unit, Department of Health Sciences, "Magna Græcia" University of Catanzaro-"Mater Domini" Teaching Hospital, 88100 Catanzaro, Italy
| | - Aida Giancotti
- Clinical Microbiology Unit, Department of Health Sciences, "Magna Græcia" University of Catanzaro-"Mater Domini" Teaching Hospital, 88100 Catanzaro, Italy
| | - Giorgio Settimo Barreca
- Clinical Microbiology Unit, Department of Health Sciences, "Magna Græcia" University of Catanzaro-"Mater Domini" Teaching Hospital, 88100 Catanzaro, Italy
| | - Maria Mazzitelli
- Infectious and Tropical Diseases Unit, Padua University Hospital, 35128 Padua, Italy
| | - Enrico Maria Trecarichi
- Infectious and Tropical Diseases Unit, Department of Medical and Surgical Sciences, "Magna Graecia" University-"Mater Domini" Teaching Hospital, 88100 Catanzaro, Italy
| | - Carlo Torti
- Infectious and Tropical Diseases Unit, Department of Medical and Surgical Sciences, "Magna Graecia" University-"Mater Domini" Teaching Hospital, 88100 Catanzaro, Italy
| | - Francesca Perandin
- Department of Infectious, Tropical Diseases and Microbiology, IRCCS Sacro Cuore Don Calabria Hospital, Negrar di Valpolicella, 37024 Verona, Italy
| | - Zeno Bisoffi
- Department of Infectious, Tropical Diseases and Microbiology, IRCCS Sacro Cuore Don Calabria Hospital, Negrar di Valpolicella, 37024 Verona, Italy
| | - Angela Quirino
- Clinical Microbiology Unit, Department of Health Sciences, "Magna Græcia" University of Catanzaro-"Mater Domini" Teaching Hospital, 88100 Catanzaro, Italy
| | - Giovanni Matera
- Clinical Microbiology Unit, Department of Health Sciences, "Magna Græcia" University of Catanzaro-"Mater Domini" Teaching Hospital, 88100 Catanzaro, Italy
| |
Collapse
|
26
|
Morales-Ruiz V, López-Recinos D, Castañeda MG, Guevara-Salinas A, Parada-Colin C, Gómez-Fuentes S, Espitia-Pinzón C, Hernández-González M, Adalid-Peralta L. Characterization of excretory/secretory products of the Taenia crassiceps cysticercus involved in the induction of regulatory T cells in vivo. Parasitol Res 2023:10.1007/s00436-023-07847-x. [PMID: 37115316 DOI: 10.1007/s00436-023-07847-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 04/08/2023] [Indexed: 04/29/2023]
Abstract
The ability to modulate the host immune response has allowed some parasites to establish themselves in the tissues of an immunocompetent organism. While some parasite excretion/secretion products (ESPs) were recently reported to induce differentiation of regulatory T cells (Tregs), their identity is not known. This work is aimed to identify and characterize ESPs of Taenia crassiceps cysticerci linked with Treg induction in vivo. ESPs were obtained from cultures of T. crassiceps cysticerci and inoculated in mice, measuring Treg levels by flow cytometry. Proteins in ESPs were analyzed by electrophoresis; then, ESPs were classified as either differential or conserved. Differentially included proteins were MS-sequenced and functionally characterized. Only 4 of 10 ESPs induced Tregs. Proteins with catalytic activity and those involved in immunological processes predominated, supporting the idea that these molecules could play an important role in the induction of Tregs.
Collapse
Affiliation(s)
- Valeria Morales-Ruiz
- Unidad Periférica Para El Estudio de La Neuroinflamación en Patologías Neurológicas del Instituto de Investigaciones Biomédicas de La UNAM en El Instituto Nacional de Neurología Y Neurocirugía. Insurgentes Sur, 3877, Col. La Fama, 14269, Mexico City, Mexico
| | - Dina López-Recinos
- Unidad Periférica Para El Estudio de La Neuroinflamación en Patologías Neurológicas del Instituto de Investigaciones Biomédicas de La UNAM en El Instituto Nacional de Neurología Y Neurocirugía. Insurgentes Sur, 3877, Col. La Fama, 14269, Mexico City, Mexico
| | - María Gracia Castañeda
- Unidad Periférica Para El Estudio de La Neuroinflamación en Patologías Neurológicas del Instituto de Investigaciones Biomédicas de La UNAM en El Instituto Nacional de Neurología Y Neurocirugía. Insurgentes Sur, 3877, Col. La Fama, 14269, Mexico City, Mexico
- Laboratorio de Biología Molecular y Genética del Centro Médico Naval, Cirujano Mayor Santiago Távara de La Marina de Guerra del Perú, Avenida S/N, Avenida República de Venezuela, Bellavista, Peru
| | - Adrián Guevara-Salinas
- Unidad Periférica Para El Estudio de La Neuroinflamación en Patologías Neurológicas del Instituto de Investigaciones Biomédicas de La UNAM en El Instituto Nacional de Neurología Y Neurocirugía. Insurgentes Sur, 3877, Col. La Fama, 14269, Mexico City, Mexico
| | - Cristina Parada-Colin
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Circuito Escolar S/N, Ciudad Universitaria, 04510, Mexico City, Mexico
| | - Sandra Gómez-Fuentes
- Unidad Periférica Para El Estudio de La Neuroinflamación en Patologías Neurológicas del Instituto de Investigaciones Biomédicas de La UNAM en El Instituto Nacional de Neurología Y Neurocirugía. Insurgentes Sur, 3877, Col. La Fama, 14269, Mexico City, Mexico
| | - Clara Espitia-Pinzón
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Circuito Escolar S/N, Ciudad Universitaria, 04510, Mexico City, Mexico
| | - Marisela Hernández-González
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Circuito Escolar S/N, Ciudad Universitaria, 04510, Mexico City, Mexico
| | - Laura Adalid-Peralta
- Unidad Periférica Para El Estudio de La Neuroinflamación en Patologías Neurológicas del Instituto de Investigaciones Biomédicas de La UNAM en El Instituto Nacional de Neurología Y Neurocirugía. Insurgentes Sur, 3877, Col. La Fama, 14269, Mexico City, Mexico.
- Instituto Nacional de Neurología Y Neurocirugía, Insurgentes Sur 3877, Col. La Fama, 14269, Mexico City, Mexico.
| |
Collapse
|
27
|
Atagozli T, Elliott DE, Ince MN. Helminth Lessons in Inflammatory Bowel Diseases (IBD). Biomedicines 2023; 11:1200. [PMID: 37189818 PMCID: PMC10135676 DOI: 10.3390/biomedicines11041200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/10/2023] [Accepted: 04/11/2023] [Indexed: 05/17/2023] Open
Abstract
Helminths are multicellular invertebrates that colonize the gut of many vertebrate animals including humans. This colonization can result in pathology, which requires treatment. It can also lead to a commensal and possibly even a symbiotic relationship where the helminth and the host benefit from each other's presence. Epidemiological data have linked helminth exposure to protection from immune disorders that include a wide range of diseases, such as allergies, autoimmune illnesses, and idiopathic inflammatory disorders of the gut, which are grouped as inflammatory bowel diseases (IBD). Treatment of moderate to severe IBD involves the use of immune modulators and biologics, which can cause life-threatening complications. In this setting, their safety profile makes helminths or helminth products attractive as novel therapeutic approaches to treat IBD or other immune disorders. Helminths stimulate T helper-2 (Th2) and immune regulatory pathways, which are targeted in IBD treatment. Epidemiological explorations, basic science studies, and clinical research on helminths can lead to the development of safe, potent, and novel therapeutic approaches to prevent or treat IBD in addition to other immune disorders.
Collapse
Affiliation(s)
- Tyler Atagozli
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Iowa Carver College of Medicine, Iowa City, IA 52246, USA
| | - David E. Elliott
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Iowa Carver College of Medicine, Iowa City, IA 52246, USA
- Iowa City Veterans Affairs Medical Center, Iowa City, IA 52246, USA
| | - Mirac Nedim Ince
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Iowa Carver College of Medicine, Iowa City, IA 52246, USA
- Iowa City Veterans Affairs Medical Center, Iowa City, IA 52246, USA
| |
Collapse
|
28
|
Filarial infections compromise influenza vaccination efficacy: Lessons from the mouse. Immunol Lett 2023; 255:62-66. [PMID: 36889363 DOI: 10.1016/j.imlet.2023.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 02/24/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023]
Abstract
Helminth parasites infect more than a quarter of the human population and inflict significant changes to the immunological status of their hosts. Several human studies report impaired responses to vaccinations in helminth-infected individuals. Analysing the impact of helminth infections on the efficacy of influenza vaccinations in the mouse system helps to elucidate the underlying immunological processes. Concurrent infection with the parasitic nematode Litomosoides sigmodontis reduced the quantity and quality of antibody responses to vaccination against seasonal influenza in BALB/c and C57BL/6 mice. This led to impaired vaccination-induced protection against challenge infections with the human pathogenic 2009 pandemic H1N1 influenza A virus in helminth-infected mice. Impaired responses were also observed if vaccinations were performed after immune-driven or drug-induced clearance of a previous helminth infection. Mechanistically, the suppression was associated with a systemic and sustained expansion of IL-10-producing CD4+CD49b+LAG-3+ type 1 regulatory T cells and partially abrogated by in vivo blockade of the IL-10 receptor. In summary, these findings raise the concern that individuals in helminth-endemic areas may not always benefit from vaccinations, even in the absence of an acute and diagnosable helminth infection.
Collapse
|
29
|
Andrade MCR, Lemos BRP, Silva LM, Pecotte JK. Eliminating Potential Effects of Other Infections During Selection of Nonhuman Primates for COVID-19 Research. Comp Med 2023; 73:45-57. [PMID: 36744555 PMCID: PMC9948906 DOI: 10.30802/aalas-cm-21-000086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The study of nonhuman primates (NHP) can provide significant insights into our understanding numerous infectious agents. The etiological agent of COVID-19, SARS-CoV-2 virus, first emerged in 2019 and has so far been responsible for the deaths of over 4 million people globally. In the frenzied search to understand its pathogenesis and immunology and to find measures for prevention and control of this pandemic disease, NHP, particularly macaques, are the preferred model because they manifest similar clinical signs and immunologic features as humans. However, possible latent, subclinical, and opportunistic infections not previously detected in animals participating in a study may obscure experimental results and confound data interpretations in testing treatments and vaccine studies for COVID-19. Certain pathophysiologic changes that occur with SARS-CoV-2 virus infection are similar to those of simian pathogens. The current review discusses numerous coinfections of COVID-19 with other diseases and describes possible outcomes and mechanisms in COVID-19 studies of NHP that have coinfections. Due to the urgency triggered by the pandemic, screening that is more rigorous than usual is necessary to limit background noise and maximize the reliability of data from NHP COVID-19 studies. Screening for influenza virus, selected respiratory bacteria, and regional endemic pathogens such as vector-borne agents, together with the animal's individual exposure history, should be the main considerations in selecting a NHP for a COVID-19 study. In addition, because NHP are susceptible to the SARS-CoV-2 virus, management and surveillance measures should be established to prevent transmission to healthy animals from infected colony animals and husbandry staff. This review presents compiled data on the use of NHP in COVID-19 studies, emphasizing the need to create the most reliable NHP model for those studies by extensive screening for other pathogens.
Collapse
Key Words
- absl, animal biosafety level
- ace-2, angiotensin-converting enzyme
- ards, acute respiratory distress syndrome
- cnprc, california national primate research center
- e, envelope
- esr, erythrocyte sedimentation rate
- hav, hepatitis a virus
- hbv, hepatitis b virus
- hgf hepatocyte growth factor
- htlv, human t-cell lymphotropic virus
- ifn, interferon
- il, interleukin
- ip, inducible protein
- m, matrix
- mcp, monocyte chemotactic proteins
- mcsf, macrophage colony-stimulating factor
- mip, macrophage inflammatory protein
- n, nucleocapsid
- nsp, non-structural proteins
- rdrp, rna-dependent rna polymerase
- s, spike
- sars-cov-2, severe acute respiratory syndrome-coronavirus-2
- sfv, simian foamy virus
- sop, standard operating procedures
- srv/d, simian retrovirus type d
- stlv, simian t-lymphotropic virus
- tb, tuberculosis
- tgf, transforming growth factor
- tmprss2 transmembrane serine protease 2
Collapse
Affiliation(s)
- Marcia C R Andrade
- Institute of Science and Technology on Biomodels, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil;,
| | - Bárbara R P Lemos
- Institute of Science and Technology on Biomodels, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil; Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Larissa M Silva
- Institute of Science and Technology on Biomodels, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil; University of Grande Rio, Duque de Caxias, Brazil
| | | |
Collapse
|
30
|
Hodžić A, Dheilly NM, Cabezas-Cruz A, Berry D. The helminth holobiont: a multidimensional host-parasite-microbiota interaction. Trends Parasitol 2023; 39:91-100. [PMID: 36503639 DOI: 10.1016/j.pt.2022.11.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/24/2022] [Accepted: 11/25/2022] [Indexed: 12/13/2022]
Abstract
Gastrointestinal helminths have developed multiple mechanisms by which they manipulate the host microbiome to make a favorable environment for their long-term survival. While the impact of helminth infections on vertebrate host immunity and its gut microbiota is relatively well studied, little is known about the structure and functioning of microbial populations supported by metazoan parasites. Here we argue that an integrated understanding of the helminth-associated microbiome and its role in the host disease pathogenesis may facilitate the discovery of specific microbial and/or genetic patterns critical for parasite biology and subsequently pave the way for the development of alternative control strategies against parasites and parasitic disease.
Collapse
Affiliation(s)
- Adnan Hodžić
- Centre for Microbiology and Environmental Systems Science (CMESS), Department of Microbiology and Ecosystem Science, Division of Microbial Ecology (DoME), University of Vienna, Djerassiplatz 1, 1030 Vienna, Austria.
| | - Nolwenn M Dheilly
- ANSES, INRAE, Ecole Nationale Vétérinaire d'Alfort, UMR VIROLOGIE, Laboratoire de Santé Animale, 14 rue Pierre et Marie Curie, 94706 Maisons-Alfort, France
| | - Alejandro Cabezas-Cruz
- UMR BIPAR, INRAE, ANSES, Ecole Nationale Vétérinaire d'Alfort, 14 rue Pierre et Marie Curie, 94706 Maisons-Alfort, France
| | - David Berry
- Centre for Microbiology and Environmental Systems Science (CMESS), Department of Microbiology and Ecosystem Science, Division of Microbial Ecology (DoME), University of Vienna, Djerassiplatz 1, 1030 Vienna, Austria
| |
Collapse
|
31
|
Quinteros SL, von Krusenstiern E, Snyder NW, Tanaka A, O’Brien B, Donnelly S. The helminth derived peptide FhHDM-1 redirects macrophage metabolism towards glutaminolysis to regulate the pro-inflammatory response. Front Immunol 2023; 14:1018076. [PMID: 36761766 PMCID: PMC9905698 DOI: 10.3389/fimmu.2023.1018076] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 01/09/2023] [Indexed: 01/26/2023] Open
Abstract
We have previously identified an immune modulating peptide, termed FhHDM-1, within the secretions of the liver fluke, Fasciola hepatica, which is sufficiently potent to prevent the progression of type 1 diabetes and multiple sclerosis in murine models of disease. Here, we have determined that the FhHDM-1 peptide regulates inflammation by reprogramming macrophage metabolism. Specifically, FhHDM-1 switched macrophage metabolism to a dependence on oxidative phosphorylation fuelled by fatty acids and supported by the induction of glutaminolysis. The catabolism of glutamine also resulted in an accumulation of alpha ketoglutarate (α-KG). These changes in metabolic activity were associated with a concomitant reduction in glycolytic flux, and the subsequent decrease in TNF and IL-6 production at the protein level. Interestingly, FhHDM-1 treated macrophages did not express the characteristic genes of an M2 phenotype, thereby indicating the specific regulation of inflammation, as opposed to the induction of an anti-inflammatory phenotype per se. Use of an inactive derivative of FhHDM-1, which did not modulate macrophage responses, revealed that the regulation of immune responses was dependent on the ability of FhHDM-1 to modulate lysosomal pH. These results identify a novel functional association between the lysosome and mitochondrial metabolism in macrophages, and further highlight the significant therapeutic potential of FhHDM-1 to prevent inflammation.
Collapse
Affiliation(s)
- Susel Loli Quinteros
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia
| | | | - Nathaniel W. Snyder
- Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Akane Tanaka
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia
| | - Bronwyn O’Brien
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia
| | - Sheila Donnelly
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia,*Correspondence: Sheila Donnelly,
| |
Collapse
|
32
|
The Immune Response to Nematode Infection. Int J Mol Sci 2023; 24:ijms24032283. [PMID: 36768605 PMCID: PMC9916427 DOI: 10.3390/ijms24032283] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/18/2023] [Accepted: 01/20/2023] [Indexed: 01/26/2023] Open
Abstract
Nematode infection is a major threat to the health of humans, domestic animals and wildlife. Nematodes vary in their effect on the host and in the mechanisms underlying immunity but the general features are becoming clear. There is considerable variation among individuals in resistance to infection and much of this variation is due to genetic variation in the immune response. The major histocompatibility complex has a strong influence on resistance to infection but other genes are collectively more important. Resistant individuals produce more IgA, eosinophils, IgE and mast cells than susceptible individuals and this is a consequence of stronger type 2 (Th2) immune responses. A variety of factors promote Th2 responses including genetic background, diet, molecules produced by the parasite and the location of the infection. A variety of cells and molecules including proteins, glycolipids and RNA act in concert to promote responses and to regulate the response. Nematodes themselves also modulate the host response and over 20 parasite-derived immunomodulatory molecules have been identified. Different species of nematodes modulate the immune response in different ways and probably use multiple molecules. The reasons for this are unclear and the interactions among immunomodulators have still to be investigated.
Collapse
|
33
|
Ruiz-Campillo MT, Barrero-Torres DM, Abril N, Pérez J, Zafra R, Buffoni L, Martínez-Moreno Á, Martínez-Moreno FJ, Molina-Hernández V. Fasciola hepatica primoinfections and reinfections in sheep drive distinct Th1/Th2/Treg immune responses in liver and hepatic lymph node at early and late stages. Vet Res 2023; 54:2. [PMID: 36627694 PMCID: PMC9832625 DOI: 10.1186/s13567-022-01129-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 10/24/2022] [Indexed: 01/12/2023] Open
Abstract
The expression of proinflammatory (IL-1β, IFN-γ, TNF-α) and regulatory (IL-10, TGF-β, IL-4) cytokines, as well as the transcription factor FoxP3, was quantified in the liver and hepatic lymph node (HLN) of sheep primoinfected and reinfected with Fasciola hepatica at early (4, 8 and 16 days post-infection [dpi]) and late (100 dpi) stages. The liver exerted a Th2 immune response at very early stages after the primoinfection with F. hepatica that induced the downregulation of IFN-γ, followed by a Th1/Th2/Treg response although the late stages were characterised by the expression of Th1/Th2 immune mediators. Contrarily, in reinfected sheep a robust mixed Th1/Th2/Treg immune response was found at very early stages meanwhile at late stages we observed a Th2/Treg immune response overcoming the expression of Th1 immune mediators. However, the HLN displayed a completely different Th1/Th2/Treg expression profile compared to the liver. Primoinfections with F. hepatica in HLN induced a mixed Th1/Th2/Treg environment from early stages, establishing a Th2 immune response at a late stage. However, the reinfected sheep exerted a Th2 immune response at early stages led by the IL-4 expression in opposition to the Th1/Th2/Treg found in the liver, meanwhile at late stages the HLN of reinfected sheep exerted a mixed Th1/Th2/Treg immune response. This is the first work publishing the expression of immune mediators in the liver and HLN from reinfected sheep with F. hepatica. The study of the immune responses exerted by the natural host in the target organs directly implied in the development of F. hepatica are crucial to better understand the immunopathogenesis of the fasciolosis being a key factor to develop effective vaccines.
Collapse
Affiliation(s)
- María Teresa Ruiz-Campillo
- Departamento de Anatomía y Anatomía Patológica Comparadas y Toxicología, Facultad de Veterinaria, UIC Zoonosis y Enfermedades Emergentes ENZOEM, Universidad de Córdoba, Edificio de Sanidad Animal, Campus de Rabanales, Ctra. Madrid-Cádiz Km 396, 14014, Córdoba, Spain
| | - Diana María Barrero-Torres
- Departamento de Anatomía y Anatomía Patológica Comparadas y Toxicología, Facultad de Veterinaria, UIC Zoonosis y Enfermedades Emergentes ENZOEM, Universidad de Córdoba, Edificio de Sanidad Animal, Campus de Rabanales, Ctra. Madrid-Cádiz Km 396, 14014, Córdoba, Spain
| | - Nieves Abril
- Departamento de Bioquímica y Biología Molecular, Universidad de Córdoba, Edificio Severo Ochoa, Campus de Rabanales, Ctra. Madrid-Cádiz Km 396, 14014, Córdoba, Spain
| | - José Pérez
- Departamento de Anatomía y Anatomía Patológica Comparadas y Toxicología, Facultad de Veterinaria, UIC Zoonosis y Enfermedades Emergentes ENZOEM, Universidad de Córdoba, Edificio de Sanidad Animal, Campus de Rabanales, Ctra. Madrid-Cádiz Km 396, 14014, Córdoba, Spain
| | - Rafael Zafra
- Departamento de Sanidad Animal (Parasitología), Facultad de Veterinaria, UIC Zoonosis y Enfermedades Emergentes ENZOEM, Universidad de Córdoba, Edificio de Sanidad Animal, Campus de Rabanales, Ctra. Madrid-Cádiz Km 396, 14014, Córdoba, Spain
| | - Leandro Buffoni
- Departamento de Sanidad Animal (Parasitología), Facultad de Veterinaria, UIC Zoonosis y Enfermedades Emergentes ENZOEM, Universidad de Córdoba, Edificio de Sanidad Animal, Campus de Rabanales, Ctra. Madrid-Cádiz Km 396, 14014, Córdoba, Spain
| | - Álvaro Martínez-Moreno
- Departamento de Sanidad Animal (Parasitología), Facultad de Veterinaria, UIC Zoonosis y Enfermedades Emergentes ENZOEM, Universidad de Córdoba, Edificio de Sanidad Animal, Campus de Rabanales, Ctra. Madrid-Cádiz Km 396, 14014, Córdoba, Spain
| | - Francisco Javier Martínez-Moreno
- Departamento de Sanidad Animal (Parasitología), Facultad de Veterinaria, UIC Zoonosis y Enfermedades Emergentes ENZOEM, Universidad de Córdoba, Edificio de Sanidad Animal, Campus de Rabanales, Ctra. Madrid-Cádiz Km 396, 14014, Córdoba, Spain
| | - Verónica Molina-Hernández
- Departamento de Anatomía y Anatomía Patológica Comparadas y Toxicología, Facultad de Veterinaria, UIC Zoonosis y Enfermedades Emergentes ENZOEM, Universidad de Córdoba, Edificio de Sanidad Animal, Campus de Rabanales, Ctra. Madrid-Cádiz Km 396, 14014, Córdoba, Spain.
| |
Collapse
|
34
|
Schneider-Crease IA, Feder JA, Baniel A, McCann C, Haile AA, Abebe B, Fitzgerald L, Gomery MA, Simberloff RA, Petrie ZL, Gabriel S, Dorny P, Fashing PJ, Nguyen N, Bergman TJ, Beehner JC, Snyder-Mackler N, Lu A. Urinary neopterin reflects immunological variation associated with age, helminth parasitism, and the microbiome in a wild primate. Sci Rep 2022; 12:21307. [PMID: 36494454 PMCID: PMC9734142 DOI: 10.1038/s41598-022-25298-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 11/28/2022] [Indexed: 12/13/2022] Open
Abstract
Neopterin, a product of activated white blood cells, is a marker of nonspecific inflammation that can capture variation in immune investment or disease-related immune activity and can be collected noninvasively in urine. Mounting studies in wildlife point to lifetime patterns in neopterin related to immune development, aging, and certain diseases, but rarely are studies able to assess whether neopterin can capture multiple concurrent dimensions of health and disease in a single system. We assessed the relationship between urinary neopterin stored on filter paper and multiple metrics of health and disease in wild geladas (Theropithecus gelada), primates endemic to the Ethiopian highlands. We tested whether neopterin captures age-related variation in inflammation arising from developing immunity in infancy and chronic inflammation in old age, inflammation related to intramuscular tapeworm infection, helminth-induced anti-inflammatory immunomodulation, and perturbations in the gastrointestinal microbiome. We found that neopterin had a U-shaped relationship with age, no association with larval tapeworm infection, a negative relationship with metrics related to gastrointestinal helminth infection, and a negative relationship with microbial diversity. Together with growing research on neopterin and specific diseases, our results demonstrate that urinary neopterin can be a powerful tool for assessing multiple dimensions of health and disease in wildlife.
Collapse
Affiliation(s)
- India A Schneider-Crease
- School of Human Evolution and Social Change, Arizona State University, Tempe, AZ, USA.
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ, USA.
| | - Jacob A Feder
- Interdepartmental Doctoral Program in Anthropological Sciences, Stony Brook University, Stony Brook, NY, USA
| | - Alice Baniel
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ, USA
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Colleen McCann
- Department of Mammals, Bronx Zoo, Wildlife Conservation Society, New York, NY, USA
- New York Consortium in Evolutionary Primatology, New York, NY, USA
| | | | - Belayneh Abebe
- African Wildlife Foundation, Simien Mountains Landscape Conservation and Management Project, Debark, Ethiopia
| | | | - Megan A Gomery
- Simien Mountains Gelada Research Project, Debark, Ethiopia
| | - Ruth A Simberloff
- Department of Ecology and Evolutionary Biology, University of Tennessee, Knoxville, TN, USA
| | | | - Sarah Gabriel
- Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium
| | - Pierre Dorny
- Department of Biomedical Sciences, Institute for Tropical Medicine, Antwerp, Belgium
| | - Peter J Fashing
- Department of Anthropology, California State University Fullerton, Fullerton, CA, USA
- Centre for Ecological and Evolutionary Synthesis, Department of Biosciences, University of Oslo, Oslo, Norway
| | - Nga Nguyen
- Department of Anthropology, California State University Fullerton, Fullerton, CA, USA
- Centre for Ecological and Evolutionary Synthesis, Department of Biosciences, University of Oslo, Oslo, Norway
| | - Thore J Bergman
- Department of Ecology and Evolution, University of Michigan, Ann Arbor, MI, USA
- Department of Psychology, University of Michigan, Ann Arbor, MI, USA
| | - Jacinta C Beehner
- Department of Psychology, University of Michigan, Ann Arbor, MI, USA
- Department of Anthropology, University of Michigan, Ann Arbor, MI, USA
| | - Noah Snyder-Mackler
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ, USA
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Amy Lu
- Department of Anthropology, Stony Brook University, Stony Brook, NY, USA
| |
Collapse
|
35
|
Zhao X, Wang S, Wang S, Xie J, Cui D. mTOR signaling: A pivotal player in Treg cell dysfunction in systemic lupus erythematosus. Clin Immunol 2022; 245:109153. [DOI: 10.1016/j.clim.2022.109153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/30/2022] [Accepted: 10/04/2022] [Indexed: 11/03/2022]
|
36
|
Wesołowska A. Sex—the most underappreciated variable in research: insights from helminth-infected hosts. Vet Res 2022; 53:94. [PMID: 36397174 PMCID: PMC9672581 DOI: 10.1186/s13567-022-01103-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 07/29/2022] [Indexed: 11/18/2022] Open
Abstract
The sex of a host affects the intensity, prevalence, and severity of helminth infection. In many cases, one sex has been found to be more susceptible than the other, with the prevalence and intensity of helminth infections being generally higher among male than female hosts; however, many exceptions exist. This observed sex bias in parasitism results primarily from ecological, behavioural, and physiological differences between males and females. Complex interactions between these influences modulate the risk of infection. Indeed, an interplay among sex hormones, sex chromosomes, the microbiome and the immune system significantly contributes to the generation of sex bias among helminth-infected hosts. However, sex hormones not only can modulate the course of infection but also can be exploited by the parasites, and helminths appear to have developed molecules and pathways for this purpose. Furthermore, host sex may influence the efficacy of anti-helminth vaccines; however, although little data exist regarding this sex-dependent efficacy, host sex is known to influence the response to vaccines. Despite its importance, host sex is frequently overlooked in parasitological studies. This review focuses on the key contributors to sex bias in the case of helminth infection. The precise nature of the mechanisms/factors determining these sex-specific differences generally remains largely unknown, and this represents an obstacle in the development of control methods. There is an urgent need to identify any protective elements that could be targeted in future therapies to provide optimal disease management with regard to host sex. Hence, more research is needed into the impact of host sex on immunity and protection.
Collapse
|
37
|
Cerutti A, Filipska M, Fa XM, Tachó-Piñot R. Impact of the mucosal milieu on antibody responses to allergens. J Allergy Clin Immunol 2022; 150:503-512. [PMID: 36075636 DOI: 10.1016/j.jaci.2022.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 11/30/2022]
Abstract
Respiratory and digestive mucosal surfaces are continually exposed to common environmental antigens, which include potential allergens. Although innocuous in healthy individuals, allergens cause allergy in predisposed subjects and do so by triggering a pathologic TH2 cell response that induces IgE class switching and somatic hypermutation in allergen-specific B cells. The ensuing affinity maturation and plasma cell differentiation lead to the abnormal release of high-affinity IgE that binds to powerful FcεRI receptors on basophils and mast cells. When cross-linked by allergen, FcεRI-bound IgE instigates the release of prestored and de novo-induced proinflammatory mediators. Aside from causing type I hypersensitivity reactions underlying allergy, IgE affords protection against nematodes or venoms from insects and snakes, which raises questions as to the fundamental differences between protective and pathogenic IgE responses. In this review, we discuss the impact of the mucosal environment, including the epithelial and mucus barriers, on the induction of protective IgE responses against environmental antigens. We further discuss how perturbations of these barriers may contribute to the induction of pathogenic IgE production.
Collapse
Affiliation(s)
- Andrea Cerutti
- Catalan Institute for Research and Advanced Studies, Barcelona, Spain; Institut Hospital del Mar d'Investigacions Mèdiques, Barcelona Biomedical Research Park, Barcelona, Spain; Division of Clinical Immunology, Department of Medicine, Mount Sinai School of Medicine, New York.
| | - Martyna Filipska
- Institut Hospital del Mar d'Investigacions Mèdiques, Barcelona Biomedical Research Park, Barcelona, Spain
| | - Xavi Marcos Fa
- Institut Hospital del Mar d'Investigacions Mèdiques, Barcelona Biomedical Research Park, Barcelona, Spain
| | - Roser Tachó-Piñot
- Lydia Becher Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
38
|
Investigating the Potential Effects of COVID-19 Pandemic on Intestinal Coccidian Infections. JOURNAL OF PURE AND APPLIED MICROBIOLOGY 2022. [DOI: 10.22207/jpam.16.3.51] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
New infectious agents pose a global threat to the healthcare system, and studies are conducted to estimate their health and epidemiological outcomes in the long run. The SARS-CoV-2 virus, which has caused the COVID-19 disease, was formerly assumed to be a respiratory virus; however, it can have serious systemic effects, affecting organs such as the gastrointestinal tract (GIT). Viral RNA was reported in the stool in a subset of patients, indicating another mode of transmission and diagnosis. In COVID-19, prolonged GIT symptoms, especially diarrhea, were associated with reduced diversity and richness of gut microbiota, immunological dysregulation, and delayed viral clearance. Intestinal coccidian parasites are intracellular protozoa that are most typically transmitted to humans by oocysts found in fecally contaminated food and water. Their epidemiological relevance is coupled to opportunistic infections, which cause high morbidity and mortality among immunocompromised individuals. Among immunocompetent people, intestinal coccidia is also involved in acute diarrhea, which is usually self-limiting. Evaluating the available evidence provided an opportunity to carefully consider that; the COVID-19 virus and coccidian protozoan parasites: namely, Cryptosporidium spp., Cyclospora cayetanensis, and Isospora belli, could mutually influence each other from the microbiological, clinical, diagnostic, and elimination aspects. We further systemically highlighted the possible shared pathogenesis mechanisms, transmission routes, clinical manifestations, parasite-driven immune regulation, and intestinal microbiota alteration. Finally, we showed how this might impact developing and developed countries prevention and vaccination strategies. To the best of our knowledge, there is no review that has discussed the reciprocal effect between coccidian parasites and COVID-19 coinfection.
Collapse
|
39
|
Zawistowska-Deniziak A, Lambooij JM, Kalinowska A, Patente TA, Łapiński M, van der Zande HJP, Basałaj K, de Korne CM, Chayé MAM, Gasan TA, Norbury LJ, Giera M, Zaldumbide A, Smits HH, Guigas B. Fasciola hepatica Fatty Acid Binding Protein 1 Modulates T cell Polarization by Promoting Dendritic Cell Thrombospondin-1 Secretion Without Affecting Metabolic Homeostasis in Obese Mice. Front Immunol 2022; 13:884663. [PMID: 35720355 PMCID: PMC9204345 DOI: 10.3389/fimmu.2022.884663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 04/19/2022] [Indexed: 11/24/2022] Open
Abstract
Background The parasitic trematode Fasciola hepatica evades host immune defenses through secretion of various immunomodulatory molecules. Fatty Acid Binding Proteins (fhFABPs) are among the main excreted/secreted proteins and have been shown to display anti-inflammatory properties. However, little is currently known regarding their impact on dendritic cells (DCs) and their subsequent capacity to prime specific CD4+ T cell subsets. Methodology/Principal Findings The immunomodulatory effects of both native F. hepatica extracts and recombinant fhFABPs were assessed on monocyte-derived human DCs (moDCs) and the underlying mechanism was next investigated using various approaches, including DC-allogenic T cell co-culture and DC phenotyping through transcriptomic, proteomic and FACS analyses. We mainly showed that fhFABP1 induced a tolerogenic-like phenotype in LPS-stimulated moDCs characterized by a dose-dependent increase in the cell-surface tolerogenic marker CD103 and IL-10 secretion, while DC co-stimulatory markers were not affected. A significant decrease in secretion of the pro-inflammatory cytokines IL-12p70 and IL-6 was also observed. In addition, these effects were associated with an increase in both Th2-on-Th1 ratio and IL-10 secretion by CD4+ T cells following DC-T cell co-culture. RNA sequencing and targeted proteomic analyses identified thrombospondin-1 (TSP-1) as a non-canonical factor highly expressed and secreted by fhFABP1-primed moDCs. The effect of fhFABP1 on T cell skewing was abolished when using a TSP-1 blocking antibody during DC-T cell co-culture. Immunomodulation by helminth molecules has been linked to improved metabolic homeostasis during obesity. Although fhFABP1 injection in high-fat diet-fed obese mice induced a potent Th2 immune response in adipose tissue, it did not improved insulin sensitivity or glucose homeostasis. Conclusions/Significance We show that fhFABP1 modulates T cell polarization, notably by promoting DC TSP-1 secretion in vitro, without affecting metabolic homeostasis in a mouse model of type 2 diabetes.
Collapse
Affiliation(s)
- Anna Zawistowska-Deniziak
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
- Witold Stefański Institute of Parasitology, Polish Academy of Sciences, Warsaw, Poland
- Leiden University Center for Infectious Diseases (LU-CID), Leiden, Netherlands
| | - Joost M. Lambooij
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
- Leiden University Center for Infectious Diseases (LU-CID), Leiden, Netherlands
| | - Alicja Kalinowska
- Witold Stefański Institute of Parasitology, Polish Academy of Sciences, Warsaw, Poland
| | - Thiago A. Patente
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
- Leiden University Center for Infectious Diseases (LU-CID), Leiden, Netherlands
| | - Maciej Łapiński
- International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Hendrik J. P. van der Zande
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
- Leiden University Center for Infectious Diseases (LU-CID), Leiden, Netherlands
| | - Katarzyna Basałaj
- Witold Stefański Institute of Parasitology, Polish Academy of Sciences, Warsaw, Poland
| | - Clarize M. de Korne
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
- Leiden University Center for Infectious Diseases (LU-CID), Leiden, Netherlands
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| | - Mathilde A. M. Chayé
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
- Leiden University Center for Infectious Diseases (LU-CID), Leiden, Netherlands
| | - Thomas A. Gasan
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
- Leiden University Center for Infectious Diseases (LU-CID), Leiden, Netherlands
| | - Luke J. Norbury
- Witold Stefański Institute of Parasitology, Polish Academy of Sciences, Warsaw, Poland
- School of Science, STEM College, Royal Melbourne Institute of Technology (RMIT) University, Bundoora, VIC, Australia
| | - Martin Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - Arnaud Zaldumbide
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Hermelijn H. Smits
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
- Leiden University Center for Infectious Diseases (LU-CID), Leiden, Netherlands
| | - Bruno Guigas
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
- Leiden University Center for Infectious Diseases (LU-CID), Leiden, Netherlands
| |
Collapse
|
40
|
Castañeda S, Paniz-Mondolfi A, Ramírez JD. Detangling the Crosstalk Between Ascaris, Trichuris and Gut Microbiota: What´s Next? Front Cell Infect Microbiol 2022; 12:852900. [PMID: 35694539 PMCID: PMC9174645 DOI: 10.3389/fcimb.2022.852900] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 04/21/2022] [Indexed: 11/25/2022] Open
Abstract
Helminth infections remain a global public health issue, particularly in low- and middle-income countries, where roundworms from theTrichuris and Ascaris genera are most prevalent. These geohelminths not only impact human health but most importantly also affect animal well-being, in particular the swine industry. Host-helminth parasite interactions are complex and at the same time essential to understand the biology, dynamics and pathophysiology of these infections. Within these interactions, the immunomodulatory capacity of these helminths in the host has been extensively studied. Moreover, in recent years a growing interest on how helminths interact with the intestinal microbiota of the host has sparked, highlighting how this relationship plays an essential role in the establishment of initial infection, survival and persistence of the parasite, as well as in the development of chronic infections. Identifying the changes generated by these helminths on the composition and structure of the host intestinal microbiota constitutes a field of great scientific interest, since this can provide essential and actionable information for designing effective control and therapeutic strategies. Helminths like Trichuris and Ascaris are a focus of special importance due to their high prevalence, higher reinfection rates, resistance to anthelmintic therapy and unavailability of vaccines. Therefore, characterizing interactions between these helminths and the host intestinal microbiota represents an important approach to better understand the nature of this dynamic interface and explore novel therapeutic alternatives based on management of host microbiota. Given the extraordinary impact this may have from a biological, clinical, and epidemiological public health standpoint, this review aims to provide a comprehensive overview of current knowledge and future perspectives examining the parasite-microbiota interplay and its impact on host immunity.
Collapse
Affiliation(s)
- Sergio Castañeda
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Alberto Paniz-Mondolfi
- Molecular Microbiology Laboratory, Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Juan David Ramírez
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
- Molecular Microbiology Laboratory, Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- *Correspondence: Juan David Ramírez, ;
| |
Collapse
|
41
|
Swart Z, Duong TA, Wingfield BD, Postma A, Slippers B. The relevance of studying insect-nematode interactions for human disease. Pathog Glob Health 2022; 116:140-145. [PMID: 34726122 PMCID: PMC9090338 DOI: 10.1080/20477724.2021.1996796] [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] [Indexed: 10/19/2022] Open
Abstract
Vertebrate-parasitic nematodes cause debilitating, chronic infections in millions of people worldwide. The burden of these so-called 'neglected tropical diseases' is often carried by poorer socioeconomic communities in part because research on parasitic nematodes and their vertebrate hosts is challenging and costly. However, complex biological and pathological processes can be modeled in simpler organisms. Here, we consider how insight into the interactions between entomopathogenic nematodes (EPN), their insect hosts and bacterial symbionts may reveal novel treatment targets for parasitic nematode infections. We argue that a combination of approaches that target nematodes, as well as the interaction of pathogens with insect vectors and bacterial symbionts, offer potentially effective, but underexplored opportunities.
Collapse
Affiliation(s)
- Zorada Swart
- Department of Biochemistry Genetics and Microbiology, Forestry and Agricultural Biotechnology Institute, University of Pretoria, South Africa
| | - Tuan A. Duong
- Department of Biochemistry Genetics and Microbiology, Forestry and Agricultural Biotechnology Institute, University of Pretoria, South Africa
| | - Brenda D. Wingfield
- Department of Biochemistry Genetics and Microbiology, Forestry and Agricultural Biotechnology Institute, University of Pretoria, South Africa
| | - Alisa Postma
- Department of Biochemistry Genetics and Microbiology, Forestry and Agricultural Biotechnology Institute, University of Pretoria, South Africa
| | - Bernard Slippers
- Department of Biochemistry Genetics and Microbiology, Forestry and Agricultural Biotechnology Institute, University of Pretoria, South Africa
| |
Collapse
|
42
|
Mukundan A, Byeon CH, Hinck CS, Cunningham K, Campion T, Smyth DJ, Maizels RM, Hinck AP. Convergent evolution of a parasite-encoded complement control protein-scaffold to mimic binding of mammalian TGF-β to its receptors, TβRI and TβRII. J Biol Chem 2022; 298:101994. [PMID: 35500648 PMCID: PMC9163516 DOI: 10.1016/j.jbc.2022.101994] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 04/22/2022] [Accepted: 04/23/2022] [Indexed: 11/02/2022] Open
Abstract
The mouse intestinal helminth Heligmosomoides polygyrus modulates host immune responses by secreting a transforming growth factor (TGF)-β mimic (TGM), to expand the population of Foxp3+ Tregs. TGM comprises five complement control protein (CCP)-like domains, designated D1-D5. Though lacking homology to TGF-β, TGM binds directly to the TGF-β receptors TβRI and TβRII and stimulates the differentiation of naïve T-cells into Tregs. However, the molecular determinants of binding are unclear. Here, we used surface plasmon resonance, isothermal calorimetry, NMR spectroscopy, and mutagenesis to investigate how TGM binds the TGF-β receptors. We demonstrate that binding is modular, with D1-D2 binding to TβRI and D3 binding to TβRII. D1-D2 and D3 were further shown to compete with TGF-β(TβRII)2 and TGF-β for binding to TβRI and TβRII, respectively. The solution structure of TGM-D3 revealed that TGM adopts a CCP-like fold but is also modified to allow the C-terminal strand to diverge, leading to an expansion of the domain and opening potential interaction surfaces. TGM-D3 also incorporates a long structurally ordered hypervariable loop, adding further potential interaction sites. Through NMR shift perturbations and binding studies of TGM-D3 and TβRII variants, TGM-D3 was shown to occupy the same site of TβRII as bound by TGF-β using both a novel interaction surface and the hypervariable loop. These results, together with the identification of other secreted CCP-like proteins with immunomodulatory activity in H. polygyrus, suggest that TGM is part of a larger family of evolutionarily plastic parasite effector molecules that mediate novel interactions with their host.
Collapse
Affiliation(s)
- Ananya Mukundan
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania USA
| | - Chang-Hyeock Byeon
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania USA
| | - Cynthia S Hinck
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania USA
| | - Kyle Cunningham
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Tiffany Campion
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Danielle J Smyth
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Rick M Maizels
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Andrew P Hinck
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania USA.
| |
Collapse
|
43
|
Localization and RNA Interference-Driven Inhibition of a Brugia malayi-Encoded Interleukin-5 Receptor Binding Protein. Infect Immun 2022; 90:e0031721. [PMID: 35467360 DOI: 10.1128/iai.00317-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A molecule we termed Brugia malayi IL-5 receptor (IL-5R) binding protein (BmIL5Rbp; also known as Bm8757) was identified from B. malayi filarial worms and found to inhibit human interleukin-5 (IL-5) binding to its human receptor competitively. After the expression and purification of a recombinant BmIL5Rbp and generation of BmIL5Rbp-specific rabbit antibody, we localized the molecule on B. malayi worms through immunohistochemistry and immunoelectron microscopy. RNA interference (RNAi) was used to inhibit BmIL5Rbp mRNA and protein production. BmIL5Rbp was shown to localize to the cuticle of Brugia malayi and to be released in its excretory/secretory products. RNAi inhibited BmIL5Rbp mRNA production by 33%, reduced the surface protein expression by ~50%, and suppressed the release of BmIL5Rbp in the excretory/secretory products. RNAi has been used successfully to knock down the mRNA and protein expression of BmIL5Rbp in the early larval stages of B. malayi and provided a proof of principle for the local inhibition of the human IL-5R. These findings provide evidence that a parasite-encoded IL-5R antagonist may locally inhibit a vital host innate immune activation of IL-5 on eosinophils.
Collapse
|
44
|
Kaur R, Arora N, Nair MG, Prasad A. The interplay of helminthic neuropeptides and proteases in parasite survival and host immunomodulation. Biochem Soc Trans 2022; 50:107-118. [PMID: 35076687 PMCID: PMC9042389 DOI: 10.1042/bst20210405] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 12/22/2021] [Accepted: 12/23/2021] [Indexed: 11/17/2022]
Abstract
Neuropeptides comprise a diverse and broad group of neurotransmitters in vertebrates and invertebrates, with critical roles in neuronal signal transduction. While their role in controlling learning and memory in the brains of mammals is known, their extra-synaptic function in infection and inflammation with effects on distinct tissues and immune cells is increasingly recognized. Helminth infections especially of the central nervous system (CNS), such as neurocysticercosis, induce neuropeptide production by both host and helminth, but their role in host-parasite interplay or host inflammatory response is unclear. Here, we review the neurobiology of helminths, and discuss recent studies on neuropeptide synthesis and function in the helminth as well as the host CNS and immune system. Neuropeptides are summarized according to structure and function, and we discuss the complex enzyme processing for mature neuropeptides, focusing on helminth enzymes as potential targets for novel anthelminthics. We next describe known immunomodulatory effects of mammalian neuropeptides discovered from mouse infection models and draw functional parallels with helminth neuropeptides. Last, we discuss the anti-microbial properties of neuropeptides, and how they may be involved in host-microbiota changes in helminth infection. Overall, a better understanding of the biology of helminth neuropeptides, and whether they affect infection outcomes could provide diagnostic and therapeutic opportunities for helminth infections.
Collapse
Affiliation(s)
- Rimanpreet Kaur
- School of Basic Sciences, Indian Institute of Technology Mandi, Mandi, Himachal Pradesh 175005, India
| | - Naina Arora
- School of Basic Sciences, Indian Institute of Technology Mandi, Mandi, Himachal Pradesh 175005, India
| | - Meera G. Nair
- Division of Biomedical Sciences, University of California Riverside, Riverside, CA 92521, U.S.A
| | - Amit Prasad
- School of Basic Sciences, Indian Institute of Technology Mandi, Mandi, Himachal Pradesh 175005, India
| |
Collapse
|
45
|
Dai M, Yang X, Yu Y, Pan W. Helminth and Host Crosstalk: New Insight Into Treatment of Obesity and Its Associated Metabolic Syndromes. Front Immunol 2022; 13:827486. [PMID: 35281054 PMCID: PMC8913526 DOI: 10.3389/fimmu.2022.827486] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 02/02/2022] [Indexed: 12/16/2022] Open
Abstract
Obesity and its associated Metabolic Syndromes (Mets) represent a global epidemic health problem. Metabolic inflammation, lipid accumulation and insulin resistance contribute to the progression of these diseases, thereby becoming targets for drug development. Epidemiological data have showed that the rate of helminth infection negatively correlates with the incidence of obesity and Mets. Correspondingly, numerous animal experiments and a few of clinic trials in human demonstrate that helminth infection or its derived molecules can mitigate obesity and Mets via induction of macrophage M2 polarization, inhibition of adipogenesis, promotion of fat browning, and improvement of glucose tolerance, insulin resistance and metabolic inflammation. Interestingly, sporadic studies also uncover that several helminth infections can reshape gut microbiota of hosts, which is intimately implicated in the pathogenesis of obesity and Mets. Overall, these findings indicate that the crosstalk between helminth and hosts may be a novel direction for obesity and Mets therapy. The present article reviews the molecular mechanism of how helminth masters immunity and metabolism in obesity.
Collapse
Affiliation(s)
- Mengyu Dai
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
- The Second Clinical Medicine, Xuzhou Medical University, Xuzhou, China
- National Demonstration Center for Experimental Basic Medical Science Education (Xuzhou Medical University), Xuzhou, China
| | - Xiaoying Yang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| | - Yinghua Yu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
- *Correspondence: Wei Pan, ; Yinghua Yu,
| | - Wei Pan
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
- *Correspondence: Wei Pan, ; Yinghua Yu,
| |
Collapse
|
46
|
Abstract
Viral infections are often studied in model mammalian organisms under specific pathogen-free conditions. However, in nature, coinfections are common, and infection with one organism can alter host susceptibility to infection with another. Helminth parasites share a long coevolutionary history with mammalian hosts and have shaped host physiology, metabolism, immunity, and the composition of the microbiome. Published studies suggest that helminth infection can either be beneficial or detrimental during viral infection. Here, we discuss coinfection studies in mouse models and use them to define key determinants that impact outcomes, including the type of antiviral immunity, the tissue tropism of both the helminth and the virus, and the timing of viral infection in relation to the helminth lifecycle. We also explore the current mechanistic understanding of how helminth-virus coinfection impacts host immunity and viral pathogenesis. While much attention has been placed on the impact of the gut bacterial microbiome on immunity to infection, we suggest that enteric helminths, as a part of the eukaryotic macrobiome, also represent an important modulator of disease pathogenesis and severity following virus infection.
Collapse
Affiliation(s)
- Pritesh Desai
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, United States
| | - Michael S. Diamond
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, United States,Pathology & Immunology, Washington University School of Medicine, Saint Louis, MO, United States,Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO, United States,The Andrew M. And Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO, United States
| | - Larissa B. Thackray
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, United States,CONTACT Larissa B. Thackray Department of Medicine, Washington University School of Medicine, Saint Louis, MO63110, United States
| |
Collapse
|
47
|
Schneider-Crease IA, Blackwell AD, Kraft TS, Emery Thompson M, Maldonado Suarez I, Cummings DK, Stieglitz J, Snyder-Mackler N, Gurven M, Kaplan H, Trumble BC. Helminth infection is associated with dampened cytokine responses to viral and bacterial stimulations in Tsimane forager-horticulturalists. Evol Med Public Health 2021; 9:349-359. [PMID: 34868595 PMCID: PMC8634526 DOI: 10.1093/emph/eoab035] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 10/19/2021] [Indexed: 02/07/2023] Open
Abstract
Background Soil-transmitted helminths (STHs) and humans share long co-evolutionary histories over which STHs have evolved strategies to permit their persistence by downregulating host immunity. Understanding the interactions between STHs and other pathogens can inform our understanding of human evolution and contemporary disease patterns. Methodology We worked with Tsimane forager-horticulturalists in the Bolivian Amazon, where STHs are prevalent. We tested whether STHs and eosinophil levels—likely indicative of infection in this population—are associated with dampened immune responses to in vitro stimulation with H1N1 and lipopolysaccharide (LPS) antigens. Whole blood samples (n = 179) were treated with H1N1 vaccine and LPS and assayed for 13 cytokines (INF-γ, IL-1β, IL-2, IL-4, IL-5, IL-6, IL-7, IL-8, IL-10, IL-12p70, IL-13, GM-CSF and TNF-ɑ). We evaluated how STHs and eosinophil levels affected cytokine responses and T helper (Th) 1 and Th2-cytokine suite responses to stimulation. Results Infection with Ascaris lumbricoides was significantly (P ≤ 0.05) associated with lower response of some cytokines to H1N1 and LPS in women. Eosinophils were significantly negatively associated with some cytokine responses to H1N1 and LPS, with the strongest effects in women, and associated with a reduced Th1- and Th2-cytokine response to H1N1 and LPS in women and men. Conclusions and implications Consistent with the ‘old friends’ and hygiene hypotheses, we find that STHs were associated with dampened cytokine responses to certain viral and bacterial antigens. This suggests that STH infections may play an essential role in immune response regulation and that the lack of STH immune priming in industrialized populations may increase the risk of over-reactive immunity. Lay Summary: Indicators of helminth infection were associated with dampened cytokine immune responses to in vitro stimulation with viral and bacterial antigens in Tsimane forager-horticulturalists in the Bolivian Amazon, consistent with the ‘old friends’ and hygiene hypotheses.
Collapse
Affiliation(s)
| | - Aaron D Blackwell
- Department of Anthropology, Washington State University, Pullman, WA, USA
| | - Thomas S Kraft
- Department of Anthropology, University of California Santa Barbara, Santa Barbara, CA, USA
| | | | | | | | | | - Noah Snyder-Mackler
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ, USA.,School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Michael Gurven
- Department of Anthropology, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Hillard Kaplan
- Economic Science Institute, Chapman University, Orange, CA, USA
| | - Benjamin C Trumble
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ, USA.,School of Life Sciences, Arizona State University, Tempe, AZ, USA.,School of Human Evolution and Social Change, Arizona State University, Tempe, AZ, USA
| |
Collapse
|
48
|
The yin and yang of human soil-transmitted helminth infections. Int J Parasitol 2021; 51:1243-1253. [PMID: 34774540 PMCID: PMC9145206 DOI: 10.1016/j.ijpara.2021.11.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/01/2021] [Accepted: 11/02/2021] [Indexed: 12/22/2022]
Abstract
The major soil-transmitted helminths that infect humans are the roundworms, whipworms and hookworms. Soil-transmitted helminth infections rank among the most important neglected tropical diseases in terms of morbidity, and almost one billion people are still infected with at least one species. While anthelmintic drugs are available, they do not offer long term protection against reinfection, precipitating the need for vaccines that provide long-term immunologic defense. Vaccine discovery and development is in advanced clinical development for hookworm infection, with a bivalent human hookworm vaccine in clinical trials in Brazil and Africa, but is in its infancy for both roundworm (ascariasis) and whipworm (trichuriasis) infections. One of the greatest hurdles to developing soil-transmitted helminth vaccines is the potent immunoregulatory properties of these helminths, creating a barrier to the induction of meaningful long-term protective immunity. While challenging for vaccinologists, this phenomenon presents unique opportunities to develop an entirely new class of anti-inflammatory drugs that capitalise on these immunomodulatory strategies. Epidemiologic studies and clinical trials employing experimental soil-transmitted helminth challenge models, when coupled with findings from animal models, show that at least some soil-transmitted helminth-derived molecules can protect against the onset of autoimmune, allergic and metabolic disorders, and several natural products with the desired bioactivity have been isolated and tested in pre-clinical settings. The yin and yang of soil-transmitted helminth infections reflect both the urgency for effective vaccines and the potential for new immunoregulatory molecules from parasite products.
Collapse
|
49
|
Gao YR, Sun XZ, Li R, Tang CL, Zhang RH, Zhu YW, Li XR, Pan Q. The effect of regulatory T cells in Schistosoma-mediated protection against type 2 diabetes. Acta Trop 2021; 224:106073. [PMID: 34487719 DOI: 10.1016/j.actatropica.2021.106073] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 07/22/2021] [Accepted: 07/26/2021] [Indexed: 12/28/2022]
Abstract
In western societies, the prevalence of type 2 diabetes (T2D) is related to the hygiene hypothesis, which implies that reduced exposure to infectious factors results in a loss of the immune stimulation necessary to form the immune system during development. In fact, it has been reported that parasites, such as Schistosoma, can improve or prevent the development of T2D, which may be related to the activity of immune cells, including regulatory T cells (Tregs). Hence, Schistosoma, Tregs, and T2D share a close relationship. Schistosoma infection and the molecules released can lead to an increase in Tregs, which play an important role in the suppression of T2D. In this review, we provide an overview of the role of Tregs in the response to Schistosoma infection and the protective mechanism of Schistosoma-related molecular products against T2D.
Collapse
Affiliation(s)
- Yan-Ru Gao
- Medical Department, Wuhan City College, Wuhan, 430083, China
| | - Xue-Zhi Sun
- Wuhan Pulmonary Hospital, Wuhan Institute for Tuberculosis Control, Wuhan 430030, China
| | - Ru Li
- Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Wuhan 430063, China
| | - Chun-Lian Tang
- Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Wuhan 430063, China
| | - Rong-Hui Zhang
- Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Wuhan 430063, China
| | - Ya-Wen Zhu
- Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Wuhan 430063, China
| | - Xiu-Rong Li
- Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Wuhan 430063, China.
| | - Qun Pan
- Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Wuhan 430063, China.
| |
Collapse
|
50
|
Drurey C, Maizels RM. Helminth extracellular vesicles: Interactions with the host immune system. Mol Immunol 2021; 137:124-133. [PMID: 34246032 PMCID: PMC8636279 DOI: 10.1016/j.molimm.2021.06.017] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 06/14/2021] [Accepted: 06/23/2021] [Indexed: 12/21/2022]
Abstract
As long-lived parasites, helminths depend upon immunomodulation of their hosts for survival. The release of excretory-secretory (ES) products, including proteins, lipids and RNAs is how successful host manipulation is achieved. It has recently been discovered that the ES products of helminths contain extracellular vesicles (EVs), with every species investigated found to secrete these lipid-bound structures. EVs are perfect for packaging and delivering immune modulators to target cell types. This review outlines the research carried out on helminth EVs and their constituents thus far, as well as their interaction with components of the mammalian immune system. We discuss how targeting EVs will aid treatment of helminth infection and consider how EVs and their immunomodulatory cargo could be used as therapeutics as we progress through this exciting era.
Collapse
Affiliation(s)
- Claire Drurey
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, UK
| | - Rick M Maizels
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, UK.
| |
Collapse
|