1
|
Hussain S, Ain QU, Aamir M, Alsyaad KM, Ahmed AE, Zakai JG, Zakai HA, Hou Y. Deciphering Host-Pathogen Interactions: Role of Cryptosporidium in Tumorigenesis. Pathogens 2025; 14:208. [PMID: 40137692 PMCID: PMC11946830 DOI: 10.3390/pathogens14030208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/11/2025] [Accepted: 02/18/2025] [Indexed: 03/29/2025] Open
Abstract
Cryptosporidium, a protozoan parasite affecting the gastrointestinal system, is primarily known for causing diarrhea, especially in those with weakened immune systems. However, there is increasingly persuasive evidence that it may be directly involved in tumorigenesis. This review examines some of the potential mechanisms through which Cryptosporidium infections can induce cancer, specifically chronic inflammation, manipulation of the immune system, and alteration of cell signaling pathways. Persistent inflammation with immune system changes due to chronic infection, particularly among immunocompromised hosts, leads to a microenvironment that facilitates tumorigenesis. Cryptosporidium manipulates important cellular pathways such as PI3K, NF-κB, Wnt, and p38/MAPK to promote cell survival, regulate immune responses, and foster tissue remodeling, all of which contribute to a tumor-friendly microenvironment. Moreover, Cryptosporidium virulence factors such as ROP1, sPLA2, and microRNAs disrupt host cellular stability and significantly alter host cellular gene expression, which also exacerbates inflammation and tissue damage. Epidemiological data have indicated higher rates of Cryptosporidium infection in cancer patients, especially patients with gastrointestinal cancers. This, among other observations, raises the possibility that the infection may be connected to cancer progression. In animal models, especially studies with C. parvum-challenged rodents, chronic inflammation, immune repression, and genetic mutations related to neoplasia have been reported. While this has provided us with valuable information, we still have a long way to go to fully understand the long-term ramifications of Cryptosporidium infection. These cover aspects such as the contribution of latent infections and the genetic diversity of Cryptosporidium strains in cancer. Further investigation is urgently needed to understand the molecular processes by which Cryptosporidium might contribute to carcinogenesis and explore potential strategies for therapy and prevention especially among immunocompromised populations.
Collapse
Affiliation(s)
- Shakeel Hussain
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, China;
| | - Qurrat ul Ain
- School Education Department, Government of Punjab, Mailsi 61200, Pakistan;
| | - Muhammad Aamir
- Chemical Engineering Department, School of Sciences, Harbin Institute of Technology, Shenzhen 518000, China;
| | - Khalid M. Alsyaad
- Department of Biology, College of Science, King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia; (K.M.A.); (A.E.A.)
| | - Ahmed Ezzat Ahmed
- Department of Biology, College of Science, King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia; (K.M.A.); (A.E.A.)
- Prince Sultan Bin Abdelaziz for Environmental Research and Natural Resources Sustainability Center, King Khalid University, Abha 61421, Saudi Arabia
| | - Jude G. Zakai
- Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Haytham Ahmed Zakai
- Medical Laboratory Sciences Department, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Yongzhong Hou
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, China;
| |
Collapse
|
2
|
Wang BN, Zhang XZ, Wu JY, Zhang ZY, Cong PK, Zheng WW, Long SR, Liu RD, Cui J, Wang ZQ. Vaccination of mice with Trichinella spiralis C-type lectin elicited the protective immunity and enhanced gut epithelial barrier function. PLoS Negl Trop Dis 2025; 19:e0012825. [PMID: 39841790 PMCID: PMC11761079 DOI: 10.1371/journal.pntd.0012825] [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: 08/23/2024] [Revised: 01/24/2025] [Accepted: 01/06/2025] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND C-type lectin (CTL) plays an important act in parasite adhesion, host's cell invasion and immune escape. Our previous studies showed that recombinant Trichinella spiralis C-type lectin (rTsCTL) mediated larval invasion of enteral mucosal epithelium. The aim of this study was to investigate protective immunity produced by vaccination with rTsCTL and its effect on gut epithelial barrier function in a mouse model. METHODOLOGY/PRINCIPAL FINDING The ELISA results showed that subcutaneous vaccination of mice with rTsCTL elicited a systemic humoral response (high levels of serum IgG, IgG1/IgG2a and IgA) and significant gut mucosal sIgA responses. The levels of Th1/Th2 cytokines (IFN-γ/IL-4) secreted from spleen, mesenteric lymph nodes and Peyer's patches were distinctly increased at 6 weeks following vaccination (P < 0.05). At one week after challenge, the numbers of goblet cells and expression level of Muc2, Muc5ac and pro-inflammatory cytokines (TNF-α and IL-1β) in gut tissues of vaccinated mice were obviously decreased, while expression of anti-inflammatory cytokines (IL-4 and IL-10) was evidently increased, compared to the infected PBS group. It is interesting that expression levels of gut epithelial tight junctions (TJs; occludin, claudin-1 and E-cad) were prominently elevated and intestinal permeability was interestingly declined in vaccinated mice. The rTsCTL-vaccinated mice exhibited a 51.69 and 48.19% reduction of intestinal adult and muscle larva burdens, respectively. The female fecundity in rTsCTL vaccinated mice was reduced by 40.51%. These findings indicated that rTsCTL vaccination impeded larval invasion and improved gut epithelial integrity and barrier function, reduced worm burdens, and relieved gut and muscle inflammation. CONCLUSIONS Vaccination of mice with rTsCTL elicited an obvious protective immunity against larval challenge, impeded larval invasion of gut mucosa, enhanced gut epithelial integrity and barrier function, reduced worm burdens; it also alleviated gut and muscle inflammation. TsCTL might be a novel candidate target molecule for anti-Trichinella vaccines.
Collapse
Affiliation(s)
- Bo Ning Wang
- Department of Parasitology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Xin Zhuo Zhang
- Department of Parasitology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jin Yi Wu
- Department of Parasitology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Zhao Yu Zhang
- Department of Parasitology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Pei Kun Cong
- Department of Parasitology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Wen Wen Zheng
- Department of Parasitology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Shao Rong Long
- Department of Parasitology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Ruo Dan Liu
- Department of Parasitology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jing Cui
- Department of Parasitology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Zhong Quan Wang
- Department of Parasitology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
3
|
Huang W, He W, Huang Y, Tang Y, Chen M, Sun L, Yang Z, Hou T, Liu H, Chen H, Wang T, Li N, Guo Y, Xiao L, Feng Y. Multicopy subtelomeric genes underlie animal infectivity of divergent Cryptosporidium hominis subtypes. Nat Commun 2024; 15:10774. [PMID: 39737947 PMCID: PMC11685829 DOI: 10.1038/s41467-024-54995-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 11/27/2024] [Indexed: 01/01/2025] Open
Abstract
The anthroponotic Cryptosporidium hominis differs from the zoonotic C. parvum in its lack of infectivity to animals, but several divergent subtypes have recently been found in nonhuman primates and equines. Here, we sequence 17 animal C. hominis isolates and generate a new IbA12G3 genome at the chromosome level. Comparative analysis with 222 human isolates shows significant genetic divergence of the animal isolates, with genetic recombination among them. They have additional subtelomeric insulinase and MEDLE genes. In interferon-γ knockout mice, three monkey isolates show differences in infectivity and induce higher and longer oocyst shedding than a reference C. parvum isolate. Deletion of the MEDLE genes significantly reduces the growth and pathogenicity of a virulent strain in mice. Co-infection of two fluorescence-tagged C. hominis subtypes produces bicolored oocysts, supporting the conclusion that mixed subtype infections can lead to genetic recombination. These data provide insight into potential determinants of host infectivity in Cryptosporidium, and a convenient animal model for biological studies of C. hominis.
Collapse
Affiliation(s)
- Wanyi Huang
- State Key Laboratory for Animal Disease Control and Prevention, Center for Emerging and Zoonotic Diseases, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Wei He
- State Key Laboratory for Animal Disease Control and Prevention, Center for Emerging and Zoonotic Diseases, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yue Huang
- State Key Laboratory for Animal Disease Control and Prevention, Center for Emerging and Zoonotic Diseases, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yongping Tang
- State Key Laboratory for Animal Disease Control and Prevention, Center for Emerging and Zoonotic Diseases, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Ming Chen
- State Key Laboratory for Animal Disease Control and Prevention, Center for Emerging and Zoonotic Diseases, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Lianbei Sun
- State Key Laboratory for Animal Disease Control and Prevention, Center for Emerging and Zoonotic Diseases, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Zuwei Yang
- State Key Laboratory for Animal Disease Control and Prevention, Center for Emerging and Zoonotic Diseases, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Tianyi Hou
- State Key Laboratory for Animal Disease Control and Prevention, Center for Emerging and Zoonotic Diseases, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Huimin Liu
- State Key Laboratory for Animal Disease Control and Prevention, Center for Emerging and Zoonotic Diseases, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Haoyu Chen
- State Key Laboratory for Animal Disease Control and Prevention, Center for Emerging and Zoonotic Diseases, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Tianpeng Wang
- State Key Laboratory for Animal Disease Control and Prevention, Center for Emerging and Zoonotic Diseases, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Na Li
- State Key Laboratory for Animal Disease Control and Prevention, Center for Emerging and Zoonotic Diseases, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yaqiong Guo
- State Key Laboratory for Animal Disease Control and Prevention, 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, Center for Emerging and Zoonotic Diseases, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.
| | - Yaoyu Feng
- State Key Laboratory for Animal Disease Control and Prevention, Center for Emerging and Zoonotic Diseases, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.
| |
Collapse
|
4
|
Wang BN, Zhang XZ, Cong PK, Zheng WW, Wu JY, Long SR, Liu RD, Zhang X, Cui J, Wang ZQ. Trichinellaspiralis C-type lectin mediates larva invasion of gut mucosa via binding to syndecan-1 and damaging epithelial integrity in mice. Int J Biol Macromol 2024; 280:135958. [PMID: 39322156 DOI: 10.1016/j.ijbiomac.2024.135958] [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: 08/24/2024] [Revised: 09/11/2024] [Accepted: 09/21/2024] [Indexed: 09/27/2024]
Abstract
C-type lectin (CTL) plays a vital role in parasite adhesion, invading host's cells and immune escape. The objective of this research was to explore whether recombinant T. spiralis CTL (rTsCTL) binding with syndecan-1 damages intestine epithelial integrity and mediates T. spiralis intrusion in mice. The results showed that rTsCTL interacted with syndecan-1 and activated STAT3 pathway in gut epithelium, decreased tight junctions (TJs) expressions and damaged gut epithelium integrity, promoted T. spiralis intrusion, and increased expression level of inflammatory cytokine and mucin. The syndecan-1 inhibitor (β-xyloside) and STAT3 phosphorylation inhibitor (Stattic) significantly suppressed syndecan-1 expression and STAT3 pathway activation, reduced the expression levels of TJs, pro-inflammatory cytokines (TNF-α and IL-1β), Muc2 and Muc5ac, and declined intestinal permeability in T. spiralis-infected mice. These results revealed that the inhibitors suppressed T. spiralis invasion and development in gut mucosa, decreased intestinal adult burdens and relieved gut inflammation. These findings further testified that the in vivo binding of TsCTL with syndecan-1 destroyed enteral mucosal epithelial integrity and promoted T. spiralis intrusion of gut mucosa via activating STAT3 pathway and decreasing TJs expression. TsCTL could be deemed as a promising vaccine target to interrupt T. spiralis infection.
Collapse
Affiliation(s)
- Bo Ning Wang
- Department of Parasitology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Xin Zhuo Zhang
- Department of Parasitology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Pei Kun Cong
- Department of Parasitology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Wen Wen Zheng
- Department of Parasitology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Jin Yi Wu
- Department of Parasitology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Shao Rong Long
- Department of Parasitology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Ruo Dan Liu
- Department of Parasitology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Xi Zhang
- Department of Parasitology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Jing Cui
- Department of Parasitology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Zhong Quan Wang
- Department of Parasitology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
5
|
Zhang X, Sun S, Zhao W, Wang L, Liang G, Wang Y, Cai B, Zhang L, Li X, Zhang S. A single-pass type I membrane protein, mannose-specific L-type lectin, potentially involved in the adhesion and invasion of Cryptosporidium parvum. Parasite 2024; 31:51. [PMID: 39212528 PMCID: PMC11363900 DOI: 10.1051/parasite/2024051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024] Open
Abstract
Cryptosporidium is a globally distributed zoonotic protozoan parasite that can cause severe diarrhea in humans and animals. L-type lectins are carbohydrate-binding proteins involved in multiple pathways in animals and plants, including protein transportation, secretion, innate immunity, and the unfolded protein response signaling pathway. However, the biological function of the L-type lectins remains unknown in Cryptosporidium parvum. Here, we preliminarily characterized an L-type lectin in C. parvum (CpLTL) that contains a lectin-leg-like domain. Immunofluorescence assay confirmed that CpLTL is located on the wall of oocysts, the surface of the mid-anterior region of the sporozoite and the cytoplasm of merozoites. The involvement of CpLTL in parasite invasion is partly supported by experiments showing that an anti-CpLTL antibody could partially block the invasion of C. parvum sporozoites into host cells. Moreover, the recombinant CpLTL showed binding ability with mannose and the surface of host cells, and competitively inhibited the invasion of C. parvum. Two host cell proteins were identified by proteomics which should be prioritized for future validation of CpLTL-binding. Our data indicated that CpLTL is potentially involved in the adhesion and invasion of C. parvum.
Collapse
Affiliation(s)
- Xiaotian Zhang
- College of Veterinary Medicine, Henan Agricultural University Zhengzhou 450046 Henan PR China
- International Joint Research Laboratory for Zoonotic Diseases of Henan Zhengzhou 450046 Henan PR China
- Key Laboratory of Quality and Safety Control of Poultry Products, Ministry of Agriculture and Rural Affairs Zhengzhou 450046 Henan PR China
| | - Songying Sun
- College of Veterinary Medicine, Henan Agricultural University Zhengzhou 450046 Henan PR China
- International Joint Research Laboratory for Zoonotic Diseases of Henan Zhengzhou 450046 Henan PR China
- Key Laboratory of Quality and Safety Control of Poultry Products, Ministry of Agriculture and Rural Affairs Zhengzhou 450046 Henan PR China
| | - Wenchao Zhao
- College of Veterinary Medicine, Henan Agricultural University Zhengzhou 450046 Henan PR China
- International Joint Research Laboratory for Zoonotic Diseases of Henan Zhengzhou 450046 Henan PR China
- Key Laboratory of Quality and Safety Control of Poultry Products, Ministry of Agriculture and Rural Affairs Zhengzhou 450046 Henan PR China
| | - Luyang Wang
- College of Veterinary Medicine, Henan Agricultural University Zhengzhou 450046 Henan PR China
- International Joint Research Laboratory for Zoonotic Diseases of Henan Zhengzhou 450046 Henan PR China
- Key Laboratory of Quality and Safety Control of Poultry Products, Ministry of Agriculture and Rural Affairs Zhengzhou 450046 Henan PR China
| | - Guanda Liang
- College of Veterinary Medicine, Henan Agricultural University Zhengzhou 450046 Henan PR China
- International Joint Research Laboratory for Zoonotic Diseases of Henan Zhengzhou 450046 Henan PR China
- Key Laboratory of Quality and Safety Control of Poultry Products, Ministry of Agriculture and Rural Affairs Zhengzhou 450046 Henan PR China
| | - Yuexin Wang
- College of Veterinary Medicine, Henan Agricultural University Zhengzhou 450046 Henan PR China
- International Joint Research Laboratory for Zoonotic Diseases of Henan Zhengzhou 450046 Henan PR China
- Key Laboratory of Quality and Safety Control of Poultry Products, Ministry of Agriculture and Rural Affairs Zhengzhou 450046 Henan PR China
| | - Baiyi Cai
- Department of Medicine, University of Alabama at Birmingham Birmingham AL 35249 USA
| | - Longxian Zhang
- College of Veterinary Medicine, Henan Agricultural University Zhengzhou 450046 Henan PR China
- International Joint Research Laboratory for Zoonotic Diseases of Henan Zhengzhou 450046 Henan PR China
- Key Laboratory of Quality and Safety Control of Poultry Products, Ministry of Agriculture and Rural Affairs Zhengzhou 450046 Henan PR China
| | - Xiaoying Li
- College of Veterinary Medicine, Henan Agricultural University Zhengzhou 450046 Henan PR China
- International Joint Research Laboratory for Zoonotic Diseases of Henan Zhengzhou 450046 Henan PR China
- Key Laboratory of Quality and Safety Control of Poultry Products, Ministry of Agriculture and Rural Affairs Zhengzhou 450046 Henan PR China
| | - Sumei Zhang
- College of Veterinary Medicine, Henan Agricultural University Zhengzhou 450046 Henan PR China
- International Joint Research Laboratory for Zoonotic Diseases of Henan Zhengzhou 450046 Henan PR China
- Key Laboratory of Quality and Safety Control of Poultry Products, Ministry of Agriculture and Rural Affairs Zhengzhou 450046 Henan PR China
| |
Collapse
|
6
|
Najer A. Pathogen-binding nanoparticles to inhibit host cell infection by heparan sulfate and sialic acid dependent viruses and protozoan parasites. SMART MEDICINE 2024; 3:e20230046. [PMID: 39188697 PMCID: PMC11235646 DOI: 10.1002/smmd.20230046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 01/25/2024] [Indexed: 08/28/2024]
Abstract
Global health faces an immense burden from infectious diseases caused by viruses and intracellular protozoan parasites such as the coronavirus disease (COVID-19) and malaria, respectively. These pathogens propagate through the infection of human host cells. The first stage of this host cell infection mechanism is cell attachment, which typically involves interactions between the infectious agent and surface components on the host cell membranes, specifically heparan sulfate (HS) and/or sialic acid (SA). Hence, nanoparticles (NPs) which contain or mimic HS/SA that can directly bind to the pathogen surface and inhibit cell infection are emerging as potential candidates for an alternative anti-infection therapeutic strategy. These NPs can be prepared from metals, soft matter (lipid, polymer, and dendrimer), DNA, and carbon-based materials among others and can be designed to include aspects of multivalency, broad-spectrum activity, biocidal mechanisms, and multifunctionality. This review provides an overview of such anti-pathogen nanomedicines beyond drug delivery. Nanoscale inhibitors acting against viruses and obligate intracellular protozoan parasites are discussed. In the future, the availability of broadly applicable nanotherapeutics would allow early tackling of existing and upcoming viral diseases. Invasion inhibitory NPs could also provide urgently needed effective treatments for protozoan parasitic infections.
Collapse
Affiliation(s)
- Adrian Najer
- Institute of Pharmaceutical ScienceKing's College LondonLondonUK
| |
Collapse
|
7
|
Huang W, Tang K, Chen C, Arrowood MJ, Chen M, Guo Y, Li N, Roellig DM, Feng Y, Xiao L. Sequence introgression from exogenous lineages underlies genomic and biological differences among Cryptosporidium parvum IOWA lines. WATER RESEARCH 2024; 254:121333. [PMID: 38402753 PMCID: PMC10994760 DOI: 10.1016/j.watres.2024.121333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 02/15/2024] [Accepted: 02/16/2024] [Indexed: 02/27/2024]
Abstract
The IOWA strain of Cryptosporidium parvum is widely used in studies of the biology and detection of the waterborne pathogens Cryptosporidium spp. While several lines of the strain have been sequenced, IOWA-II, the only reference of the original subtype (IIaA15G2R1), exhibits significant assembly errors. Here we generated a fully assembled genome of IOWA-CDC of this subtype using PacBio and Illumina technologies. In comparative analyses of seven IOWA lines maintained in different laboratories (including two sequenced in this study) and 56 field isolates, IOWA lines (IIaA17G2R1) with less virulence had mixed genomes closely related to IOWA-CDC but with multiple sequence introgressions from IOWA-II and unknown lineages. In addition, the IOWA-IIaA17G2R1 lines showed unique nucleotide substitutions and loss of a gene associated with host infectivity, which were not observed in other isolates analyzed. These genomic differences among IOWA lines could be the genetic determinants of phenotypic traits in C. parvum. These data provide a new reference for comparative genomic analyses of Cryptosporidium spp. and rich targets for the development of advanced source tracking tools.
Collapse
Affiliation(s)
- Wanyi Huang
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou 510642, China
| | - Kevin Tang
- Division of Scientific Resources, Centers for Disease Control and Prevention, Atlanta, GA 30341, USA
| | - Chengyi Chen
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou 510642, China
| | - Michael J Arrowood
- Division of Foodborne, Waterborne, and Environmental Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia 30341, USA
| | - Ming Chen
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou 510642, China
| | - Yaqiong Guo
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou 510642, China
| | - Na Li
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou 510642, China
| | - Dawn M Roellig
- Division of Foodborne, Waterborne, and Environmental Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia 30341, USA.
| | - Yaoyu Feng
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou 510642, China.
| | - Lihua Xiao
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
8
|
Wu TL, Wang BN, Yang AJ, Wang L, You YN, Zhou RQ. C-type lectin 4 of Toxocara canis activates NF-ĸB and MAPK pathways by modulating NOD1/2 and RIP2 in murine macrophages in vitro. Parasitol Res 2024; 123:189. [PMID: 38639821 DOI: 10.1007/s00436-024-08212-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 04/15/2024] [Indexed: 04/20/2024]
Abstract
Toxocara canis is a parasitic zoonose that is distributed worldwide and is one of the two pathogens causing toxocariasis. After infection, it causes serious public health and safety problems, which pose significant veterinary and medical challenges. To better understand the regulatory effects of T. canis infection on the host immune cells, murine macrophages (RAW264.7) were incubated with recombinant T. canis C-type lectin 4 (rTc-CTL-4) protein in vitro. The quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot were used to analyze the nucleotide-binding oligomerization domain-containing protein 1/2 (NOD1/2), receptor-interacting protein 2 (RIP2), nuclear factor kappa-light-chain enhancer of activated B cells (NF-κB), and mitogen-activated protein kinase (MAPK) on mRNA level and protein expression level in macrophages. Our results indicated that 10 μg/mL rTc-CTL-4 protein could modulate the expression of NOD1, NOD2, and RIP2 at both the transcriptional and translational levels. The protein translation levels of NF-κB, P-p65, p38, and P-p38 in macrophages were also modulated by rTc-CTL-4 protein. Macrophages were co-incubated with rTc-CTL-4 protein after siRNA silencing of NOD1, NOD2, and RIP2. The expression levels of NF-κB, P-p65, p38, and P-p38 were significantly changed compared with the negative control groups (Neg. Ctrl.). Taken together, rTc-CTL-4 protein seemed to act on NOD1/2-RIP2-NF-κB and MAPK signaling pathways in macrophages and might activate MAPK and NF-κB signaling pathways by regulating NOD1, NOD2, and RIP2. The insights from the above studies could contribute to our understanding of immune recognition and regulatory mechanisms of T. canis infection in the host animals.
Collapse
Affiliation(s)
- Tian-Le Wu
- College of Veterinary Medicine, Southwest University, Chongqing, 402460, China
| | - Bing-Nan Wang
- College of Veterinary Medicine, Southwest University, Chongqing, 402460, China
| | - Ai-Jia Yang
- College of Veterinary Medicine, Southwest University, Chongqing, 402460, China
| | - Lei Wang
- College of Veterinary Medicine, Southwest University, Chongqing, 402460, China
| | - Yi-Ning You
- College of Veterinary Medicine, Southwest University, Chongqing, 402460, China
| | - Rong-Qiong Zhou
- College of Veterinary Medicine, Southwest University, Chongqing, 402460, China.
| |
Collapse
|
9
|
Wang Z, Lu QQ, Weng MM, Li YL, Han LL, Song YY, Shi YL, Liu RD, Cui J, Wang ZQ. Binding of Trichinella spiralis C-type lectin with syndecan-1 on intestinal epithelial cells mediates larval invasion of intestinal epithelium. Vet Res 2023; 54:86. [PMID: 37784173 PMCID: PMC10546719 DOI: 10.1186/s13567-023-01217-2] [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: 07/14/2023] [Accepted: 08/18/2023] [Indexed: 10/04/2023] Open
Abstract
C-type lectin (CTL) is a protein that binds to saccharides and plays an important role in parasite adhesion, host cell invasion and immune evasion. Previous studies showed that recombinant T. spiralis C-type lectin (rTsCTL) promotes larval invasion of intestinal epithelium cells (IEC), whereas anti-rTsCTL antibodies inhibits larval invasion. Syndecan-1 (SDC-1) is a member of the heparan sulfate proteoglycan family which is mainly expressed on the surface of IEC and in extracellular matrices where they interact with a plethora of ligands. SDC-1 has a principal role in maintaining cell morphogenesis, establishing cell-cell adhesions, and regulating the gut mucosal barrier. The aim of this study was to investigate whether rTsCTL binds to SDC-1 on IEC, and the binding of rTsCTL with SDC-1 promotes larval invasion and its mechanism. IFA results show that rTsCTL and SDC-1 co-localized on Caco-2 cell membrane. GST pull-down and Co-IP verified the direct interaction between rTsCTL and SDC-1 on Caco-2 cells. qPCR and Western blotting revealed that rTsCTL binding to SDC-1 increased the expression of SDC-1 and claudin-2, and reduced the expression of occludin and claudin-1 in Caco-2 cells incubated with rTsCTL via the STAT3 pathway. β-Xyloside (a syndecan-1 synthesis inhibitor) and Stattic (a STAT3 inhibitor) significantly inhibited rTsCTL binding to syndecan-1 in Caco-2 cells and activation of the STAT3 pathway, abrogated the effects of rTsCTL on the expression of gut tight junctions, and impeded larval invasion. The results demonstrate that binding of rTsCTL to SDC-1 on Caco-2 cells activated the STAT3 pathway, decreased gut tight junction expression, damaged the integrity of the gut epithelial barrier, and mediated T. spiralis invasion of the gut mucosa. TsCTL might be regarded as a candidate vaccine target against T. spiralis invasion and infection.
Collapse
Affiliation(s)
- Zhen Wang
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, 450052, China
| | - Qi Qi Lu
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, 450052, China
| | - Min Min Weng
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, 450052, China
| | - Yang Li Li
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, 450052, China
| | - Lu Lu Han
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, 450052, China
| | - Yan Yan Song
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, 450052, China
| | - Yu Long Shi
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, 450052, China
| | - Ruo Dan Liu
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, 450052, China
| | - Jing Cui
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, 450052, China.
| | - Zhong Quan Wang
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
10
|
Mayerberger EA, Yazdanparast Tafti S, Jedlicka SS, Jellison KL. Effect of Glycosaminoglycans on Cryptosporidium Oocyst Attachment and Excystation. Appl Environ Microbiol 2023; 89:e0173722. [PMID: 36790186 PMCID: PMC10056967 DOI: 10.1128/aem.01737-22] [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] [Received: 10/18/2022] [Accepted: 01/11/2023] [Indexed: 02/16/2023] Open
Abstract
Cryptosporidium causes severe gastrointestinal disease resulting from the ingestion of oocysts, followed by oocyst excystation in the small intestine and the release of infective sporozoites. An understudied strategy for Cryptosporidium inactivation is purposeful oocyst excystation, as sporozoites do not survive long in the environment. This study showed that C. parvum oocyst excystation was induced by direct contact with various glycosaminoglycans (GAGs), including heparin (Hep), chondroitin sulfate A (CSA), and hyaluronan (HA), assembled on polydopamine (PD)-functionalized surfaces. PD surfaces elicited 97.9 ± 3.6% oocyst attachment, with some of the attached oocysts partially (7.3 ± 1.3%) or fully (4.0 ± 0.6%) excysted after 4 days. The PD-GAG surfaces (GAG concentration = 2 mg/mL) elicited similarly high attachment (>97%) and higher oocyst excystation efficiencies after 4 days. The PD-Hep surfaces elicited the highest number of attached excysted oocysts (11.8 ± 0.63% partially excysted; 11.9 ± 0.49% fully excysted), and the PD-HA surfaces elicited the lowest (8.8 ± 2.1% partially excysted; 7.8 ± 1.2% fully excysted). Surface characterization revealed that the addition of GAGs to the PD surface changed both the surface roughness as well as the surface wettability. Treatment of oocysts with an enzyme that degraded the surface glycocalyx markedly reduced excystation (to <2%) of the oocysts attached to the PD and PD-GAG surfaces. These findings suggest that GAGs provide an important local signal for the excystation of C. parvum oocysts and that certain surface-expressed oocyst receptors are necessary for efficient excystation. These oocyst-receptor relationships may be useful in the design of functionalized surfaces for the purposeful inactivation of oocysts in the environment or in water treatment systems. IMPORTANCE Polydopamine surfaces functionalized with glycosaminoglycans were shown to facilitate the attachment and excystation of Cryptosporidium parvum oocysts. Our findings suggest that a surface-expressed receptor on the oocyst wall plays a key role in excystation, with glycosaminoglycans serving as ligands that trigger the initiation of the process. Future technologies and treatment strategies designed to promote premature excystation of oocysts will minimize the ingestion of sporozoites that initiate infection. Therefore, the results from this study have important implications for the protection of public health from waterborne cryptosporidiosis and may serve as a foundation for engineered surfaces designed to remove oocysts from surface waters or inactivate oocysts in water treatment systems.
Collapse
Affiliation(s)
- Elisa A. Mayerberger
- Department of Civil and Environmental Engineering, Lehigh University, Bethlehem, Pennsylvania, USA
| | | | - Sabrina S. Jedlicka
- Department of Bioengineering, Lehigh University, Bethlehem, Pennsylvania, USA
- Department of Materials Science and Engineering, Lehigh University, Bethlehem, Pennsylvania, USA
| | - Kristen L. Jellison
- Department of Civil and Environmental Engineering, Lehigh University, Bethlehem, Pennsylvania, USA
| |
Collapse
|
11
|
Hao HN, Lu QQ, Wang Z, Li YL, Long SR, Dan Liu R, Cui J, Wang ZQ. Mannose facilitates Trichinella spiralis expulsion from the gut and alleviates inflammation of intestines and muscles in mice. Acta Trop 2023; 241:106897. [PMID: 36931335 DOI: 10.1016/j.actatropica.2023.106897] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 10/25/2022] [Accepted: 03/13/2023] [Indexed: 03/17/2023]
Abstract
Trichinellosis is a major zoonotic parasitosis which is a vital risk to meat food safety. It is requisite to exploit new strategy to interdict food animal Trichinella infection and to obliterate Trichinella from food animals to ensure meat safety. Mannose is an oligosaccharide that specifically binds to the carbohydrate-recognition domain of C-type lectin; it has many physiological functions including reliving inflammation and regulating immune reaction. The purpose of this study was to investigate the suppressive role of mannose on T. spiralis larval invasion and infection, its effect on intestinal and muscle inflammation, and immune responses after challenge. The results showed that compared to the saline-treated infected mice, the mannose-treated infected mice had less intestinal adult and muscle worm burdens, mild inflammation of intestine and muscle of infected mice. The levels of specific anti-Trichinella IgG (IgG1/IgG2a), IgA and sIgA in mannose-treated infected mice were obviously inferior to saline-treated infected mice (P < 0.01). Furthermore, the levels of two cytokines (IFN-γ and IL-4) in mannose-treated infected mice were also significantly lower than the saline-treated infected mice (P < 0.01). The protective effect of the mannose against Trichinella infection might be not related to specific antibody and cellular immune responses. The above results demonstrated that mannose could be considered as a novel adjuvant therapeutic agent for anti-Trichinella drugs to block larval invasion at early stage of Trichinella infection.
Collapse
Affiliation(s)
- Hui Nan Hao
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou 450052, China
| | - Qi Qi Lu
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou 450052, China
| | - Zhen Wang
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou 450052, China
| | - Yang Li Li
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou 450052, China
| | - Shao Rong Long
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou 450052, China
| | - Ruo Dan Liu
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou 450052, China
| | - Jing Cui
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou 450052, China.
| | - Zhong Quan Wang
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou 450052, China.
| |
Collapse
|
12
|
Huang W, Guo Y, Lysen C, Wang Y, Tang K, Seabolt MH, Yang F, Cebelinski E, Gonzalez-Moreno O, Hou T, Chen C, Chen M, Wan M, Li N, Hlavsa MC, Roellig DM, Feng Y, Xiao L. Multiple introductions and recombination events underlie the emergence of a hyper-transmissible Cryptosporidium hominis subtype in the USA. Cell Host Microbe 2023; 31:112-123.e4. [PMID: 36521488 PMCID: PMC10124589 DOI: 10.1016/j.chom.2022.11.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/20/2022] [Accepted: 11/22/2022] [Indexed: 12/15/2022]
Abstract
The parasite Cryptosporidium hominis is a leading cause of the diarrheal disease cryptosporidiosis, whose incidence in the United States has increased since 2005. Here, we show that the newly emerged and hyper-transmissible subtype IfA12G1R5 is now dominant in the United States. In a comparative analysis of 127 newly sequenced and 95 published C. hominis genomes, IfA12G1R5 isolates from the United States place into three of the 14 clusters (Pop6, Pop13, and Pop14), indicating that this subtype has multiple ancestral origins. Pop6 (IfA12G1R5a) has an East Africa origin and has recombined with autochthonous subtypes after its arrival. Pop13 (IfA12G1R5b) is imported from Europe, where it has recombined with the prevalent local subtype, whereas Pop14 (IfA12G1R5c) is a progeny of secondary recombination between Pop6 and Pop13. Selective sweeps in invasion-associated genes have accompanied the emergence of the dominant Pop14. These observations offer insights into the emergence and evolution of hyper-transmissible pathogens.
Collapse
Affiliation(s)
- Wanyi Huang
- Guangdong Laboratory for Lingnan Modern Agriculture, Center for Emerging and Zoonotic Diseases, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Yaqiong Guo
- Guangdong Laboratory for Lingnan Modern Agriculture, Center for Emerging and Zoonotic Diseases, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Colleen Lysen
- Division of Foodborne, Waterborne, and Environmental Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30341, USA
| | - Yuanfei Wang
- Division of Foodborne, Waterborne, and Environmental Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30341, USA
| | - Kevin Tang
- Division of Scientific Resources, Centers for Disease Control and Prevention, Atlanta, GA 30341, USA
| | - Matthew H Seabolt
- Division of Foodborne, Waterborne, and Environmental Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30341, USA
| | - Fengkun Yang
- Division of Foodborne, Waterborne, and Environmental Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30341, USA
| | - Elizabeth Cebelinski
- Infectious Disease Laboratory, Minnesota Department of Health, St. Paul, MN 55101, USA
| | | | - Tianyi Hou
- Guangdong Laboratory for Lingnan Modern Agriculture, Center for Emerging and Zoonotic Diseases, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Chengyi Chen
- Guangdong Laboratory for Lingnan Modern Agriculture, Center for Emerging and Zoonotic Diseases, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Ming Chen
- Guangdong Laboratory for Lingnan Modern Agriculture, Center for Emerging and Zoonotic Diseases, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Muchun Wan
- Guangdong Laboratory for Lingnan Modern Agriculture, Center for Emerging and Zoonotic Diseases, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Na Li
- Guangdong Laboratory for Lingnan Modern Agriculture, Center for Emerging and Zoonotic Diseases, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Michele C Hlavsa
- Division of Foodborne, Waterborne, and Environmental Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30341, USA
| | - Dawn M Roellig
- Division of Foodborne, Waterborne, and Environmental Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30341, USA.
| | - Yaoyu Feng
- Guangdong Laboratory for Lingnan Modern Agriculture, Center for Emerging and Zoonotic Diseases, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Lihua Xiao
- Guangdong Laboratory for Lingnan Modern Agriculture, Center for Emerging and Zoonotic Diseases, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
13
|
Hao HN, Song YY, Ma KN, Wang BN, Long SR, Liu RD, Zhang X, Wang ZQ, Cui J. A novel C-type lectin from Trichinella spiralis mediates larval invasion of host intestinal epithelial cells. Vet Res 2022; 53:85. [PMID: 36258242 PMCID: PMC9580147 DOI: 10.1186/s13567-022-01104-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 09/05/2022] [Indexed: 11/10/2022] Open
Abstract
The aim of this study was to investigate the characteristics of a novel type C lectin from Trichinella spiralis (TsCTL) and its role in larval invasion of intestinal epithelial cells (IECs). TsCTL has a carbohydrate recognition domain (CRD) of C-type lectin. The full-length TsCTL cDNA sequence was cloned and expressed in Escherichia coli BL21. The results of qPCR, Western blotting and immunofluorescence assays (IFAs) showed that TsCTL was a surface and secretory protein that was highly expressed at the T. spiralis intestinal infective larva (IIL) stages and primarily located at the cuticle, stichosome and embryos of the parasite. rTsCTL could specifically bind with IECs, and the binding site was localized in the IEC nucleus and cytoplasm. The IFA results showed that natural TsCTL was secreted and bound to the enteral epithelium at the intestinal stage of T. spiralis infection. The rTsCTL had a haemagglutinating effect on murine erythrocytes, while mannose was able to inhibit the rTsCTL agglutinating effect for mouse erythrocytes. rTsCTL accelerated larval intrusion into the IECs, whereas anti-rTsCTL antibodies and mannose significantly impeded larval intrusion in a dose-dependent manner. The results indicated that TsCTL specifically binds to IECs and promotes larval invasion of intestinal epithelium, and it might be a potential target of vaccines against T. spiralis enteral stages.
Collapse
Affiliation(s)
- Hui Nan Hao
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, 450052 China
| | - Yan Yan Song
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, 450052 China
| | - Kai Ning Ma
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, 450052 China
| | - Bo Ning Wang
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, 450052 China
| | - Shao Rong Long
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, 450052 China
| | - Ruo Dan Liu
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, 450052 China
| | - Xi Zhang
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, 450052 China
| | - Zhong Quan Wang
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, 450052 China
| | - Jing Cui
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, 450052 China
| |
Collapse
|
14
|
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
|
15
|
He X, Huang W, Sun L, Hou T, Wan Z, Li N, Guo Y, Kváč M, Xiao L, Feng Y. A productive immunocompetent mouse model of cryptosporidiosis with long oocyst shedding duration for immunological studies. J Infect 2022; 84:710-721. [PMID: 35192895 DOI: 10.1016/j.jinf.2022.02.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/14/2022] [Accepted: 02/16/2022] [Indexed: 11/28/2022]
Abstract
OBJECTIVES Studies on the pathogenesis and immune responses of Cryptosporidium infection and development of drugs and vaccines use mostly immunocompromised mouse models. In this study, we establish an immunocompetent mouse model of cryptosporidiosis with high intensity and long duration of infection. METHODS We have obtained a Cryptosporidium tyzzeri isolate from laboratory mice, and infect adult C57BL/6J mice experimentally with the isolate for determinations of infectivity, infection patterns, pathological changes, and transcriptomic responses. RESULTS The isolate has an ID50 of 5.2 oocysts, with oocyst shedding lasting at high levels for >2 months. The oocyst shedding is boosted by immunosuppression of animals and suppressed by paromomycin treatment. The isolate induces strong inflammatory and acquired immune responses, but down-regulates the expression of α-defensins in epithelium. Comparative genomics analysis has revealed significant sequence differences from other isolates in subtelomeric genes. The down-regulation of the expression of α-defensins may be responsible for the high-intensity and long-lasting infection in this animal model. CONCLUSIONS The immunocompetent mouse model of cryptosporidiosis developed has the advantages of high oocyst shedding intensity and long oocyst shedding duration. It provides an effective mechanism for the propagation of Cryptosporidium, evaluations of potential therapeutics, and studies of pathogen biology and immune responses.
Collapse
Affiliation(s)
- Xi He
- Center for Emerging and Zoonotic Diseases, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture. Guangzhou, Guangdong 510642, China.
| | - Wanyi Huang
- Center for Emerging and Zoonotic Diseases, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Lianbei Sun
- Center for Emerging and Zoonotic Diseases, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Tianyi Hou
- Center for Emerging and Zoonotic Diseases, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Zhuowei Wan
- Center for Emerging and Zoonotic Diseases, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Na Li
- Center for Emerging and Zoonotic Diseases, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Yaqiong Guo
- Center for Emerging and Zoonotic Diseases, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Martin Kváč
- Institute of Parasitology, Biology Centre of the Academy of Sciences of the Czech Republic, České Budějovice 370 05, Czech Republic.
| | - Lihua Xiao
- Center for Emerging and Zoonotic Diseases, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture. Guangzhou, Guangdong 510642, China.
| | - Yaoyu Feng
- Center for Emerging and Zoonotic Diseases, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture. Guangzhou, Guangdong 510642, China.
| |
Collapse
|
16
|
The Oesophageal Squamous Cell Carcinoma Cell Line COLO-680N Fails to Support Sustained Cryptosporidium parvum Proliferation. Pathogens 2021; 11:pathogens11010049. [PMID: 35055997 PMCID: PMC8778297 DOI: 10.3390/pathogens11010049] [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: 11/29/2021] [Revised: 12/28/2021] [Accepted: 12/30/2021] [Indexed: 11/16/2022] Open
Abstract
Cryptosporidium parvum is an important diarrhoea-associated protozoan, which is difficult to propagate in vitro. In 2017, a report described a continuous culture of C. parvum Moredun strain, in the oesophageal squamous cell carcinoma cell line COLO-680N, as an easy-to-use system for C. parvum propagation and continuous production of oocysts. Here, we report that—using the Köllitsch strain of C. parvum—even though COLO-680N cells, indeed, allowed parasite invasion and early asexual parasite replication, C. parvum proliferation decreased after the second day post infection. Considering recurring studies, reporting on successful production of newly generated Cryptosporidium oocysts in the past, and the subsequent replication failure by other research groups, the current data stand as a reminder of the importance of reproducibility of in vitro systems in cryptosporidiosis research. This is of special importance since it will only be possible to develop promising strategies to fight cryptosporidiosis and its ominous consequences for both human and animal health by a continuous and reliable methodological progress.
Collapse
|
17
|
Bauer S, Zhang F, Linhardt RJ. Implications of Glycosaminoglycans on Viral Zoonotic Diseases. Diseases 2021; 9:85. [PMID: 34842642 PMCID: PMC8628766 DOI: 10.3390/diseases9040085] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/10/2021] [Accepted: 11/15/2021] [Indexed: 11/21/2022] Open
Abstract
Zoonotic diseases are infectious diseases that pass from animals to humans. These include diseases caused by viruses, bacteria, fungi, and parasites and can be transmitted through close contact or through an intermediate insect vector. Many of the world's most problematic zoonotic diseases are viral diseases originating from animal spillovers. The Spanish influenza pandemic, Ebola outbreaks in Africa, and the current SARS-CoV-2 pandemic are thought to have started with humans interacting closely with infected animals. As the human population grows and encroaches on more and more natural habitats, these incidents will only increase in frequency. Because of this trend, new treatments and prevention strategies are being explored. Glycosaminoglycans (GAGs) are complex linear polysaccharides that are ubiquitously present on the surfaces of most human and animal cells. In many infectious diseases, the interactions between GAGs and zoonotic pathogens correspond to the first contact that results in the infection of host cells. In recent years, researchers have made progress in understanding the extraordinary roles of GAGs in the pathogenesis of zoonotic diseases, suggesting potential therapeutic avenues for using GAGs in the treatment of these diseases. This review examines the role of GAGs in the progression, prevention, and treatment of different zoonotic diseases caused by viruses.
Collapse
Affiliation(s)
- Sarah Bauer
- Department of Chemistry and Chemical Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA;
| | - Fuming Zhang
- Department of Chemistry and Chemical Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA;
- Department of Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Robert J. Linhardt
- Department of Chemistry and Chemical Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA;
- Department of Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
- Departments of Biological Science, Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| |
Collapse
|
18
|
Establishment of a Newborn Lamb Gut-Loop Model to Evaluate New Methods of Enteric Disease Control and Reduce Experimental Animal Use. Vet Sci 2021; 8:vetsci8090170. [PMID: 34564564 PMCID: PMC8472880 DOI: 10.3390/vetsci8090170] [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: 07/30/2021] [Revised: 08/20/2021] [Accepted: 08/20/2021] [Indexed: 11/16/2022] Open
Abstract
Enteric infectious diseases are not all well controlled, which leads to animal suffering and sometimes death in the most severe cases, in addition to economic losses for farmers. Typical symptoms of enteric infections include watery diarrhea, stomach cramps or pain, dehydration, nausea, vomiting, fever and weight loss. Evaluation of new control methods against enteric infections requires the use of many animals. We aimed to develop a new method for an initial in vivo screen of promising compounds against neonatal diseases such as cryptosporidiosis while limiting experimental animal use. We therefore adapted an in vivo method of multiple consecutive but independent intestinal loops to newborn lambs delivered by cesarean section, in which endotoxin responsiveness is retained. This new method allowed for the screening of natural yeast fractions for their ability to stimulate immune responses and to limit early Cryptosporidium parvum development. This model may also be used to investigate host–pathogen interactions and immune responses in a neonatal controlled environment.
Collapse
|
19
|
Zhang T, Gao X, Wang D, Zhao J, Zhang N, Li Q, Zhu G, Yin J. A Single-Pass Type I Membrane Protein from the Apicomplexan Parasite Cryptosporidium parvum with Nanomolar Binding Affinity to Host Cell Surface. Microorganisms 2021; 9:microorganisms9051015. [PMID: 34066754 PMCID: PMC8151451 DOI: 10.3390/microorganisms9051015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/04/2021] [Accepted: 05/06/2021] [Indexed: 11/16/2022] Open
Abstract
Cryptosporidium parvum is a globally recognized zoonotic parasite of medical and veterinary importance. This parasite mainly infects intestinal epithelial cells and causes mild to severe watery diarrhea that could be deadly in patients with weakened or defect immunity. However, its molecular interactions with hosts and pathogenesis, an important part in adaptation of parasitic lifestyle, remain poorly understood. Here we report the identification and characterization of a C. parvum T-cell immunomodulatory protein homolog (CpTIPH). CpTIPH is a 901-aa single-pass type I membrane protein encoded by cgd5_830 gene that also contains a short Vibrio, Colwellia, Bradyrhizobium and Shewanella (VCBS) repeat and relatively long integrin alpha (ITGA) N-terminus domain. Immunofluorescence assay confirmed the location of CpTIPH on the cell surface of C. parvum sporozoites. In congruence with the presence of VCBS repeat and ITGA domain, CpTIPH displayed high, nanomolar binding affinity to host cell surface (i.e., Kd(App) at 16.2 to 44.7 nM on fixed HCT-8 and CHO-K1 cells, respectively). The involvement of CpTIPH in the parasite invasion is partly supported by experiments showing that an anti-CpTIPH antibody could partially block the invasion of C. parvum sporozoites into host cells. These observations provide a strong basis for further investigation of the roles of CpTIPH in parasite-host cell interactions.
Collapse
Affiliation(s)
- Tianyu Zhang
- Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun 130062, China; (T.Z.); (X.G.); (D.W.); (N.Z.); (Q.L.)
- Peking-Tsinghua Center for Life Sciences and Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Xin Gao
- Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun 130062, China; (T.Z.); (X.G.); (D.W.); (N.Z.); (Q.L.)
| | - Dongqiang Wang
- Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun 130062, China; (T.Z.); (X.G.); (D.W.); (N.Z.); (Q.L.)
| | - Jixue Zhao
- Department of Pediatric Surgery, First Hospital of Jilin University, Changchun 130021, China;
| | - Nan Zhang
- Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun 130062, China; (T.Z.); (X.G.); (D.W.); (N.Z.); (Q.L.)
| | - Qiushi Li
- Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun 130062, China; (T.Z.); (X.G.); (D.W.); (N.Z.); (Q.L.)
- Hospital of Stomatology, Jilin University, Changchun 130041, China
| | - Guan Zhu
- Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun 130062, China; (T.Z.); (X.G.); (D.W.); (N.Z.); (Q.L.)
- Correspondence: (G.Z.); (J.Y.)
| | - Jigang Yin
- Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun 130062, China; (T.Z.); (X.G.); (D.W.); (N.Z.); (Q.L.)
- Correspondence: (G.Z.); (J.Y.)
| |
Collapse
|
20
|
Martínez-Ocaña J, Maravilla P, Olivo-Díaz A. Interaction between human mucins and parasite glycoproteins: the role of lectins and glycosidases in colonization by intestinal protozoa. Rev Inst Med Trop Sao Paulo 2020; 62:e64. [PMID: 32901761 PMCID: PMC7477959 DOI: 10.1590/s1678-9946202062064] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 08/17/2020] [Indexed: 08/30/2023] Open
Abstract
Intestinal mucins are the first line of defense against microorganisms. Although knowledge about the mechanisms involved in the establishment of intestinal protozoa is limited, there is evidence that these parasites produce lectin-like molecules and glycosidases, that exert both, constitutive and secretory functions, promoting the establishment of these microorganisms. In the present review, we analyse the main interactions between mucins of the host intestine and the four main protozoan parasites in humans and their implications in intestinal colonization. There are lectin-like molecules that contain complex oligosaccharide structures and N-acetylglucosamine (GlcNAc), mannose and sialic acid as main components, which are excreted/secreted by Giardia intestinalis, and recognized by the host using mannose-binding lectins (MBL). Entamoeba histolytica and Cryptosporidium spp. express the lectin galactose/N-acetyl-D-galactosamine, which facilitates their adhesion to cells. In Cryptosporidium, the glycoproteins gp30, gp40/15 and gp900 and the glycoprotein lectin CpClec are involved in protozoan adhesion to intestinal cells, forming an adhesion-attack complex. G. intestinalis and E. histolytica can also produce glycosidases such as β-N-acetyl-D-glucosaminidase, α-d-glucosidase, β-d-galactosidase, β-l-fucosidase, α-N-acetyl-d-galactosaminidase and β-mannosidase. In Blastocystis, α-D-mannose, α-D-glucose, GlcNAc, α-D-fucose, chitin and sialic acid that have been identified on their surface. Fucosidases, hexosaminidases and polygalacturonases, which may be involved in the mucin degradation process, have also been described in the Blastocystis secretoma. Similarly, symbiotic coexistence with the intestinal microbiota promotes the survival of parasites facilitating cell invasion and nutrients obtention. Furthermore, it is necessary to identify and characterize more glycosidases, which have been only partially described by in silico analyses of the parasite genome.
Collapse
Affiliation(s)
- Joel Martínez-Ocaña
- Hospital General "Dr. Manuel Gea González", Departamento de Ecología de Agentes Patógenos, Ciudad de México, Mexico
| | - Pablo Maravilla
- Hospital General "Dr. Manuel Gea González", Subdirección de Investigación, Ciudad de México, Mexico
| | - Angélica Olivo-Díaz
- Hospital General "Dr. Manuel Gea González", Departamento de Biología Molecular e Histocompatibilidad, Hospital General "Dr. Manuel Gea González", Ciudad de México, Mexico
| |
Collapse
|
21
|
Costa RB, Campana PT, Chambergo FS, Napoleão TH, Paiva PMG, Pereira HJV, Oliva MLV, Gomes FS. Purification and characterization of a lectin with refolding ability from Genipa americana bark. Int J Biol Macromol 2018; 119:517-523. [DOI: 10.1016/j.ijbiomac.2018.07.178] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 07/24/2018] [Accepted: 07/28/2018] [Indexed: 12/29/2022]
|
22
|
Zhang L, Mou L, Chen X, Yang Y, Hu M, Li X, Suo X, Zhu XQ, Du A. Identification and preliminary characterization of Hc-clec-160, a novel C-type lectin domain-containing gene of the strongylid nematode Haemonchus contortus. Parasit Vectors 2018; 11:430. [PMID: 30029661 PMCID: PMC6054721 DOI: 10.1186/s13071-018-3005-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Accepted: 07/09/2018] [Indexed: 11/10/2022] Open
Abstract
Background The strongylid parasite Haemonchus contortus causes severe anemia in domestic animals worldwide. Effective preventive and therapeutical agents are lacking, because of drug resistance and that little is known about the molecular mechanism of the interaction between H. contortus and host cells. Methods A new gene, Hc-clec-160, was discovered with RT-PCR. Transcriptional levels of Hc-clec-160 and Ce-clec-160 throughout different growth phases of corresponding nematodes were assayed by qPCR. Immunofluorescence staining of paraffin section were performed to determine the protein localization in adult worms of H. contortus. To monitor the promoter capacity of the 5'-flanking region of Ce-clec-160, micro-injection was used. Overexpression and RNAi constructs was carried out in the N2 strain of Caenorhabditis elegans to find out the gene function of Hc-clec-160. Results The full-length cDNA of 1224 bp of Hc-clec-160 was cloned by RT-PCR. The corresponding gene contained twelve exons. Its transcripts peaked in male adult worms. Hc-CLEC-160 was predicted to have a Willebrand factor type A (vWA) domain and a C-type lectin domain. The proteins were not detected by expression in C. elegans or paraffin section experiments in adult of H. contortus. Knockdown of Ce-clec-160 expression in C. elegans by RNAi resulted in shortened body length and decreased brood size. Conclusions In this experiment, a new gene Hc-clec-160 was obtained in H. contortus and its function was addressed using a model organism: C. elegans. Our study showed that Hc-clec-160 possesses characteristics similar to those of Ce-clec-160 and plays an important role in the growth and reproduction of this parasitic nematode. Electronic supplementary material The online version of this article (10.1186/s13071-018-3005-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ling Zhang
- College of Animal Sciences, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Zhejiang University, Hangzhou, China
| | - Lingyun Mou
- College of Animal Sciences, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Zhejiang University, Hangzhou, China
| | - Xueqiu Chen
- College of Animal Sciences, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Zhejiang University, Hangzhou, China
| | - Yi Yang
- College of Animal Sciences, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Zhejiang University, Hangzhou, China
| | - Min Hu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xiangrui Li
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xun Suo
- State Key Laboratory of Agrobiotechnology, Key Laboratory of Zoonosis of Ministry of Agriculture, National Animal Protozoa Laboratory and College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xing-Quan Zhu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, Gansu Province, China
| | - Aifang Du
- College of Animal Sciences, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
23
|
Dagher Z, Xu S, Negoro PE, Khan NS, Feldman MB, Reedy JL, Tam JM, Sykes DB, Mansour MK. Fluorescent Tracking of Yeast Division Clarifies the Essential Role of Spleen Tyrosine Kinase in the Intracellular Control of Candida glabrata in Macrophages. Front Immunol 2018; 9:1058. [PMID: 29868018 PMCID: PMC5964189 DOI: 10.3389/fimmu.2018.01058] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 04/27/2018] [Indexed: 01/07/2023] Open
Abstract
Macrophages play a critical role in the elimination of fungal pathogens. They are sensed via cell surface pattern-recognition receptors and are phagocytosed into newly formed organelles called phagosomes. Phagosomes mature through the recruitment of proteins and lysosomes, resulting in addition of proteolytic enzymes and acidification of the microenvironment. Our earlier studies demonstrated an essential role of Dectin-1-dependent activation of spleen tyrosine kinase (Syk) in the maturation of fungal containing phagosomes. The absence of Syk activity interrupted phago-lysosomal fusion resulting in arrest at an early phagosome stage. In this study, we sought to define the contribution of Syk to the control of phagocytosed live Candida glabrata in primary macrophages. To accurately measure intracellular yeast division, we designed a carboxyfluorescein succinimidyl ester (CFSE) yeast division assay in which bright fluorescent parent cells give rise to dim daughter cells. The CFSE-labeling of C. glabrata did not affect the growth rate of the yeast. Following incubation with macrophages, internalized CFSE-labeled C. glabrata were retrieved by cellular lysis, tagged using ConA-647, and the amount of residual CFSE fluorescence was assessed by flow cytometry. C. glabrata remained undivided (CFSE bright) for up to 18 h in co-culture with primary macrophages. Treatment of macrophages with R406, a specific Syk inhibitor, resulted in loss of intracellular control of C. glabrata with initiation of division within 4 h. Delayed Syk inhibition after 8 h was less effective indicating that Syk is critically required at early stages of macrophage–fungal interaction. In conclusion, we demonstrate a new method of tracking division of C. glabrata using CFSE labeling. Our results suggest that early Syk activation is essential for macrophage control of phagocytosed C. glabrata.
Collapse
Affiliation(s)
- Zeina Dagher
- Division of Infectious Disease, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Shuying Xu
- Division of Infectious Disease, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Paige E Negoro
- Division of Infectious Disease, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Nida S Khan
- Division of Infectious Disease, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States.,Biomedical Engineering and Biotechnology, University of Massachusetts Medical School, Worcester, MA, United States
| | - Michael B Feldman
- Division of Pulmonary and Critical Care, Massachusetts General Hospital, Boston, MA, United States
| | - Jennifer L Reedy
- Division of Infectious Disease, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Jenny M Tam
- Division of Infectious Disease, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - David B Sykes
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, United States
| | - Michael K Mansour
- Division of Infectious Disease, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
24
|
Vong M, Ludington JG, Ward HD, Nibert ML. Complete cryspovirus genome sequences from Cryptosporidium parvum isolate Iowa. Arch Virol 2017; 162:2875-2879. [PMID: 28477174 PMCID: PMC5671912 DOI: 10.1007/s00705-017-3385-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 04/21/2017] [Indexed: 10/19/2022]
Abstract
Bisegmented dsRNA viruses that infect most or all isolates of apicomplexan parasite Cryptosporidium parvum are currently assigned to a single species, Cryptosporidium parvum virus 1, in genus Cryspovirus, family Partitiviridae. An analysis of existing sequence data suggested that the complete sequences of both cryspovirus genome segments, dsRNA1 and dsRNA2, had yet to be determined. We therefore set out to accomplish this for the virus strain that infects C. parvum isolate Iowa. The results suggest that several previous cryspovirus sequences are indeed truncated at one or both segment termini and also identify sequences at or near the termini that are conserved in both segments. Complete sequences of other cryspovirus strains, including ones from other Cryptosporidium species, are needed for refining their classification into one or more virus species.
Collapse
Affiliation(s)
- Minh Vong
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Jacob G Ludington
- Division of Geographic Medicine and Infectious Disease, Tufts Medical Center, Boston, MA, 02116, USA
| | - Honorine D Ward
- Division of Geographic Medicine and Infectious Disease, Tufts Medical Center, Boston, MA, 02116, USA.
| | - Max L Nibert
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
25
|
Li B, Wu H, Li N, Su J, Jia R, Jiang J, Feng Y, Xiao L. Preliminary Characterization of MEDLE-2, a Protein Potentially Involved in the Invasion of Cryptosporidium parvum. Front Microbiol 2017; 8:1647. [PMID: 28912761 PMCID: PMC5583231 DOI: 10.3389/fmicb.2017.01647] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 08/15/2017] [Indexed: 11/13/2022] Open
Abstract
Cryptosporidium spp. are important causes of diarrhea in humans, ruminants, and other mammals. Comparative genomic analysis indicated that genetically related and host-adapted Cryptosporidium species have different numbers of subtelomeric genes encoding the Cryptosporidium-specific MEDLE family of secreted proteins, which could contribute to differences in host specificity. In this study, a Cryptosporidium parvum-specific member of the protein family MEDLE-2 encoded by cgd5_4590 was cloned and expressed in Escherichia coli. Immunofluorescent staining with antibodies generated from the recombinant protein showed the expression of the protein in sporozoites and development stages. In vitro neutralization assay with the antibodies partially blocked the invasion of sporozoites. These results support the potential involvement of MEDLE-2 in the invasion of host cells.
Collapse
Affiliation(s)
- Baoling Li
- State Key Laboratory of Bioreactor Engineering, School of Resources and Environmental Engineering, East China University of Science and TechnologyShanghai, China
| | - Haizhen Wu
- School of Biotechnology, East China University of Science and TechnologyShanghai, China
| | - Na Li
- College of Veterinary Medicine, South China Agricultural UniversityGuangzhou, China
| | - Jiayuan Su
- State Key Laboratory of Bioreactor Engineering, School of Resources and Environmental Engineering, East China University of Science and TechnologyShanghai, China
| | - Ruilian Jia
- State Key Laboratory of Bioreactor Engineering, School of Resources and Environmental Engineering, East China University of Science and TechnologyShanghai, China
| | - Jianlin Jiang
- Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, AtlantaGA, United States
| | - Yaoyu Feng
- State Key Laboratory of Bioreactor Engineering, School of Resources and Environmental Engineering, East China University of Science and TechnologyShanghai, China.,College of Veterinary Medicine, South China Agricultural UniversityGuangzhou, China
| | - Lihua Xiao
- Division of Foodborne, Waterborne, and Environmental Diseases, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, AtlantaGA, United States
| |
Collapse
|
26
|
Comparative genomic analysis of the IId subtype family of Cryptosporidium parvum. Int J Parasitol 2017; 47:281-290. [PMID: 28192123 DOI: 10.1016/j.ijpara.2016.12.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 12/19/2016] [Accepted: 12/22/2016] [Indexed: 11/20/2022]
Abstract
Host adaptation is known to occur in Cryptosporidium parvum, with IIa and IId subtype families preferentially infecting calves and lambs, respectively. To improve our understanding of the genetic basis of host adaptation in Cryptosporidium parvum, we sequenced the genomes of two IId specimens and one IIa specimen from China and Egypt using the Illumina technique and compared them with the published IIa IOWA genome. Sequence data were obtained for >99.3% of the expected genome. Comparative genomic analysis identified differences in numbers of three subtelomeric gene families between sequenced genomes and the reference genome, including those encoding SKSR secretory proteins, the MEDLE family of secretory proteins, and insulinase-like proteases. These gene gains and losses compared with the reference genome were confirmed by PCR analysis. Altogether, 5,191-5,766 single nucleotide variants were seen between genomes sequenced in this study and the reference genome, with most SNVs occurring in subtelomeric regions of chromosomes 1, 4, and 6. The most highly polymorphic genes between IIa and IId encode mainly invasion-associated and immunodominant mucin proteins, and other families of secretory proteins. Further studies are needed to verify the biological significance of these genomic differences.
Collapse
|
27
|
Blondel CJ, Park JS, Hubbard TP, Pacheco AR, Kuehl CJ, Walsh MJ, Davis BM, Gewurz BE, Doench JG, Waldor MK. CRISPR/Cas9 Screens Reveal Requirements for Host Cell Sulfation and Fucosylation in Bacterial Type III Secretion System-Mediated Cytotoxicity. Cell Host Microbe 2016; 20:226-37. [PMID: 27453484 DOI: 10.1016/j.chom.2016.06.010] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 05/24/2016] [Accepted: 06/17/2016] [Indexed: 12/11/2022]
Abstract
Type III secretion systems (T3SSs) inject bacterial effector proteins into host cells and underlie the virulence of many gram-negative pathogens. Studies have illuminated bacterial factors required for T3SS function, but the required host processes remain largely undefined. We coupled CRISPR/Cas9 genome editing technology with the cytotoxicity of two Vibrio parahaemolyticus T3SSs (T3SS1 and T3SS2) to identify human genome disruptions conferring resistance to T3SS-dependent cytotoxicity. We identity non-overlapping genes required for T3SS1- and T3SS2-mediated cytotoxicity. Genetic ablation of cell surface sulfation reduces bacterial adhesion and thereby alters the kinetics of T3SS1-mediated cytotoxicity. Cell surface fucosylation is required for T3SS2-dependent killing, and genetic inhibition of fucosylation prevents membrane insertion of the T3SS2 translocon complex. These findings reveal the importance of ubiquitous surface modifications for T3SS function, potentially explaining the broad tropism of V. parahaemolyticus, and highlight the utility of genome-wide CRISPR/Cas9 screens to discover processes underlying host-pathogen interactions.
Collapse
Affiliation(s)
- Carlos J Blondel
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Joseph S Park
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, Boston, MA 02115, USA; Boston University School of Medicine, Boston, MA 02118, USA
| | - Troy P Hubbard
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Alline R Pacheco
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Carole J Kuehl
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Michael J Walsh
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Brigid M Davis
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Benjamin E Gewurz
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - John G Doench
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Matthew K Waldor
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, Boston, MA 02115, USA.
| |
Collapse
|