1
|
Guo Y, Song J, Chen Y, Lü Y, Yu W. Impact of NLRP6 inflammasome on neuroinflammation in temporal lobe epilepsy. Neurochem Int 2025; 188:105994. [PMID: 40398748 DOI: 10.1016/j.neuint.2025.105994] [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: 03/12/2025] [Revised: 04/28/2025] [Accepted: 05/18/2025] [Indexed: 05/23/2025]
Abstract
Epilepsy is one of the most common and severe chronic brain diseases, affecting up to 70 million people worldwide. Neuroinflammation plays a central role in the progression of the disease. The Nod-Like Receptor Protein 6 (NLRP6) inflammasome assembles with apoptosis-associated speck-like protein (ASC) to cleave pro-caspase-1 into caspase-1, thus forming the NLRP6 inflammasome. This process promotes the maturation and release of downstream interleukins (IL)-18 and IL-1β, exacerbating pathological processes in various diseases. In this study, we demonstrated significantly enhanced NLRP6 expression in the cortex and hippocampus of epileptic mice, suggesting a role for the inflammasome in epilepsy. Immunofluorescence staining further revealed that NLRP6 was predominantly expressed in hippocampal neurons of these mice. Additionally, knockdown of NLRP6 reduced susceptibility to epilepsy, alleviated post-seizure neuronal damage, and decreased levels of pro-inflammatory cytokines, including IL-18, IL-1β, and IL-6. Conversely, NLRP6 overexpression produced opposite effects, which were effectively reversed by treatment with the caspase-1 inhibitor VX765. To the best of our knowledge, this is the first study to demonstrate a link between NLRP6 and the activation of the caspase-1/IL-1β/IL-18 signaling pathway in a kainic acid (KA)-induced epilepsy mouse model. Administration of VX765 alleviated pathological alterations and exerted neuroprotective effects. These findings suggest that NLRP6 plays a critical role in the initiation and progression of epilepsy.
Collapse
Affiliation(s)
- Yiming Guo
- Institute of Neuroscience, Chongqing Medical University, Chongqing, 400016, China
| | - Jiaqi Song
- Institute of Neuroscience, Chongqing Medical University, Chongqing, 400016, China
| | - Yingxi Chen
- Institute of Neuroscience, Chongqing Medical University, Chongqing, 400016, China
| | - Yang Lü
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Weihua Yu
- Institute of Neuroscience, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
2
|
Merolle M, Striepen B, Hunter CA. Parasite and host immune factors that impact the development of a mucosal vaccine for Cryptosporidium. Mucosal Immunol 2025:S1933-0219(25)00049-2. [PMID: 40379259 DOI: 10.1016/j.mucimm.2025.05.002] [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: 12/23/2024] [Revised: 03/28/2025] [Accepted: 05/07/2025] [Indexed: 05/19/2025]
Abstract
The parasite Cryptosporidium is a leading cause of diarrhea and death in malnourished children and immunocompromised individuals and an important enteric pathogen of livestock. A mucosal vaccine to mitigate clinical disease and decrease transmission would address the public health impact of this organism, but current options are limited. The development of a rational strategy for vaccination requires an appreciation of the parasite life cycle, how Cryptosporidium interacts with its host cell (the enterocyte), and the immune mechanisms that act locally to control this organism. Here we review current knowledge of the adaptive immune mechanisms that mediate resistance to Cryptosporidium, their relevance to vaccine design, and how recent advances in parasite genetics inform vaccine development.
Collapse
Affiliation(s)
- Maria Merolle
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 380 South University Avenue, Philadelphia, PA 19104, United States
| | - Boris Striepen
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 380 South University Avenue, Philadelphia, PA 19104, United States
| | - Christopher A Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 380 South University Avenue, Philadelphia, PA 19104, United States.
| |
Collapse
|
3
|
Bai Y, Pan Y, Liu X. Mechanistic insights into gasdermin-mediated pyroptosis. Nat Rev Mol Cell Biol 2025:10.1038/s41580-025-00837-0. [PMID: 40128620 DOI: 10.1038/s41580-025-00837-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/29/2025] [Indexed: 03/26/2025]
Abstract
Pyroptosis, a novel mode of inflammatory cell death, is executed by membrane pore-forming gasdermin (GSDM) family members in response to extracellular or intracellular injury cues and is characterized by a ballooning cell morphology, plasma membrane rupture and the release of inflammatory mediators such as interleukin-1β (IL-1β), IL-18 and high mobility group protein B1 (HMGB1). It is a key effector mechanism for host immune defence and surveillance against invading pathogens and aberrant cancerous cells, and contributes to the onset and pathogenesis of inflammatory and autoimmune diseases. Manipulating the pore-forming activity of GSDMs and pyroptosis could lead to novel therapeutic strategies. In this Review, we discuss the current knowledge regarding how GSDM-mediated pyroptosis is initiated, executed and regulated, its roles in physiological and pathological processes, and the crosstalk between different modes of programmed cell death. We also highlight the development of drugs that target pyroptotic pathways for disease treatment.
Collapse
Affiliation(s)
- Yang Bai
- Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Youdong Pan
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Xing Liu
- Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China.
- Shanghai Academy of Natural Sciences (SANS), Shanghai, China.
| |
Collapse
|
4
|
Winsor NJ, Bayer G, Singh O, Chan JK, Li LY, Lieng BY, Foerster E, Popovic A, Tsankov BK, Maughan H, Lemire P, Tam E, Streutker C, Chen L, Heaver SL, Ley RE, Parkinson J, Montenegro-Burke JR, Birchenough GMH, Philpott DJ, Girardin SE. Cross-kingdom-mediated detection of intestinal protozoa through NLRP6. Cell Host Microbe 2025; 33:388-407.e9. [PMID: 40043701 DOI: 10.1016/j.chom.2025.02.008] [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: 07/08/2024] [Revised: 12/13/2024] [Accepted: 02/10/2025] [Indexed: 03/15/2025]
Abstract
Intestinal protists are detected by the host innate immune system through mechanisms that remain poorly understood. Here, we demonstrate that Tritrichomonas protozoa induce thickening of the colonic mucus in an NLRP6-, ASC-, and caspase-11-dependent manner, consistent with the activation of sentinel goblet cells. Mucus growth is recapitulated with cecal extracts from Tritrichomonas-infected mice but not purified protozoa, suggesting that NLRP6 may detect infection-induced microbial dysbiosis. In agreement, Tritrichomonas infection causes a shift in the microbiota with the expansion of Bacteroides and Prevotella, and untargeted metabolomics reveals a dramatic increase in several classes of metabolites, including sphingolipids. Finally, using a combination of gnotobiotic mice and ex vivo mucus analysis, we demonstrate that wild-type, but not sphingolipid-deficient, B. thetaiotaomicron is sufficient to induce NLRP6-dependent sentinel goblet cell function, with the greatest effect observed in female mice. Thus, we propose that NLRP6 is a sensor of intestinal protozoa infection through monitoring microbial sphingolipids.
Collapse
Affiliation(s)
- Nathaniel J Winsor
- Department of Immunology, University of Toronto, Toronto, ON, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Giuliano Bayer
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Ojas Singh
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Jeremy K Chan
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Lu Yi Li
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Brandon Y Lieng
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | | | - Ana Popovic
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada; Molecular Medicine, Hospital for Sick Children, Toronto, ON, Canada
| | - Boyan K Tsankov
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | | | - Paul Lemire
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Elaine Tam
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | | | - Lina Chen
- Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Stacey L Heaver
- Department of Microbiome Science, Max Planck Institute for Biology, Tübingen, Germany
| | - Ruth E Ley
- Department of Microbiome Science, Max Planck Institute for Biology, Tübingen, Germany
| | - John Parkinson
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada; Department of Biochemistry, University of Toronto, Toronto, ON, Canada; Molecular Medicine, Hospital for Sick Children, Toronto, ON, Canada
| | - J Rafael Montenegro-Burke
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada; Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - George M H Birchenough
- Department of Medical Biochemistry, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Dana J Philpott
- Department of Immunology, University of Toronto, Toronto, ON, Canada.
| | - Stephen E Girardin
- Department of Immunology, University of Toronto, Toronto, ON, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
5
|
Wang Y, Chang Y, Yin F, Kang C, Meng Y, Xu F, Liu Y, Zhang Y, Wu C, Fan S, Zhao J. Structural analyses of Cryptosporidium parvum epitopes reveal a novel scheme of decapeptide binding to H-2K b. J Struct Biol 2025; 217:108168. [PMID: 39809366 DOI: 10.1016/j.jsb.2025.108168] [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: 11/12/2024] [Revised: 01/07/2025] [Accepted: 01/11/2025] [Indexed: 01/16/2025]
Abstract
Cryptosporidium has gained much attention as a major cause of diarrhea worldwide. Here, we present the first structure of H-2Kb complexed with a decapeptide from Cryptosporidium parvum Gp40/15 protein (Gp40/15-VTF10). In contrast to all published structures, the aromatic residue P3-Phe of Gp40/15-VTF10 is anchored in pocket C rather than the canonical Y/F at P5 or P6 reported for octapeptides and nonapeptides. The results of in vitro refolding assays and circular dichroism experiments showed that the side chains of P3 and P5 play key roles in Gp40/15-VTF10 peptide binding. However, functional analysis of decapeptide epitopes revealed that the Gp40/15-VTF10 peptide did not elicit a strong CD8+T immune response, whereas the decapeptide epitope MEDLE2-INF10 induced a significant CD8+ T-cell response in peptide-immunized C57BL/6 mice. Using a model structure of H-2Kb-INF10 complex, we found that the antigenic decapeptide INF10 exhibits a completely different conformation, with the aromatic anchors P3F and P7F docked into the D and C pockets, respectively, while similar peptide conformation and hydrogen bond interactions between the peptide and major histocompatibility complex were found in the resolved H-2Kb-SVF9 complex. As the H-2Kb molecule predominantly prefers octapeptides with a strong anchor of P5 Y/F (or P6 Y/F for nonapeptides) binding to the C pocket, we propose that P7 Y/F in the C pocket may represent a novel binding mode for decapeptides. The results should increase the accuracy of T-cell epitope prediction and support the development of T-cell epitope vaccines against cryptosporidiosis.
Collapse
Affiliation(s)
- Yongli Wang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, PR China; College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou, PR China
| | - Yankai Chang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, PR China
| | - Fangyuan Yin
- College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou, PR China; Institute of Neuroscience and Translational Medicine, Zhoukou Normal University, Zhoukou, PR China
| | - Chunliu Kang
- College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou, PR China; Institute of Neuroscience and Translational Medicine, Zhoukou Normal University, Zhoukou, PR China
| | - Yao Meng
- College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou, PR China; Institute of Neuroscience and Translational Medicine, Zhoukou Normal University, Zhoukou, PR China
| | - Fukang Xu
- College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou, PR China; Institute of Neuroscience and Translational Medicine, Zhoukou Normal University, Zhoukou, PR China
| | - Yiran Liu
- College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou, PR China; Institute of Neuroscience and Translational Medicine, Zhoukou Normal University, Zhoukou, PR China
| | - Yunxia Zhang
- College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou, PR China; Institute of Neuroscience and Translational Medicine, Zhoukou Normal University, Zhoukou, PR China
| | - Changjing Wu
- College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou, PR China; Institute of Neuroscience and Translational Medicine, Zhoukou Normal University, Zhoukou, PR China
| | - Shuhua Fan
- College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou, PR China; Institute of Neuroscience and Translational Medicine, Zhoukou Normal University, Zhoukou, PR China.
| | - Junlong Zhao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, PR China.
| |
Collapse
|
6
|
M Bader S, Scherer L, Schaefer J, Cooney JP, Mackiewicz L, Dayton M, Georgy SR, Davidson KC, Allison CC, Herold MJ, Strasser A, Pellegrini M, Doerflinger M. IL-1β drives SARS-CoV-2-induced disease independently of the inflammasome and pyroptosis signalling. Cell Death Differ 2025:10.1038/s41418-025-01459-x. [PMID: 40016339 DOI: 10.1038/s41418-025-01459-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 01/22/2025] [Accepted: 02/10/2025] [Indexed: 03/01/2025] Open
Abstract
Excessive inflammation and cytokine release are hallmarks of severe COVID-19. Certain programmed cell death processes can drive inflammation, however, their role in the pathogenesis of severe COVID-19 is unclear. Pyroptosis is a pro-inflammatory form of regulated cell death initiated by inflammasomes and executed by the pore-forming protein gasdermin D (GSDMD). Using an established mouse adapted SARS-CoV-2 virus and a panel of gene-targeted mice we found that deletion of the inflammasome (NLRP1/3 and the adaptor ASC) and pore forming proteins involved in pyroptosis (GSDMA/C/D/E) only marginally reduced IL-1β levels and did not impact disease outcome or viral loads. Furthermore, we found that SARS-CoV-2 infection did not trigger GSDMD activation in mouse lungs. Finally, we did not observe any difference between WT animals and mice with compound deficiencies in the pro-inflammatory initiator caspases (C1/11/12-/-). This indicates that the classical canonical and non-canonical pro-inflammatory caspases known to process and activate pro-IL-1β, pro-IL-18 and GSDMD do not substantially contribute to SARS-CoV-2 pathogenesis. However, the loss of IL-1β, but not the absence of IL-18, ameliorated disease and enhanced survival in SARS-CoV-2 infected animals compared to wildtype mice. Collectively, these findings demonstrate that IL-1β is an important factor contributing to severe SARS-CoV-2 disease, but its release was largely independent of inflammasome and pyroptotic pathways.
Collapse
Affiliation(s)
- Stefanie M Bader
- The Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Lena Scherer
- The Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC, 3052, Australia
| | - Jan Schaefer
- The Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - James P Cooney
- The Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Liana Mackiewicz
- The Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC, 3052, Australia
| | - Merle Dayton
- The Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC, 3052, Australia
| | - Smitha Rose Georgy
- Anatomic Pathology-Veterinary Biosciences, Melbourne Veterinary School, University of Melbourne, Werribee, VIC, 3030, Australia
| | - Kathryn C Davidson
- The Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Cody C Allison
- The Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC, 3052, Australia
| | - Marco J Herold
- The Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, Australia
| | - Andreas Strasser
- The Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Marc Pellegrini
- The Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC, 3052, Australia.
- Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia.
- Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW, Australia.
| | - Marcel Doerflinger
- The Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC, 3052, Australia.
- Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
7
|
Ali M, Xu C, Wang M, Hina Q, Ji Y, Anwar S, Lu S, He Q, Qiu Y, Li K. Gut Barrier Dysfunction and Microbiota Variations in Cryptosporidiosis: A Comprehensive Review. Vet Sci 2025; 12:85. [PMID: 40005845 PMCID: PMC11861801 DOI: 10.3390/vetsci12020085] [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: 12/26/2024] [Revised: 01/21/2025] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
Cryptosporidiosis is a zoonotic protozoan parasite-born disease, equally significant in both animals and humans, especially affecting immunocompromised individuals (e.g., AIDS patients) and neonates. The prime concerns of this review article are to demonstrate the disruption of the intestinal barrier and variations in the gut microbiome during cryptosporidiosis, and to explore host gut-parasite interactions that can lead to the development of novel therapeutics. The review concluded that the enteric barrier is particularly maintained by tight junction proteins (e.g., occludin, claudin, and ZO-1, etc.) and mucosal immunity, both of which are severely compromised during Cryptosporidium spp. infections, resulting in increased intestinal barrier permeability, inflammatory responses, diarrhea, and ultimately death in severe cases. Cryptosporidium-induced dysbiosis is characterized by reduced microbial diversity and richness, a shift from commensal to pathogenic bacteria, as evidenced by increased pro-inflammatory taxa like Proteobacteria, and reduced proportions of beneficial SCFAs producing bacteria, e.g., Firmicutes. Recent investigations have highlighted the interrelations between gut microbiota and epithelial barrier integrity, especially during cryptosporidiosis, demonstrating the modulations regarding tight junctions (TJs), immune reactions, and SCFA production, all of which are main players in alleviating this protozoal parasitic infection. This review comprehensively describes the fine details underlying these impairments, including autophagy-mediated TJs' degradation, inflammasome activation, and gut microbiome-driven alterations in metabolic pathways, providing the latest relevant, and well-organized piece of knowledge regarding intestinal barrier alterations and microbial shifts during cryptosporidiosis. This work emphasizes the future need for longitudinal studies and advanced sequencing techniques to understand host gut microbiota-parasite interactions, aiming to formulate innovative strategies to mitigate cryptosporidiosis.
Collapse
Affiliation(s)
- Munwar Ali
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (M.A.)
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Chang Xu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (M.A.)
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Mingyue Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (M.A.)
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Qazal Hina
- Department of Animal Nutrition, University of Veterinary and Animal Sciences, Lahore 54000, Pakistan
| | - Yaru Ji
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (M.A.)
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Subiha Anwar
- Department of Animal Husbandry, University of Agriculture, Faisalabad 38000, Pakistan
| | - Sijia Lu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (M.A.)
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Qing He
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (M.A.)
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Yawei Qiu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (M.A.)
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Kun Li
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (M.A.)
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
8
|
Pandey A, Li Z, Gautam M, Ghosh A, Man SM. Molecular mechanisms of emerging inflammasome complexes and their activation and signaling in inflammation and pyroptosis. Immunol Rev 2025; 329:e13406. [PMID: 39351983 PMCID: PMC11742652 DOI: 10.1111/imr.13406] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Inflammasomes are multi-protein complexes that assemble within the cytoplasm of mammalian cells in response to pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs), driving the secretion of the pro-inflammatory cytokines IL-1β and IL-18, and pyroptosis. The best-characterized inflammasome complexes are the NLRP3, NAIP-NLRC4, NLRP1, AIM2, and Pyrin canonical caspase-1-containing inflammasomes, and the caspase-11 non-canonical inflammasome. Newer inflammasome sensor proteins have been identified, including NLRP6, NLRP7, NLRP9, NLRP10, NLRP11, NLRP12, CARD8, and MxA. These inflammasome sensors can sense PAMPs from bacteria, viruses and protozoa, or DAMPs in the form of mitochondrial damage, ROS, stress and heme. The mechanisms of action, physiological relevance, consequences in human diseases, and avenues for therapeutic intervention for these novel inflammasomes are beginning to be realized. Here, we discuss these emerging inflammasome complexes and their putative activation mechanisms, molecular and signaling pathways, and physiological roles in health and disease.
Collapse
Affiliation(s)
- Abhimanu Pandey
- Division of Immunology and Infectious Diseases, The John Curtin School of Medical ResearchThe Australian National UniversityCanberraAustralia
| | - Zheyi Li
- Division of Immunology and Infectious Diseases, The John Curtin School of Medical ResearchThe Australian National UniversityCanberraAustralia
| | - Manjul Gautam
- Division of Immunology and Infectious Diseases, The John Curtin School of Medical ResearchThe Australian National UniversityCanberraAustralia
| | - Aritra Ghosh
- Division of Immunology and Infectious Diseases, The John Curtin School of Medical ResearchThe Australian National UniversityCanberraAustralia
| | - Si Ming Man
- Division of Immunology and Infectious Diseases, The John Curtin School of Medical ResearchThe Australian National UniversityCanberraAustralia
| |
Collapse
|
9
|
Huang Y, Pei S, Lv X, Yang F, Gong X, Li N, Guo Y, Feng Y, Xiao L. Stage-specific expression and divergent functions of two insulinase-like proteases associated with host infectivity in Cryptosporidium. PLoS Negl Trop Dis 2025; 19:e0012777. [PMID: 39804945 PMCID: PMC11760560 DOI: 10.1371/journal.pntd.0012777] [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: 08/24/2024] [Revised: 01/24/2025] [Accepted: 12/10/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND The determinants of differences in host infectivity among Cryptosporidium species and subtypes are poorly understood. Results from recent comparative genomic studies suggest that gains and losses of multicopy subtelomeric genes encoding insulinase-like proteases (INS-19 and INS-20 in Cryptosporidium parvum and their orthologs in closely related species) may potentially contribute to these differences. METHODOLOGY/PRINCIPAL FINDINGS In this study, we investigated the expression and biological function of the INS-19 and INS-20 of C. parvum. CRISPR/Cas9 was used to endogenously tag both genes with the hemagglutinin epitope. Immunofluorescence analysis revealed that INS-19 and INS-20 are expressed at different developmental stages of the pathogen. Although knockout of either had no detectable effect on the in vitro growth of C. parvum, knockout of INS-20, deletion of its multiple domains, or mutation of the active motif in the functional domain reduced the intensity of C. parvum infection in IFN-γ knockout mice. Consistent with this, mice infected with the INS-20-deleted mutant had reduced intestinal damage and parasite burden. CONCLUSIONS/SIGNIFICANCE These results suggest that INS-19 and INS-20 have stage-specific expression with distinct biological functions, and that the presence of the INS-20 in zoonotic C. parvum contributes to its infectivity and fitness in mice.
Collapse
Affiliation(s)
- Yue Huang
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
| | - Shifeng Pei
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
| | - Xin Lv
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
| | - Fuxian Yang
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
| | - Xiaoqing Gong
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
| | - Na Li
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
| | - Yaqiong Guo
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
| | - Yaoyu Feng
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Center for Emerging and Zoonotic Diseases, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Lihua Xiao
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Center for Emerging and Zoonotic Diseases, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| |
Collapse
|
10
|
Wang ZX, Jiao WJ, Yang Y, Liu HL, Wang HL. Role of inflammasomes in Toxoplasma and Plasmodium infections. Parasit Vectors 2024; 17:466. [PMID: 39548522 PMCID: PMC11566176 DOI: 10.1186/s13071-024-06529-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 10/08/2024] [Indexed: 11/18/2024] Open
Abstract
BACKGROUND The detection of pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs) by multimeric protein complexes, known as inflammasomes, triggers an inflammatory response, which is a critical component of the innate immune system. This inflammatory response plays a pivotal role in host resistance against parasitic infections, presenting a significant global health challenge. METHODS We systematically searched for relevant articles from the Pubmed and the Web of Science database to summarize current insights into how inflammasomes function in preventing infections caused by the apicomplexan parasites Toxoplasma and Plasmodium. RESULTS In vivo and in vitro studies have extensively explored inflammasomes such as the absent in melanoma 2 (AIM2), NLR family pyrin-containing protein 1 (NLRP1), NLRP3, and NLRP12 inflammasomes, alongside noncanonical inflammasomes, with particular emphasis on the NLRP1 and the NLRP3 inflammasome during Toxoplasma gondii infection or the AIM2 and the NLRP3 inflammasome at various stages of Plasmodium infection. Toxoplasma gondii interacts with inflammasomes to activate or inhibit immune responses. CONCLUSIONS Inflammasomes control parasite burden and parasite-induced cell death, contribute to immune recognition and inflammatory responses and thus influence apicomplexan parasite-associated pathogenesis and the severity of clinical outcomes. Hence, inflammasomes play crucial roles in the progression and outcomes of toxoplasmosis and malaria. A comprehensive understanding of how parasitic infections modulate inflammasome activity enhances insight into host immune responses against parasites.
Collapse
Affiliation(s)
- Zhi-Xin Wang
- School of Basic Medicine, Basic Medical Sciences Center, Shanxi Medical University, Jinzhong, 030600, Shanxi, China
| | - Wan-Jun Jiao
- School of Basic Medicine, Basic Medical Sciences Center, Shanxi Medical University, Jinzhong, 030600, Shanxi, China
| | - Yong Yang
- School of Basic Medicine, Basic Medical Sciences Center, Shanxi Medical University, Jinzhong, 030600, Shanxi, China
| | - Hong-Li Liu
- School of Basic Medicine, Basic Medical Sciences Center, Shanxi Medical University, Jinzhong, 030600, Shanxi, China.
| | - Hai-Long Wang
- School of Basic Medicine, Basic Medical Sciences Center, Shanxi Medical University, Jinzhong, 030600, Shanxi, China.
| |
Collapse
|
11
|
Babcock IW, Sibley LA, Labuzan SA, Cowan MN, Sethi I, Alemu S, Kelly AG, Kovacs MA, Lukens JR, Harris TH. Caspase-1 in Cx3cr1-expressing cells drives an IL-18-dependent T cell response that promotes parasite control during acute Toxoplasma gondii infection. PLoS Pathog 2024; 20:e1012006. [PMID: 39446964 PMCID: PMC11537422 DOI: 10.1371/journal.ppat.1012006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 11/05/2024] [Accepted: 10/13/2024] [Indexed: 10/26/2024] Open
Abstract
Inflammasome activation is a robust innate immune mechanism that promotes inflammatory responses through the release of alarmins and leaderless cytokines, including IL-1α, IL-1β, and IL-18. Various stimuli, including infectious agents and cellular stress, cause inflammasomes to assemble and activate caspase-1. Then, caspase-1 cleaves targets that lead to pore formation and leaderless cytokine activation and release. Toxoplasma gondii has been shown to promote inflammasome formation, but the cell types utilizing caspase-1 and the downstream effects on immunological outcomes during acute in vivo infection have not been explored. Here, using knockout mice, we examine the role of caspase-1 responses during acute T. gondii infection globally and in Cx3cr1-positive populations. We provide in vivo evidence that caspase-1 expression is critical for, IL-18 release, optimal interferon-γ (IFN-γ) production, monocyte and neutrophil recruitment to the site of infection, and parasite control. Specifically, we find that caspase-1 expression in Cx3cr1-positive cells drives IL-18 release, which potentiates CD4+ T cell IFN-γ production and parasite control. Notably, our Cx3cr1-Casp1 knockouts exhibited a selective T cell defect, mirroring the phenotype observed in Il18 knockouts. In further support of this finding, treatment of Cx3cr1-Casp1 knockout mice with recombinant IL-18 restored CD4+ T cell IFN-γ responses and parasite control. Additionally, we show that neutrophil recruitment is dependent on IL-1 receptor accessory protein (IL-1RAP) signaling but is dispensable for parasite control. Overall, these experiments highlight the multifaceted role of caspase-1 in multiple cell populations contributing to specific pathways that collectively contribute to caspase-1 dependent immunity to T. gondii.
Collapse
Affiliation(s)
- Isaac W. Babcock
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Lydia A. Sibley
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Sydney A. Labuzan
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Maureen N. Cowan
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Ish Sethi
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Seblework Alemu
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Abigail G. Kelly
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Michael A. Kovacs
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - John R. Lukens
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Tajie H. Harris
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| |
Collapse
|
12
|
Wallbank BA, Pardy RD, Brodsky IE, Hunter CA, Striepen B. Cryptosporidium impacts epithelial turnover and is resistant to induced death of the host cell. mBio 2024; 15:e0172024. [PMID: 38995074 PMCID: PMC11323733 DOI: 10.1128/mbio.01720-24] [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: 06/10/2024] [Accepted: 06/13/2024] [Indexed: 07/13/2024] Open
Abstract
Infection with the apicomplexan parasite Cryptosporidium is a leading cause of diarrheal disease. Cryptosporidiosis is of particular importance in infants and shows a strong association with malnutrition, both as a risk factor and as a consequence. Cryptosporidium invades and replicates within the small intestine epithelial cells. This is a highly dynamic tissue that is developmentally stratified along the villus axis. New cells emerge from a stem cell niche in the crypt and differentiate into mature epithelial cells while moving toward the villus tip, where they are ultimately shed. Here, we studied the impact of Cryptosporidium infection on this dynamic architecture. Tracing DNA synthesis in pulse-chase experiments in vivo, we quantified the genesis and migration of epithelial cells along the villus. We found proliferation and epithelial migration to be elevated in response to Cryptosporidium infection. Infection also resulted in significant cell loss documented by imaging and molecular assays. Consistent with these observations, single-cell RNA sequencing of infected intestines showed a gain of young and a loss of mature cells. Interestingly, enhanced epithelial cell loss was not a function of enhanced apoptosis of infected cells. To the contrary, Cryptosporidium-infected cells were less likely to be apoptotic than bystanders, and experiments in tissue culture demonstrated that infection provided enhanced resistance to chemically induced apoptosis to the host but not bystander cells. Overall, this study suggests that Cryptosporidium may modulate cell apoptosis and documents pronounced changes in tissue homeostasis due to parasite infection, which may contribute to its long-term impact on the developmental and nutritional state of children. IMPORTANCE The intestine must balance its roles in digestion and nutrient absorption with the maintenance of an effective barrier to colonization and breach by numerous potential pathogens. An important component of this balance is its constant turnover, which is modulated by a gain of cells due to proliferation and loss due to death or extrusion. Here, we report that Cryptosporidium infection changes the dynamics of this process increasing both gain and loss of enterocytes speeding up the villus elevator. This leads to a much more immature epithelium and a reduction of the number of those cells typically found toward the villus apex best equipped to take up key nutrients including carbohydrates and lipids. These changes in the cellular architecture and physiology of the small intestine may be linked to the profound association between cryptosporidiosis and malnutrition.
Collapse
Affiliation(s)
- Bethan A. Wallbank
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ryan D. Pardy
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Igor E. Brodsky
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Christopher A. Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Boris Striepen
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
13
|
Cohn IS, Wallbank BA, Haskins BE, O’Dea KM, Pardy RD, Shaw S, Merolle MI, Gullicksrud JA, Christian DA, Striepen B, Hunter CA. Intestinal cDC1s provide cues required for CD4+ T cell-mediated resistance to Cryptosporidium. J Exp Med 2024; 221:e20232067. [PMID: 38829369 PMCID: PMC11148471 DOI: 10.1084/jem.20232067] [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: 11/10/2023] [Revised: 04/01/2024] [Accepted: 05/06/2024] [Indexed: 06/05/2024] Open
Abstract
Cryptosporidium is an enteric pathogen and a prominent cause of diarrheal disease worldwide. Control of Cryptosporidium requires CD4+ T cells, but how protective CD4+ T cell responses are generated is poorly understood. Here, Cryptosporidium parasites that express MHCII-restricted model antigens were generated to understand the basis for CD4+ T cell priming and effector function. These studies revealed that parasite-specific CD4+ T cells are primed in the draining mesenteric lymph node but differentiate into Th1 cells in the gut to provide local parasite control. Although type 1 conventional dendritic cells (cDC1s) were dispensable for CD4+ T cell priming, they were required for CD4+ T cell gut homing and were a source of IL-12 at the site of infection that promoted local production of IFN-γ. Thus, cDC1s have distinct roles in shaping CD4+ T cell responses to an enteric infection: first, to promote gut homing from the mesLN, and second, to drive effector responses in the intestine.
Collapse
Affiliation(s)
- Ian S. Cohn
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bethan A. Wallbank
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Breanne E. Haskins
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Keenan M. O’Dea
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ryan D. Pardy
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sebastian Shaw
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Maria I. Merolle
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jodi A. Gullicksrud
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - David A. Christian
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Boris Striepen
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christopher A. Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
14
|
Alipour S, Mardi A, Shajari N, Kazemi T, Sadeghi MR, Ahmadian Heris J, Masoumi J, Baradaran B. Unmasking the NLRP3 inflammasome in dendritic cells as a potential therapeutic target for autoimmunity, cancer, and infectious conditions. Life Sci 2024; 348:122686. [PMID: 38710282 DOI: 10.1016/j.lfs.2024.122686] [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: 01/17/2024] [Revised: 04/13/2024] [Accepted: 05/03/2024] [Indexed: 05/08/2024]
Abstract
Proper and functional immune response requires a complex interaction between innate and adaptive immune cells, which dendritic cells (DCs) are the primary actors in this coordination as professional antigen-presenting cells. DCs are armed with numerous pattern recognition receptors (PRRs) such as nucleotide-binding and oligomerization domain-like receptors (NLRs) like NLRP3, which influence the development of their activation state upon sensation of ligands. NLRP3 is a crucial component of the immune system for protection against tumors and infectious agents, because its activation leads to the assembly of inflammasomes that cause the formation of active caspase-1 and stimulate the maturation and release of proinflammatory cytokines. But, when NLRP3 becomes overactivated, it plays a pathogenic role in the progression of several autoimmune disorders. So, NLRP3 activation is strictly regulated by diverse signaling pathways that are mentioned in detail in this review. Furthermore, the role of NLRP3 in all of the diverse immune cells' subsets is briefly mentioned in this study because NLRP3 plays a pivotal role in modulating other immune cells which are accompanied by DCs' responses and subsequently influence differentiation of T cells to diverse T helper subsets and even impact on cytotoxic CD8+ T cells' responses. This review sheds light on the functional and therapeutic role of NLRP3 in DCs and its contribution to the occurrence and progression of autoimmune disorders, prevention of diverse tumors' development, and recognition and annihilation of various infectious agents. Furthermore, we highlight NLRP3 targeting potential for improving DC-based immunotherapeutic approaches, to be used for the benefit of patients suffering from these disorders.
Collapse
Affiliation(s)
- Shiva Alipour
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amirhossein Mardi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Neda Shajari
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tohid Kazemi
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Reza Sadeghi
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Javad Masoumi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
15
|
Haskins BE, Gullicksrud JA, Wallbank BA, Dumaine JE, Guérin A, Cohn IS, O'Dea KM, Pardy RD, Merolle MI, Shallberg LA, Hunter EN, Byerly JH, Smith EJ, Buenconsejo GY, McLeod BI, Christian DA, Striepen B, Hunter CA. Dendritic cell-mediated responses to secreted Cryptosporidium effectors promote parasite-specific CD8 + T cell responses. Mucosal Immunol 2024; 17:387-401. [PMID: 38508522 PMCID: PMC11193387 DOI: 10.1016/j.mucimm.2024.03.003] [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: 08/16/2023] [Revised: 03/04/2024] [Accepted: 03/05/2024] [Indexed: 03/22/2024]
Abstract
Cryptosporidium causes debilitating diarrheal disease in patients with primary and acquired defects in T cell function. However, it has been a challenge to understand how this infection generates T cell responses and how they mediate parasite control. Here, Cryptosporidium was engineered to express a parasite effector protein (MEDLE-2) that contains the major histocompatibility complex-I restricted SIINFEKL epitope which is recognized by T cell receptor transgenic OT-I(OVA-TCR-I) clusters of differentiation (CD)8+ T cells. These modified parasites induced expansion of endogenous SIINFEKL-specific and OT-I CD8+ T cells that were a source of interferon-gamma (IFN-γ) that could restrict growth of Cryptosporidium. This T cell response was dependent on the translocation of the effector and similar results were observed with another secreted parasite effector (rhoptry protein 1). Although infection and these translocated effector proteins are restricted to intestinal epithelial cells, type 1 conventional dendritic cells were required to generate CD8+ T cell responses to these model antigens. These data sets highlight Cryptosporidium effectors as potential targets of the immune system and suggest that crosstalk between enterocytes and type 1 conventional dendritic cells is crucial for CD8+ T cell responses to Cryptosporidium.
Collapse
Affiliation(s)
- Breanne E Haskins
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, USA
| | - Jodi A Gullicksrud
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, USA; Cell Press, Cambridge, Massachusetts, USA
| | - Bethan A Wallbank
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, USA
| | - Jennifer E Dumaine
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, USA
| | - Amandine Guérin
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, USA
| | - Ian S Cohn
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, USA
| | - Keenan M O'Dea
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, USA
| | - Ryan D Pardy
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, USA
| | - Maria I Merolle
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, USA
| | - Lindsey A Shallberg
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, USA
| | - Emma N Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, USA
| | - Jessica H Byerly
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, USA
| | - Eleanor J Smith
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, USA
| | - Gracyn Y Buenconsejo
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, USA
| | - Briana I McLeod
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, USA
| | - David A Christian
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, USA
| | - Boris Striepen
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, USA
| | - Christopher A Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, USA.
| |
Collapse
|
16
|
Pardy RD, Walzer KA, Wallbank BA, Byerly JH, O’Dea KM, Cohn IS, Haskins BE, Roncaioli JL, Smith EJ, Buenconsejo GY, Striepen B, Hunter CA. Analysis of intestinal epithelial cell responses to Cryptosporidium highlights the temporal effects of IFN-γ on parasite restriction. PLoS Pathog 2024; 20:e1011820. [PMID: 38718306 PMCID: PMC11078546 DOI: 10.1371/journal.ppat.1011820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 04/14/2024] [Indexed: 05/12/2024] Open
Abstract
The production of IFN-γ is crucial for control of multiple enteric infections, but its impact on intestinal epithelial cells (IEC) is not well understood. Cryptosporidium parasites exclusively infect epithelial cells and the ability of interferons to activate the transcription factor STAT1 in IEC is required for parasite clearance. Here, the use of single cell RNA sequencing to profile IEC during infection revealed an increased proportion of mid-villus enterocytes during infection and induction of IFN-γ-dependent gene signatures that was comparable between uninfected and infected cells. These analyses were complemented by in vivo studies, which demonstrated that IEC expression of the IFN-γ receptor was required for parasite control. Unexpectedly, treatment of Ifng-/- mice with IFN-γ showed the IEC response to this cytokine correlates with a delayed reduction in parasite burden but did not affect parasite development. These data sets provide insight into the impact of IFN-γ on IEC and suggest a model in which IFN-γ signalling to uninfected enterocytes is important for control of Cryptosporidium.
Collapse
Affiliation(s)
- Ryan D. Pardy
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Katelyn A. Walzer
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Bethan A. Wallbank
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jessica H. Byerly
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Keenan M. O’Dea
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Ian S. Cohn
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Breanne E. Haskins
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Justin L. Roncaioli
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Eleanor J. Smith
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Gracyn Y. Buenconsejo
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Boris Striepen
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Christopher A. Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
17
|
Deng Q, Yang S, Huang K, Zhu Y, Sun L, Cao Y, Dong K, Li Y, Wu S, Huang R. NLRP6 induces RIP1 kinase-dependent necroptosis via TAK1-mediated p38 MAPK/MK2 phosphorylation in S. typhimurium infection. iScience 2024; 27:109339. [PMID: 38500819 PMCID: PMC10945251 DOI: 10.1016/j.isci.2024.109339] [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: 04/23/2023] [Revised: 11/16/2023] [Accepted: 02/22/2024] [Indexed: 03/20/2024] Open
Abstract
Programmed cell death (PCD) is tightly orchestrated by molecularly defined executors and signaling pathways. NLRP6, a member of cytoplasmic pattern recognition receptors, has a multifaceted role in host resistance to bacterial infection. However, whether and how NLRP6 may contribute to regulate host PCD during Gram-negative bacterial infection remain to be illuminated. Here, we report that NLRP6 promotes RIP1 kinase-mediated necroptosis, a form of lytic PCD, in both an in vitro and in vivo model of Salmonella typhimurium infection. By downregulating TAK1-mediated p38MAPK/MK2 phosphorylation, NLRP6 decreased RIP1 phosphorylation at residue S321 and subsequently increased RIP1 kinase-dependent MLKL phosphorylation. Suppression of p38MAPK/MK2 cascade not only reduced the number of dead cells caused by NLRP6 but also decreased the systemic dissemination of S. typhimurium resulting from NLRP6. Taken together, our findings provide new insights into the role and regulatory mechanism of NLRP6-associated antimicrobial responses by revealing a function for NLRP6 in regulating necroptosis.
Collapse
Affiliation(s)
- Qifeng Deng
- Department of Medical Microbiology, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Key Laboratory of Pathogen Bioscience and Anti-infective Medicine, School of Biology & Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215123, P.R. China
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510275, P.R. China
| | - Sidi Yang
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong 510005, P.R. China
| | - Kai Huang
- Orthopaedic Institute, Wuxi 9th People’s Hospital Affiliated to Soochow University, Wuxi, Jiangsu 214062, P.R. China
| | - Yuan Zhu
- Department of Medical Microbiology, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Key Laboratory of Pathogen Bioscience and Anti-infective Medicine, School of Biology & Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215123, P.R. China
- Department of Laboratory Medicine, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, P.R. China
| | - Lanqing Sun
- Department of Medical Microbiology, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Key Laboratory of Pathogen Bioscience and Anti-infective Medicine, School of Biology & Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215123, P.R. China
- Department of Laboratory Medicine, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214000, P.R. China
| | - Yu Cao
- Department of Medical Microbiology, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Key Laboratory of Pathogen Bioscience and Anti-infective Medicine, School of Biology & Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Kedi Dong
- Department of Medical Microbiology, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Key Laboratory of Pathogen Bioscience and Anti-infective Medicine, School of Biology & Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215123, P.R. China
- Department of Blood Transfusion, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, P.R. China
| | - Yuanyuan Li
- Experimental Center, Suzhou Medical College of Soochow University, No. 199, Ren Ai Road, Suzhou, Jiangsu 215123, P.R. China
| | - Shuyan Wu
- Department of Medical Microbiology, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Key Laboratory of Pathogen Bioscience and Anti-infective Medicine, School of Biology & Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Rui Huang
- Department of Medical Microbiology, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Key Laboratory of Pathogen Bioscience and Anti-infective Medicine, School of Biology & Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215123, P.R. China
| |
Collapse
|
18
|
Ewald S, Nasuhidehnavi A, Feng TY, Lesani M, McCall LI. The intersection of host in vivo metabolism and immune responses to infection with kinetoplastid and apicomplexan parasites. Microbiol Mol Biol Rev 2024; 88:e0016422. [PMID: 38299836 PMCID: PMC10966954 DOI: 10.1128/mmbr.00164-22] [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] [Indexed: 02/02/2024] Open
Abstract
SUMMARYProtozoan parasite infection dramatically alters host metabolism, driven by immunological demand and parasite manipulation strategies. Immunometabolic checkpoints are often exploited by kinetoplastid and protozoan parasites to establish chronic infection, which can significantly impair host metabolic homeostasis. The recent growth of tools to analyze metabolism is expanding our understanding of these questions. Here, we review and contrast host metabolic alterations that occur in vivo during infection with Leishmania, trypanosomes, Toxoplasma, Plasmodium, and Cryptosporidium. Although genetically divergent, there are commonalities among these pathogens in terms of metabolic needs, induction of the type I immune responses required for clearance, and the potential for sustained host metabolic dysbiosis. Comparing these pathogens provides an opportunity to explore how transmission strategy, nutritional demand, and host cell and tissue tropism drive similarities and unique aspects in host response and infection outcome and to design new strategies to treat disease.
Collapse
Affiliation(s)
- Sarah Ewald
- Department of Microbiology, Immunology, and Cancer Biology at the Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Azadeh Nasuhidehnavi
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma, USA
| | - Tzu-Yu Feng
- Department of Microbiology, Immunology, and Cancer Biology at the Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Mahbobeh Lesani
- Department of Microbiology and Plant Biology, University of Oklahoma, Norman, Oklahoma, USA
| | - Laura-Isobel McCall
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma, USA
- Department of Microbiology and Plant Biology, University of Oklahoma, Norman, Oklahoma, USA
- Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, Oklahoma, USA
- Department of Chemistry and Biochemistry, San Diego State University, San Diego, California, USA
| |
Collapse
|
19
|
Shalaby NE, Shoheib ZS, Yassin NA, El-Kaliny HH, Hasby Saad MA. Pyroptosis Tuning in Intestinal Cryptosporidiosis via the Natural Histone Deacetylase Inhibitor Romidepsin. Parasite Immunol 2024; 46:e13032. [PMID: 38497997 DOI: 10.1111/pim.13032] [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: 09/04/2023] [Revised: 02/13/2024] [Accepted: 02/17/2024] [Indexed: 03/19/2024]
Abstract
Cryptosporidium is an opportunistic protozoan, with many species of cross-human infectivity. It causes life-threatening diarrhoea in children and CD4-defective patients. Despite its limited efficacy, nitazoxanide remains the primary anti-cryptosporidial drug. Cryptosporidium infects the intestinal brush border (intracellular-extracytoplasmic) and down-regulates pyroptosis to prevent expulsion. Romidepsin is a natural histone deacetylase inhibitor that triggers pyroptosis. Romidepsin's effect on cryptosporidiosis was assessed in immunocompromised mice via gasdermin-D (GSDM-D) immunohistochemical expression, IFN-γ, IL-1β and IL-18 blood levels by ELISA, and via parasite scanning by modified Ziehl-Neelsen staining and scanning electron microscopy (SEM). Oocyst deformity and local cytokines were also assessed in ex vivo ileal explants. Following intraperitoneal injection of romidepsin, oocyst shedding significantly reduced at the 9th, 12th and 15th d.p.i. compared with infected-control and drug-control (nitazoxanide-treated) mice. H&E staining of intestinal sections from romidepsin-treated mice showed significantly low intestinal scoring with marked reduction in epithelial hyperplasia, villous blunting and cellular infiltrate. SEM revealed marked oocyst blebbing and paucity (in vivo and ex vivo) after romidepsin compared with nitazoxanide. Regarding pyroptosis, romidepsin triggered significantly higher intestinal GSDM-D expression in vivo, and higher serum/culture IFN-γ, IL-1β and IL-18 levels in romidepsin-treated mice than in the control groups. Collectively, in cryptosporidiosis, romidepsin succeeded in enhancing pyroptosis in the oocysts and infected epithelium, reducing infection and shifting the brush border towards normalisation.
Collapse
Affiliation(s)
- Noha E Shalaby
- Medical Parasitology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Zeinab S Shoheib
- Medical Parasitology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Nabila A Yassin
- Medical Parasitology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Heba H El-Kaliny
- Histology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
- Anatomy and Histology Department, Mutah University, Mutah, Jordan
| | - Marwa A Hasby Saad
- Medical Parasitology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| |
Collapse
|
20
|
Guo Y, Song J, Yan M, Chen Y, Huang L, Liu J, He Y, Lü Y, Yu W. The role of NLRP6 in the development and progression of neurological diseases. Mol Biol Rep 2024; 51:351. [PMID: 38400865 DOI: 10.1007/s11033-024-09293-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 01/29/2024] [Indexed: 02/26/2024]
Abstract
The nervous system possesses the remarkable ability to undergo changes in order to store information; however, it is also susceptible to damage caused by invading pathogens or neurodegenerative processes. As a member of nucleotide-binding oligomerization domain-like receptor (NLR) family, the NLRP6 inflammasome serves as a cytoplasmic innate immune sensor responsible for detecting microbe-associated molecular patterns. Upon activation, NLRP6 can recruit the adapter protein apoptosis-associated speck-like protein (ASC) and the inflammatory factors caspase-1 or caspase-11. Consequently, inflammasomes are formed, facilitating the maturation and secretion of pro-inflammatory cytokines such as inflammatory factors-18 (IL-18) and inflammatory factors-1β (IL-1β). Precise regulation of NLRP6 is crucial for maintaining tissue homeostasis, as dysregulated inflammasome activation can contribute to the development of various diseases. Furthermore, NLRP6 may also play a role in the regulation of extraintestinal diseases. In cells of the brain, such as astrocytes and neurons, NLRP6 inflammasome are also present. Here, the assembly and subsequent activation of caspase-1 mediated by NLRP6 contribute to disease progression. This review aims to discuss the structure and function of NLRP6, explain clearly the mechanisms that induce and activate NLRP6, and explore its role within the central and peripheral nervous system.
Collapse
Affiliation(s)
- Yiming Guo
- Institute of Neuroscience, Chongqing Medical University, No. 1, Yixuayuan Road, Yuzhong District, Chongqing, 400016, China
| | - Jiaqi Song
- Institute of Neuroscience, Chongqing Medical University, No. 1, Yixuayuan Road, Yuzhong District, Chongqing, 400016, China
| | - Mengyu Yan
- Institute of Neuroscience, Chongqing Medical University, No. 1, Yixuayuan Road, Yuzhong District, Chongqing, 400016, China
| | - Yingxi Chen
- Institute of Neuroscience, Chongqing Medical University, No. 1, Yixuayuan Road, Yuzhong District, Chongqing, 400016, China
| | - Lihong Huang
- Institute of Neuroscience, Chongqing Medical University, No. 1, Yixuayuan Road, Yuzhong District, Chongqing, 400016, China
| | - Jiarui Liu
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yurou He
- Institute of Neuroscience, Chongqing Medical University, No. 1, Yixuayuan Road, Yuzhong District, Chongqing, 400016, China
| | - Yang Lü
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Weihua Yu
- Institute of Neuroscience, Chongqing Medical University, No. 1, Yixuayuan Road, Yuzhong District, Chongqing, 400016, China.
| |
Collapse
|
21
|
Pardy RD, Wallbank BA, Striepen B, Hunter CA. Immunity to Cryptosporidium: insights into principles of enteric responses to infection. Nat Rev Immunol 2024; 24:142-155. [PMID: 37697084 PMCID: PMC11881751 DOI: 10.1038/s41577-023-00932-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/28/2023] [Indexed: 09/13/2023]
Abstract
Cryptosporidium parasites replicate within intestinal epithelial cells and are an important cause of diarrhoeal disease in young children and in patients with primary and acquired defects in T cell function. This Review of immune-mediated control of Cryptosporidium highlights advances in understanding how intestinal epithelial cells detect this infection, the induction of innate resistance and the processes required for activation of T cell responses that promote parasite control. The development of a genetic tool set to modify Cryptosporidium combined with tractable mouse models provide new opportunities to understand the principles that govern the interface between intestinal epithelial cells and the immune system that mediate resistance to enteric pathogens.
Collapse
Affiliation(s)
- Ryan D Pardy
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bethan A Wallbank
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Boris Striepen
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Christopher A Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
22
|
Greigert V, Saraav I, Son J, Zhu Y, Dayao D, Antia A, Tzipori S, Witola WH, Stappenbeck TS, Ding S, Sibley LD. Cryptosporidium infection of human small intestinal epithelial cells induces type III interferon and impairs infectivity of Rotavirus. Gut Microbes 2024; 16:2297897. [PMID: 38189373 PMCID: PMC10793699 DOI: 10.1080/19490976.2023.2297897] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 12/18/2023] [Indexed: 01/09/2024] Open
Abstract
Cryptosporidiosis is a major cause of severe diarrheal disease in infants from resource poor settings. The majority of infections are caused by the human-specific pathogen C. hominis and absence of in vitro growth platforms has limited our understanding of host-pathogen interactions and development of effective treatments. To address this problem, we developed a stem cell-derived culture system for C. hominis using human enterocytes differentiated under air-liquid interface (ALI) conditions. Human ALI cultures supported robust growth and complete development of C. hominis in vitro including all life cycle stages. Cryptosporidium infection induced a strong interferon response from enterocytes, possibly driven, in part, by an endogenous dsRNA virus in the parasite. Prior infection with Cryptosporidium induced type III IFN secretion and consequently blunted infection with Rotavirus, including live attenuated vaccine strains. The development of hALI provides a platform for further studies on human-specific pathogens, including clinically important coinfections that may alter vaccine efficacy.
Collapse
Affiliation(s)
- Valentin Greigert
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Iti Saraav
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Juhee Son
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Yinxing Zhu
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Denise Dayao
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA, USA
| | - Avan Antia
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Saul Tzipori
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA, USA
| | - William H. Witola
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Thaddeus S. Stappenbeck
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Siyuan Ding
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - L. David Sibley
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
23
|
Pardy RD, Walzer KA, Wallbank BA, Byerly JH, O’Dea KM, Cohn IS, Haskins BE, Roncaioli JL, Smith EJ, Buenconsejo GY, Striepen B, Hunter CA. Analysis of intestinal epithelial cell responses to Cryptosporidium highlights the temporal effects of IFN-γ on parasite restriction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.14.567008. [PMID: 38014210 PMCID: PMC10680692 DOI: 10.1101/2023.11.14.567008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
The production of IFN-γ is crucial for control of multiple enteric infections, but its impact on intestinal epithelial cells (IEC) is not well understood. Cryptosporidium parasites exclusively infect epithelial cells and the ability of interferons to activate the transcription factor STAT1 in IEC is required for parasite clearance. The use of single cell RNA sequencing to profile IEC during infection revealed induction of IFN-γ-dependent gene signatures that was comparable between uninfected and infected cells, and IEC expression of the IFN-γ receptor was required for parasite control. Unexpectedly, treatment of Ifng-/- mice with IFN-γ demonstrated the IEC response to this cytokine correlates with a delayed reduction in parasite burden but did not affect parasite development. These data sets provide insight into the impact of IFN-γ on IEC and suggest a model in which IFN-γ-mediated bystander activation of uninfected enterocytes is important for control of Cryptosporidium.
Collapse
Affiliation(s)
- Ryan D. Pardy
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Katelyn A. Walzer
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bethan A. Wallbank
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jessica H. Byerly
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Keenan M. O’Dea
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ian S. Cohn
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Breanne E. Haskins
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Justin L. Roncaioli
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Eleanor J. Smith
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gracyn Y. Buenconsejo
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Boris Striepen
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christopher A. Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
24
|
Cohn IS, Wallbank BA, Haskins BE, O’Dea KM, Pardy RD, Shaw S, Merolle MI, Gullicksrud JA, Christian DA, Striepen B, Hunter CA. Intestinal cDC1s provide IL-12 dependent and independent functions required for CD4 + T cell-mediated resistance to Cryptosporidium. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.11.566669. [PMID: 38014026 PMCID: PMC10680586 DOI: 10.1101/2023.11.11.566669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Cryptosporidium is an enteric pathogen that is a prominent cause of diarrheal disease. Control of this infection requires CD4+ T cells, though the processes that lead to T cell-mediated resistance have been difficult to assess. Here, Cryptosporidium parasites that express MHCII-restricted model antigens were generated to dissect the early events that influence CD4+ T cell priming and effector function. These studies highlight that parasite-specific CD4+ T cells are primed in the draining mesenteric lymph node (mesLN) and differentiate into Th1 cells in the gut, where they mediate IFN-γ-dependent control of the infection. Although type 1 conventional dendritic cells (cDC1s) were not required for initial priming of CD4+ T cells, cDC1s were required for CD4+ T cell expansion and gut homing. cDC1s were also a major source of IL-12 that was not required for priming but promoted full differentiation of CD4+ T cells and local production of IFN-γ. Together, these studies reveal distinct roles for cDC1s in shaping CD4+ T cell responses to enteric infection: first to drive early expansion in the mesLN and second to drive effector responses in the gut.
Collapse
Affiliation(s)
- Ian S. Cohn
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bethan A. Wallbank
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Breanne E. Haskins
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Keenan M. O’Dea
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ryan D. Pardy
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sebastian Shaw
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Maria I. Merolle
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jodi A. Gullicksrud
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - David A. Christian
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Boris Striepen
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christopher A. Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
25
|
Chang Y, Li S, Wang L, Wang K, Li J, Li X, Jian F, Wang R, Zhang S, Zhang L. Micro-RNA expression profile of BALB/c mouse glandular stomach in the early phase of Cryptosporidium muris infection. Exp Parasitol 2023; 253:108603. [PMID: 37633513 DOI: 10.1016/j.exppara.2023.108603] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/28/2023] [Accepted: 08/21/2023] [Indexed: 08/28/2023]
Abstract
Cryptosporidiosis is a zoonotic disease in humans and animals that is caused by infection with the oocysts of Cryptosporidium. MicroRNAs (miRNAs) are important players in regulating the innate immune response against parasitic infection. Public miRNAs data for studying pathogenic mechanisms of cryptosporidiosis, particularly in natural hosts, are scarce. Here, we compared miRNA profiles of the glandular stomach of C. muris-infected and uninfected BALB/c mice using microarray sequencing. A total of 10 miRNAs (including 3 upregulated and 7 downregulated miRNAs) with significant differential expression (|FC| ≥ 2 and P value < 0.05) were identified in the glandular stomach of BALB/c mice 8 h after infection with C. muris. MiRWalk and miRDB online bioinformatics tools were used to predict the target genes of differentially expressed miRNAs. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were performed to annotate the target genes. GO analysis indicate that gene transcription-related and ion transport-related GO terms were significantly enriched. In addition, the KEGG analyses showed that the target genes were strongly related to diverse types of tumor disease progression and anti-pathogen immunity pathways. In the current study, we firstly report changes in miRNA expression profiles in the glandular stomach of BALB/c mice at the early phase of C. muris invasion. This dysregulation in miRNA expression may contribute to our understanding of cryptosporidiosis pathology. This study provides a new perspective on the miRNA regulatory mechanisms of cryptosporidiosis, which may help in the development of effective control strategies against this pathogen.
Collapse
Affiliation(s)
- Yankai Chang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, 450046, China; International Joint Research Laboratory for Zoonotic Diseases of Henan, Zhengzhou, Henan, 450046, China; Key Laboratory of Quality and Safety Control of Poultry Products (Zhengzhou), Ministry of Agriculture and Rural Affairs, Zhengzhou, Henan, 450046, China.
| | - Songrui Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, 450046, China; International Joint Research Laboratory for Zoonotic Diseases of Henan, Zhengzhou, Henan, 450046, China; Key Laboratory of Quality and Safety Control of Poultry Products (Zhengzhou), Ministry of Agriculture and Rural Affairs, Zhengzhou, Henan, 450046, China.
| | - Luyang Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, 450046, China; International Joint Research Laboratory for Zoonotic Diseases of Henan, Zhengzhou, Henan, 450046, China; Key Laboratory of Quality and Safety Control of Poultry Products (Zhengzhou), Ministry of Agriculture and Rural Affairs, Zhengzhou, Henan, 450046, China.
| | - Ke Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, 450046, China; International Joint Research Laboratory for Zoonotic Diseases of Henan, Zhengzhou, Henan, 450046, China; Key Laboratory of Quality and Safety Control of Poultry Products (Zhengzhou), Ministry of Agriculture and Rural Affairs, Zhengzhou, Henan, 450046, China.
| | - Junqiang Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, 450046, China; International Joint Research Laboratory for Zoonotic Diseases of Henan, Zhengzhou, Henan, 450046, China; Key Laboratory of Quality and Safety Control of Poultry Products (Zhengzhou), Ministry of Agriculture and Rural Affairs, Zhengzhou, Henan, 450046, China.
| | - Xiaoying Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, 450046, China; International Joint Research Laboratory for Zoonotic Diseases of Henan, Zhengzhou, Henan, 450046, China; Key Laboratory of Quality and Safety Control of Poultry Products (Zhengzhou), Ministry of Agriculture and Rural Affairs, Zhengzhou, Henan, 450046, China.
| | - Fuchun Jian
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, 450046, China; International Joint Research Laboratory for Zoonotic Diseases of Henan, Zhengzhou, Henan, 450046, China; Key Laboratory of Quality and Safety Control of Poultry Products (Zhengzhou), Ministry of Agriculture and Rural Affairs, Zhengzhou, Henan, 450046, China.
| | - Rongjun Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, 450046, China; International Joint Research Laboratory for Zoonotic Diseases of Henan, Zhengzhou, Henan, 450046, China; Key Laboratory of Quality and Safety Control of Poultry Products (Zhengzhou), Ministry of Agriculture and Rural Affairs, Zhengzhou, Henan, 450046, China.
| | - Sumei Zhang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, 450046, China; International Joint Research Laboratory for Zoonotic Diseases of Henan, Zhengzhou, Henan, 450046, China; Key Laboratory of Quality and Safety Control of Poultry Products (Zhengzhou), Ministry of Agriculture and Rural Affairs, Zhengzhou, Henan, 450046, China.
| | - Longxian Zhang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, 450046, China; International Joint Research Laboratory for Zoonotic Diseases of Henan, Zhengzhou, Henan, 450046, China; Key Laboratory of Quality and Safety Control of Poultry Products (Zhengzhou), Ministry of Agriculture and Rural Affairs, Zhengzhou, Henan, 450046, China.
| |
Collapse
|
26
|
Greigert V, Saraav I, Son J, Dayao D, Antia A, Tzipori S, Witola WH, Stappenbeck TS, Ding S, Sibley LD. Cryptosporidium infection of human small intestinal epithelial cells induces type III interferon and impairs infectivity of Rotavirus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.30.555581. [PMID: 37693422 PMCID: PMC10491271 DOI: 10.1101/2023.08.30.555581] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Cryptosporidiosis is a major cause of severe diarrheal disease in infants from resource poor settings. The majority of infections are caused by the human-specific pathogen C. hominis and absence of in vitro growth platforms has limited our understanding of host-pathogen interactions and development of effective treatments. To address this problem, we developed a stem cell-derived culture system for C. hominis using human enterocytes differentiated under air-liquid interface (ALI) conditions. Human ALI cultures supported robust growth and complete development of C. hominis in vitro including all life cycle stages. C. hominis infection induced a strong interferon response from enterocytes, likely driven by an endogenous dsRNA virus in the parasite. Prior infection with Cryptosporidium induced type III IFN secretion and consequently blunted infection with Rotavirus, including live attenuated vaccine strains. The development of hALI provides a platform for further studies on human-specific pathogens, including clinically important coinfections that may alter vaccine efficacy.
Collapse
Affiliation(s)
- Valentin Greigert
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Iti Saraav
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Juhee Son
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Denise Dayao
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA, 01536, USA
| | - Avan Antia
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Saul Tzipori
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA, 01536, USA
| | - William H. Witola
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, 61802, USA
| | - Thaddeus S. Stappenbeck
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Siyuan Ding
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO, 63110, USA
| | - L. David Sibley
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO, 63110, USA
| |
Collapse
|
27
|
Lv X, He FL, Dai Y, Dai X. IFNγ synergies with cold atmospheric plasma in triggering colorectal cancer cell ferroptosis via the IFNγ/IFNR2/APC/TCF4/GPX4 axis. Aging (Albany NY) 2023; 15:8692-8711. [PMID: 37671945 PMCID: PMC10522381 DOI: 10.18632/aging.204985] [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: 04/25/2023] [Accepted: 08/04/2023] [Indexed: 09/07/2023]
Abstract
Colorectal cancer accounts for the second most common cancer-related lethality. Intestinal stem cells are responsible for enteric homeostasis maintenance that, once being transformed, become colorectal cancer stem cells. Arresting cancer stemness represents an innovative strategy for colorectal cancer management. Using intestinal stem cell organoids as the primary model, we screened common inflammatory cytokines to identify key players targeting cancer stemness. We also explored the downstream signaling that drives the functionalities of the identified cytokine through both experimental investigations and computational predictions. As the results, we identified IFNγ as the key cytokine capable of arresting intestinal stem cells via the IFNγ/IFNGR2/APC/TCF4/GPX4 axis, proposed its role in killing colorectal cancer stem cells via triggering GPX4-dependent ferroptosis, and demonstrated its synergistic anti-cancer effect with cold atmospheric plasma in killing colorectal cancer cells that is worthy to be experimentally validated.
Collapse
Affiliation(s)
- Xinyu Lv
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Fu-le He
- Zhejiang Chinese Medicine Museum, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Yilin Dai
- National Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi, China
| | - Xiaofeng Dai
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu, China
- National Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi, China
| |
Collapse
|
28
|
Tomal F, Sadrin G, Gaboriaud P, Guitton E, Sedano L, Lallier N, Rossignol C, Larcher T, Rouille E, Ledevin M, Guabiraba R, Silvestre A, Lacroix-Lamandé S, Schouler C, Laurent F, Bussière FI. The caecal microbiota promotes the acute inflammatory response and the loss of the intestinal barrier integrity during severe Eimeria tenella infection. Front Cell Infect Microbiol 2023; 13:1250080. [PMID: 37680750 PMCID: PMC10482108 DOI: 10.3389/fcimb.2023.1250080] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 07/28/2023] [Indexed: 09/09/2023] Open
Abstract
Introduction Coccidiosis, a disease caused by intestinal apicomplexan parasites Eimeria, is a threat to poultry production. Eimeria tenella is one of the most pathogenic species, frequently causing a high prevalence of opportunistic infections. Objective The objective of this study is to investigate the role of the microbiota in the pathogenesis of severe Eimeria tenella infection. Methods We have previously shown that microbiota can promote parasite development. To study the effect of the microbiota on the pathogenesis of this infection, we used an experimental condition (inoculum of 10 000 oocysts E. tenella INRAE) in which the parasite load is similar between germ-free and conventional broilers at 7 days post-infection (pi). Thirteen conventional and 24 germ-free chickens were infected. Among this latter group, 12 remained germ-free and 12 received a microbiota from conventional healthy chickens at 4 days pi. Caeca and spleens were collected at 7 days pi. Results Our results demonstrated caecal lesions and epithelium damage in conventional chickens at 7 days pi but not in germ-free infected chickens. Administration of conventional microbiota to germ-free chickens partially restored these deleterious effects. At day 7 pi, both infected conventional and germ-free chickens exhibited increased gene expression of inflammatory mediators, including IL15, IFNγ, TNFα and the anti-inflammatory mediator SOCS1, whereas the inflammatory mediators CXCLi2, CCL20, IL18, CSF1, NOS2, PTGS2, IL1β, IL6, the receptor CCR2, and the anti-inflammatory mediators TGFβ1 and IL10 were upregulated only in infected conventional chickens. Notably, the IL18, PTGS2 gene expression was significantly higher in the infected conventional group. Overall, the inflammatory response enhanced by the microbiota might be in part responsible for higher lesion scores. Epithelial tight junction protein gene expression analysis revealed a significant upregulation of CLDN1 with the infection and microbiota, indicating a potential loss of the intestinal barrier integrity. Conclusion These observations imply that, during E. tenella infection, the caecal microbiota could trigger an acute inflammatory response, resulting in a loss of intestinal integrity. Increase in bacterial translocation can then lead to the likelihood of opportunistic infections. Hence, modulating the microbiota may offer a promising strategy for improving poultry gut health and limiting caecal coccidiosis.
Collapse
Affiliation(s)
- Florian Tomal
- INRAE, Université de Tours, UMR ISP, Nouzilly, France
| | | | | | | | - Laura Sedano
- INRAE, Université de Tours, UMR ISP, Nouzilly, France
| | | | | | | | - Elodie Rouille
- INRAE, Oniris, PAnTher, APEX, Nantes, France
- Laboratoire IHP VETO, Nantes, France
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Haskins BE, Gullicksrud JA, Wallbank BA, Dumaine JE, Guérin A, Cohn IS, O'Dea KM, Pardy RD, Merolle MI, Shallberg LA, Hunter EN, Byerly JH, Smith EJ, Buenconsejo GY, McLeod BI, Christian DA, Striepen B, Hunter CA. Dendritic cell-mediated responses to secreted Cryptosporidium effectors are required for parasite-specific CD8 + T cell responses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.16.553566. [PMID: 37645924 PMCID: PMC10462095 DOI: 10.1101/2023.08.16.553566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Cryptosporidium causes debilitating diarrheal disease in patients with primary and acquired defects in T cell function. However, it has been a challenge to understand how this infection generates T cell responses and how they mediate parasite control. Here, Cryptosporidium was engineered to express a parasite effector protein (MEDLE-2) that contains the MHC-I restricted SIINFEKL epitope which is recognized by TCR transgenic OT-I CD8 + T cells. These modified parasites induced expansion of endogenous SIINFEKL-specific and OT-I CD8 + T cells that were a source of IFN-γ that could restrict growth of Cryptosporidium . This T cell response was dependent on the translocation of the effector and similar results were observed with another secreted parasite effector (ROP1). Although infection and these translocated effector proteins are restricted to intestinal epithelial cells (IEC), type I dendritic cells (cDC1) were required to generate CD8 + T cell responses to these model antigens. These data sets highlight Cryptosporidium effectors as targets of the immune system and suggest that crosstalk between enterocytes and cDC1s is crucial for CD8 + T cell responses to Cryptosporidium .
Collapse
|
30
|
Saraav I, Sibley LD. Dendritic Cells and Cryptosporidium: From Recognition to Restriction. Microorganisms 2023; 11:1056. [PMID: 37110479 PMCID: PMC10144555 DOI: 10.3390/microorganisms11041056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 03/29/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
Host immune responses are required for the efficient control of cryptosporidiosis. Immunity against Cryptosporidium infection has been best studied in mice, where it is mediated by both innate and adaptive immune responses. Dendritic cells are the key link between innate and adaptive immunity and participate in the defense against Cryptosporidium infection. While the effector mechanism varies, both humans and mice rely on dendritic cells for sensing parasites and restricting infection. Recently, the use of mouse-adapted strains C. parvum and mouse-specific strain C. tyzzeri have provided tractable systems to study the role of dendritic cells in mice against this parasite. In this review, we provide an overview of recent advances in innate immunity acting during infection with Cryptosporidium with a major focus on the role of dendritic cells in the intestinal mucosa. Further work is required to understand the role of dendritic cells in the activation of T cells and to explore associated molecular mechanisms. The identification of Cryptosporidium antigen involved in the activation of Toll-like receptor signaling in dendritic cells during infection is also a matter of future study. The in-depth knowledge of immune responses in cryptosporidiosis will help develop targeted prophylactic and therapeutic interventions.
Collapse
Affiliation(s)
| | - L. David Sibley
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
31
|
Clark JT, Weizman OE, Aldridge DL, Shallberg LA, Eberhard J, Lanzar Z, Wasche D, Huck JD, Zhou T, Ring AM, Hunter CA. IL-18BP mediates the balance between protective and pathological immune responses to Toxoplasma gondii. Cell Rep 2023; 42:112147. [PMID: 36827187 PMCID: PMC10131179 DOI: 10.1016/j.celrep.2023.112147] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 12/02/2022] [Accepted: 02/07/2023] [Indexed: 02/25/2023] Open
Abstract
Interleukin-18 (IL-18) promotes natural killer (NK) and T cell production of interferon (IFN)-γ, a key factor in resistance to Toxoplasma gondii, but previous work has shown a limited role for endogenous IL-18 in control of this parasite. Although infection with T. gondii results in release of IL-18, the production of IFN-γ induces high levels of the IL-18 binding protein (IL-18BP). Antagonism of IL-18BP with a "decoy-to-the-decoy" (D2D) IL-18 construct that does not signal but rather binds IL-18BP results in enhanced innate lymphoid cell (ILC) and T cell responses and improved parasite control. In addition, the use of IL-18 resistant to IL-18BP ("decoy-resistant" IL-18 [DR-18]) is more effective than exogenous IL-18 at promoting innate resistance to infection. DR-18 enhances CD4+ T cell production of IFN-γ but results in CD4+ T cell-mediated pathology. Thus, endogenous IL-18BP restrains aberrant immune pathology, and this study highlights strategies that can be used to tune this regulatory pathway for optimal anti-pathogen responses.
Collapse
Affiliation(s)
- Joseph T Clark
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - Orr-El Weizman
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06519, USA
| | - Daniel L Aldridge
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - Lindsey A Shallberg
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - Julia Eberhard
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - Zachary Lanzar
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - Devon Wasche
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06519, USA
| | - John D Huck
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06519, USA
| | - Ting Zhou
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06519, USA
| | - Aaron M Ring
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06519, USA.
| | - Christopher A Hunter
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
32
|
Structural Analyses of a Dominant Cryptosporidium parvum Epitope Presented by H-2K b Offer New Options To Combat Cryptosporidiosis. mBio 2023; 14:e0266622. [PMID: 36602309 PMCID: PMC9973275 DOI: 10.1128/mbio.02666-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Cryptosporidium parvum has gained much attention as a major cause of diarrhea in the world, particularly in those with compromised immune systems. The data currently available on how the immune system recognizes C. parvum are growing rapidly, but we lack data on the interactions among host major histocompatibility complex (MHC) diversity and parasitic T-cell epitopes. To identify antigenic epitopes in a murine model, we performed systematic profiling of H-2Kb-restricted peptides by screening the dominant Cryptosporidium antigens. The results revealed that the glycoprotein-derived epitope Gp40/15-SVF9 induced an immunodominant response in C. parvum-recovered C57BL/6 mice, and injection of the cytotoxic-T-lymphocyte (CTL) peptide with the adjuvant activated peptide-specific CD8+ T cells. Notably, the SVF9 epitope was highly conserved across Cryptosporidium hominis, C. parvum, and many other Cryptosporidium species. SVF9 also formed stable peptide-MHC class I (MHC I) complexes with HLA-A*0201, suggesting cross-reactivity between H-2Kb and human MHC I specificities. Crystal structure analyses revealed that the interactions of peptide-MHC surface residues of H-2Kb and HLA-A*0201 are highly conserved. The hydrogen bonds of H-2Kb-SVF9 are similar to those of a dominant epitope presented by HLA-A*0201, which can be recognized by a public human T-cell receptor (TCR). Notably, we found double conformations in position 4 (P4), 5 (P5) of the SVF9 peptide, which showed high flexibility, and multiple peptide conformations generated more molecular surfaces that can potentially be recognized by TCRs. Our findings demonstrate that an immunodominant C. parvum epitope and its homologs from different Cryptosporidium species and subtypes can benefit vaccine development to combat cryptosporidiosis. IMPORTANCE Adaptive immune responses and T lymphocytes have been implicated as important mechanisms of parasite-induced protection. However, the role of CD8+ T lymphocytes in the resolution of C. parvum infection is largely unresolved. Our results revealed that the glycoprotein-derived epitope Gp40/15-SVF9 induced an immunodominant CD8+ T-cell response in C57BL/6 mice. Crystal structure analyses revealed that the interactions of the H-2Kb-SVF9 peptide are similar to those of a dominant epitope presented by HLA-A*0201, which can be recognized by human TCRs. In addition, we found double conformations of the SVF9 peptide, which showed high flexibility and multiple peptide conformations that can potentially be recognized by TCRs.
Collapse
|
33
|
Yang X, Wu X, Huang S, Yao Q, Chen X, Song J, Fan Y, Zhao G. C3a/C3aR Affects the Propagation of Cryptosporidium parvum in the Ileum Tissues of Mice by Regulating the Gut Barrier, Cell Proliferation, and CD4 + T Cell Main Effectors. Animals (Basel) 2023; 13:ani13050837. [PMID: 36899694 PMCID: PMC10000055 DOI: 10.3390/ani13050837] [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: 12/20/2022] [Revised: 02/15/2023] [Accepted: 02/23/2023] [Indexed: 03/02/2023] Open
Abstract
Cryptosporidium parvum is an important zoonotic protozoon that threatens the health of humans and animals, but the interaction mechanisms between C. parvum and hosts are poorly understood. Our previous study indicated that the expression levels of C3a and C3aR were up-regulated in mice during C. parvum infection, but the mechanisms of C3a/C3aR signaling during C. parvum infection have not been elucidated. In the present study, an optimized BALB/c suckling mouse model infected with C. parvum was used to explore the function of C3a/C3aR signaling during C. parvum infection. The expression levels of C3aR in the ileum tissues of mice infected with C. parvum were analyzed using real-time PCR, Western blot and immunohistochemistry. The mRNA levels of the Cryptosporidium 18S rRNA gene, tight junction proteins (zo-1, claudin 3, and occludin), intestinal stem cell marker lgr5, cell proliferation marker ki67, Th1 cell-related cytokine ifn-γ, and Treg cell-related cytokine tgf-β in mouse ileum tissues were analyzed by real-time PCR. The pathological injury of ileal mucosa was examined by histopathology analysis. The mRNA expression levels of Cryptosporidium 18S rRNA gene were significantly up-regulated in the ileum tissues of C3aR-inhibited mice during C. parvum infection. Meanwhile, histopathology analysis of ileal mucosa in mice showed that inhibition of C3aR significantly aggravated the changes in villus length, villus diameter, mucosal thickness and the ratio of villus length to crypt depth during C. parvum infection. Further studies found inhibition of C3aR aggravated the down-regulation of occludin at most time points during C. parvum infection. The mRNA levels of ki67 and lgr5 in the ileum tissues of mice infected with C. parvum were significantly down-regulated. Inhibition of C3aR significantly down-regulated the mRNA expression levels of lgr5 at most time points, but significantly up-regulated the mRNA expression levels of ki67 at most time points. The mRNA expression levels of ifn-γ and tgf-β were significantly up-regulated and down-regulated in the ileum tissues of mice infected with C. parvum, respectively. However, inhibition of C3aR significantly increased the mRNA expression levels of ifn-γ and tgf-β in the ileum tissues of mice infected with C. parvum. Taken together, C3a/C3aR signaling could possibly affect the propagation of C. parvum in mouse ileum tissues by regulating the gut barrier, cell proliferation and CD4+ T cell main effectors, which would contribute to our understanding of the interaction between Cryptosporidium and hosts.
Collapse
|
34
|
Wang H, Zhang B, Shang Y, Chen F, Fan Y, Tan K. A novel risk score model based on pyroptosis-related genes for predicting survival and immunogenic landscape in hepatocellular carcinoma. Aging (Albany NY) 2023; 15:1412-1444. [PMID: 36920176 PMCID: PMC10042690 DOI: 10.18632/aging.204544] [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: 10/25/2022] [Accepted: 02/13/2023] [Indexed: 03/15/2023]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the third leading cause of cancer worldwide, with high incidence and mortality. Pyroptosis, a form of inflammatory-regulated cell death, is closely associated with oncogenesis. METHODS Expression profiles of HCC were downloaded from the TCGA database and validated using the ICGC and GEO databases. Consensus clustering analysis was used to determine distinct clusters. The pyroptosis-related genes (PRGs) included in the pyroptosis-related signature were selected by univariate Cox regression and LASSO regression analysis. Kaplan-Meier and receiver operating characteristic (ROC) analyses were performed to estimate the prognostic potential of the model. The characteristics of infiltration of immune cells between different groups of HCC were explored. RESULTS Two independent clusters were identified according to PRG expression. Cluster 2 showed upregulated expression, poor prognosis, increased immune cell infiltration and worse immunotherapy response than cluster 1. A prognostic risk signature consisting of five genes (GSDME, NOD1, PLCG1, NLRP6 and NLRC4) was identified. In the high-risk score group, HCC patients showed decreased survival rates. In particular, multiple clinicopathological characteristics and immune cell infiltration were significantly associated with the risk score. Notably, the 5 PRGs in the risk score have been implicated in carcinogenesis, immunological pathways and drug sensitivity. CONCLUSIONS A prognostic signature comprising five PRGs can be used as a potential prognostic factor for HCC. The PRG-related signature provides an in-depth understanding of the association between pyroptosis and chemotherapy or immunotherapy for HCC patients.
Collapse
Affiliation(s)
- Hongyu Wang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, Hebei, China
| | - Bo Zhang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, Hebei, China
| | - Yanan Shang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, Hebei, China
| | - Fei Chen
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, Hebei, China
| | - Yumei Fan
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, Hebei, China
| | - Ke Tan
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, Hebei, China
| |
Collapse
|
35
|
Mead JR. Early immune and host cell responses to Cryptosporidium infection. FRONTIERS IN PARASITOLOGY 2023; 2:1113950. [PMID: 37325809 PMCID: PMC10269812 DOI: 10.3389/fpara.2023.1113950] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 01/13/2023] [Indexed: 06/17/2023]
Abstract
Cryptosporidium spp. are opportunistic protozoan parasites that infect epithelial cells of the small intestine and cause diarrheal illness in both immunocompetent and immunodeficient individuals. These infections may be more severe in immunocompromised individuals and young children, especially in children under 2 in developing countries. The parasite has a global distribution and is an important cause of childhood diarrhea where it may result in cognitive impairment and growth deficits. Current therapies are limited with nitazoxanide being the only FDA-approved drug. However, it is not efficacious in immunocompromised patients. Additionally, there are no vaccines for cryptosporidiosis available. While acquired immunity is needed to clear Cryptosporidium parasites completely, innate immunity and early responses to infection are important in keeping the infection in check so that adaptive responses have time to develop. Infection is localized to the epithelial cells of the gut. Therefore, host cell defenses are important in the early response to infection and may be triggered through toll receptors or inflammasomes which induce a number of signal pathways, interferons, cytokines, and other immune mediators. Chemokines and chemokine receptors are upregulated which recruit immune cells such neutrophils, NK cells, and macrophages to the infection site to help in host cell defense as well as dendritic cells that are an important bridge between innate and adaptive responses. This review will focus on the host cell responses and the immune responses that are important in the early stages of infection.
Collapse
Affiliation(s)
- Jan R. Mead
- Department of Pediatrics, Children’s Healthcare Organization of Atlanta, Emory University, Atlanta, GA, United States
- Atlanta Veterans Affairs Medical Center, Decatur, GA, United States
| |
Collapse
|
36
|
Cohn IS, Henrickson SE, Striepen B, Hunter CA. Immunity to Cryptosporidium: Lessons from Acquired and Primary Immunodeficiencies. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:2261-2268. [PMID: 36469846 PMCID: PMC9731348 DOI: 10.4049/jimmunol.2200512] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 08/30/2022] [Indexed: 01/04/2023]
Abstract
Cryptosporidium is a ubiquitous protozoan parasite that infects gut epithelial cells and causes self-limited diarrhea in immunocompetent individuals. However, in immunocompromised hosts with global defects in T cell function, this infection can result in chronic, life-threatening disease. In addition, there is a subset of individuals with primary immunodeficiencies associated with increased risk for life-threatening cryptosporidiosis. These patients highlight MHC class II expression, CD40-CD40L interactions, NF-κB signaling, and IL-21 as key host factors required for resistance to this enteric pathogen. Understanding which immune deficiencies do (or do not) lead to increased risk for severe Cryptosporidium may reveal mechanisms of parasite restriction and aid in the identification of novel strategies to manage this common pathogen in immunocompetent and deficient hosts.
Collapse
Affiliation(s)
- Ian S. Cohn
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sarah E. Henrickson
- Institute for Immunology, University of Pennsylvania, Philadelphia, PA, USA
- Division of Allergy Immunology, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Boris Striepen
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christopher A. Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
37
|
Gong W, Yang K, Zhao W, Zheng J, Yu J, Guo K, Sun X. Intestinal Gasdermins for regulation of inflammation and tumorigenesis. Front Immunol 2022; 13:1052111. [PMID: 36505474 PMCID: PMC9732009 DOI: 10.3389/fimmu.2022.1052111] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 11/14/2022] [Indexed: 11/27/2022] Open
Abstract
Gasdermins (GSDMs) protein family express in intestinal epithelial cells or lamina propria immune cells, and play a nonnegligible function during gut homeostasis. With the gradually in-depth investigation of GSDMs protein family, the proteases that cleave GSDMA-E have been identified. Intestinal GSDMs-induced pyroptosis is demonstrated to play a crucial role in the removal of self-danger molecules and clearance of pathogenic organism infection by mediating inflammatory reaction and collapsing the protective niche for pathogens. Simultaneously, excessive pyroptosis leading to the release of cellular contents including inflammatory mediators into the extracellular environment, enhancing the mucosal immune response. GSDMs-driver pyroptosis also participates in a novel inflammatory cell death, PANoptosis, which makes a significant sense to the initiation and progression of gut diseases. Moreover, GSDMs are expressed in healthy intestinal tissue without obvious pyroptosis and inflammation, indicating the potential intrinsic physiological functions of GSDMs that independent of pyroptotic cell death during maintenance of intestinal homeostasis. This review provides an overview of the latest advances in the physiological and pathological properties of GSDMs, including its mediated pyroptosis, related PANoptosis, and inherent functions independent of pyroptosis, with a focus on their roles involved in intestinal inflammation and tumorigenesis.
Collapse
Affiliation(s)
- Wenbin Gong
- Department of General Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Kui Yang
- Department of General Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Wei Zhao
- Department of General Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Jianbao Zheng
- Department of General Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Junhui Yu
- Department of General Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China,*Correspondence: Junhui Yu, ; Kun Guo, ; Xuejun Sun,
| | - Kun Guo
- Department of General Surgery, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China,*Correspondence: Junhui Yu, ; Kun Guo, ; Xuejun Sun,
| | - Xuejun Sun
- Department of General Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China,*Correspondence: Junhui Yu, ; Kun Guo, ; Xuejun Sun,
| |
Collapse
|
38
|
Differential Response to the Course of Cryptosporidium parvum Infection and Its Impact on Epithelial Integrity in Differentiated versus Undifferentiated Human Intestinal Enteroids. Infect Immun 2022; 90:e0039722. [PMID: 36286526 PMCID: PMC9671013 DOI: 10.1128/iai.00397-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cryptosporidium is a leading cause of diarrhea and death in young children and untreated AIDS patients and causes waterborne outbreaks. Pathogenic mechanisms underlying diarrhea and intestinal dysfunction are poorly understood. We previously developed stem-cell derived human intestinal enteroid (HIE) models for Cryptosporidium parvum which we used in this study to investigate the course of infection and its effect on intestinal epithelial integrity. By immunofluorescence and confocal microscopy, there was robust infection of undifferentiated and differentiated HIEs in two and three-dimensional (2D, 3D) models. Infection of differentiated HIEs in the 2D model was greater than that of undifferentiated HIEs but lasted only for 3 days, whereas infection persisted for 21 days and resulted in completion of the life cycle in undifferentiated HIEs. Infection of undifferentiated HIE monolayers suggest that C. parvum infects LGR5+ stem cells. Transepithelial electrical resistance measurement of HIEs in the 2D model revealed that infection resulted in decreased epithelial integrity which persisted in differentiated HIEs but recovered in undifferentiated HIEs. Compromised epithelial integrity was reflected in disorganization of the tight and adherens junctions as visualized using the markers ZO-1 and E-cadherin, respectively. Quantitation using the image analysis tools Tight Junction Organizational Rate and Intercellular Junction Organization Quantification, measurement of monolayer height, and RNA transcripts of both proteins by quantitative reverse transcription PCR confirmed that disruption persisted in differentiated HIEs but recovered in undifferentiated HIEs. These models, which more accurately recapitulate human infection, will be useful tools to dissect pathogenic mechanisms underlying diarrhea and intestinal dysfunction in cryptosporidiosis.
Collapse
|
39
|
Sardinha-Silva A, Alves-Ferreira EVC, Grigg ME. Intestinal immune responses to commensal and pathogenic protozoa. Front Immunol 2022; 13:963723. [PMID: 36211380 PMCID: PMC9533738 DOI: 10.3389/fimmu.2022.963723] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 08/15/2022] [Indexed: 11/17/2022] Open
Abstract
The physical barrier of the intestine and associated mucosal immunity maintains a delicate homeostatic balance between the host and the external environment by regulating immune responses to commensals, as well as functioning as the first line of defense against pathogenic microorganisms. Understanding the orchestration and characteristics of the intestinal mucosal immune response during commensal or pathological conditions may provide novel insights into the mechanisms underlying microbe-induced immunological tolerance, protection, and/or pathogenesis. Over the last decade, our knowledge about the interface between the host intestinal mucosa and the gut microbiome has been dominated by studies focused on bacterial communities, helminth parasites, and intestinal viruses. In contrast, specifically how commensal and pathogenic protozoa regulate intestinal immunity is less well studied. In this review, we provide an overview of mucosal immune responses induced by intestinal protozoa, with a major focus on the role of different cell types and immune mediators triggered by commensal (Blastocystis spp. and Tritrichomonas spp.) and pathogenic (Toxoplasma gondii, Giardia intestinalis, Cryptosporidium parvum) protozoa. We will discuss how these various protozoa modulate innate and adaptive immune responses induced in experimental models of infection that benefit or harm the host.
Collapse
|
40
|
Olajide JS, Xiong L, Yang S, Qu Z, Xu X, Yang B, Wang J, Liu B, Ma X, Cai J. Eimeria falciformis secretes extracellular vesicles to modulate proinflammatory response during interaction with mouse intestinal epithelial cells. Parasit Vectors 2022; 15:245. [PMID: 35804396 PMCID: PMC9270845 DOI: 10.1186/s13071-022-05364-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 06/15/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Protozoan parasite secretions can be triggered by various modified media and diverse physicochemical stressors. Equally, host-parasite interactions are known to co-opt the exchange and secretion of soluble biochemical components. Analysis of Eimeria falciformis sporozoite secretions in response to interaction with mouse intestinal epithelial cells (MIECs) may reveal parasite secretory motifs, protein composition and inflammatory activities of E. falciformis extracellular vesicles (EVs). METHODS Eimeria falciformis sporozoites were allowed to interact with inactivated MIECs. Parasite secretions were separated into EV and vesicle-free (VF) fractions by discontinuous centrifugation and ultracentrifugation. Secreted EVs were purified in an iodixanol density gradient medium and the protein composition of both EV and VF fractions were analyzed by liquid chromatoraphy-tandem mass spectroscopy. The inflammatory activities of E. falciformis sporozoite EV on MIECs were then investigated. RESULTS During the interaction of E. falciformis sporozoites with inactivated MIECs, the parasite secreted VF and vesicle-bound molecules. Eimeria falciformis vesicles are typical pathogenic protozoan EVs with a mean diameter of 264 ± 2 nm, and enclosed heat shock protein (Hsp) 70 as classical EV marker. Refractile body-associated aspartyl proteinase (or eimepsin), GAP45 and aminopeptidase were the main components of E. falciformis sporozoite EVs, while VF proteins include Hsp90, actin, Vps54 and kinases, among others. Proteomic data revealed that E. falciformis EV and VF proteins are aggregates of bioactive, antigenic and immunogenic molecules which act in concert for E. falciformis sporozoite motility, pathogenesis and survival. Moreover, in MIECs, E. falciformis EVs induced upregulation of gene expression and secretion of IL-1β, IL-6, IL-17, IL-18, MCP1 as well as pyroptosis-dependent caspase 11 and NLRP6 inflammasomes with the concomitant secretion of lactate dehydrogenase. CONCLUSIONS Eimeria falciformis sporozoite interaction with MIECs triggered the secretion of immunogenic and antigenic proteins. In addition, E. falciformis sporozoite EVs constitute parasite-associated molecular pattern that induced inflammatory response and cell death. This study offers additional insight in the secretion and protein composition of E. falciformis secretomes as well as the proinflammatory functions of E. falciformis sporozoite EVs.
Collapse
Affiliation(s)
- Joshua Seun Olajide
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046 China
- Centre for Distance Learning, Obafemi Awolowo University, Ile-Ife, Nigeria
| | - Ling Xiong
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046 China
| | - Shunli Yang
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046 China
| | - Zigang Qu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046 China
| | - Xiao Xu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046 China
| | - Bin Yang
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046 China
| | - Jing Wang
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046 China
| | - Baohong Liu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046 China
| | - Xueting Ma
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046 China
| | - Jianping Cai
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046 China
| |
Collapse
|
41
|
Dhal AK, Panda C, Yun SIL, Mahapatra RK. An update on Cryptosporidium biology and therapeutic avenues. J Parasit Dis 2022; 46:923-939. [PMID: 35755159 PMCID: PMC9215156 DOI: 10.1007/s12639-022-01510-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 06/07/2022] [Indexed: 12/02/2022] Open
Abstract
Cryptosporidium species has been identified as an important pediatric diarrheal pathogen in resource-limited countries, particularly in very young children (0–24 months). However, the only available drug (nitazoxanide) has limited efficacy and can only be prescribed in a medical setting to children older than one year. Many drug development projects have started to investigate new therapeutic avenues. Cryptosporidium’s unique biology is challenging for the traditional drug discovery pipeline and requires novel drug screening approaches. Notably, in recent years, new methods of oocyst generation, in vitro processing, and continuous three-dimensional cultivation capacities have been developed. This has enabled more physiologically pertinent research assays for inhibitor discovery. In a short time, many great strides have been made in the development of anti-Cryptosporidium drugs. These are expected to eventually turn into clinical candidates for cryptosporidiosis treatment in the future. This review describes the latest development in Cryptosporidium biology, genomics, transcriptomics of the parasite, assay development, and new drug discovery.
Collapse
Affiliation(s)
- Ajit Kumar Dhal
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar, Odisha 751024 India
| | - Chinmaya Panda
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar, Odisha 751024 India
| | - Soon-IL Yun
- Department of Food Science and Technology, Jeonbuk National University, Jeonju, 54896 Republic of Korea
- Department of Agricultural Convergence Technology, Jeonbuk National University, Jeonju, 54896 Republic of Korea
| | | |
Collapse
|
42
|
Angosto-Bazarra D, Molina-López C, Pelegrín P. Physiological and pathophysiological functions of NLRP6: pro- and anti-inflammatory roles. Commun Biol 2022; 5:524. [PMID: 35650327 PMCID: PMC9160023 DOI: 10.1038/s42003-022-03491-w] [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: 01/20/2022] [Accepted: 05/12/2022] [Indexed: 12/26/2022] Open
Abstract
The nucleotide-binding oligomerization and leucine-rich repeat receptor (NLR) protein family consists of important immune sensors that form inflammasomes, a cytosolic multi-protein platform that induces caspase-1 activation and is involved in different inflammatory pathologies. The NLR family pyrin domain containing 6 (NLRP6) is a receptor that can signal by forming inflammasomes, but which can also play an important role without forming inflammasomes. NLRP6 regulates intestinal homeostasis and inflammation, but also is involved in cancer, the nervous system or liver diseases, with both protective and deleterious consequences. In the present article, we review the different roles of NLRP6 in these processes and offer new insights into NLRP6 activation. This review discusses emerging roles for the NLR family pyrin domain containing 6 receptor (NLRP6) in intestinal homeostasis, inflammation, cancer, the nervous system and liver disease.
Collapse
Affiliation(s)
- Diego Angosto-Bazarra
- Línea de Inflamación Molecular, Instituto Murciano de Investigación Biosanitaria IMIB-Arrixaca, Hospital Clínico Universitario Virgen de la Arrixaca, 30120, Murcia, Spain.
| | - Cristina Molina-López
- Línea de Inflamación Molecular, Instituto Murciano de Investigación Biosanitaria IMIB-Arrixaca, Hospital Clínico Universitario Virgen de la Arrixaca, 30120, Murcia, Spain
| | - Pablo Pelegrín
- Línea de Inflamación Molecular, Instituto Murciano de Investigación Biosanitaria IMIB-Arrixaca, Hospital Clínico Universitario Virgen de la Arrixaca, 30120, Murcia, Spain. .,Department of Biochemistry and Molecular Biology B and Immunology, Faculty of Medicine, University of Murcia, 30120, Murcia, Spain.
| |
Collapse
|
43
|
Gibson AR, Sateriale A, Dumaine JE, Engiles JB, Pardy RD, Gullicksrud JA, O’Dea KM, Doench JG, Beiting DP, Hunter CA, Striepen B. A genetic screen identifies a protective type III interferon response to Cryptosporidium that requires TLR3 dependent recognition. PLoS Pathog 2022; 18:e1010003. [PMID: 35584177 PMCID: PMC9154123 DOI: 10.1371/journal.ppat.1010003] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 05/31/2022] [Accepted: 04/11/2022] [Indexed: 11/18/2022] Open
Abstract
Cryptosporidium is a leading cause of severe diarrhea and diarrheal-related death in children worldwide. As an obligate intracellular parasite, Cryptosporidium relies on intestinal epithelial cells to provide a niche for its growth and survival, but little is known about the contributions that the infected cell makes to this relationship. Here we conducted a genome wide CRISPR/Cas9 knockout screen to discover host genes that influence Cryptosporidium parvum infection and/or host cell survival. Gene enrichment analysis indicated that the host interferon response, glycosaminoglycan (GAG) and glycosylphosphatidylinositol (GPI) anchor biosynthesis are important determinants of susceptibility to C. parvum infection and impact on the viability of host cells in the context of parasite infection. Several of these pathways are linked to parasite attachment and invasion and C-type lectins on the surface of the parasite. Evaluation of transcript and protein induction of innate interferons revealed a pronounced type III interferon response to Cryptosporidium in human cells as well as in mice. Treatment of mice with IFNλ reduced infection burden and protected immunocompromised mice from severe outcomes including death, with effects that required STAT1 signaling in the enterocyte. Initiation of this type III interferon response was dependent on sustained intracellular growth and mediated by the pattern recognition receptor TLR3. We conclude that host cell intrinsic recognition of Cryptosporidium results in IFNλ production critical to early protection against this infection.
Collapse
Affiliation(s)
- Alexis R. Gibson
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Adam Sateriale
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jennifer E. Dumaine
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Julie B. Engiles
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Pathobiology, New Bolton Center, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Ryan D. Pardy
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jodi A. Gullicksrud
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Keenan M. O’Dea
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - John G. Doench
- Genetic Perturbation Platform, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Daniel P. Beiting
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Christopher A. Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Boris Striepen
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
44
|
Churchill MJ, Mitchell PS, Rauch I. Epithelial Pyroptosis in Host Defense. J Mol Biol 2022; 434:167278. [PMID: 34627788 PMCID: PMC10010195 DOI: 10.1016/j.jmb.2021.167278] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 09/17/2021] [Accepted: 09/25/2021] [Indexed: 12/28/2022]
Abstract
Pyroptosis is a lytic form of cell death that is executed by a family of pore-forming proteins called gasdermins (GSDMs). GSDMs are activated upon proteolysis by host proteases including the proinflammatory caspases downstream of inflammasome activation. In myeloid cells, GSDM pore formation serves two primary functions in host defense: the selective release of processed cytokines to initiate inflammatory responses, and cell death, which eliminates a replicative niche of the pathogen. Barrier epithelia also undergo pyroptosis. However, unique mechanisms are required for the removal of pyroptotic epithelial cells to maintain epithelial barrier integrity. In the following review, we discuss the role of epithelial inflammasomes and pyroptosis in host defense against pathogens. We use the well-established role of inflammasomes in intestinal epithelia to highlight principles of epithelial pyroptosis in host defense of barrier tissues, and discuss how these principles might be shared or distinctive across other epithelial sites.
Collapse
Affiliation(s)
- Madeline J Churchill
- Department of Molecular Microbiology & Immunology, Oregon Health and Science University, Portland, OR, USA
| | | | - Isabella Rauch
- Department of Molecular Microbiology & Immunology, Oregon Health and Science University, Portland, OR, USA.
| |
Collapse
|
45
|
Gullicksrud JA, Sateriale A, Engiles JB, Gibson AR, Shaw S, Hutchins ZA, Martin L, Christian DA, Taylor GA, Yamamoto M, Beiting DP, Striepen B, Hunter CA. Enterocyte-innate lymphoid cell crosstalk drives early IFN-γ-mediated control of Cryptosporidium. Mucosal Immunol 2022; 15:362-372. [PMID: 34750455 PMCID: PMC8881313 DOI: 10.1038/s41385-021-00468-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 08/14/2021] [Accepted: 09/03/2021] [Indexed: 02/04/2023]
Abstract
The intestinal parasite, Cryptosporidium, is a major contributor to global child mortality and causes opportunistic infection in immune deficient individuals. Innate resistance to Cryptosporidium, which specifically invades enterocytes, is dependent on the production of IFN-γ, yet whether enterocytes contribute to parasite control is poorly understood. In this study, utilizing a mouse-adapted strain of C. parvum, we show that epithelial-derived IL-18 synergized with IL-12 to stimulate innate lymphoid cell (ILC) production of IFN-γ required for early parasite control. The loss of IFN-γ-mediated STAT1 signaling in enterocytes, but not dendritic cells or macrophages, antagonized early parasite control. Transcriptional profiling of enterocytes from infected mice identified an IFN-γ signature and enrichment of the anti-microbial effectors IDO, GBP, and IRG. Deletion experiments identified a role for Irgm1/m3 in parasite control. Thus, enterocytes promote ILC production of IFN-γ that acts on enterocytes to restrict the growth of Cryptosporidium.
Collapse
Affiliation(s)
- Jodi A Gullicksrud
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Adam Sateriale
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
- The Francis Crick Institute, London, UK
| | - Julie B Engiles
- Department of Pathobiology, New Bolton Center, School of Veterinary Medicine, University of Pennsylvania, Kennett Square, PA, USA
| | - Alexis R Gibson
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sebastian Shaw
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zachary A Hutchins
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Jill Robests Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, New York, NY, USA
| | - Lindsay Martin
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - David A Christian
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gregory A Taylor
- Departments of Medicine, Molecular Genetics and Microbiology and Immunology and Center for the Study of Aging and Human Development, Duke University Medical Center, Durham, NC, USA
- Geriatric Research, Education, and Clinical Center, Durham VA Health Care System, Durham, NC, USA
| | - Masahiro Yamamoto
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Daniel P Beiting
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Boris Striepen
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christopher A Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
46
|
Constant DA, Nice TJ, Rauch I. Innate immune sensing by epithelial barriers. Curr Opin Immunol 2021; 73:1-8. [PMID: 34392232 PMCID: PMC8648961 DOI: 10.1016/j.coi.2021.07.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 02/02/2023]
Abstract
Epithelial cells in barrier tissues perform a critical immune function by detecting, restricting, and often directly eliminating extrinsic pathogens. Membrane-bound and cytosolic pattern recognition receptors in epithelial cells bind to diverse ligands, detecting pathogen components and behaviors and stimulating cell-autonomous immunity. In addition to directly acting as first-responders to pathogens, epithelial cells detect commensal-derived and diet-derived products to promote homeostasis. Recent advances have clarified the array of molecular sensors expressed by epithelial cells, and how epithelial cells responses are wired to promote homeostatic balance while simultaneously allowing elimination of pathogens. These new studies emphatically position epithelial cells as central to an effective innate immune response.
Collapse
Affiliation(s)
- David A Constant
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR 97239, United States
| | - Timothy J Nice
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR 97239, United States
| | - Isabella Rauch
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR 97239, United States.
| |
Collapse
|
47
|
Babamale AO, Chen ST. Nod-like Receptors: Critical Intracellular Sensors for Host Protection and Cell Death in Microbial and Parasitic Infections. Int J Mol Sci 2021; 22:11398. [PMID: 34768828 PMCID: PMC8584118 DOI: 10.3390/ijms222111398] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/04/2021] [Accepted: 10/19/2021] [Indexed: 12/14/2022] Open
Abstract
Cell death is an essential immunological apparatus of host defense, but dysregulation of mutually inclusive cell deaths poses severe threats during microbial and parasitic infections leading to deleterious consequences in the pathological progression of infectious diseases. Nucleotide-binding oligomerization domain (NOD)-Leucine-rich repeats (LRR)-containing receptors (NLRs), also called nucleotide-binding oligomerization (NOD)-like receptors (NLRs), are major cytosolic pattern recognition receptors (PRRs), their involvement in the orchestration of innate immunity and host defense against bacteria, viruses, fungi and parasites, often results in the cleavage of gasdermin and the release of IL-1β and IL-18, should be tightly regulated. NLRs are functionally diverse and tissue-specific PRRs expressed by both immune and non-immune cells. Beyond the inflammasome activation, NLRs are also involved in NF-κB and MAPK activation signaling, the regulation of type I IFN (IFN-I) production and the inflammatory cell death during microbial infections. Recent advancements of NLRs biology revealed its possible interplay with pyroptotic cell death and inflammatory mediators, such as caspase 1, caspase 11, IFN-I and GSDMD. This review provides the most updated information that caspase 8 skews the NLRP3 inflammasome activation in PANoptosis during pathogen infection. We also update multidimensional roles of NLRP12 in regulating innate immunity in a content-dependent manner: novel interference of NLRP12 on TLRs and NOD derived-signaling cascade, and the recently unveiled regulatory property of NLRP12 in production of type I IFN. Future prospects of exploring NLRs in controlling cell death during parasitic and microbial infection were highlighted.
Collapse
Affiliation(s)
- Abdulkareem Olarewaju Babamale
- Taiwan International Graduate Program in Molecular Medicine, National Yang-Ming Chiao Tung University and Academia Sinica, Taipei 11266, Taiwan;
- Parasitology Unit, Faculty of Life Sciences, University of Ilorin, Ilorin 240003, Nigeria
| | - Szu-Ting Chen
- Taiwan International Graduate Program in Molecular Medicine, National Yang-Ming Chiao Tung University and Academia Sinica, Taipei 11266, Taiwan;
- Institute of Clinical Medicine, National Yang-Ming Chiao Tung University, Taipei 11266, Taiwan
- Cancer Progression Research Center, National Yang-Ming Chiao Tung University, Taipei 11266, Taiwan
| |
Collapse
|
48
|
Anticryptosporidium Efficacy of Olea europaea and Ficus carica Leaves Extract in Immunocompromised Mice Associated with Biochemical Characters and Antioxidative System. Cells 2021; 10:cells10092419. [PMID: 34572068 PMCID: PMC8471006 DOI: 10.3390/cells10092419] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/10/2021] [Accepted: 09/10/2021] [Indexed: 12/17/2022] Open
Abstract
Cryptosporidiosis is caused by an opportunistic protozoan parasite (Cryptosporidium parvum and C. hominis) known as a parasite of humans, especially children and immunocompromised patients. The current study was designed to evaluate the therapeutic efficacy of a mixture of fig and olive leaf extracts as an alternative medicinal plant. Parasitological examination for oocysts in the stool and histopathological alterations in the small intestines were examined. Additionally, biochemical analyses of liver and kidney functions in addition to antioxidant parameters such as superoxide dismutase (SOD), glutathione peroxidase (GSH) and catalase (CAT) in the plasma were evaluated. Our results showed that marked reduction in oocysts shedding and amelioration in intestinal histopathological changes and hepatic or renal functions were detected in all treated groups compared to the control infected group. Additionally, the treated groups with tested extracts at ratios 1:3 and 1:5 showed a significant decrease in the number of oocysts compared to the other treated groups. Results exhibited a significant increase in the plasma SOD, CAT and GSH levels in treated groups compared to the infected control one. This study suggested that a mixture of fig and olive leaf extracts is a convenient promising therapeutic agent for Cryptosporidiosis.
Collapse
|
49
|
Guérin A, Roy NH, Kugler EM, Berry L, Burkhardt JK, Shin JB, Striepen B. Cryptosporidium rhoptry effector protein ROP1 injected during invasion targets the host cytoskeletal modulator LMO7. Cell Host Microbe 2021; 29:1407-1420.e5. [PMID: 34348092 PMCID: PMC8475647 DOI: 10.1016/j.chom.2021.07.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/19/2021] [Accepted: 07/02/2021] [Indexed: 12/30/2022]
Abstract
The parasite Cryptosporidium invades and replicates in intestinal epithelial cells and is a leading cause of diarrheal disease and early childhood mortality. The molecular mechanisms that underlie infection and pathogenesis are largely unknown. Here, we delineate the events of host cell invasion and uncover a mechanism unique to Cryptosporidium. We developed a screen to identify parasite effectors, finding the injection of multiple parasite proteins into the host from the rhoptry organelle. These factors are targeted to diverse locations within the host cell and its interface with the parasite. One identified effector, rhoptry protein 1 (ROP1), accumulates in the terminal web of enterocytes through direct interaction with the host protein LIM domain only 7 (LMO7) an organizer of epithelial cell polarity and cell-cell adhesion. Genetic ablation of LMO7 or ROP1 in mice or parasites, respectively, impacts parasite burden in vivo in opposite ways. Taken together, these data provide molecular insight into how Cryptosporidium manipulates its intestinal host niche.
Collapse
Affiliation(s)
- Amandine Guérin
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nathan H Roy
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Emily M Kugler
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Laurence Berry
- LPHI, CNRS, Université de Montpellier, Montpellier 34095, France
| | - Janis K Burkhardt
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jung-Bum Shin
- Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA
| | - Boris Striepen
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
50
|
Yang J, Yan H. Mucosal epithelial cells: the initial sentinels and responders controlling and regulating immune responses to viral infections. Cell Mol Immunol 2021; 18:1628-1630. [PMID: 33927362 PMCID: PMC8082051 DOI: 10.1038/s41423-021-00650-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 01/30/2021] [Indexed: 12/25/2022] Open
Affiliation(s)
- Jingyi Yang
- Vaccine and Immunology Research Center, Translational Medical Research Institute, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Huimin Yan
- Vaccine and Immunology Research Center, Translational Medical Research Institute, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China.
| |
Collapse
|