1
|
Burton AM, Else KJ, Irving J, Mair I, Shultz S. Antibodies and Inflammation: Fecal Biomarkers of Gut Health in Domestic Ruminants. JOURNAL OF EXPERIMENTAL ZOOLOGY. PART A, ECOLOGICAL AND INTEGRATIVE PHYSIOLOGY 2025. [PMID: 39840509 DOI: 10.1002/jez.2896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 12/08/2024] [Accepted: 12/22/2024] [Indexed: 01/23/2025]
Abstract
Gastrointestinal infections present major challenges to ruminant livestock systems, and gut health is a key constraint on fitness, welfare, and productivity. Fecal biomarkers present opportunities to monitor animal health without using invasive methods, and with greater resolution compared to observational metrics. Here we developed enzyme-linked immunosorbent assays for three potential fecal biomarkers of gut health in domestic ruminants: two immunological (total immunoglobulin [Ig]A and total IgG) and one inflammatory (lactoferrin). We analytically validated the assays, then evaluated whether they could be used as a biomarker of clinically diagnosed gastrointestinal pathologies in cattle (Bos taurus), and finally compared them with helminth fecal egg counts in sheep (Ovis aries). The analytes were detected above the lower limits of detection in cattle, sheep, and goats. Fecal IgA and lactoferrin were higher in cattle with infectious pathologies (strongyles, coccidiosis and symptomatic Johne's disease) compared to healthy controls. Lactoferrin was additionally higher in animals with infectious pathologies compared to noninfectious pathologies, and to asymptomatic Johne's cases. No significant relationships were found with sheep fecal egg counts. These initial findings suggest that fecal IgA and lactoferrin may be useful biomarkers of poor gastrointestinal health in cattle, and that fecal lactoferrin is specific to active inflammation caused by infectious agents. These could be incorporated into the growing suite of noninvasive ecoimmunological tools and used to understand ruminant gut health in a range of species. Applications include improving treatment regimens for gastrointestinal infections, and understanding wildlife physiological responses to infectious challenges.
Collapse
Affiliation(s)
- A M Burton
- Department of Earth and Environmental Science, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, Manchester, UK
| | - K J Else
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - J Irving
- Department of Earth and Environmental Science, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, Manchester, UK
| | - I Mair
- Institute of Ecology and Evolution, Institute of Immunology and Infection Research, School of Biological Sciences, The University of Edinburgh, Edinburgh, UK
| | - S Shultz
- Department of Earth and Environmental Science, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, Manchester, UK
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| |
Collapse
|
2
|
Johnson H, Banakis S, Chung M, Ghedin E, Voronin D. MicroRNAs secreted by the parasitic nematode Brugia malayi disrupt lymphatic endothelial cell integrity. PLoS Negl Trop Dis 2024; 18:e0012803. [PMID: 39739969 PMCID: PMC11706539 DOI: 10.1371/journal.pntd.0012803] [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: 08/15/2024] [Revised: 01/07/2025] [Accepted: 12/20/2024] [Indexed: 01/02/2025] Open
Abstract
Lymphatic filariasis (LF) is a neglected tropical disease affecting over 51 million people in 72 endemic countries. Causative agents of LF are mosquito-borne parasitic nematodes Wuchereria bancrofti, Brugia malayi, and Brugia timori. The adult parasites impact the integrity of lymphatic vessels and damage valves, leading to a remodeling of the lymphatic system and lymphatic dilation. Chronic infections can develop into severe clinical manifestations, primarily lymphedema, hydrocoele, and elephantiasis. Mechanistic studies on the underlying pathology due to the parasite are necessary to better manage human filariasis. Since parasite molecules, such as microRNAs (miRNAs), can be found in secreted extracellular vesicles (EVs) and are transported between parasite and host cells, we hypothesized that these could also play a role in the development of pathology in LF. In this study, we tested two B. malayi miRNAs previously detected in vitro in the culture media of microfilarial stages of worms. While one is Brugia-specific (bma-miR-5864) and the other nematode-specific (bma-miR-86), both miRNAs are secreted in high abundance. We first examined the in vitro response by transcriptomic profiling of human lymphatic endothelial cells to treatment with these miRNAs, which allowed us to identify genes involved in maintaining the integrity of the lymphatic endothelium. We then measured the effect of these miRNAs on the regulation of proteins necessary for cell integrity, demonstrating downregulation leading to a significant increase in the permeability of the endothelium monolayer. With this study we identify parasite miRNAs involved in undermining the integrity of endothelial cells, thus potentially contributing to the development of pathology. These findings could pave the way for a novel treatment strategy where the inhibition of parasite-secreted molecules could slow the progression of LF pathology. From a broader perspective, the miRNAs secreted by filarial parasites could potentially be used in the future for diagnosing and monitoring disease progression or treatment efficacy.
Collapse
Affiliation(s)
- Hailey Johnson
- Systems Genomics Section, Laboratory of Parasitic Diseases, Division of Intramural Research, NIAID, NIH, Bethesda, Maryland, United States of America
| | - Stephanie Banakis
- Systems Genomics Section, Laboratory of Parasitic Diseases, Division of Intramural Research, NIAID, NIH, Bethesda, Maryland, United States of America
| | - Matthew Chung
- Systems Genomics Section, Laboratory of Parasitic Diseases, Division of Intramural Research, NIAID, NIH, Bethesda, Maryland, United States of America
| | - Elodie Ghedin
- Systems Genomics Section, Laboratory of Parasitic Diseases, Division of Intramural Research, NIAID, NIH, Bethesda, Maryland, United States of America
| | - Denis Voronin
- Systems Genomics Section, Laboratory of Parasitic Diseases, Division of Intramural Research, NIAID, NIH, Bethesda, Maryland, United States of America
| |
Collapse
|
3
|
Flores-Sotelo I, Juárez N, González MI, Chávez A, Vannan DT, Eksteen B, Terrazas LI, Reyes JL. Endogenous innate sensor NLRP3 is a key component in peritoneal macrophage dynamics required for cestode establishment. Immunol Res 2024; 72:948-963. [PMID: 38842647 PMCID: PMC11564225 DOI: 10.1007/s12026-024-09496-3] [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: 11/23/2023] [Accepted: 05/21/2024] [Indexed: 06/07/2024]
Abstract
The NLRP3 receptor can assemble inflammasome platforms to trigger inflammatory responses; however, accumulating evidence suggests that it can also display anti-inflammatory properties. Here, we explored the role of nucleotide-binding oligomerization domain pyrin-containing protein 3 (NLRP3) in Taenia crassiceps experimental infection, which requires immune polarization into a Th2-type profile and peritoneal influx of suppressive macrophages for successful colonization. NLRP3 deficient mice (NLRP3-/-) were highly resistant against T. crassiceps, relative to wild-type (WT) mice. Resistance in NLRP3-/- mice was associated with a diminished IL-4 output, high levels of IL-15, growth factor for both innate and adaptive lymphocytes, and a dramatic decrease in peritoneum-infiltrating suppressive macrophages. Also, a transcriptional analysis on bone marrow-derived macrophages exposed to Taenia-secreted antigens and IL-4 revealed that NLRP3-/- macrophages express reduced transcripts of relm-α and PD-1 ligands, markers of alternative activation and suppressive ability, respectively. Finally, we found that the resistance displayed by NLRP3-/- mice is transferred through intestinal microbiota exchange, since WT mice co-housed with NLRP3-/- mice were significantly more resistant than WT animals preserving their native microbiota. Altogether, these data demonstrate that NLRP3 is a component of innate immunity required for T. crassiceps to establish, most likely contributing to macrophage recruitment, and controlling lymphocyte-stimulating cytokines such as IL-15.
Collapse
Affiliation(s)
- Irán Flores-Sotelo
- Laboratorio de Inmunología Experimental y Regulación de la Inflamación Hepato-Intestinal, UBIMED, FES, Iztacala, UNAM, Tlalnepantla de Baz, Estado de México, Mexico
| | - Natalia Juárez
- Laboratorio de Inmunología Experimental y Regulación de la Inflamación Hepato-Intestinal, UBIMED, FES, Iztacala, UNAM, Tlalnepantla de Baz, Estado de México, Mexico
| | - Marisol I González
- Laboratorio de Inmunología Experimental y Regulación de la Inflamación Hepato-Intestinal, UBIMED, FES, Iztacala, UNAM, Tlalnepantla de Baz, Estado de México, Mexico
| | - Auraamellaly Chávez
- Laboratorio de Inmunología Experimental y Regulación de la Inflamación Hepato-Intestinal, UBIMED, FES, Iztacala, UNAM, Tlalnepantla de Baz, Estado de México, Mexico
| | - Danielle T Vannan
- Boston Scientific Corporation, Urology Division, 200 Boston Scientific Way, Marlborough, MA, USA
| | | | - Luis I Terrazas
- Laboratorio de Inmunoparasitología, UBIMED, FES Iztacala, UNAM, Tlalnepantla de Baz, Estado de México, Mexico
| | - José L Reyes
- Laboratorio de Inmunología Experimental y Regulación de la Inflamación Hepato-Intestinal, UBIMED, FES, Iztacala, UNAM, Tlalnepantla de Baz, Estado de México, Mexico.
| |
Collapse
|
4
|
Schreiber F, Balas I, Robinson MJ, Bakdash G. Border Control: The Role of the Microbiome in Regulating Epithelial Barrier Function. Cells 2024; 13:477. [PMID: 38534321 PMCID: PMC10969408 DOI: 10.3390/cells13060477] [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: 02/16/2024] [Revised: 03/01/2024] [Accepted: 03/03/2024] [Indexed: 03/28/2024] Open
Abstract
The gut mucosal epithelium is one of the largest organs in the body and plays a critical role in regulating the crosstalk between the resident microbiome and the host. To this effect, the tight control of what is permitted through this barrier is of high importance. There should be restricted passage of harmful microorganisms and antigens while at the same time allowing the absorption of nutrients and water. An increased gut permeability, or "leaky gut", has been associated with a variety of diseases ranging from infections, metabolic diseases, and inflammatory and autoimmune diseases to neurological conditions. Several factors can affect gut permeability, including cytokines, dietary components, and the gut microbiome. Here, we discuss how the gut microbiome impacts the permeability of the gut epithelial barrier and how this can be harnessed for therapeutic purposes.
Collapse
Affiliation(s)
| | | | | | - Ghaith Bakdash
- Microbiotica Ltd., Cambridge CB10 1XL, UK; (F.S.); (I.B.); (M.J.R.)
| |
Collapse
|
5
|
Han LL, Lu QQ, Zheng WW, Li YL, Song YY, Zhang XZ, Long SR, Liu RD, Wang ZQ, Cui J. A novel trypsin of Trichinella spiralis mediates larval invasion of gut epithelium via binding to PAR2 and activating ERK1/2 pathway. PLoS Negl Trop Dis 2024; 18:e0011874. [PMID: 38166153 PMCID: PMC10786404 DOI: 10.1371/journal.pntd.0011874] [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: 10/03/2023] [Revised: 01/12/2024] [Accepted: 12/19/2023] [Indexed: 01/04/2024] Open
Abstract
BACKGROUND Proteases secreted by Trichinella spiralis intestinal infective larvae (IIL) play an important role in larval invasion and pathogenesis. However, the mechanism through which proteases mediate larval invasion of intestinal epithelial cells (IECs) remains unclear. A novel T. spiralis trypsin (TsTryp) was identified in IIL excretory/secretory (ES) proteins. It was an early and highly expressed protease at IIL stage, and had the potential as an early diagnostic antigen. The aim of this study was to investigate the biological characteristics of this novel TsTryp, its role in larval invasion of gut epithelium, and the mechanisms involved. METHODOLOGY/PRINCIPAL FINDING TsTryp with C-terminal domain was cloned and expressed in Escherichia coli BL21 (DE3), and the rTsTryp had the enzymatic activity of natural trypsin, but it could not directly degrade gut tight junctions (TJs) proteins. qPCR and western blotting showed that TsTryp was highly expressed at the invasive IIL stage. Immunofluorescence assay (IFA), ELISA and Far Western blotting revealed that rTsTryp specifically bound to IECs, and confocal microscopy showed that the binding of rTsTryp with IECs was mainly localized in the cytomembrane. Co-immunoprecipitation (Co-IP) confirmed that rTsTryp bound to protease activated receptors 2 (PAR2) in Caco-2 cells. rTsTryp binding to PAR2 resulted in decreased expression levels of ZO-1 and occludin and increased paracellular permeability in Caco-2 monolayers by activating the extracellular regulated protein kinases 1/2 (ERK1/2) pathway. rTsTryp decreased TJs expression and increased epithelial permeability, which could be abrogated by the PAR2 antagonist AZ3451 and ERK1/2 inhibitor PD98059. rTsTryp facilitated larval invasion of IECs, and anti-rTsTryp antibodies inhibited invasion. Both inhibitors impeded larval invasion and alleviated intestinal inflammation in vitro and in vivo. CONCLUSIONS TsTryp binding to PAR2 activated the ERK1/2 pathway, decreased the expression of gut TJs proteins, disrupted epithelial integrity and barrier function, and consequently mediated larval invasion of the gut mucosa. Therefore, rTsTryp could be regarded as a potential vaccine target for blocking T. spiralis invasion and infection.
Collapse
Affiliation(s)
- Lu Lu Han
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, China
| | - Qi Qi Lu
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, China
| | - Wen Wen Zheng
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, China
| | - Yang Li Li
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, China
| | - Yan Yan Song
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, China
| | - Xin Zhuo Zhang
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, China
| | - Shao Rong Long
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, China
| | - Ruo Dan Liu
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, China
| | - Zhong Quan Wang
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, China
| | - Jing Cui
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
6
|
Maizels RM, Gause WC. Targeting helminths: The expanding world of type 2 immune effector mechanisms. J Exp Med 2023; 220:e20221381. [PMID: 37638887 PMCID: PMC10460967 DOI: 10.1084/jem.20221381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 06/24/2023] [Accepted: 08/09/2023] [Indexed: 08/29/2023] Open
Abstract
In this new review, Rick Maizels and Bill Gause summarize how type 2 immune responses combat helminth parasites through novel mechanisms, coordinating multiple innate and adaptive cell and molecular players that can eliminate infection and repair-resultant tissue damage.
Collapse
Affiliation(s)
- Rick M. Maizels
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - William C. Gause
- Center for Immunity and Inflammation, Rutgers Biomedical Health Sciences Institute for Infectious and Inflammatory Diseases, New Jersey Medical School, Rutgers Biomedical Health Sciences, Newark, NJ, USA
| |
Collapse
|
7
|
Liang W, Feng Y, Yang D, Qin J, Zhi X, Wu W, Jie Q. Oral probiotics increased the proportion of Treg, Tfr, and Breg cells to inhibit the inflammatory response and impede gestational diabetes mellitus. Mol Med 2023; 29:122. [PMID: 37684563 PMCID: PMC10492300 DOI: 10.1186/s10020-023-00716-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 08/14/2023] [Indexed: 09/10/2023] Open
Abstract
BACKGROUND Children of mothers with gestational diabetes mellitus (GDM) are more prone to acquire type 2 diabetes and obesity as adults. Due to this link, early intervention strategies that alter the gut microbiome may benefit the mother and kid long-term. This work uses metagenomic and transcriptome sequencing to investigate how probiotics affect gut microbiota dysbiosis and inflammation in GDM. METHODS GDM and control metagenomic sequencing data were obtained from the SRA database. This metagenomic data helped us understand gut microbiota abundance and function. KEGG detected and extracted functional pathway genes. Transcriptome sequencing data evaluated GDM-related gene expression. Finally, GDM animal models were given probiotics orally to evaluate inflammatory response, regulatory immune cell fractions, and leptin protein levels. RESULTS GDM patients had more Fusobacteria and Firmicutes, while healthy people had more Bacteroidetes. Gut microbiota composition may affect GDM by altering the L-aspartate and L-asparagine super pathways. Mannan degradation and the super pathway of L-aspartate and L-asparagine synthesis enhanced in GDM mice with leptin protein overexpression. Oral probiotics prevent GDM by lowering leptin. Oral probiotics increased Treg, Tfr, and Breg cells, which decreased TNF-α and IL-6 and increased TGF-β and IL-10, preventing inflammation and preserving mouse pregnancy. CONCLUSION Dysbiosis of the gut microbiota may increase leptin expression and cause GDM. Oral probiotics enhance Treg, Tfr, and Breg cells, which limit the inflammatory response and assist mice in sustaining normal pregnancy. Thus, oral probiotics may prevent GDM, enabling targeted gut microbiota modulation and maternal and fetal health.
Collapse
Affiliation(s)
- Weijie Liang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, People's Republic of China
- Department of Geriatric Endocrinology, Guangdong Provincial Geriatrics Institute (East Zone), Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, No. 3, Chanchugang, Zhongshan 2nd Road, Yuexiu District, Guangzhou, 510080, Guangdong Province, People's Republic of China
- Department of Cardiology, Panyu Central Hospital, Cardiovascular Institute of Panyu District, No. 8, Fuyu East Road, Qiaonan Street, Panyu District, Guangzhou, 511400, Guangdong Province, People's Republic of China
| | - Yuanyi Feng
- Department of Geriatrics, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital), Shenzhen, 518052, People's Republic of China
| | - Dongmei Yang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, People's Republic of China
- Department of Geriatric Endocrinology, Guangdong Provincial Geriatrics Institute (East Zone), Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, No. 3, Chanchugang, Zhongshan 2nd Road, Yuexiu District, Guangzhou, 510080, Guangdong Province, People's Republic of China
| | - Jiajun Qin
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, People's Republic of China
- Department of Geriatric Endocrinology, Guangdong Provincial Geriatrics Institute (East Zone), Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, No. 3, Chanchugang, Zhongshan 2nd Road, Yuexiu District, Guangzhou, 510080, Guangdong Province, People's Republic of China
| | - Ximei Zhi
- Department of Geriatric Endocrinology, Guangdong Provincial Geriatrics Institute (East Zone), Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, No. 3, Chanchugang, Zhongshan 2nd Road, Yuexiu District, Guangzhou, 510080, Guangdong Province, People's Republic of China
| | - Wen Wu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, People's Republic of China.
- Department of Geriatric Endocrinology, Guangdong Provincial Geriatrics Institute (East Zone), Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, No. 3, Chanchugang, Zhongshan 2nd Road, Yuexiu District, Guangzhou, 510080, Guangdong Province, People's Republic of China.
| | - Qiang Jie
- Department of Cardiology, Panyu Central Hospital, Cardiovascular Institute of Panyu District, No. 8, Fuyu East Road, Qiaonan Street, Panyu District, Guangzhou, 511400, Guangdong Province, People's Republic of China.
| |
Collapse
|
8
|
Piazzesi A, Putignani L. Impact of helminth-microbiome interactions on childhood health and development-A clinical perspective. Parasite Immunol 2023; 45:e12949. [PMID: 36063358 DOI: 10.1111/pim.12949] [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: 06/30/2022] [Revised: 08/26/2022] [Accepted: 09/01/2022] [Indexed: 12/01/2022]
Abstract
Humans have co-existed with parasites for virtually the entirety of our existence as a species. Today, nearly one third of the human population is infected with at least one helminthic species, most of which reside in the intestinal tract, where they have co-evolved alongside the human gut microbiota (GM). Appreciation for the interconnected relationship between helminths and GM has increased in recent years. Here, we review the evidence of how helminths and GM can influence various aspects of childhood development and the onset of paediatric diseases. We discuss the emerging evidence of how many of the changes that parasitic worms inflict on their host is enacted through gut microbes. In this light, we argue that helminth-induced microbiota modifications are of great importance in both facing the global challenge of overcoming parasitic infections, and in replicating helminthic protective effects against inflammatory diseases. We propose that deepening our knowledge of helminth-microbiota interactions will uncover novel, safer and more effective therapeutic strategies in combatting an array of childhood disorders.
Collapse
Affiliation(s)
- Antonia Piazzesi
- Multimodal Laboratory Medicine Research Area, Unit of Human Microbiome, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Lorenza Putignani
- Department of Diagnostic and Laboratory Medicine, Unit of Microbiology and Diagnostic Immunology, Unit of Microbiomics and Multimodal Laboratory Medicine Research Area, Unit of Human Microbiome, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| |
Collapse
|
9
|
Petrellis G, Piedfort O, Katsandegwaza B, Dewals BG. Parasitic worms affect virus coinfection: a mechanistic overview. Trends Parasitol 2023; 39:358-372. [PMID: 36935340 DOI: 10.1016/j.pt.2023.02.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 02/16/2023] [Accepted: 02/18/2023] [Indexed: 03/19/2023]
Abstract
Helminths are parasitic worms that coevolve with their host, usually resulting in long-term persistence through modulating host immunity. The multifarious mechanisms altering the immune system induced by helminths have significant implications on the control of coinfecting pathogens such as viruses. Here, we explore the recent literature to highlight the main immune alterations and mechanisms that affect the control of viral coinfection. Insights from these mechanisms are valuable in the understanding of clinical observations in helminth-prevalent areas and in the design of new therapeutic and vaccination strategies to control viral diseases.
Collapse
Affiliation(s)
- Georgios Petrellis
- Laboratory of Parasitology, FARAH, University of Liège, Liège, Belgium; Laboratory of Immunology-Vaccinology, FARAH, University of Liège, Liège, Belgium
| | - Ophélie Piedfort
- Laboratory of Parasitology, FARAH, University of Liège, Liège, Belgium; Laboratory of Immunology-Vaccinology, FARAH, University of Liège, Liège, Belgium
| | - Brunette Katsandegwaza
- Laboratory of Parasitology, FARAH, University of Liège, Liège, Belgium; Laboratory of Immunology-Vaccinology, FARAH, University of Liège, Liège, Belgium
| | - Benjamin G Dewals
- Laboratory of Parasitology, FARAH, University of Liège, Liège, Belgium; Laboratory of Immunology-Vaccinology, FARAH, University of Liège, Liège, Belgium.
| |
Collapse
|
10
|
Hoby S, Berenguer Veiga I, Olias P, Küchler L, Schönbächler K, Blatter H, Schmidt-Posthaus H. Fatal Diplostomum phoxini infection in captive Atlantic puffin Fratercula arctica chicks following ingestion of infected European minnows Phoxinus phoxinus. DISEASES OF AQUATIC ORGANISMS 2022; 150:161-167. [PMID: 35979990 DOI: 10.3354/dao03683] [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: 06/15/2023]
Abstract
Conservation of endangered animal species is a major task of zoos. Husbandry and breeding of Atlantic puffins Fratercula arctica in captivity is challenging. In 2019, the entire chick population (n = 4 chicks) in Berne Animal Park's Atlantic puffin colony (Bern, Switzerland) died within 7 d. Due to supply constraints, the chicks had been fed with wild-caught European minnows Phoxinus phoxinus. At necropsy, the main pathological finding in all deceased puffin chicks was a multifocal, moderate to severe subacute heterophilic and granulomatous enteritis with intralesional adult trematodes and eggs. Metacercariae surrounded by few necrotic cells and scattered macrophages were found in the brain and spinal cord of the food fish. Additional microbiological analyses of both the puffin chicks and fish were unremarkable. Diplostomum phoxini DNA could be identified in formalin-fixed paraffin-embedded tissue from the small intestine of all puffin chicks and European minnows following PCR and sequencing of the 18S ribosomal RNA gene and the internal transcribed spacer (ITS1) region. This report illustrates the importance of intensive health checks of food fish for animal species kept in captivity.
Collapse
Affiliation(s)
- Stefan Hoby
- Berne Animal Park, Tierparkweg 1, 3005 Bern, Switzerland
| | | | | | | | | | | | | |
Collapse
|
11
|
Schälter F, Frech M, Dürholz K, Lucas S, Sarter K, Lebon L, Esser-von Bieren J, Dubey LK, Voehringer D, Schett G, Harris NL, Zaiss MM. Acetate, a metabolic product of Heligmosomoides polygyrus, facilitates intestinal epithelial barrier breakdown in a FFAR2-dependent manner. Int J Parasitol 2022; 52:591-601. [PMID: 35671792 DOI: 10.1016/j.ijpara.2022.04.004] [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: 01/27/2022] [Revised: 04/11/2022] [Accepted: 04/11/2022] [Indexed: 11/05/2022]
Abstract
Approximately 2 billion people worldwide and a significant part of the domestic livestock are infected with soil-transmitted helminths, of which many establish chronic infections causing substantial economic and welfare burdens. Beside intensive research on helminth-triggered mucosal and systemic immune responses, the local mechanism that enables infective larvae to cross the intestinal epithelial barrier and invade mucosal tissue remains poorly addressed. Here, we show that Heligmosomoides polygyrus infective L3s secrete acetate and that acetate potentially facilitates paracellular epithelial tissue invasion by changed epithelial tight junction claudin expression. In vitro, impedance-based real-time epithelial cell line barrier measurements together with ex vivo functional permeability assays in intestinal organoid cultures revealed that acetate decreased intercellular barrier function via the G-protein coupled free fatty acid receptor 2 (FFAR2, GPR43). In vivo validation experiments in FFAR2-/- mice showed lower H. polygyrus burdens, whereas oral acetate-treated C57BL/6 wild type mice showed higher burdens. These data suggest that locally secreted acetate - as a metabolic product of the energy metabolism of H. polygyrus L3s - provides a significant advantage to the parasite in crossing the intestinal epithelial barrier and invading mucosal tissues. This is the first and a rate-limiting step for helminths to establish chronic infections in their hosts and if modulated could have profound consequences for their life cycle.
Collapse
Affiliation(s)
- Fabian Schälter
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Michael Frech
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Kerstin Dürholz
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Sébastien Lucas
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Kerstin Sarter
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Luc Lebon
- Global Health Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland
| | - Julia Esser-von Bieren
- Global Health Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland; Center of Allergy and Environment, Technical University of Munich and Helmholtz Zentrum München, Munich, Germany
| | - Lalit K Dubey
- Global Health Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland; Centre of Microvascular Research, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, United Kingdom
| | - David Voehringer
- Department of Infection Biology, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Georg Schett
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Nicola L Harris
- Global Health Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland; Department of Immunology, Monash University, Clayton, Victoria, Australia
| | - Mario M Zaiss
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany; Global Health Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland.
| |
Collapse
|
12
|
Bąska P, Norbury LJ. The Role of Nuclear Factor Kappa B (NF-κB) in the Immune Response against Parasites. Pathogens 2022; 11:pathogens11030310. [PMID: 35335634 PMCID: PMC8950322 DOI: 10.3390/pathogens11030310] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/17/2022] [Accepted: 02/25/2022] [Indexed: 12/28/2022] Open
Abstract
The immune system consists of various cells, organs, and processes that interact in a sophisticated manner to defend against pathogens. Upon initial exposure to an invader, nonspecific mechanisms are raised through the activation of macrophages, monocytes, basophils, mast cells, eosinophils, innate lymphoid cells, or natural killer cells. During the course of an infection, more specific responses develop (adaptive immune responses) whose hallmarks include the expansion of B and T cells that specifically recognize foreign antigens. Cell to cell communication takes place through physical interactions as well as through the release of mediators (cytokines, chemokines) that modify cell activity and control and regulate the immune response. One regulator of cell states is the transcription factor Nuclear Factor kappa B (NF-κB) which mediates responses to various stimuli and is involved in a variety of processes (cell cycle, development, apoptosis, carcinogenesis, innate and adaptive immune responses). It consists of two protein classes with NF-κB1 (p105/50) and NF-κB2 (p100/52) belonging to class I, and RelA (p65), RelB and c-Rel belonging to class II. The active transcription factor consists of a dimer, usually comprised of both class I and class II proteins conjugated to Inhibitor of κB (IκB). Through various stimuli, IκB is phosphorylated and detached, allowing dimer migration to the nucleus and binding of DNA. NF-κB is crucial in regulating the immune response and maintaining a balance between suppression, effective response, and immunopathologies. Parasites are a diverse group of organisms comprised of three major groups: protozoa, helminths, and ectoparasites. Each group induces distinct effector immune mechanisms and is susceptible to different types of immune responses (Th1, Th2, Th17). This review describes the role of NF-κB and its activity during parasite infections and its contribution to inducing protective responses or immunopathologies.
Collapse
Affiliation(s)
- Piotr Bąska
- Division of Pharmacology and Toxicology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences-SGGW, 02-786 Warsaw, Poland
- Correspondence:
| | - Luke J. Norbury
- Department of Biosciences and Food Technology, School of Science, STEM College, RMIT University, Bundoora, VIC 3083, Australia;
| |
Collapse
|
13
|
Myhill LJ, Stolzenbach S, Mejer H, Krych L, Jakobsen SR, Kot W, Skovgaard K, Canibe N, Nejsum P, Nielsen DS, Thamsborg SM, Williams AR. Parasite-Probiotic Interactions in the Gut: Bacillus sp. and Enterococcus faecium Regulate Type-2 Inflammatory Responses and Modify the Gut Microbiota of Pigs During Helminth Infection. Front Immunol 2022; 12:793260. [PMID: 35069576 PMCID: PMC8766631 DOI: 10.3389/fimmu.2021.793260] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 12/07/2021] [Indexed: 01/18/2023] Open
Abstract
Dietary probiotics may enhance gut health by directly competing with pathogenic agents and through immunostimulatory effects. These properties are recognized in the context of bacterial and viral pathogens, but less is known about interactions with eukaryotic pathogens such as parasitic worms (helminths). In this study we investigated whether two probiotic mixtures (comprised of Bacillus amyloliquefaciens, B. subtilis, and Enterococcus faecium [BBE], or Lactobacillus rhamnosus LGG and Bifidobacterium animalis subspecies Lactis Bb12 [LB]) could modulate helminth infection kinetics as well as the gut microbiome and intestinal immune responses in pigs infected with the nodular worm Oesophagostomum dentatum. We observed that neither probiotic mixture influenced helminth infection levels. BBE, and to a lesser extent LB, changed the alpha- and beta-diversity indices of the colon and fecal microbiota, notably including an enrichment of fecal Bifidobacterium spp. by BBE. However, these effects were muted by concurrent O. dentatum infection. BBE (but not LB) significantly attenuated the O. dentatum-induced upregulation of genes involved in type-2 inflammation and restored normal lymphocyte ratios in the ileo-caecal lymph nodes that were altered by infection. Moreover, inflammatory cytokine release from blood mononuclear cells and intestinal lymphocytes was diminished by BBE. Collectively, our data suggest that selected probiotic mixtures can play a role in maintaining immune homeostasis during type 2-biased inflammation. In addition, potentially beneficial changes in the microbiome induced by dietary probiotics may be counteracted by helminths, highlighting the complex inter-relationships that potentially exist between probiotic bacteria and intestinal parasites.
Collapse
Affiliation(s)
- Laura J Myhill
- Department of Veterinary and Animal Sciences, University of Copenhagen, Faculty of Health and Medical Sciences, Frederiksberg, Denmark
| | - Sophie Stolzenbach
- Department of Veterinary and Animal Sciences, University of Copenhagen, Faculty of Health and Medical Sciences, Frederiksberg, Denmark
| | - Helena Mejer
- Department of Veterinary and Animal Sciences, University of Copenhagen, Faculty of Health and Medical Sciences, Frederiksberg, Denmark
| | - Lukasz Krych
- Department of Food Science, University of Copenhagen, Frederiksberg, Denmark
| | - Simon R Jakobsen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Faculty of Health and Medical Sciences, Frederiksberg, Denmark
| | - Witold Kot
- Department of Plant and Environmental Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Kerstin Skovgaard
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Nuria Canibe
- Department of Animal Science - Immunology and Microbiology, Aarhus University, Tjele, Denmark
| | - Peter Nejsum
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Dennis S Nielsen
- Department of Food Science, University of Copenhagen, Frederiksberg, Denmark
| | - Stig M Thamsborg
- Department of Veterinary and Animal Sciences, University of Copenhagen, Faculty of Health and Medical Sciences, Frederiksberg, Denmark
| | - Andrew R Williams
- Department of Veterinary and Animal Sciences, University of Copenhagen, Faculty of Health and Medical Sciences, Frederiksberg, Denmark
| |
Collapse
|
14
|
The Role of the Intestinal Epithelium in the "Weep and Sweep" Response during Gastro-Intestinal Helminth Infections. Animals (Basel) 2022; 12:ani12020175. [PMID: 35049796 PMCID: PMC8772803 DOI: 10.3390/ani12020175] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/25/2021] [Accepted: 01/10/2022] [Indexed: 02/08/2023] Open
Abstract
Simple Summary The immune system actively combats intruders such as bacteria, viruses, fungi, and protozoan and metazoan parasites using leukocytes. During an infection white blood cells are activated to internalize bacteria or viruses and release a number of molecules to kill pathogens. Unfortunately, those mechanisms are ineffective against larger intruders like helminths, which are too large to be killed by a single immune cell. To eliminate gastro-intestinal helminths an integrated response involving the nervous, endocrine, and immune systems are used to expel the parasites. This is achieved through increased gut hydration and muscle contractions which detach worms from the gut and lead to release outside the body in a “weep and sweep” response. Epithelial cells of the intestine are significant players in this process, being responsible for detecting the presence of helminths in the gut and participating in the regulation of parasite expulsion. This paper describes the role of the gut epithelium in detecting and eliminating helminths from the intestine. Abstract Helminths are metazoan parasites infecting around 1.5 billion people all over the world. During coevolution with hosts, worms have developed numerous ways to trick and evade the host immune response, and because of their size, they cannot be internalized and killed by immune cells in the same way as bacteria or viruses. During infection, a substantial Th2 component to the immune response is evoked which helps restrain Th1-mediated tissue damage. Although an enhanced Th2 response is often not enough to kill the parasite and terminate an infection in itself, when tightly coordinated with the nervous, endocrine, and motor systems it can dislodge parasites from tissues and expel them from the gut. A significant role in this “weep and seep” response is attributed to intestinal epithelial cells (IEC). This review highlights the role of various IEC lineages (enterocytes, tuft cells, Paneth cells, microfold cells, goblet cells, and intestine stem cells) during the course of helminth infections and summarizes their roles in regulating gut architecture and permeability, and muscle contractions and interactions with the immune and nervous system.
Collapse
|
15
|
Drurey C, Lindholm HT, Coakley G, Poveda MC, Löser S, Doolan R, Gerbe F, Jay P, Harris N, Oudhoff MJ, Maizels RM. Intestinal epithelial tuft cell induction is negated by a murine helminth and its secreted products. J Exp Med 2022; 219:e20211140. [PMID: 34779829 PMCID: PMC8597987 DOI: 10.1084/jem.20211140] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 09/20/2021] [Accepted: 10/27/2021] [Indexed: 02/06/2023] Open
Abstract
Helminth parasites are adept manipulators of the immune system, using multiple strategies to evade the host type 2 response. In the intestinal niche, the epithelium is crucial for initiating type 2 immunity via tuft cells, which together with goblet cells expand dramatically in response to the type 2 cytokines IL-4 and IL-13. However, it is not known whether helminths modulate these epithelial cell populations. In vitro, using small intestinal organoids, we found that excretory/secretory products (HpES) from Heligmosomoides polygyrus blocked the effects of IL-4/13, inhibiting tuft and goblet cell gene expression and expansion, and inducing spheroid growth characteristic of fetal epithelium and homeostatic repair. Similar outcomes were seen in organoids exposed to parasite larvae. In vivo, H. polygyrus infection inhibited tuft cell responses to heterologous Nippostrongylus brasiliensis infection or succinate, and HpES also reduced succinate-stimulated tuft cell expansion. Our results demonstrate that helminth parasites reshape their intestinal environment in a novel strategy for undermining the host protective response.
Collapse
Affiliation(s)
- Claire Drurey
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, UK
| | - Håvard T. Lindholm
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Gillian Coakley
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, UK
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Australia
| | - Marta Campillo Poveda
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, UK
| | - Stephan Löser
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, UK
| | - Rory Doolan
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Australia
| | - François Gerbe
- Institut de Génomique Fonctionnelle, University of Montpellier, Centre national de la recherche scientifique UMR-5203, Institut National de la Santé et de la Recherche Médicale U1191, Montpellier, France
| | - Philippe Jay
- Institut de Génomique Fonctionnelle, University of Montpellier, Centre national de la recherche scientifique UMR-5203, Institut National de la Santé et de la Recherche Médicale U1191, Montpellier, France
| | - Nicola Harris
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Australia
| | - Menno J. Oudhoff
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Rick M. Maizels
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, UK
| |
Collapse
|
16
|
Corrêa PS, Mendes LW, Lemos LN, Sampaio ACK, Issakowicz J, McManus CM, Tsai SM, Faciola AP, Abdalla AL, Louvandini H. The effect of Haemonchus contortus and Trichostrongylus colubriforms infection on the ruminal microbiome of lambs. Exp Parasitol 2021; 231:108175. [PMID: 34740587 DOI: 10.1016/j.exppara.2021.108175] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 09/14/2021] [Accepted: 10/31/2021] [Indexed: 12/16/2022]
Abstract
We evaluated Haemonchus contortus (HC) and Trichostrongylus colubriformis (TC) infection on the ruminal microbial community of Santa Ines lambs to better understand the pathophysiology of parasite infections and the interactions among gastrointestinal nematodes and gut resident microbiota. In this study, 18 six months of age lambs were maintained for 34 days in individual pens divided into three treatments that included animals infected with HC and TC, and control (infection-free). Haematological, ruminal parameter and microbial nitrogen absorbed by pune derivatives, as well as enteric methane emission (CH4), were analysed, and the rumen microbial taxonomic and functional profile assessed by shotgun metagenomics. The analysis showed that total protein, albumin, urea, and butyrate level were lower in animals infected by both parasites, while HC infection also decreased the haemoglobin level. Both infected groups (TC and HC) increased the enteric methane emission (CH4). TC and HC infections increased the diversity and richness of functional microbial genes. Most alterations in the rumen microbiome composition of infected groups are associated with the suppression of microbes involved in microbial homeostasis maintenance and expansion of the archaeal community in the infected animals. Infection led to an increased abundance of nitrogen, amino acid, protein, and energy metabolism genes. Overall, TC and HC infection increased the enteric methane emission, negatively affected taxon's responsible for maintenance de rumen homeostasis and modulated some important genes related to protein and energy metabolism.
Collapse
Affiliation(s)
- Patricia Spoto Corrêa
- University of São Paulo, Centre for Nuclear Energy in Agriculture, Laboratory of Animal Nutrition, Piracicaba, SP, 13400-970, Brazil; University of Florida, Department of Animal Sciences, United States of America, Gainesville, FL, 32611, USA.
| | - Lucas William Mendes
- University of São Paulo, Centre for Nuclear Energy in Agriculture, Laboratory of Molecular Cell Biology, Piracicaba, SP, 13400-970, Brazil
| | - Leandro Nascimento Lemos
- Department of Bioinformatics, National Laboratory of Scientific Computing, Petrópolis, RJ, 25651-076, Brazil
| | - Ana Claudia Koki Sampaio
- University of São Paulo, Centre for Nuclear Energy in Agriculture, Laboratory of Animal Nutrition, Piracicaba, SP, 13400-970, Brazil
| | - Juliano Issakowicz
- University of São Paulo, Centre for Nuclear Energy in Agriculture, Laboratory of Animal Nutrition, Piracicaba, SP, 13400-970, Brazil
| | | | - Siu Mui Tsai
- University of São Paulo, Centre for Nuclear Energy in Agriculture, Laboratory of Molecular Cell Biology, Piracicaba, SP, 13400-970, Brazil
| | - Antonio Pinheiro Faciola
- University of Florida, Department of Animal Sciences, United States of America, Gainesville, FL, 32611, USA
| | - Adibe Luiz Abdalla
- University of São Paulo, Centre for Nuclear Energy in Agriculture, Laboratory of Animal Nutrition, Piracicaba, SP, 13400-970, Brazil
| | - Helder Louvandini
- University of São Paulo, Centre for Nuclear Energy in Agriculture, Laboratory of Animal Nutrition, Piracicaba, SP, 13400-970, Brazil; University of Florida, Department of Animal Sciences, United States of America, Gainesville, FL, 32611, USA
| |
Collapse
|
17
|
Doyen V, Corazza F, Nhu Thi H, Le Chi T, Truyens C, Nagant C, Tran Thi Mong H, Fils JF, Thi Ngoc Huynh P, Michel O. Hookworm treatment induces a decrease of suppressive regulatory T cell associated with a Th2 inflammatory response. PLoS One 2021; 16:e0252921. [PMID: 34111180 PMCID: PMC8191899 DOI: 10.1371/journal.pone.0252921] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 05/25/2021] [Indexed: 01/04/2023] Open
Abstract
Background Like other helminths, hookworms (HW) induce a regulatory immune response able to modulate and dampen reactivity of the host to antigens. No data about the evolution of the immune response after treatment are available. We aim to phenotype the regulatory immune response during natural HW infection and its evolution after treatment. Methodology Twenty hookworm infected (HW+) and 14 non-infected subjects HW–from endemic area in the periphery of Ho Chi Minh City were included. Blood and feces samples were obtained before, 2 and 4 weeks after treatment with Albendazole 400mg. Additional samples were obtained at 3 and 12 months in the HW+ group. Hematological parameters, Treg (CD4+CD25hiFoxP3hi) and surface molecules (CD39, CD62L, ICOS, PD-1, CD45RA) were measured as well as inflammatory and lymphocytes differentiation cytokines such as IL-1β, IL-6, IFNγ, IL-4, IL-17, IL-10, IL-2 and TGFβ. Results HW+ subjects showed higher Treg, TregICOS+, Treg PD1-, TregCD62L+ and CD45RA+FoxP3lo resting Treg (rTreg). CD45RA-FoxP3lo non-suppressive Treg cells were also increased. No preferential Th1/Th2 orientation was observed, nor difference for IL-10 between two groups. After treatment, Treg, TregICOS+, TregCD62L+, Treg PD1- and rTreg decreased while IL-4 and IL-6 cytokines increased. Conclusion During HW infection, Treg are increased and characterized by a heterogeneous population: a highly suppressive as well as a non-suppressive T cells phenotype. After treatment, Treg with immune-suppressive phenotype exhibited a decrease parallel to an inflammatory Th2 response.
Collapse
Affiliation(s)
- Virginie Doyen
- Laboratory of Translational Research, ULB223, CHU Brugmann, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Clinic of Immunoallergology, CHU Brugmann, ULB, Brussels, Belgium
- * E-mail:
| | - Francis Corazza
- Laboratory of Translational Research, ULB223, CHU Brugmann, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Immunology Laboratory, LHUB-ULB, Brussels, Belgium
| | - Hoa Nhu Thi
- Parasitology and Mycology Department, Pham Ngoc Thach University of Medicine, Ho Chi Minh, Vietnam
| | - Thanh Le Chi
- Immunology Laboratory, Pasteur Institute, Ho Chi Minh, Vietnam
| | - Carine Truyens
- Parasitology Laboratory, ULB Center for Research in immunology (U-CRI), Université Libre de Bruxelles, Brussels, Belgium
| | - Carole Nagant
- Laboratory of Translational Research, ULB223, CHU Brugmann, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Immunology Laboratory, LHUB-ULB, Brussels, Belgium
| | - Hiep Tran Thi Mong
- Department of Family Medicine, Pham Ngoc Thach University of Medicine, Ho Chi Minh, Vietnam
| | | | | | - Olivier Michel
- Clinic of Immunoallergology, CHU Brugmann, ULB, Brussels, Belgium
| |
Collapse
|
18
|
Transcriptome Analysis and Autophagy Investigation of LoVo Cells Stimulated with Exosomes Derived from T. asiatica Adult Worms. Microorganisms 2021; 9:microorganisms9050994. [PMID: 34062985 PMCID: PMC8147967 DOI: 10.3390/microorganisms9050994] [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: 04/18/2021] [Revised: 04/25/2021] [Accepted: 04/28/2021] [Indexed: 11/17/2022] Open
Abstract
Taenia asiatica is a zoonotic parasite found in the human intestine and pig liver that evolved various strategies to survive the host’s defenses. Exosomes are membranous vesicles released by cells and are an important vehicle in parasite-host interactions. However, no literature exists on the specific infection mechanisms of T. asiatica against the host defense response, and further research is required to understand the parasite-host interaction. In this study, we investigated the host’s differentially expressed genes (DEGs) while stimulating them with exosomes derived from the T. asiatica adult worm (Tas-exo) on LoVo by RNA-seq analysis. Our results identified 348 genes as being significantly differentially expressed for the Tas-exo group when comparing with that of the NC group. Some of these genes are related to modulation of cell proliferation and cell autophagy. Surprisingly, autophagy and cell proliferation have crucial roles in the defense against parasites; accordingly, we detected cell proliferation and autophagy in LoVo cells by CCK8, immunofluorescence, and Western blotting, demonstrating that Tas-exo could inhibit LoVo cell proliferation and autophagy via AMPK pathway. When P62 and p-mTOR/mTOR expression were significantly increased, BeclinI and pAMPK/AMPK were significantly decreased. These results expand our understanding of parasite-host interactions mediated by exosomes.
Collapse
|
19
|
Dela Justina V, Gama LA, Schönholzer T, Bressan AF, Lima VV, Americo MF, Giachini FR. Resistance mesenteric arteries display hypercontractility in the resolution time of Strongyloides venezuelensis infection. Exp Parasitol 2021; 222:108078. [PMID: 33485874 DOI: 10.1016/j.exppara.2021.108078] [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: 06/08/2020] [Revised: 10/23/2020] [Accepted: 01/17/2021] [Indexed: 10/22/2022]
Abstract
The blood flow in the mesenteric region is crucial for nutrient absorption and immune response in the gastrointestinal tract. The presence of nematodes or their excreted/secreted products seems to provoke vascular dysfunction. However, it is unclear whether and how the intestinal nematodes with habitat in the intestinal niche could affect the mesenteric vascular resistance. In this study, male Wistar rats were infected with 2000 larvae of S. venezuelensis, and experiments were conducted at 0 (non-infected control), 10 or 30 days post-infection (DPI). Eggs were counted in rats' feces and adult worms recovered from the small intestine. Second- or third-order mesenteric arteries were extracted for concentration-response curves (CRC) to phenylephrine [PE; in the presence or absence of L-NAME or indomethacin] and acetylcholine. The number of eggs and adult worms were significantly higher in the 10 DPI group than those of 30 DPI group. Augmented PE-induced contraction was seen after 30 DPI compared to 10 DPI or control group. Hypercontractility to PE was partially prevented by L-NAME and wholly abolished by indomethacin incubation. Endothelium-dependent relaxation and endothelial nitric oxide synthase expression were unchanged among groups. COX-1 and COX-2 display a different pattern of expression over the infection. Hypercontractility observed in mesenteric resistance arteries in the resolution time of S. venezuelensis infection may represent systemic damage, which can generate significant cardiovascular and gastrointestinal repercussions.
Collapse
Affiliation(s)
- Vanessa Dela Justina
- Institute of Biological Sciences, Federal University of Goias, Goiânia, GO, Brazil
| | - Loyane Almeida Gama
- Institute of Biological Sciences and Health, Federal University of Mato Grosso, Barra Do Garças, MT, Brazil; Institute of Biosciences, São Paulo State University - UNESP, Botucatu, SP, Brazil
| | - Tatiane Schönholzer
- Institute of Biological Sciences and Health, Federal University of Mato Grosso, Barra Do Garças, MT, Brazil
| | - Alecsander F Bressan
- Institute of Biological Sciences and Health, Federal University of Mato Grosso, Barra Do Garças, MT, Brazil; Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Victor Vitorino Lima
- Institute of Biological Sciences and Health, Federal University of Mato Grosso, Barra Do Garças, MT, Brazil
| | - Madileine F Americo
- Institute of Biological Sciences and Health, Federal University of Mato Grosso, Barra Do Garças, MT, Brazil; Institute of Biosciences, São Paulo State University - UNESP, Botucatu, SP, Brazil
| | - Fernanda R Giachini
- Institute of Biological Sciences, Federal University of Goias, Goiânia, GO, Brazil; Institute of Biological Sciences and Health, Federal University of Mato Grosso, Barra Do Garças, MT, Brazil.
| |
Collapse
|
20
|
Macleod T, Ainscough JS, Hesse C, Konzok S, Braun A, Buhl AL, Wenzel J, Bowyer P, Terao Y, Herrick S, Wittmann M, Stacey M. The Proinflammatory Cytokine IL-36γ Is a Global Discriminator of Harmless Microbes and Invasive Pathogens within Epithelial Tissues. Cell Rep 2020; 33:108515. [PMID: 33326792 PMCID: PMC7758160 DOI: 10.1016/j.celrep.2020.108515] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 10/05/2020] [Accepted: 11/19/2020] [Indexed: 12/20/2022] Open
Abstract
Epithelial tissues represent vital interfaces between organisms and their environment. As they are constantly exposed to harmful pathogens, innocuous commensals, and environmental microbes, it is essential they sense and elicit appropriate responses toward these different types of microbes. Here, we demonstrate that the epithelial cytokine interleukin-36γ (IL-36γ) acts as a global discriminator of pathogenic and harmless microbes via cell damage and proteolytic activation. We show that intracellular pro-IL-36γ is upregulated by both fungal and bacterial epithelial microbes; yet, it is only liberated from cells, and subsequently processed to its mature, potent, proinflammatory form, by pathogen-mediated cell damage and pathogen-derived proteases. This work demonstrates that IL-36γ senses pathogen-induced cell damage and proteolytic activity and is a key initiator of immune responses and pathological inflammation within epithelial tissues. As an apically located epithelial proinflammatory cytokine, we therefore propose that IL-36γ is critical as the initial discriminator of harmless microbes and invasive pathogens within epithelial tissues. Epithelial pathogens induce expression and release of IL-36γ Proteases secreted by several epithelial pathogens activate IL-36γ The A. fumigatus and S. pyogenes virulence factors Asp F13 and SpeB activate IL-36γ IL-36γ is a global sensor of pathogen-derived proteases during epithelial infection
Collapse
Affiliation(s)
- Thomas Macleod
- Faculty of Biological Sciences, School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, UK
| | - Joseph S Ainscough
- Faculty of Biological Sciences, School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, UK
| | - Christina Hesse
- Fraunhofer Institute for Toxicology and Experimental Medicine, Nikolai-Fuchs-Straße 1, 30625 Hannover, Germany; Member of the German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH) Research Network, Hannover, Germany
| | - Sebastian Konzok
- Fraunhofer Institute for Toxicology and Experimental Medicine, Nikolai-Fuchs-Straße 1, 30625 Hannover, Germany; Member of the German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH) Research Network, Hannover, Germany
| | - Armin Braun
- Fraunhofer Institute for Toxicology and Experimental Medicine, Nikolai-Fuchs-Straße 1, 30625 Hannover, Germany; Member of the German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH) Research Network, Hannover, Germany
| | - Anna-Lena Buhl
- Department of Dermatology and Allergy, University of Bonn, Bonn 53012, Germany
| | - Joerg Wenzel
- Department of Dermatology and Allergy, University of Bonn, Bonn 53012, Germany
| | - Paul Bowyer
- Division of Infection, Immunity & Respiratory Medicine, University of Manchester, Manchester M13 9PL, UK
| | - Yutaka Terao
- Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Sarah Herrick
- Faculty of Biology Medicine and Health, School of Biological Sciences, University of Manchester and Manchester Academic Health Science Centre, Manchester M13 9PL, UK
| | - Miriam Wittmann
- Faculty of Biological Sciences, School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, UK; Faculty of Medicine and Health, Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds LS2 9JT, UK; Leeds Biomedical Research Centre, National Institute for Health Research, Leeds Teaching Hospitals, Leeds, UK.
| | - Martin Stacey
- Faculty of Biological Sciences, School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, UK.
| |
Collapse
|
21
|
Wilhelm FR, Krautwald-Junghanns ME, Ortín-Piqueras V, Junnila J, Cramer K, Forsgård RA, Frias R, Spillmann T, Schmidt V. Iohexol-based measurement of intestinal permeability in birds. J Exot Pet Med 2020. [DOI: 10.1053/j.jepm.2020.04.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
22
|
Shute A, Wang A, Jayme TS, Strous M, McCoy KD, Buret AG, McKay DM. Worm expulsion is independent of alterations in composition of the colonic bacteria that occur during experimental Hymenolepis diminuta-infection in mice. Gut Microbes 2020; 11:497-510. [PMID: 31928118 PMCID: PMC7524392 DOI: 10.1080/19490976.2019.1688065] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The tapeworm Hymenolepis diminuta fails to establish in mice. Given the potential for helminth-bacteria interaction in the gut and the influence that commensal bacteria exert on host immunity, we tested if worm expulsion was related to alterations in the gut microbiota. Specific pathogen-free (SPF) mice, treated with broad-spectrum antibiotics, or germ-free wild-type mice were infected with H. diminuta, gut bacterial composition assessed by 16S rRNA gene sequencing, and worm counts, blood eosinophilia, goblet cells, splenic IL-4, -5 and -10, and colonic cytokines/chemokines mRNA were assessed. Effects of a PBS-soluble extract of adult H. diminuta on bacterial growth in vitro was tested. H. diminuta-infected mice displayed increased α and β diversity in colonic mucosa-associated and fecal bacterial communities, characterized by increased Lachnospiraceae and clostridium cluster XIVa. In vitro analysis revealed that the worm extract promoted the growth of anaerobic bacteria on M2GSC agar. H. diminuta-infection was accompanied by increased Th2 immune responses, and colon from infected mice had increased levels of IL-10, IL-25, Muc2, trefoil factor 3, and β2-defensin mRNA. SPF-mice treated with antibiotics, or germ-free mice, expelled H. diminuta with kinetics similar to control SPF mice. In both settings, measurements of Th2-immune responses were not significantly different across the groups. Thus, while infection with H. diminuta results in subtle but distinct changes to the colonic microbiota, we have no evidence to support an essential role for gut bacteria in the expulsion of the worm from the mouse host.
Collapse
Affiliation(s)
- Adam Shute
- Host-Parasite Interactions Program, University of Calgary, Calgary, Alberta, Canada,Gastrointestinal Research Group and Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Joan and Phoebe Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Arthur Wang
- Gastrointestinal Research Group and Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Joan and Phoebe Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Timothy S. Jayme
- Host-Parasite Interactions Program, University of Calgary, Calgary, Alberta, Canada,Gastrointestinal Research Group and Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Joan and Phoebe Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Marc Strous
- Department of Geoscience, University of Calgary, Calgary, Alberta, Canada
| | - Kathy D. McCoy
- Gastrointestinal Research Group and Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Joan and Phoebe Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Andre G. Buret
- Host-Parasite Interactions Program, University of Calgary, Calgary, Alberta, Canada,Gastrointestinal Research Group and Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Joan and Phoebe Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada,Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Derek M. McKay
- Host-Parasite Interactions Program, University of Calgary, Calgary, Alberta, Canada,Gastrointestinal Research Group and Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Joan and Phoebe Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada,CONTACT Derek M. McKay Department of Physiology & Pharmacology, 1877 HSC, University of Calgary, 3330 Hospital Drive NW, Calgary, AlbertaT2N 4N1, Canada
| |
Collapse
|
23
|
Hussain Z, El-Omar E, Lee YY. Dual infective burden of Helicobacter pylori and intestinal parasites: Good or bad news for the host? Indian J Gastroenterol 2020; 39:111-116. [PMID: 32372188 DOI: 10.1007/s12664-020-01045-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Zahid Hussain
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Emad El-Omar
- Microbiome Research Centre, St George and Sutherland Clinical School, University of New South Wales, Sydney, Australia
| | - Yeong Yeh Lee
- Microbiome Research Centre, St George and Sutherland Clinical School, University of New South Wales, Sydney, Australia. .,School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, Malaysia.
| |
Collapse
|
24
|
Maizels RM. Regulation of immunity and allergy by helminth parasites. Allergy 2020; 75:524-534. [PMID: 31187881 DOI: 10.1111/all.13944] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 06/04/2019] [Accepted: 06/05/2019] [Indexed: 12/26/2022]
Abstract
There is increasing interest in helminth parasite modulation of the immune system, both from the fundamental perspective of the "arms race" between host and parasite, and equally importantly, to understand if parasites offer new pathways to abate and control untoward immune responses in humans. This article reviews the epidemiological and experimental evidence for parasite down-regulation of host immunity and immunopathology, in allergy and other immune disorders, and recent progress towards defining the mechanisms and molecular mediators which parasites exploit in order to modulate their host. Among these are novel products that interfere with epithelial cell alarmins, dendritic cell activation, macrophage function and T-cell responsiveness through the promotion of an immunoregulatory environment. These modulatory effects assist parasites to establish and survive, while dampening immune reactivity to allergens, autoantigens and microbiome determinants.
Collapse
Affiliation(s)
- Rick M. Maizels
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunology and Inflammation University of Glasgow Glasgow UK
| |
Collapse
|
25
|
Toychiev A, Mirzoeva M, Davis N, Islamova J, Osipova S. Pityriasis alba: Possible associations with intestinal helminths and pathogenic protozoa. Int J Clin Pract 2020; 74:e13441. [PMID: 31633268 DOI: 10.1111/ijcp.13441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 07/22/2019] [Accepted: 10/16/2019] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND The aetiology of pityriasis alba (PA) remains uncertain, and children are at risk for PA and intestinal parasites. AIM To determine the prevalence of intestinal parasites in PA patients and to evaluate their possible role in PA pathogenesis. METHODS Overall, 180 PA patients and 100 healthy individuals were enrolled. Intestinal parasites were diagnosed by triple coproscopy, and the total serum Immunoglobulin E (IgE) levels were determined by ELISA. PA patients with intestinal parasites were treated with conventional antiparasitic drugs. Patient response to antiparasitic therapy was evaluated after 6 weeks. RESULTS The prevalence of intestinal parasites in PA patients and controls was 60 ± 3.6% and 32 ± 4.6%, respectively (P < .0001). Regardless of the parasite species among the PA patients and control individuals, the total IgE levels were significantly higher in PA patients (P ≤ .05). The highest values of IgE were found in PA patients with Hymenolepis nana (641.7 ± 46.3 IU/mL). The total IgE level in PA patients with parasites decreased after antiparasitic therapy, but the reduction was only significant in patients with H. nana (P < .05). Complete disappearance of hypopigmented patches was observed after the elimination of H. nana, Giardia lamblia and Enterobius vermicularis in 65 ± 10.6%, 48.7 ± 8.0% and 33.3 ± 8.2% of cases, respectively. In total, a positive clinical response to antiparasitic therapy was achieved in 60 ± 4.7% of infected PA patients. CONCLUSION A positive clinical response to antiparasitic therapy indicates the role of intestinal parasites in PA pathogenesis. Parasitological examination is justified by the recovery of 60 ± 4.7% of infected PA patients after the elimination of intestinal parasites.
Collapse
Affiliation(s)
- Abdurakhim Toychiev
- Department of Immunology of Parasitic Diseases, the Research Institute of Epidemiology, Microbiology and Infectious Diseases, Tashkent, Uzbekistan
| | - Mexriniso Mirzoeva
- Department of Infectious Diseases, Bukhara state medical institute, Bukhara, Uzbekistan
| | - Nikolay Davis
- Department of Immunology of Parasitic Diseases, the Research Institute of Epidemiology, Microbiology and Infectious Diseases, Tashkent, Uzbekistan
| | - Jannat Islamova
- Department of Pharmacology and Toxicology, the Institute of the Chemistry of Plant Substances named acad. S. Yu. Yunusov, Tashkent, Uzbekistan
| | - Svetlana Osipova
- Department of Immunology of Parasitic Diseases, the Research Institute of Epidemiology, Microbiology and Infectious Diseases, Tashkent, Uzbekistan
| |
Collapse
|
26
|
Ahmed N, Heitlinger E, Affinass N, Kühl AA, Xenophontos N, Jarquin VH, Jost J, Steinfelder S, Hartmann S. A Novel Non-invasive Method to Detect RELM Beta Transcript in Gut Barrier Related Changes During a Gastrointestinal Nematode Infection. Front Immunol 2019; 10:445. [PMID: 30915083 PMCID: PMC6423163 DOI: 10.3389/fimmu.2019.00445] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 02/19/2019] [Indexed: 12/16/2022] Open
Abstract
Currently, methods for monitoring changes of gut barrier integrity and the associated immune response via non-invasive means are limited. Therefore, we aimed to develop a novel non-invasive technique to investigate immunological host responses representing gut barrier changes in response to infection. We identified the mucous layer on feces from mice to be mainly composed of exfoliated intestinal epithelial cells. Expression of RELM-β, a gene prominently expressed in intestinal nematode infections, was used as an indicator of intestinal cellular barrier changes to infection. RELM-β was detected as early as 6 days post-infection (dpi) in exfoliated epithelial cells. Interestingly, RELM-β expression also mirrored the quality of the immune response, with higher amounts being detectable in a secondary infection and in high dose nematode infection in laboratory mice. This technique was also applicable to captured worm-infected wild house mice. We have therefore developed a novel non-invasive method reflecting gut barrier changes associated with alterations in cellular responses to a gastrointestinal nematode infection.
Collapse
Affiliation(s)
- Norus Ahmed
- Department of Veterinary Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| | - Emanuel Heitlinger
- Research Group Ecology and Evolution of Molecular Parasite Host Interactions, Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany.,Institute for Biology, Molecular Parasitology, Humboldt Universität, Berlin, Germany
| | - Nicole Affinass
- Department of Veterinary Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| | - Anja A Kühl
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, iPATH.Berlin, Berlin, Germany
| | - Natasa Xenophontos
- Department of Veterinary Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| | - Victor Hugo Jarquin
- Research Group Ecology and Evolution of Molecular Parasite Host Interactions, Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany.,Institute for Biology, Molecular Parasitology, Humboldt Universität, Berlin, Germany
| | - Jenny Jost
- Research Group Ecology and Evolution of Molecular Parasite Host Interactions, Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany
| | - Svenja Steinfelder
- Department of Veterinary Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany.,Department of Neuroscience, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Susanne Hartmann
- Department of Veterinary Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
27
|
Sharkey KA, Beck PL, McKay DM. Neuroimmunophysiology of the gut: advances and emerging concepts focusing on the epithelium. Nat Rev Gastroenterol Hepatol 2018; 15:765-784. [PMID: 30069036 DOI: 10.1038/s41575-018-0051-4] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The epithelial lining of the gastrointestinal tract serves as the interface for digestion and absorption of nutrients and water and as a defensive barrier. The defensive functions of the intestinal epithelium are remarkable considering that the gut lumen is home to trillions of resident bacteria, fungi and protozoa (collectively, the intestinal microbiota) that must be prevented from translocation across the epithelial barrier. Imbalances in the relationship between the intestinal microbiota and the host lead to the manifestation of diseases that range from disorders of motility and sensation (IBS) and intestinal inflammation (IBD) to behavioural and metabolic disorders, including autism and obesity. The latest discoveries shed light on the sophisticated intracellular, intercellular and interkingdom signalling mechanisms of host defence that involve epithelial and enteroendocrine cells, the enteric nervous system and the immune system. Together, they maintain homeostasis by integrating luminal signals, including those derived from the microbiota, to regulate the physiology of the gastrointestinal tract in health and disease. Therapeutic strategies are being developed that target these signalling systems to improve the resilience of the gut and treat the symptoms of gastrointestinal disease.
Collapse
Affiliation(s)
- Keith A Sharkey
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada. .,Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada. .,Gastrointestinal Research Group, University of Calgary, Calgary, Alberta, Canada. .,Department of Physiology & Pharmacology, University of Calgary, Calgary, Alberta, Canada.
| | - Paul L Beck
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada.,Gastrointestinal Research Group, University of Calgary, Calgary, Alberta, Canada.,Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada.,Division of Gastroenterology and Hepatology, University of Calgary, Calgary, Alberta, Canada.,Department of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Derek M McKay
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada.,Gastrointestinal Research Group, University of Calgary, Calgary, Alberta, Canada.,Department of Physiology & Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
28
|
Rausch S, Midha A, Kuhring M, Affinass N, Radonic A, Kühl AA, Bleich A, Renard BY, Hartmann S. Parasitic Nematodes Exert Antimicrobial Activity and Benefit From Microbiota-Driven Support for Host Immune Regulation. Front Immunol 2018; 9:2282. [PMID: 30349532 PMCID: PMC6186814 DOI: 10.3389/fimmu.2018.02282] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 09/14/2018] [Indexed: 12/04/2022] Open
Abstract
Intestinal parasitic nematodes live in intimate contact with the host microbiota. Changes in the microbiome composition during nematode infection affect immune control of the parasites and shifts in the abundance of bacterial groups have been linked to the immunoregulatory potential of nematodes. Here we asked if the small intestinal parasite Heligmosomoides polygyrus produces factors with antimicrobial activity, senses its microbial environment and if the anti-nematode immune and regulatory responses are altered in mice devoid of gut microbes. We found that H. polygyrus excretory/secretory products exhibited antimicrobial activity against gram+/− bacteria. Parasites from germ-free mice displayed alterations in gene expression, comprising factors with putative antimicrobial functions such as chitinase and lysozyme. Infected germ-free mice developed increased small intestinal Th2 responses coinciding with a reduction in local Foxp3+RORγt+ regulatory T cells and decreased parasite fecundity. Our data suggest that nematodes sense their microbial surrounding and have evolved factors that limit the outgrowth of certain microbes. Moreover, the parasites benefit from microbiota-driven immune regulatory circuits, as an increased ratio of intestinal Th2 effector to regulatory T cells coincides with reduced parasite fitness in germ-free mice.
Collapse
Affiliation(s)
- Sebastian Rausch
- Department of Veterinary Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| | - Ankur Midha
- Department of Veterinary Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| | - Matthias Kuhring
- Bioinformatics Unit (MF 1), Robert Koch Institute, Berlin, Germany.,Core Unit Bioinformatics, Berlin Institute of Health (BIH), Berlin, Germany.,Berlin Institute of Health Metabolomics Platform, Berlin Institute of Health (BIH), Berlin, Germany.,Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Nicole Affinass
- Department of Veterinary Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| | - Aleksandar Radonic
- Centre for Biological Threats and Special Pathogens (ZBS 1), Robert Koch Institute, Berlin, Germany.,Genome Sequencing Unit (MF 2), Robert Koch Institute, Berlin, Germany
| | - Anja A Kühl
- iPATH.Berlin, Core Unit for Immunopathology for Experimental Models, Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - André Bleich
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | | | - Susanne Hartmann
- Department of Veterinary Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
29
|
Modulation of the immune response by helminths: a role for serotonin? Biosci Rep 2018; 38:BSR20180027. [PMID: 30177522 PMCID: PMC6148219 DOI: 10.1042/bsr20180027] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 07/31/2018] [Accepted: 08/03/2018] [Indexed: 12/13/2022] Open
Abstract
The mammalian gut is a remarkable organ: with a nervous system that rivals the spinal cord, it is the body’s largest repository of immune and endocrine cells and houses an immense and complex microbiota. Infection with helminth parasites elicits a conserved program of effector and regulatory immune responses to eradicate the worm, limit tissue damage, and return the gut to homeostasis. Discrete changes in the nervous system, and to a lesser extent the enteroendocrine system, occur following helminth infection but the importance of these adaptations in expelling the worm is poorly understood. Approximately 90% of the body’s serotonin (5-hydroxytryptamine (5-HT)) is made in enterochromaffin (EC) cells in the gut, indicative of the importance of this amine in intestinal function. Signaling via a plethora of receptor subtypes, substantial evidence illustrates that 5-HT affects immunity. A small number of studies document changes in 5-HT levels following infection with helminth parasites, but these have not been complemented by an understanding of the role of 5-HT in the host–parasite interaction. In reviewing this area, the gap in knowledge of how changes in the enteric serotonergic system affects the outcome of infection with intestinal helminths is apparent. We present this as a call-to-action by investigators in the field. We contend that neuronal EC cell–immune interactions in the gut are essential in maintaining homeostasis and, when perturbed, contribute to pathophysiology. The full affect of infection with helminth parasites needs to define, and then mechanistically dissect the role of the enteric nervous and enteroendocrine systems of the gut.
Collapse
|
30
|
Cortés A, Toledo R, Cantacessi C. Classic Models for New Perspectives: Delving into Helminth–Microbiota–Immune System Interactions. Trends Parasitol 2018; 34:640-654. [DOI: 10.1016/j.pt.2018.05.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 05/29/2018] [Accepted: 05/31/2018] [Indexed: 02/07/2023]
|
31
|
The Untapped Pharmacopeic Potential of Helminths. Trends Parasitol 2018; 34:828-842. [PMID: 29954660 DOI: 10.1016/j.pt.2018.05.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 05/29/2018] [Accepted: 05/31/2018] [Indexed: 02/06/2023]
Abstract
The dramatic rise in immunological disorders that occurs with socioeconomic development is associated with alterations in microbial colonization and reduced exposure to helminths. Excretory-secretory (E/S) helminth products contain a mixture of proteins and low-molecular-weight molecules representing the primary interface between parasite and host. Research has shown great pharmacopeic potential for helminth-derived products in animal disease models and even in clinical trials. Although in its infancy, the translation of worm-derived products into therapeutics is highly promising. Here, we focus on important key aspects in the development of immunomodulatory drugs, also highlighting novel approaches that hold great promise for future development of innovative research strategies.
Collapse
|
32
|
Abdoli A, Maspi N. Commentary: Estimates of Global, Regional, and National Morbidity, Mortality, and Aetiologies of Diarrhoeal Diseases: A Systematic Analysis for the Global Burden of Disease Study 2015. Front Med (Lausanne) 2018; 5:11. [PMID: 29435450 PMCID: PMC5796900 DOI: 10.3389/fmed.2018.00011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 01/15/2018] [Indexed: 12/25/2022] Open
Affiliation(s)
- Amir Abdoli
- Department of Parasitology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Nahid Maspi
- Department of Parasitology, Faculty of Medical Sciences, Ilam University of Medical Sciences, Ilam, Iran
| |
Collapse
|