1
|
Temereva EN, Sokolova YY. Host-parasite interplay within a phoronid-microsporidia system: anti-parasitic defense in Lophophorata. J Invertebr Pathol 2025:108360. [PMID: 40389110 DOI: 10.1016/j.jip.2025.108360] [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: 02/14/2024] [Revised: 05/13/2025] [Accepted: 05/16/2025] [Indexed: 05/21/2025]
Abstract
Microsporidia (Opisthokonta: Rozellomycota: Microsporidia) are ubiquitous intracellular parasites infecting representatives of all major taxonomic groups of Animalia, from protozoans to mammals, and infecting marine, freshwater, and terrestrial hosts. A representative of the phylum Phoronida was recently added to the list of microsporidian hosts. Only one species Microsporidium phoronidi, a parasite of Phoronis embryolabi, has been recently described. The paper presents further study of this host-parasite system, specifically, the observation of an efficient anti-microsporidial defense reaction in a phoronid host, and a unique mechanism of clearing the host of infection. This defense reaction results in encapsulation of infected cells and subsequent releasing of the capsules through excretory ducts of metanephridia, together with larvae, which regularly leave the mother organism this way. We hypothesize that by encapsulation, phoronids destroy most of parasites, block spread of the infection throughout the body, and prevent horizontal transmission. At the same time, microsporidia that develop in vasoperitoneal tissue that nourish maturing oocytes and embryos, likely overcome the host defense by sporadic or regular infection of embryos. As a result, the parasite secures its persistence in host populations by vertical transmission, which, in turn, benefits evolving less pathogenic forms. Overall, such elaborated and well-balanced phoronid host-microsporidia parasite interactions may suggest long history of co-existence and deserve further studies. New data extend our knowledge about parasite-host interactions and immune response in Lophophorata.
Collapse
Affiliation(s)
- Elena N Temereva
- 119992, Russia, Moscow, Department of Invertebrate Zoology, Biological faculty, Moscow State University, Russian Federation.
| | | |
Collapse
|
2
|
Guo W, Kang H, Huo F, Zhang H, Ye X, Jashenko R, Zhang T, Ji R, Hu H. Environmental drivers affecting the dormancy of Paranosema locustae. J Appl Microbiol 2025; 136:lxaf097. [PMID: 40275519 DOI: 10.1093/jambio/lxaf097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 03/22/2025] [Accepted: 04/23/2025] [Indexed: 04/26/2025]
Abstract
AIMS As a gastrotoxic biocontrol agent employed for locust outbreak management, the infectivity of Paranosema locustae demonstrates significant dependence on pre-ingestion environmental exposure conditions, particularly temperature fluctuations, humidity levels, and UV radiation intensity, making the systematic investigation of these abiotic factors crucial for optimal field application. METHODS AND RESULTS In this study, we simulated key environmental parameters (temperature, humidity, and UV radiation) that critically influence P. locustae viability during the pre-infection phase of host exposure. Analyzed the locust growth curve post-infection, the pathogen's copy number, dormancy factor Lso2 gene expression, and phosphorylated protein levels. Results show a marked decline in lethality and infectivity of P. locustae after prolonged exposure to water, especially at 20°C for 15 days, the survival curve became similar to that of the negative control group. In contrast, drying at 40°C for 15 days preserved its pathogenicity. The pathogen exhibited strong UV resistance, remaining infectious after 24 h of UV exposure at intensities over 100 µW/cm². After 5-10 days of dry conditions, the significant increase in Lso2 gene expression highlights the entry of P. locustae into true dormancy, which subsequently returns to baseline with extended exposure. Western blot analysis supported that sustained phosphorylation is vital for P. locustae lethality. CONCLUSIONS Paranosema locustae demonstrates high-temperature tolerance, with dry heat and UV exposure maintaining infectivity, while wet environments reduce its viability.
Collapse
Affiliation(s)
- Weiqi Guo
- International Research Center of Cross-Border Pest Management in Central Asia, Xinjiang Key Laboratory of Special Species Conservation and Regulatory Biology, College of Life Sciences, Xinjiang Normal University, Xinyi Road, Urumqi, Xinjiang Province 830054, P.R. China
- Tacheng, Research Field (Migratory Biology), Observation and Research Station of Xinjiang, Xinjiang 834700, China
| | - Hanye Kang
- International Research Center of Cross-Border Pest Management in Central Asia, Xinjiang Key Laboratory of Special Species Conservation and Regulatory Biology, College of Life Sciences, Xinjiang Normal University, Xinyi Road, Urumqi, Xinjiang Province 830054, P.R. China
- Tacheng, Research Field (Migratory Biology), Observation and Research Station of Xinjiang, Xinjiang 834700, China
| | - Fan Huo
- International Research Center of Cross-Border Pest Management in Central Asia, Xinjiang Key Laboratory of Special Species Conservation and Regulatory Biology, College of Life Sciences, Xinjiang Normal University, Xinyi Road, Urumqi, Xinjiang Province 830054, P.R. China
- Tacheng, Research Field (Migratory Biology), Observation and Research Station of Xinjiang, Xinjiang 834700, China
| | - Huihui Zhang
- International Research Center of Cross-Border Pest Management in Central Asia, Xinjiang Key Laboratory of Special Species Conservation and Regulatory Biology, College of Life Sciences, Xinjiang Normal University, Xinyi Road, Urumqi, Xinjiang Province 830054, P.R. China
- Tacheng, Research Field (Migratory Biology), Observation and Research Station of Xinjiang, Xinjiang 834700, China
| | - Xiaofang Ye
- International Research Center of Cross-Border Pest Management in Central Asia, Xinjiang Key Laboratory of Special Species Conservation and Regulatory Biology, College of Life Sciences, Xinjiang Normal University, Xinyi Road, Urumqi, Xinjiang Province 830054, P.R. China
- Tacheng, Research Field (Migratory Biology), Observation and Research Station of Xinjiang, Xinjiang 834700, China
| | - Roman Jashenko
- Institute of Zoology RK, 93, Al-Farabi Ave., Almaty 050060, Republic of Kazakhstan
| | - Tinghao Zhang
- International Research Center of Cross-Border Pest Management in Central Asia, Xinjiang Key Laboratory of Special Species Conservation and Regulatory Biology, College of Life Sciences, Xinjiang Normal University, Xinyi Road, Urumqi, Xinjiang Province 830054, P.R. China
- Tacheng, Research Field (Migratory Biology), Observation and Research Station of Xinjiang, Xinjiang 834700, China
| | - Rong Ji
- International Research Center of Cross-Border Pest Management in Central Asia, Xinjiang Key Laboratory of Special Species Conservation and Regulatory Biology, College of Life Sciences, Xinjiang Normal University, Xinyi Road, Urumqi, Xinjiang Province 830054, P.R. China
- Tacheng, Research Field (Migratory Biology), Observation and Research Station of Xinjiang, Xinjiang 834700, China
| | - Hongxia Hu
- International Research Center of Cross-Border Pest Management in Central Asia, Xinjiang Key Laboratory of Special Species Conservation and Regulatory Biology, College of Life Sciences, Xinjiang Normal University, Xinyi Road, Urumqi, Xinjiang Province 830054, P.R. China
- Tacheng, Research Field (Migratory Biology), Observation and Research Station of Xinjiang, Xinjiang 834700, China
| |
Collapse
|
3
|
Hu C, Deng B, Fang W, Guo B, Chen P, Lu C, Dong Z, Pan M. Transgenic overexpression of bmo-miR-6498-5p increases resistance to Nosema bombycis in the silkworm, Bombyx mori. Appl Environ Microbiol 2024; 90:e0027024. [PMID: 39240120 PMCID: PMC11497792 DOI: 10.1128/aem.00270-24] [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: 03/12/2024] [Accepted: 06/23/2024] [Indexed: 09/07/2024] Open
Abstract
Microsporidia are unfriendly microorganisms, and their infections cause considerable damage to economically or environmentally important insects like silkworms and honeybees. Thus, the identification of measures to improve host resistance to microsporidia infections is critically needed. Here, an overexpressed miR-6498-5p transgenic silkworm line was constructed. Importantly, the survival rates and median lethal doses of the transgenic line were clearly higher after infection with Nosema bombycis. H&E staining and RT-qPCR analyses revealed an inhibitory effect on the proliferation of N. bombycis in the transgenic larvae. Metabolomics analysis further revealed the presence of 56 differential metabolites between the two lines. KEGG analysis of these 56 metabolites found that they were involved in various amino acid and vitamin metabolism pathways. Notably, VB6 metabolism was enriched among the metabolites, and the pathway was well known for its involvement in the synthesis, interconversion, and degradation of amino acids. These suggest that miR-6498-5p modifies parasitic environments to inhibit the proliferation of N. bombycis by affecting the host amino acid metabolism. These results demonstrate the potential of microRNAs as biomolecules that can promote resistance to microsporidia and provide new insights and a new approach to generate microsporidia-resistant biological materials.IMPORTANCEMicrosporidia have an extremely wide host range and are capable of infecting a wide variety of insects and vertebrates, including humans, and their lethality to multiple species often poses significant environmental management challenge. Here, we successfully constructed a microsporidium-resistant line in the silkworm, based on the overexpression of miR-6498-5p. Our results strongly support the hypothesis that miR-6498-5p efficiently suppresses the proliferation of Nosema bombycis by regulating the host VB6 metabolism, a key pathway for enzymes involved in amino acid transport and protein metabolism. Our study provides new insights for understanding host anti-pathogen defenses toward microsporidia.
Collapse
Affiliation(s)
- Congwu Hu
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, China
- School of Chemical Engineering, Sichuan University of Science & Engineering, Zigong, Sichuan, China
| | - Boyuan Deng
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, China
| | - Wenxuan Fang
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, China
| | - Bingyu Guo
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, China
| | - Peng Chen
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, China
- Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, Southwest University, Chongqing, China
| | - Cheng Lu
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, China
- Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, Southwest University, Chongqing, China
| | - Zhanqi Dong
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, China
- Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, Southwest University, Chongqing, China
| | - Minhui Pan
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, China
- Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, Southwest University, Chongqing, China
| |
Collapse
|
4
|
Ran M, Yang W, Faryad Khan MU, Li T, Pan G. Microsporidia secretory effectors and their roles in pathogenesis. J Eukaryot Microbiol 2024; 71:e13046. [PMID: 39228342 DOI: 10.1111/jeu.13046] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/20/2024] [Accepted: 06/28/2024] [Indexed: 09/05/2024]
Abstract
Microsporidia, a group of unicellular eukaryotic parasites, rely intensely on secretory effectors for successful invasion and proliferation within host cells. This review focuses on the identification, characterization, and functional roles of effectors, including secretory proteins and microRNAs. The adhesion proteins like the Ricin-B-lectin facilitate initial invasion, which binds to the host cell surface. Once inside, microsporidia deploy a range of effectors to modulate host immune responses, such as serpin proteins, and redirect host cell metabolism to meet the parasite's nutritional needs through hexokinase. Some effectors such as microRNAs, alter the host gene expression to create a more favorable intracellular parasitic environment. In conclusion, the secretory effectors of microsporidia play a pivotal role spanning from host cell invasion to intracellular establishment. In the future, more effectors secreted by microsporidia will be studied, which will not only help to elucidate the molecular mechanism of pathogenic manipulation of the host but also help to provide the potential targets for anti-parasitic treatments.
Collapse
Affiliation(s)
- Maoshuang Ran
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, China
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing, China
| | - Wenxin Yang
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, China
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing, China
| | - Muhammad Usman Faryad Khan
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, China
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing, China
| | - Tian Li
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, China
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing, China
| | - Guoqing Pan
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, China
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing, China
| |
Collapse
|
5
|
Fang W, Zhou L, Deng B, Guo B, Chen X, Chen P, Lu C, Dong Z, Pan M. Establishment of a Secretory Protein-Inducible CRISPR/Cas9 System for Nosema bombycis in Insect Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:13175-13185. [PMID: 38817125 DOI: 10.1021/acs.jafc.3c08647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2024]
Abstract
Gene editing techniques are widely and effectively used for the control of pathogens, but it is difficult to directly edit the genes of Microsporidia due to its unique spore wall structure. Innovative technologies and methods are urgently needed to break through this limitation of microsporidia therapies. Here, we establish a microsporidia-inducible gene editing system through core components of microsporidia secreted proteins, which could edit target genes after infection with microsporidia. We identified that Nosema bombycis NB29 is a secretory protein and found to interact with itself. The NB29-N3, which lacked the nuclear localization signal, was localized in the cytoplasm, and could be tracked into the nucleus after interacting with NB29-B. Furthermore, the gene editing system was constructed with the Cas9 protein expressed in fusion with the NB29-N3. The system could edit the exogenous gene EGFP and the endogenous gene BmRpn3 after overexpression of NB29 or infection with N. bombycis.
Collapse
Affiliation(s)
- Wenxuan Fang
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400716, China
| | - Liang Zhou
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming 650091, China
| | - Boyuan Deng
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400716, China
| | - Binyu Guo
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400716, China
| | - Xue Chen
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400716, China
| | - Peng Chen
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400716, China
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming 650091, China
| | - Cheng Lu
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400716, China
- Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, Southwest University, Chongqing 400716, China
| | - Zhanqi Dong
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400716, China
- Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, Southwest University, Chongqing 400716, China
| | - Minhui Pan
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400716, China
- Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, Southwest University, Chongqing 400716, China
| |
Collapse
|
6
|
Momenilandi M, Lévy R, Sobrino S, Li J, Lagresle-Peyrou C, Esmaeilzadeh H, Fayand A, Le Floc'h C, Guérin A, Della Mina E, Shearer D, Delmonte OM, Yatim A, Mulder K, Mancini M, Rinchai D, Denis A, Neehus AL, Balogh K, Brendle S, Rokni-Zadeh H, Changi-Ashtiani M, Seeleuthner Y, Deswarte C, Bessot B, Cremades C, Materna M, Cederholm A, Ogishi M, Philippot Q, Beganovic O, Ackermann M, Wuyts M, Khan T, Fouéré S, Herms F, Chanal J, Palterer B, Bruneau J, Molina TJ, Leclerc-Mercier S, Prétet JL, Youssefian L, Vahidnezhad H, Parvaneh N, Claeys KG, Schrijvers R, Luka M, Pérot P, Fourgeaud J, Nourrisson C, Poirier P, Jouanguy E, Boisson-Dupuis S, Bustamante J, Notarangelo LD, Christensen N, Landegren N, Abel L, Marr N, Six E, Langlais D, Waterboer T, Ginhoux F, Ma CS, Tangye SG, Meyts I, Lachmann N, Hu J, Shahrooei M, Bossuyt X, Casanova JL, Béziat V. FLT3L governs the development of partially overlapping hematopoietic lineages in humans and mice. Cell 2024; 187:2817-2837.e31. [PMID: 38701783 PMCID: PMC11149630 DOI: 10.1016/j.cell.2024.04.009] [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: 07/26/2023] [Revised: 03/04/2024] [Accepted: 04/10/2024] [Indexed: 05/05/2024]
Abstract
FMS-related tyrosine kinase 3 ligand (FLT3L), encoded by FLT3LG, is a hematopoietic factor essential for the development of natural killer (NK) cells, B cells, and dendritic cells (DCs) in mice. We describe three humans homozygous for a loss-of-function FLT3LG variant with a history of various recurrent infections, including severe cutaneous warts. The patients' bone marrow (BM) was hypoplastic, with low levels of hematopoietic progenitors, particularly myeloid and B cell precursors. Counts of B cells, monocytes, and DCs were low in the patients' blood, whereas the other blood subsets, including NK cells, were affected only moderately, if at all. The patients had normal counts of Langerhans cells (LCs) and dermal macrophages in the skin but lacked dermal DCs. Thus, FLT3L is required for B cell and DC development in mice and humans. However, unlike its murine counterpart, human FLT3L is required for the development of monocytes but not NK cells.
Collapse
Affiliation(s)
- Mana Momenilandi
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France
| | - Romain Lévy
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France; Pediatric Hematology-Immunology and Rheumatology Unit, Necker Hospital for Sick Children, AP-HP, Paris, France
| | - Steicy Sobrino
- Laboratory of Chromatin and Gene Regulation During Development, Paris Cité University, UMR1163 INSERM, Imagine Institute, Paris, France; Laboratory of Human Lymphohematopoiesis, INSERM, Imagine Institute, Paris, France
| | - Jingwei Li
- Jake Gittlen Laboratories for Cancer Research, Pennsylvania State University College of Medicine, Hershey, PA, USA; Department of Pathology and Laboratory Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Chantal Lagresle-Peyrou
- Paris Cité University, Imagine Institute, Paris, France; Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, AP-HP, INSERM, Paris, France
| | - Hossein Esmaeilzadeh
- Allergy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Allergy and Clinical Immunology, Namazi Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Antoine Fayand
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France; Sorbonne University, AP-HP, Tenon Hospital, Department of Internal Medicine, Paris, France
| | - Corentin Le Floc'h
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France
| | - Antoine Guérin
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia; St. Vincent's Clinical School, Faculty of Medicine, University of NSW, Sydney, NSW, Australia
| | - Erika Della Mina
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia; St. Vincent's Clinical School, Faculty of Medicine, University of NSW, Sydney, NSW, Australia
| | - Debra Shearer
- Jake Gittlen Laboratories for Cancer Research, Pennsylvania State University College of Medicine, Hershey, PA, USA; Department of Pathology and Laboratory Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Ottavia M Delmonte
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ahmad Yatim
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - Kevin Mulder
- Gustave Roussy Cancer Campus, Villejuif, France; Paris-Saclay University, Ile-de-France, France
| | - Mathieu Mancini
- Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, QC, Canada; Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
| | - Darawan Rinchai
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - Adeline Denis
- Laboratory of Human Lymphohematopoiesis, INSERM, Imagine Institute, Paris, France
| | - Anna-Lena Neehus
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France
| | - Karla Balogh
- Jake Gittlen Laboratories for Cancer Research, Pennsylvania State University College of Medicine, Hershey, PA, USA; Department of Pathology and Laboratory Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Sarah Brendle
- Jake Gittlen Laboratories for Cancer Research, Pennsylvania State University College of Medicine, Hershey, PA, USA; Department of Pathology and Laboratory Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA; Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Hassan Rokni-Zadeh
- Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences (ZUMS), Zanjan, Iran
| | - Majid Changi-Ashtiani
- School of Mathematics, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran
| | - Yoann Seeleuthner
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France
| | - Caroline Deswarte
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France
| | - Boris Bessot
- Paris Cité University, Imagine Institute, Paris, France; Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, AP-HP, INSERM, Paris, France
| | - Cassandre Cremades
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France
| | - Marie Materna
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France
| | - Axel Cederholm
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Masato Ogishi
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - Quentin Philippot
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France
| | - Omer Beganovic
- Laboratoire d'Onco-hématologie, Necker Hospital for Sick Children, AP-HP, Paris, France
| | - Mania Ackermann
- Hannover Medical School, Department of Pediatric Pulmonology, Allergology and Neonatology, Hannover, Germany; Hannover Medical School, Cluster of Excellence RESIST (EXC 2155), Hannover, Germany; Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hannover, Germany
| | - Margareta Wuyts
- Department of Microbiology and Immunology, Clinical and Diagnostic Immunology, KU Leuven, Leuven, Belgium
| | | | - Sébastien Fouéré
- Groupe Hospitalier Saint-Louis, Lariboisière, Fernand-Widal, CeGIDD, AP-HP, Paris, France
| | - Florian Herms
- Dermatology Department, Paris-Cité University, INSERM 976, Saint Louis Hospital, Paris, France
| | - Johan Chanal
- Dermatology Department, Cochin Hospital, INSERM U1016, AP-HP, Paris, France
| | - Boaz Palterer
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Julie Bruneau
- Department of Pathology, Necker Hospital for Sick Children, AP-HP, Paris-Cité University, Paris, France
| | - Thierry J Molina
- Department of Pathology, Necker Hospital for Sick Children, AP-HP, Paris-Cité University, Paris, France
| | - Stéphanie Leclerc-Mercier
- Department of Pathology, Necker Hospital for Sick Children, AP-HP, Paris-Cité University, Paris, France
| | - Jean-Luc Prétet
- Papillomavirus National Reference Center, Besançon Hospital, Besançon, France
| | - Leila Youssefian
- Department of Pathology and Laboratory Medicine, UCLA Clinical Genomics Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Hassan Vahidnezhad
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Nima Parvaneh
- Department of Pediatrics, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Kristl G Claeys
- Department of Neurology, University Hospitals Leuven, Leuven, Belgium; Laboratory for Muscle Diseases and Neuropathies, Department of Neurosciences, KU Leuven, and Leuven Brain Institute (LBI), Leuven, Belgium
| | - Rik Schrijvers
- Department of Microbiology, Immunology and Transplantation, Allergy and Clinical Immunology Research Group, KU Leuven, Leuven, Belgium
| | - Marine Luka
- Labtech Single-Cell@Imagine, Imagine Institute, INSERM UMR 1163, 75015 Paris, France
| | - Philippe Pérot
- Pathogen Discovery Laboratory, Institut Pasteur, Paris Cité University, Paris, France
| | - Jacques Fourgeaud
- Paris Cité University, URP 7328 FETUS, Paris, France; Microbiology Department, AP-HP, Necker Hospital for Sick Children, Paris, France
| | - Céline Nourrisson
- Clermont Auvergne University, INSERM U1071, M2iSH, USC INRAE 1382, CHU Clermont-Ferrand, 3IHP, Department of Parasitology-Mycology, Clermont-Ferrand, France; National Reference Center for Cryptosporidiosis, Microsporidia and Other Digestive Protozoa, Clermont-Ferrand, France
| | - Philippe Poirier
- Clermont Auvergne University, INSERM U1071, M2iSH, USC INRAE 1382, CHU Clermont-Ferrand, 3IHP, Department of Parasitology-Mycology, Clermont-Ferrand, France; National Reference Center for Cryptosporidiosis, Microsporidia and Other Digestive Protozoa, Clermont-Ferrand, France
| | - Emmanuelle Jouanguy
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France; St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - Stéphanie Boisson-Dupuis
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France; St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - Jacinta Bustamante
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France; St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA; Study Center for Primary Immunodeficiencies, Necker Hospital for Sick Children, AP-HP, Paris, France
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Neil Christensen
- Jake Gittlen Laboratories for Cancer Research, Pennsylvania State University College of Medicine, Hershey, PA, USA; Department of Pathology and Laboratory Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Nils Landegren
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden; Centre for Molecular Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Laurent Abel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France; St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - Nico Marr
- Research Branch, Sidra Medicine, Doha, Qatar; College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Emmanuelle Six
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, AP-HP, INSERM, Paris, France
| | - David Langlais
- Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, QC, Canada; Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada; Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Tim Waterboer
- Infections and Cancer Epidemiology, Infection, Inflammation and Cancer Program, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Florent Ginhoux
- Gustave Roussy Cancer Campus, Villejuif, France; Paris-Saclay University, Ile-de-France, France
| | - Cindy S Ma
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia; St. Vincent's Clinical School, Faculty of Medicine, University of NSW, Sydney, NSW, Australia
| | - Stuart G Tangye
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia; St. Vincent's Clinical School, Faculty of Medicine, University of NSW, Sydney, NSW, Australia
| | - Isabelle Meyts
- Laboratory of Inborn Errors of Immunity, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium; Department of Pediatrics, Leuven University Hospitals, Leuven, Belgium
| | - Nico Lachmann
- Hannover Medical School, Department of Pediatric Pulmonology, Allergology and Neonatology, Hannover, Germany; Hannover Medical School, Cluster of Excellence RESIST (EXC 2155), Hannover, Germany; Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hannover, Germany
| | - Jiafen Hu
- Jake Gittlen Laboratories for Cancer Research, Pennsylvania State University College of Medicine, Hershey, PA, USA; Department of Pathology and Laboratory Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Mohammad Shahrooei
- Department of Microbiology and Immunology, Clinical and Diagnostic Immunology, KU Leuven, Leuven, Belgium; Specialized Immunology Laboratory of Dr. Shahrooei, Tehran, Iran
| | - Xavier Bossuyt
- Department of Microbiology and Immunology, Clinical and Diagnostic Immunology, KU Leuven, Leuven, Belgium; Department of Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Jean-Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France; Pediatric Hematology-Immunology and Rheumatology Unit, Necker Hospital for Sick Children, AP-HP, Paris, France; St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA; Howard Hughes Medical Institute, New York, NY, USA
| | - Vivien Béziat
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France; St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA.
| |
Collapse
|
7
|
Yuanlae S, Prasartset T, Reamtong O, Munkongwongsiri N, Panphloi M, Preechakul T, Suebsing R, Thitamadee S, Prachumwat A, Itsathitphaisarn O, Taengchaiyaphum S, Kasamechotchung C. Shrimp injection with dsRNA targeting the microsporidian EHP polar tube protein reduces internal and external parasite amplification. Sci Rep 2024; 14:4830. [PMID: 38413745 PMCID: PMC10899260 DOI: 10.1038/s41598-024-55400-2] [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: 10/18/2023] [Accepted: 02/22/2024] [Indexed: 02/29/2024] Open
Abstract
The microsporidian Enterocytozoon hepatopenaei (EHP) is a major threat to shrimp health worldwide. Severe EHP infections in shrimp cause growth retardation and increase susceptibility to opportunistic infections. EHP produces spores with a chitin wall that enables them to survive prolonged environmental exposure. Previous studies showed that polar tube extrusion is a prerequisite for EHP infection, such that inhibiting extrusion should prevent infection. Using a proteomic approach, polar tube protein 2 of EHP (EhPTP2) was found abundantly in protein extracts obtained from extruded spores. Using an immunofluorescent antibody against EhPTP2 for immunohistochemistry, extruded spores were found in the shrimp hepatopancreas (HP) and intestine, but not in the stomach. We hypothesized that presence of EhPTP2 might be required for successful EHP spore extrusion. To test this hypothesis, we injected EhPTP2-specific double-stranded RNA (dsRNA) and found that it significantly diminished EHP copy numbers in infected shrimp. This indicated reduced amplification of EHP-infected cells in the HP by spores released from previously infected cells. In addition, injection of the dsRNA into EHP-infected shrimp prior to their use in cohabitation with naïve shrimp significantly (p < 0.05) reduced the rate of EHP transmission to naïve shrimp. The results revealed that EhPTP2 plays a crucial role in the life cycle of EHP and that dsRNA targeting EHP mRNA can effectively reach the parasite developing in host cells. This approach is a model for future investigations to identify critical genes for EHP survival and spread as potential targets for preventative and therapeutic measures in shrimp.
Collapse
Affiliation(s)
- Satika Yuanlae
- Department of Biochemistry, Faculty of Science, Mahidol University, Rama VI Rd., Bangkok, 10400, Thailand
- Center for Excellence in Shrimp Molecular Biology and Biotechnology (Centex Shrimp), Faculty of Science, Mahidol University, Rama VI Rd., Bangkok, 10400, Thailand
| | - Tharinthon Prasartset
- Department of Biotechnology, Faculty of Science, Mahidol University, Rama VI Rd., Bangkok, 10400, Thailand
| | - Onrapak Reamtong
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
| | - Natthinee Munkongwongsiri
- Aquatic Animal Health Research Team (AQHT), Integrative Aquaculture Biotechnology Research Group, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Yothi Office, Rama VI Rd., Bangkok, 10400, Thailand
| | - Muthita Panphloi
- Department of Biotechnology, Faculty of Science, Mahidol University, Rama VI Rd., Bangkok, 10400, Thailand
| | - Thanchanok Preechakul
- Department of Biotechnology, Faculty of Science, Mahidol University, Rama VI Rd., Bangkok, 10400, Thailand
- Enzyme Technology Laboratory, National Center for Genetic Engineering and Biotechnology (BIOTEC), 113 Thailand Science Park, Phaholyothin Road, Khlong Luang, Pathumthani, 12120, Thailand
| | - Rungkarn Suebsing
- Center for Excellence in Shrimp Molecular Biology and Biotechnology (Centex Shrimp), Faculty of Science, Mahidol University, Rama VI Rd., Bangkok, 10400, Thailand
| | - Siripong Thitamadee
- Center for Excellence in Shrimp Molecular Biology and Biotechnology (Centex Shrimp), Faculty of Science, Mahidol University, Rama VI Rd., Bangkok, 10400, Thailand
- Department of Biotechnology, Faculty of Science, Mahidol University, Rama VI Rd., Bangkok, 10400, Thailand
- Analytical Sciences and National Doping Test Institute, Mahidol University, Rama VI Rd., Bangkok, 10400, Thailand
| | - Anuphap Prachumwat
- Aquatic Animal Health Research Team (AQHT), Integrative Aquaculture Biotechnology Research Group, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Yothi Office, Rama VI Rd., Bangkok, 10400, Thailand
| | - Ornchuma Itsathitphaisarn
- Department of Biochemistry, Faculty of Science, Mahidol University, Rama VI Rd., Bangkok, 10400, Thailand
- Center for Excellence in Shrimp Molecular Biology and Biotechnology (Centex Shrimp), Faculty of Science, Mahidol University, Rama VI Rd., Bangkok, 10400, Thailand
| | - Suparat Taengchaiyaphum
- Aquatic Animal Health Research Team (AQHT), Integrative Aquaculture Biotechnology Research Group, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Yothi Office, Rama VI Rd., Bangkok, 10400, Thailand.
| | - Chanadda Kasamechotchung
- Department of Fisheries, Faculty of Agriculture and Natural Resources, Rajamangala University of Technology Tawan-ok, Chonburi, 20110, Thailand.
| |
Collapse
|
8
|
Akossi RF, Delbac F, El Alaoui H, Wawrzyniak I, Peyretaillade E. The intracellular parasite Anncaliia algerae induces a massive miRNA down-regulation in human cells. Noncoding RNA Res 2023; 8:363-375. [PMID: 37275245 PMCID: PMC10238475 DOI: 10.1016/j.ncrna.2023.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/27/2023] [Accepted: 05/07/2023] [Indexed: 06/07/2023] Open
Abstract
Anncaliia algerae belongs to microsporidia, a group of obligate intracellular parasites related to fungi. These parasites are largely spread in water and food-webs and can infect a wide variety of hosts ranging from invertebrates to vertebrates including humans. In humans, microsporidian infections are mainly opportunistic as immunocompetent hosts can clear parasites naturally. Recent studies however have reported persistent microsporidian infections and have highlighted them as a risk factor in colon cancer. This may be a direct result of cell infection or may be an indirect effect of the infectious microenvironment and the host's response. In both cases, this raises the question of the effects of microsporidian infection at the host and host-cell levels. We aimed to address the question of human host intracellular response to microsporidian infection through a transcriptomic kinetic study of human foreskin fibroblasts (HFF) infected with A.algerae, a human infecting microsporidia with an exceptionally wide host range. We focused solely on host response studying both coding and small non-coding miRNA expression. Our study revealed a generalized down-regulation of cell miRNAs throughout infection with up to 547 different miRNAs downregulated at some timepoints and also transcriptomic dysregulations that could facilitate parasite development with immune and lipid metabolism genes modulation. We also hypothesize possible small nucleic acid expropriation explaining the miRNA downregulation. This work contributes to a better understanding of the dialogue that can occur between an intracellular parasite and its host at the cellular level, and can guide future studies on microsporidian infection biology to unravel the mode of action of these minimalist parasites at the tissue or host levels.We have also generated a kinetic and comprehensive transcriptomic data set of an infectious process that can help support comparative studies in the broader field of parasitology. Lastly, these results may warrant for caution regarding microsporidian exposure and persistent infections.
Collapse
Affiliation(s)
- Reginald Florian Akossi
- Laboratoire “Microorganismes: Génome et Environnement” (LMGE), UMR 6023, Université Clermont Auvergne and CNRS, F-63000, Clermont-Ferrand, France
| | - Fréderic Delbac
- Laboratoire “Microorganismes: Génome et Environnement” (LMGE), UMR 6023, Université Clermont Auvergne and CNRS, F-63000, Clermont-Ferrand, France
| | - Hicham El Alaoui
- Laboratoire “Microorganismes: Génome et Environnement” (LMGE), UMR 6023, Université Clermont Auvergne and CNRS, F-63000, Clermont-Ferrand, France
| | - Ivan Wawrzyniak
- Laboratoire “Microorganismes: Génome et Environnement” (LMGE), UMR 6023, Université Clermont Auvergne and CNRS, F-63000, Clermont-Ferrand, France
| | - Eric Peyretaillade
- Laboratoire “Microorganismes: Génome et Environnement” (LMGE), UMR 6023, Université Clermont Auvergne and CNRS, F-63000, Clermont-Ferrand, France
| |
Collapse
|
9
|
De la Mora A, Morfin N, Tapia-Rivera JC, Macías-Macías JO, Tapia-González JM, Contreras-Escareño F, Petukhova T, Guzman-Novoa E. The Fungus Nosema ceranae and a Sublethal Dose of the Neonicotinoid Insecticide Thiamethoxam Differentially Affected the Health and Immunity of Africanized Honey Bees. Microorganisms 2023; 11:1258. [PMID: 37317233 DOI: 10.3390/microorganisms11051258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/01/2023] [Accepted: 05/09/2023] [Indexed: 06/16/2023] Open
Abstract
Honey bees (Apis mellifera L.) are affected by different biotic and abiotic stressors, such as the fungus Nosema ceranae and neonicotinoid insecticides, that negatively impact their health. However, most studies so far conducted have focused on the effect of these stressors separately and in European honey bees. Therefore, this study was conducted to analyze the impact of both stressors, singly and in combination, on honey bees of African descent that have demonstrated resistance to parasites and pesticides. Africanized honey bees (AHBs, Apis mellifera scutellata Lepeletier) were inoculated with N. ceranae (1 × 105 spores/bee) and/or chronically exposed for 18 days to a sublethal dose of thiamethoxam (0.025 ng/bee) to evaluate their single and combined effects on food consumption, survivorship, N. ceranae infection, and immunity at the cellular and humoral levels. No significant effects by any of the stressors were found for food consumption. However, thiamethoxam was the main stressor associated to a significant decrease in AHB survivorship, whereas N. ceranae was the main stressor affecting their humoral immune response by upregulating the expression of the gene AmHym-1. Additionally, both stressors, separately and combined, significantly decreased the concentration of haemocytes in the haemolymph of the bees. These findings indicate that N. ceranae and thiamethoxam differentially affect the lifespan and immunity of AHBs and do not seem to have synergistic effects when AHBs are simultaneously exposed to both stressors.
Collapse
Affiliation(s)
- Alvaro De la Mora
- School of Environmental Sciences, University of Guelph, Guelph, ON N1G2W1, Canada
| | - Nuria Morfin
- School of Environmental Sciences, University of Guelph, Guelph, ON N1G2W1, Canada
| | - José C Tapia-Rivera
- Centro de Investigaciones en Abejas, CUSUR, Universidad de Guadalajara, Enrique Arreola Silva 883, Zapotlan el Grande 49000, Jalisco, Mexico
| | - José O Macías-Macías
- Centro de Investigaciones en Abejas, CUSUR, Universidad de Guadalajara, Enrique Arreola Silva 883, Zapotlan el Grande 49000, Jalisco, Mexico
| | - José M Tapia-González
- Centro de Investigaciones en Abejas, CUSUR, Universidad de Guadalajara, Enrique Arreola Silva 883, Zapotlan el Grande 49000, Jalisco, Mexico
| | - Francisca Contreras-Escareño
- Departamento de Producción Agricola, CUCSUR, Universidad de Guadalajara, Independencia Nal. 161, Autlan 48900, Jalisco, Mexico
| | - Tatiana Petukhova
- Department of Population Medicine, University of Guelph, Guelph, ON N1G2W1, Canada
| | - Ernesto Guzman-Novoa
- School of Environmental Sciences, University of Guelph, Guelph, ON N1G2W1, Canada
| |
Collapse
|
10
|
Ang’ang’o LM, Herren JK, Tastan Bishop Ö. Structural and Functional Annotation of Hypothetical Proteins from the Microsporidia Species Vittaforma corneae ATCC 50505 Using in silico Approaches. Int J Mol Sci 2023; 24:3507. [PMID: 36834914 PMCID: PMC9960886 DOI: 10.3390/ijms24043507] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 01/25/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023] Open
Abstract
Microsporidia are spore-forming eukaryotes that are related to fungi but have unique traits that set them apart. They have compact genomes as a result of evolutionary gene loss associated with their complete dependency on hosts for survival. Despite having a relatively small number of genes, a disproportionately high percentage of the genes in microsporidia genomes code for proteins whose functions remain unknown (hypothetical proteins-HPs). Computational annotation of HPs has become a more efficient and cost-effective alternative to experimental investigation. This research developed a robust bioinformatics annotation pipeline of HPs from Vittaforma corneae, a clinically important microsporidian that causes ocular infections in immunocompromised individuals. Here, we describe various steps to retrieve sequences and homologs and to carry out physicochemical characterization, protein family classification, identification of motifs and domains, protein-protein interaction network analysis, and homology modelling using a variety of online resources. Classification of protein families produced consistent findings across platforms, demonstrating the accuracy of annotation utilizing in silico methods. A total of 162 out of 2034 HPs were fully annotated, with the bulk of them categorized as binding proteins, enzymes, or regulatory proteins. The protein functions of several HPs from Vittaforma corneae were accurately inferred. This improved our understanding of microsporidian HPs despite challenges related to the obligate nature of microsporidia, the absence of fully characterized genes, and the lack of homologous genes in other systems.
Collapse
Affiliation(s)
- Lilian Mbaisi Ang’ang’o
- Research Unit in Bioinformatics (RUBi), Department of Biochemistry and Microbiology, Rhodes University, Makhanda 6140, South Africa
| | - Jeremy Keith Herren
- International Centre of Insect Physiology and Ecology (icipe), Nairobi P.O. Box 30772-00100, Kenya
| | - Özlem Tastan Bishop
- Research Unit in Bioinformatics (RUBi), Department of Biochemistry and Microbiology, Rhodes University, Makhanda 6140, South Africa
| |
Collapse
|
11
|
Zhou L, Guan Z, Chen C, Zhu Q, Qiu S, Liu Y, Li M, Zeng W, Wang H, Gao Y, Yuan Y, Zhang H, Ruan G, Pan X. The successful treatment of Enterocytozoon bieneusi Microsporidiosis with nitazoxanide in a patient with B-ALL: A Case Report. Front Cell Infect Microbiol 2023; 12:1072463. [PMID: 36699718 PMCID: PMC9868268 DOI: 10.3389/fcimb.2022.1072463] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 12/19/2022] [Indexed: 01/10/2023] Open
Abstract
Introduction Enterocytozoon bieneusi (E. bieneusi) Microsporidia can cause opportunistic infections in immunocompromised patients and is also an emerging disease in these individuals. Its clinical manifestations are chronic diarrhea and severe wasting syndrome, these can be extremely debilitating and carry a significant risk of death for immunocompromised patients. Often, microsporidia cannot be confirmed immediately by routine examination and culture. Effective and available treatment options are limited for infections caused by E. bieneusi in humans. Such cases are very rare in Chinese Mainland. Case presentation A 47-year-old male had recurrent, profuse watery diarrhea and abdominal discomfort for more than 7 months, with a fever for 5 days. Two years earlier, he received treatment with a modified BFM-90 protocol for acute B cell lymphoblastic leukemia and is currently in the final stages of maintenance therapy with oral methotrexate and mercaptopurine. The leukemia was assessed as still in remission two months ago. PET/CT showed massive peritoneal fluid accumulation and a high uptake area in the diffused peritoneum (SUVmax 12.57), suggesting tumor invasion or microbial infections. However, broad-spectrum antibacterial therapies were ineffective. Metagenomic sequencing of plasma and peritoneal fluid showed no suggestion of the existence of a tumor but instead showed a high sequence number of DNA and RNA of the Microsporidia. His albendazole treatment failed and subsequent treatment with nitazoxanide successfully resolved the infection. Conclusion This case shows that we should consider the possibility of atypical pathogen infection in patients with hematologic malignancy who repeatedly develop unexplained diarrhea with wasting. mNGS can help rule out malignant neoplasms and diagnose infections. Our results suggest that nitazoxanide effectively treats E. bieneusi microsporidia infections.
Collapse
|
12
|
Carriere E, Abdul Hamid AI, Feki I, Dubuffet A, Delbac F, Gueirard P. A mouse ear skin model to study the dynamics of innate immune responses against the microsporidian Encephalitozoon cuniculi. Front Microbiol 2023; 14:1168970. [PMID: 37125152 PMCID: PMC10136781 DOI: 10.3389/fmicb.2023.1168970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 03/29/2023] [Indexed: 05/02/2023] Open
Abstract
Microsporidia are obligate intracellular parasites related to fungi that cause severe infections in immunocompromised individuals. Encephalitozoon cuniculi is a microsporidian species capable of infecting mammals, including human and rodents. In response to microsporidian infection, innate immune system serves as the first line of defense and allows a partial clearance of the parasite via the innate immune cells, namely macrophages, neutrophils, dendritic cells, and Natural Killer cells. According to the literature, microsporidia bypass this response in vitro by modulating the response of macrophages. In order to study host-parasites interactions in vivo, we developed a model using the mouse ear pinna in combination with an intravital imaging approach. Fluorescent E. cuniculi spores were inoculated into the skin tissue to follow for the first time in real time in an in vivo model the recruitment dynamics of EGFP + phagocytic cells in response to the parasite. The results show that parasites induce an important inflammatory recruitment of phagocytes, with alterations of their motility properties (speed, displacement length, straightness). This cellular response persists in the injection zone, with spores detected inside the phagocytes up to 72 h post-infection. Immunostainings performed on ear tissue cryosections evoke the presence of developing infectious foci from 5 days post-infection, in favor of parasite proliferation in this tissue. Overall, the newly set up mice ear pinna model will increase our understanding of the immunobiology of microsporidia and in particular, to know how they can bypass and hijack the host immune system of an immunocompetent or immunosuppressed host.
Collapse
|
13
|
Characterizing the Proliferation Patterns of Representative Microsporidian Species Enlightens Future Studies of Infection Mechanisms. Pathogens 2022; 11:pathogens11111352. [DOI: 10.3390/pathogens11111352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/10/2022] [Accepted: 11/12/2022] [Indexed: 11/17/2022] Open
Abstract
Background: Microsporidia are a group of pathogens that infect all kinds of animals, such as humans, silkworms, honeybees, and shrimp; they, therefore, pose a severe threat to public health and the economy. There are over 1500 species of microsporidia that have been reported, among which Encephalitozoon hellem and Nosema bombycis are the representative zoonotic and insect-infecting species, respectively. Investigating their cell infection patterns is of great significance for understanding their infection mechanisms. Methods: Specific probes were designed for the ribosomal RNA sequences of microsporidia. Fluorescence in situ hybridization (FISH) was used to trace the proliferation cycle of the pathogens in different cells. Results: Here, two rRNA large subunit gene (LSUrRNA) probes specifically labeling N. bombycis were obtained. The life cycle of N. bombycis in silkworm cells and E. hellem in three kinds of host cells was graphically drawn. N. bombycis meronts were first observed at 30 hours post-infection (hpi), and they began merogony. Sporonts were observed at 42 hpi, and the first entire proliferation cycle was completed at 48 hpi. The proliferation cycle of E. hellem in RK13 and HEK293 epithelial cells was almost the same, completing the first life cycle after 24 hpi, but it was significantly delayed to 32 hpi in RAW264.7. Conclusions: Specific FISH probes were established for labeling microsporidia in multiple host cells. The proliferation characteristics of representative zoonotic and insect-infecting microsporidian species were clarified. This study provides an experimental pattern for future analyses of microsporidian infection mechanisms.
Collapse
|
14
|
Tanida K, Hahn A, Eberhardt KA, Tannich E, Landt O, Kann S, Feldt T, Sarfo FS, Di Cristanziano V, Frickmann H, Loderstädt U. Comparative Assessment of In-House Real-Time PCRs Targeting Enteric Disease-Associated Microsporidia in Human Stool Samples. Pathogens 2021; 10:pathogens10060656. [PMID: 34073403 PMCID: PMC8229491 DOI: 10.3390/pathogens10060656] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 01/04/2023] Open
Abstract
Microsporidiosis is an infection predominantly occurring in immunosuppressed patients and infrequently also in travelers. This study was performed to comparatively evaluate the diagnostic accuracy of real-time PCR assays targeting microsporidia with etiological relevance in the stool of human patients in a latent class analysis-based test comparison without a reference standard with perfect accuracy. Thereby, two one-tube real-time PCR assays and two two-tube real-time PCR assays targeting Enterocytozoon bieneusi and Encephalocytozoon spp. were included in the assessment with reference stool material (20), stool samples from Ghanaian HIV-positive patients (903), and from travelers, migrants and Colombian indigenous people (416). Sensitivity of the assays ranged from 60.4% to 97.4% and specificity from 99.1% to 100% with substantial agreement according to Cohen’s kappa of 79.6%. Microsporidia DNA was detected in the reference material and the stool of the HIV patients but not in the stool of the travelers, migrants, and the Colombian indigenous people. Accuracy-adjusted prevalence was 5.8% (n = 78) for the study population as a whole. In conclusion, reliable detection of enteric disease-associated microsporidia in stool samples by real-time PCR could be demonstrated, but sensitivity between the compared microsporidia-specific real-time PCR assays varied.
Collapse
Affiliation(s)
- Konstantin Tanida
- Department of Microbiology and Hospital Hygiene, Bundeswehr Hospital Hamburg, 20359 Hamburg, Germany; (K.T.); (H.F.)
| | - Andreas Hahn
- Department of Medical Microbiology, Virology and Hygiene, University Medicine Rostock, 18057 Rostock, Germany;
| | - Kirsten Alexandra Eberhardt
- Department of Tropical Medicine, Bernhard Nocht Institute for Tropical Medicine & I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany;
- Institute for Transfusion Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Egbert Tannich
- Bernhard Nocht Institute for Tropical Medicine Hamburg, 20359 Hamburg, Germany;
- National Reference Centre for Tropical Pathogens, 20359 Hamburg, Germany
| | | | - Simone Kann
- Medical Mission Institute, 97074 Würzburg, Germany;
| | - Torsten Feldt
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Medical Center Düsseldorf, 40225 Düsseldorf, Germany;
| | - Fred Stephen Sarfo
- Department of Medicine, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana;
| | - Veronica Di Cristanziano
- Institute of Virology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, 50935 Cologne, Germany;
| | - Hagen Frickmann
- Department of Microbiology and Hospital Hygiene, Bundeswehr Hospital Hamburg, 20359 Hamburg, Germany; (K.T.); (H.F.)
- Department of Medical Microbiology, Virology and Hygiene, University Medicine Rostock, 18057 Rostock, Germany;
| | - Ulrike Loderstädt
- Department of Hospital Hygiene & Infectious Diseases, University Medicine Göttingen, 37075 Göttingen, Germany
- Correspondence: ; Tel.: +49-551-3965709
| |
Collapse
|
15
|
Dalboni LC, Alvares Saraiva AM, Konno FTDC, Perez EC, Codeceira JF, Spadacci-Morena DD, Lallo MA. Encephalitozoon cuniculi takes advantage of efferocytosis to evade the immune response. PLoS One 2021; 16:e0247658. [PMID: 33667240 PMCID: PMC7935246 DOI: 10.1371/journal.pone.0247658] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 02/10/2021] [Indexed: 11/19/2022] Open
Abstract
Microsporidia are recognized as opportunistic pathogens in individuals with immunodeficiencies, especially related to T cells. Although the activity of CD8+ T lymphocytes is essential to eliminate these pathogens, earlier studies have shown significant participation of macrophages at the beginning of the infection. Macrophages and other innate immunity cells play a critical role in activating the acquired immunity. After programmed cell death, the cell fragments or apoptotic bodies are cleared by phagocytic cells, a phenomenon known as efferocytosis. This process has been recognized as a way of evading immunity by intracellular pathogens. The present study evaluated the impact of efferocytosis of apoptotic cells either infected or not on macrophages and subsequently challenged with Encephalitozoon cuniculi microsporidia. Macrophages were obtained from the bone marrow monocytes from C57BL mice, pre-incubated with apoptotic Jurkat cells (ACs), and were further challenged with E. cuniculi spores. The same procedures were performed using the previously infected Jurkat cells (IACs) and challenged with E. cuniculi spores before macrophage pre-incubation. The average number of spores internalized by macrophages in phagocytosis was counted. Macrophage expression of CD40, CD206, CD80, CD86, and MHCII, as well as the cytokines released in the culture supernatants, was measured by flow cytometry. The ultrastructural study was performed to analyze the multiplication types of pathogens. Macrophages pre-incubated with ACs and challenged with E. cuniculi showed a higher percentage of phagocytosis and an average number of internalized spores. Moreover, the presence of stages of multiplication of the pathogen inside the macrophages, particularly after efferocytosis of infected apoptotic bodies, was observed. In addition, pre-incubation with ACs or IACs and/or challenge with the pathogen decreased the viability of macrophages, reflected as high percentages of apoptosis. The marked expression of CD206 and the release of large amounts of IL-10 and IL-6 indicated the polarization of macrophages to an M2 profile, compatible with efferocytosis and favorable for pathogen development. We concluded that the pathogen favored efferocytosis and polarized the macrophages to an M2 profile, allowing the survival and multiplication of E. cuniculi inside the macrophages and explaining the possibility of macrophages acting as Trojan horses in microsporidiosis.
Collapse
Affiliation(s)
- Luciane Costa Dalboni
- Programa de Patologia Ambiental e Experimental da Universidade Paulista–Unip, São Paulo, Brazil
| | - Anuska Marcelino Alvares Saraiva
- Mestrado e Doutorado Interdisciplinar em Ciências da Saúde da Universidade Cruzeiro do Sul, São Paulo, Brazil
- Laboratório de Fisiopatologia, Instituto Butantan, São Paulo, Brazil
| | | | | | | | | | - Maria Anete Lallo
- Programa de Patologia Ambiental e Experimental da Universidade Paulista–Unip, São Paulo, Brazil
- * E-mail: ,
| |
Collapse
|
16
|
Prabhuling SH, Makwana P, Pradeep ANR, Vijayan K, Mishra RK. Release of Mediator Enzyme β-Hexosaminidase and Modulated Gene Expression Accompany Hemocyte Degranulation in Response to Parasitism in the Silkworm Bombyx mori. Biochem Genet 2021; 59:997-1017. [PMID: 33616803 DOI: 10.1007/s10528-021-10046-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 02/02/2021] [Indexed: 01/03/2023]
Abstract
In insects infections trigger hemocyte-mediated immune reactions including degranulation by exocytosis; however, involvement of mediator enzymes in degranulation process is unknown in insects. We report here that in silkworm Bombyx mori, infection by endoparasitoid Exorista bombycis and microsporidian Nosema bombycis activated granulation in granulocytes and promoted degranulation of accumulated structured granules. During degranulation the mediator lysosomal enzyme β-hexosaminidase showed increased activity and expression of β-hexosaminidase gene was enhanced. The events were confirmed in vitro after incubation of uninfected hemocytes with E. bombycis larval tissue protein. On infection, cytotoxicity marker enzyme lactate dehydrogenase (LDH) was released from the hemocytes illustrating cell toxicity. Strong positive correlation (R2 = 0.71) between LDH activity and β-hexosaminidase released after the infection showed parasitic-protein-induced hemocyte damage and accompanied release of the enzymes. Expression of β-hexosaminidase gene was enhanced in early stages after infection followed by down regulation. The expression showed positive correlation (R2 = 0.705) with hexosaminidase activity pattern. B. mori hexosaminidase showed 98% amino acid similarity with that of B. mandarina showing origin from same ancestral gene; however, 45-60% varied from other lepidopterans showing diversity. The observation signifies the less known association of hexosaminidase in degranulation of hemocytes induced by parasitic infection in B. mori and its divergence in different species.
Collapse
Affiliation(s)
- Shambhavi H Prabhuling
- Seribiotech Research Laboratory, CSB-Kodathi Campus, Carmelaram. P.O, Bangalore, Karnataka, 560035, India
| | - Pooja Makwana
- Seribiotech Research Laboratory, CSB-Kodathi Campus, Carmelaram. P.O, Bangalore, Karnataka, 560035, India.,Central Sericultural Research & Training Institute, Berhampore, West Bengal, India
| | - Appukuttan Nair R Pradeep
- Seribiotech Research Laboratory, CSB-Kodathi Campus, Carmelaram. P.O, Bangalore, Karnataka, 560035, India.
| | | | - Rakesh Kumar Mishra
- Seribiotech Research Laboratory, CSB-Kodathi Campus, Carmelaram. P.O, Bangalore, Karnataka, 560035, India
| |
Collapse
|
17
|
Nosema ceranae causes cellular immunosuppression and interacts with thiamethoxam to increase mortality in the stingless bee Melipona colimana. Sci Rep 2020; 10:17021. [PMID: 33046792 PMCID: PMC7550335 DOI: 10.1038/s41598-020-74209-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 09/29/2020] [Indexed: 11/09/2022] Open
Abstract
The microsporidian parasite Nosema ceranae and neonicotinoid insecticides affect the health of honey bees (Apis mellifera). However, there is limited information about the effect of these stressors on other pollinators such as stingless bees (Hymenoptera: Meliponini). We examined the separate and combined effects of N. ceranae and the neonicotinoid thiamethoxam at field-exposure levels on the survivorship and cellular immunity (hemocyte concentration) of the stingless bee Melipona colimana. Newly-emerged bees were subjected to four treatments provided in sucrose syrup: N. ceranae spores, thiamethoxam, thiamethoxam and N. ceranae, and control (bees receiving only syrup). N. ceranae developed infections of > 467,000 spores/bee in the group treated with spores only. However, in the bees subjected to both stressors, infections were < 143,000 spores/bee, likely due to an inhibitory effect of thiamethoxam on the microsporidium. N. ceranae infections did not affect bee survivorship, but thiamethoxam plus N. ceranae significantly increased mortality. Hemocyte counts were significantly lower in N. ceranae infected-bees than in the other treatments. These results suggest that N. ceranae may infect, proliferate and cause cellular immunosuppression in stingless bees, that exposure to sublethal thiamethoxam concentrations is toxic to M. colimana when infected with N. ceranae, and that thiamethoxam restrains N. ceranae proliferation. These findings have implications on pollinators' conservation.
Collapse
|
18
|
Wang L, Lv Q, He Y, Gu R, Zhou B, Chen J, Fan X, Pan G, Long M, Zhou Z. Integrated qPCR and Staining Methods for Detection and Quantification of Enterocytozoon hepatopenaei in Shrimp Litopenaeus vannamei. Microorganisms 2020; 8:microorganisms8091366. [PMID: 32906623 PMCID: PMC7565997 DOI: 10.3390/microorganisms8091366] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 09/02/2020] [Accepted: 09/04/2020] [Indexed: 12/18/2022] Open
Abstract
Enterocytozoon hepatopenaei (EHP) is an obligate, intracellular, spore-forming parasite, which mainly infects the gastrointestinal tract of shrimp. It significantly hinders the growth of shrimp, which causes substantial economic losses in farming. In this study, we established and optimized a SYBR Green I fluorescent quantitative PCR (qPCR) assay based on the polar tube protein 2 (PTP2) gene for the quantitative analysis of EHP-infected shrimp. The result showed that the optimum annealing temperature was 60 °C for the corresponding relation between the amplification quantitative (Cq) and the logarithmic of the initial template quantity (x), conformed to Cq = −3.2751x + 31.269 with a correlation coefficient R2 = 0.993. The amplification efficiency was 102%. This qPCR method also showed high sensitivity, specificity, and repeatability. Moreover, a microscopy method was developed to observe and count EHP spores in hepatopancreas tissue of EHP-infected shrimp using Fluorescent Brightener 28 staining. By comparing the PTP2-qPCR and microscopy method, the microscopic examination was easier to operate whereas PTP2-qPCR was more sensitive for analysis. And we found that there was a correspondence between the results of these two methods. In summary, the PTP2-qPCR method integrated microscopy could serve for EHP detection during the whole period of shrimp farming and satisfy different requirements for detecting EHP in shrimp farming.
Collapse
Affiliation(s)
- Lijun Wang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China; (L.W.); (Q.L.); (Y.H.); (R.G.); (B.Z.); (J.C.); (G.P.); (Z.Z.)
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing 400715, China;
| | - Qing Lv
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China; (L.W.); (Q.L.); (Y.H.); (R.G.); (B.Z.); (J.C.); (G.P.); (Z.Z.)
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing 400715, China;
| | - Yantong He
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China; (L.W.); (Q.L.); (Y.H.); (R.G.); (B.Z.); (J.C.); (G.P.); (Z.Z.)
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing 400715, China;
| | - Ruocheng Gu
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China; (L.W.); (Q.L.); (Y.H.); (R.G.); (B.Z.); (J.C.); (G.P.); (Z.Z.)
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing 400715, China;
| | - Bingqian Zhou
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China; (L.W.); (Q.L.); (Y.H.); (R.G.); (B.Z.); (J.C.); (G.P.); (Z.Z.)
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing 400715, China;
| | - Jie Chen
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China; (L.W.); (Q.L.); (Y.H.); (R.G.); (B.Z.); (J.C.); (G.P.); (Z.Z.)
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing 400715, China;
| | - Xiaodong Fan
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing 400715, China;
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, China
| | - Guoqing Pan
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China; (L.W.); (Q.L.); (Y.H.); (R.G.); (B.Z.); (J.C.); (G.P.); (Z.Z.)
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing 400715, China;
| | - Mengxian Long
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China; (L.W.); (Q.L.); (Y.H.); (R.G.); (B.Z.); (J.C.); (G.P.); (Z.Z.)
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing 400715, China;
- Correspondence:
| | - Zeyang Zhou
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China; (L.W.); (Q.L.); (Y.H.); (R.G.); (B.Z.); (J.C.); (G.P.); (Z.Z.)
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing 400715, China;
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, China
| |
Collapse
|
19
|
Tamim El Jarkass H, Reinke AW. The ins and outs of host-microsporidia interactions during invasion, proliferation and exit. Cell Microbiol 2020; 22:e13247. [PMID: 32748538 DOI: 10.1111/cmi.13247] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/15/2020] [Accepted: 07/16/2020] [Indexed: 12/12/2022]
Abstract
Microsporidia are a large group of fungal-related obligate intracellular parasites. They are responsible for infections in humans as well as in agriculturally and environmentally important animals. Although microsporidia are abundant in nature, many of the molecular mechanisms employed during infection have remained enigmatic. In this review, we highlight recent work showing how microsporidia invade, proliferate and exit from host cells. During invasion, microsporidia use spore wall and polar tube proteins to interact with host receptors and adhere to the host cell surface. In turn, the host has multiple defence mechanisms to prevent and eliminate these infections. Microsporidia encode numerous transporters and steal host nutrients to facilitate proliferation within host cells. They also encode many secreted proteins which may modulate host metabolism and inhibit host cell defence mechanisms. Spores exit the host in a non-lytic manner that is dependent on host actin and endocytic recycling proteins. Together, this work provides a fuller picture of the mechanisms that these fascinating organisms use to infect their hosts.
Collapse
Affiliation(s)
| | - Aaron W Reinke
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
20
|
Burnham AJ, De Jong E, Jones JA, Lehman HK. North American Propolis Extracts From Upstate New York Decrease Nosema ceranae ( Microsporidia) Spore Levels in Honey Bees ( Apis mellifera). Front Microbiol 2020; 11:1719. [PMID: 32793168 PMCID: PMC7387503 DOI: 10.3389/fmicb.2020.01719] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 06/30/2020] [Indexed: 12/11/2022] Open
Abstract
Nosema ceranae infections in honey bees (Apis mellifera) pose a severe threat to colony health. Beekeepers have used dicyclohexylammonium fumagillin to control Nosema apis, although it may be ineffective against N. ceranae. We investigated the ability of various propolis extracts collected from Upstate New York (United States) to decrease in vivo N. ceranae infection levels when fed ad libitum to N. ceranae-infected honey bees. Propolis extracts, most notably a dichloromethane extract, significantly lowered spore levels in a dose-dependent fashion 4 days post inoculation. When testing the in vitro anti-Nosema activity of propolis extracts, we report for the first time that spore viability was unaffected after a 24 h exposure to propolis extracts. These results present evidence that propolis extracts may effectively lower Microsporidia infections in honey bees, and that direct exposure of environmental spores to propolis alone does not kill N. ceranae.
Collapse
Affiliation(s)
- Andre J Burnham
- Department of Biology, Hamilton College, Clinton, NY, United States.,Program in Biochemistry, Hamilton College, Clinton, NY, United States.,Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Emily De Jong
- Department of Biology, Hamilton College, Clinton, NY, United States.,Program in Biochemistry, Hamilton College, Clinton, NY, United States
| | - Jayre A Jones
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Herman K Lehman
- Department of Biology, Hamilton College, Clinton, NY, United States.,Program in Biochemistry, Hamilton College, Clinton, NY, United States.,Program in Neuroscience, Hamilton College, Clinton, NY, United States
| |
Collapse
|
21
|
Burnham AJ. Scientific Advances in Controlling Nosema ceranae ( Microsporidia) Infections in Honey Bees ( Apis mellifera). Front Vet Sci 2019; 6:79. [PMID: 30931319 PMCID: PMC6428737 DOI: 10.3389/fvets.2019.00079] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 02/25/2019] [Indexed: 12/31/2022] Open
Abstract
Honey bees (Apis mellifera) are agriculturally important pollinators that have been recently at risk to severe colony losses. A variety of parasites and pathogens have been linked to colony decline, including the microsporidian parasite Nosema ceranae. While fumagillin has been used to control nosemosis in managed honey bee colonies for decades, research shows that this antibiotic poses a toxic threat and that its efficacy against N. ceranae is uncertain. There is certainly a demand for a new veterinary medication to treat honey bee colonies infected with N. ceranae. In this review, recent scientific advances in controlling N. ceranae infections in honey bees are summarized.
Collapse
|
22
|
Molecular Phylodiagnosis of Enterocytozoon bieneusi and Encephalitozoon intestinalis in Children with Cancer: Microsporidia in Malignancies as an Emerging Opportunistic Infection. Acta Parasitol 2019; 64:103-111. [PMID: 30644064 DOI: 10.2478/s11686-018-00012-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Accepted: 11/28/2018] [Indexed: 02/02/2023]
Abstract
BACKGROUND : Microsporidia may cause infection in both immunocompromised and immunocompetent populations. The best strategy to control microsporidiosis is obtaining thorough knowledge of its outbreak and pathogenicity. PURPOSE : Because of the lack of precise estimation of microsporidia prevalence among Iranian children with cancer, the current study aimed at evaluating the rate of intestinal microsporidia in children undergoing chemotherapy. METHODS Patients with cancer undergoing chemotherapy in a children's hospital in Northwestern Iran were studied; 132 stool samples were collected and stained by the Weber and Ryan-blue modified trichrome staining techniques. The extracted DNA samples were evaluated by the nested polymerase chain reaction (PCR) method. All positive isolates were sequenced for genotyping and phylogenetic analysis. RESULTS A total of 17 (12.8%) samples were microscopically positive for microsporidia infection, whereas only 14 (10.6%) cases were positive based on nested PCR results. In the positive samples detected with nested PCR, the frequency of Enterocytozoon bieneusi and Encephalitozoon intestinalis infections was 71.4% (n = 10) and 28.6% (n = 4), respectively. After sequencing and phylogenetic analysis, the genotype of E. bieneusi was type D and the sequences of the isolated species were similar to those of the registered ones. CONCLUSION E. bieneusi is a major contributor to microsporidiosis in young immunocompromised patients in Iran. Microsporidia species are well-detected when confirmatory techniques such as molecular methods are in agreement with staining. So, to ensure this, a suggestion has been made to introduce a certain diagnostic test for microsporidiosis.
Collapse
|
23
|
Han B, Moretto M, M Weiss L. Encephalitozoon: Tissue Culture, Cryopreservation, and Murine Infection. ACTA ACUST UNITED AC 2018; 52:e72. [PMID: 30444582 DOI: 10.1002/cpmc.72] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Microsporidia are eukaryotic unicellular parasites that have been studied for more than 150 years. They are found throughout the world and are capable of infecting various invertebrate and vertebrate hosts. They can cause disease in both immune-compromised and immune-competent humans. In immune-compromised individuals, infections can be severe and often fatal. Microsporidia possess a unique, highly specialized invasion mechanism that involves a structure known as the polar tube as well as the spore wall. During spore germination, the polar tube rapidly discharges from the spore and deliver the sporoplasm into the host cell. Spores are the only stage of microsporidia that can survive outside of host cells. Since the first attempt to culture microsporidia in vitro in 1930s, their cultivation has served a critical role in the study and diagnosis of these parasites. In this chapter, we include methods on the cultivation, isolation, and cryopreservation of Encephalitozoon cuniculi, which can infect humans and provides a useful model for other microsporidia. These methods can also be utilized for the culture of Encephalitozoon hellem or Encephalitozoon intestinalis. © 2018 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Bing Han
- Department of Pathology, Division of Tropical Medicine and Parasitology, Albert Einstein College of Medicine, Bronx, New York
| | - Magali Moretto
- Department of Microbiology, Immunology, and Tropical Medicine, George Washington University, Washington, D.C
| | - Louis M Weiss
- Department of Pathology, Division of Tropical Medicine and Parasitology, Albert Einstein College of Medicine, Bronx, New York.,Department of Medicine, Division of Infectious Diseases, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
24
|
Abstract
INTRODUCTION Microsporidia have been increasingly reported to infect humans. The most common presentation of microsporidiosis is chronic diarrhea, a significant mortality risk in immune-compromised patients. Albendazole, which inhibits tubulin, and fumagillin, which inhibits methionine aminopeptidase type 2 (MetAP2), are the two main therapeutic agents used for treatment of microsporidiosis. In addition, to their role as emerging pathogens in humans, microsporidia are important pathogens in insects, aquaculture, and veterinary medicine. New therapeutic targets and therapies have become a recent focus of attention for medicine, veterinary, and agricultural use. Areas covered: Herein, we discuss the detection and symptoms of microsporidiosis in humans and the therapeutic targets that have been utilized for the design of new drugs for the treatment of this infection, including triosephosphate isomerase, tubulin, MetAP2, topoisomerase IV, chitin synthases, and polyamines. Expert opinion: Enterocytozoon bieneusi is the most common microsporidia in human infection. Fumagillin has a broader anti-microsporidian activity than albendazole and is active against both Ent. bieneusi and Encephaliozoonidae. Microsporidia lack methionine aminopeptidase type 1 and are, therefore, dependent on MetAP2, while mammalian cells have both enzymes. Thus, MetAP2 is an essential enzyme in microsporidia and new inhibitors of this pathway have significant promise as therapeutic agents.
Collapse
Affiliation(s)
- Bing Han
- Department of Pathology, Division of Tropical Medicine and Parasitology, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Louis M. Weiss
- Department of Pathology, Division of Tropical Medicine and Parasitology, Albert Einstein College of Medicine, Bronx, NY 10461
- Department of Medicine, Division of Infectious Diseases, Albert Einstein College of Medicine, Bronx, NY 10461
| |
Collapse
|
25
|
Impact of the microsporidian Nosema ceranae on the gut epithelium renewal of the honeybee, Apis mellifera. J Invertebr Pathol 2018; 159:121-128. [PMID: 30268675 DOI: 10.1016/j.jip.2018.09.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 09/21/2018] [Accepted: 09/26/2018] [Indexed: 12/12/2022]
Abstract
The invasive microsporidian species, Nosema ceranae, causes nosemosis in honeybees and is suspected to be involved in Western honeybee (Apis mellifera) declines worldwide. The midgut of honeybees is the site of infection; the microsporidium can disturb the functioning of this organ and, thus, the bee physiology. Host defense against pathogens is not limited to resistance (i.e. the immune response) but also involves resilience. This process implies that the host can tolerate and repair damage inflicted by the infection- by the pathogen itself or by an excessive host immune response. Enterocyte damage caused by N. ceranae can be compensated by proliferation of intestinal stem cells (ISCs) that are under the control of multiple pathways. In the present study, we investigated the impact of N. ceranae on honeybee epithelium renewal by following the mitotic index of midgut stem cells during a 22-day N. ceranae infection. Fluorescence in situ hybridization (FISH) and immunostaining experiments were performed to follow the parasite proliferation/progression in the intestinal tissue, especially in the ISCs as they are key cells for the midgut homeostasis. We also monitored the transcriptomic profile of 7 genes coding for key proteins involved in pathways implicated in the gut epithelium renewal and homeostasis. We have shown for the first time that N. ceranae can negatively alter the gut epithelium renewal rate and disrupt some signaling pathways involved in the gut homeostasis. This alteration is correlated to a reduced longevity of N. ceranae-infected honeybees and we can assume that honeybee susceptibility to N. ceranae could be due to an impaired ability to repair gut damage.
Collapse
|
26
|
Pan G, Bao J, Ma Z, Song Y, Han B, Ran M, Li C, Zhou Z. Invertebrate host responses to microsporidia infections. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2018; 83:104-113. [PMID: 29428490 DOI: 10.1016/j.dci.2018.02.004] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 01/31/2018] [Accepted: 02/06/2018] [Indexed: 05/06/2023]
Abstract
Microsporidia are a group of fungi-like intracellular and unicellular parasites, which infect nearly all animals. As "master parasites", over 1400 microsporidian species have been described to date. Microsporidia infections in economical invertebrates (e.g., silkworm, shrimp) cause huge financial losses, while other microsporidia infections in daphnia, nematode, locust, honeybee and mosquito play important roles in the regulation of their population size. Research investigating invertebrate host responses following microsporidia infections has yielded numerous interesting results, especially pertaining to the innate immune response to these pathogens. In this review, we comparatively summarize the invertebrate host responses to various microsporidia infections. We discuss numerous critical events in host responses including ubiquitin-mediated resistance, production of reactive oxygen species, melanization and innate immune pathways, and the increased basic metabolism and the accumulation of juvenile hormone in infected hosts. Recent studies progressing our understanding of microsporidia infection are also highlighted. Collectively, these advances shed more light on general rules of invertebrate host immune responses and pathogenesis mechanisms of microsporidia, and concurrently offer valuable clues for further research on the crosstalk between hosts and intracellular pathogens.
Collapse
Affiliation(s)
- Guoqing Pan
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, PR China
| | - Jialing Bao
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, PR China
| | - Zhengang Ma
- College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China
| | - Yue Song
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, PR China
| | - Bing Han
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, PR China
| | - Maoshuang Ran
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, PR China
| | - Chunfeng Li
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, PR China
| | - Zeyang Zhou
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, PR China; College of Life Sciences, Chongqing Normal University, Chongqing 401331, PR China.
| |
Collapse
|
27
|
Abstract
Microsporidia are obligate intracellular pathogens related to Fungi. These organisms have a unique invasion organelle, the polar tube, which upon appropriate environmental stimulation rapidly discharges out of the spore, pierces a host cell's membrane, and serves as a conduit for sporoplasm passage into the host cell. Phylogenetic analysis suggests that microsporidia are related to the Fungi, being either a basal branch or sister group. Despite the description of microsporidia over 150 years ago, we still lack an understanding of the mechanism of invasion, including the role of various polar tube proteins, spore wall proteins, and host cell proteins in the formation and function of the invasion synapse. Recent advances in ultrastructural techniques are helping to better define the formation and functioning of the invasion synapse. Over the past 2 decades, proteomic approaches have helped define polar tube proteins and spore wall proteins as well as the importance of posttranslational modifications such as glycosylation in the functioning of these proteins, but the absence of genetic techniques for the manipulation of microsporidia has hampered research on the function of these various proteins. The study of the mechanism of invasion should provide fundamental insights into the biology of these ubiquitous intracellular pathogens that can be integrated into studies aimed at treating or controlling microsporidiosis.
Collapse
|
28
|
Abstract
Microsporidia comprise a phylum of obligate intracellular pathogens related to fungi that infect virtually all animals. Recently, the nematode Caenorhabditis elegans has been developed as a convenient model for studying microsporidia infection in a whole-animal host through the identification and characterization of a natural microsporidian pathogen of this commonly studied laboratory organism. The C. elegans natural microsporidian pathogen is named Nematocida parisii, and it causes a lethal intestinal infection in C. elegans. Comparison of the genomes of N. parisii and its closely related species Nematocida sp. 1, together with the genomes of other microsporidian species, has provided insight into the evolutionary events that led to the emergence of the large, specialized microsporidia phylum. Cell biology studies of N. parisii infection in C. elegans have shown how N. parisii restructures host intestinal cells and, in particular, how it hijacks host exocytosis for nonlytic exit to facilitate transmission. Recent results also show how the host responds to infection with ubiquitin-mediated responses, and how a natural variant of C. elegans is able to clear N. parisii infection, but only during early life. Altogether, these studies provide insight into the mechanisms of microsporidia pathogenesis using a whole-animal host.
Collapse
|
29
|
Jaroenlak P, Sanguanrut P, Williams BAP, Stentiford GD, Flegel TW, Sritunyalucksana K, Itsathitphaisarn O. A Nested PCR Assay to Avoid False Positive Detection of the Microsporidian Enterocytozoon hepatopenaei (EHP) in Environmental Samples in Shrimp Farms. PLoS One 2016; 11:e0166320. [PMID: 27832178 PMCID: PMC5104377 DOI: 10.1371/journal.pone.0166320] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 10/26/2016] [Indexed: 01/29/2023] Open
Abstract
Hepatopancreatic microsporidiosis (HPM) caused by Enterocytozoon hepatopenaei (EHP) is an important disease of cultivated shrimp. Heavy infections may lead to retarded growth and unprofitable harvests. Existing PCR detection methods target the EHP small subunit ribosomal RNA (SSU rRNA) gene (SSU-PCR). However, we discovered that they can give false positive test results due to cross reactivity of the SSU-PCR primers with DNA from closely related microsporidia that infect other aquatic organisms. This is problematic for investigating and monitoring EHP infection pathways. To overcome this problem, a sensitive and specific nested PCR method was developed for detection of the spore wall protein (SWP) gene of EHP (SWP-PCR). The new SWP-PCR method did not produce false positive results from closely related microsporidia. The first PCR step of the SWP-PCR method was 100 times (104 plasmid copies per reaction vial) more sensitive than that of the existing SSU-PCR method (106 copies) but sensitivity was equal for both in the nested step (10 copies). Since the hepatopancreas of cultivated shrimp is not currently known to be infected with microsporidia other than EHP, the SSU-PCR methods are still valid for analyzing hepatopancreatic samples despite the lower sensitivity than the SWP-PCR method. However, due to its greater specificity and sensitivity, we recommend that the SWP-PCR method be used to screen for EHP in feces, feed and environmental samples for potential EHP carriers.
Collapse
Affiliation(s)
- Pattana Jaroenlak
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand
- Center of Excellence for Shrimp Molecular Biology and Biotechnology (Centex Shrimp), Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Piyachat Sanguanrut
- Center of Excellence for Shrimp Molecular Biology and Biotechnology (Centex Shrimp), Faculty of Science, Mahidol University, Bangkok, Thailand
- Shrimp Pathogen Interaction Laboratory (SPI), National Center for Genetic Engineering and Biotechnology (BIOTEC), Bangkok, Thailand
| | - Bryony A. P. Williams
- Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom
| | - Grant D. Stentiford
- European Community Reference Laboratory for Crustacean Diseases, Center for Environment, Fisheries and Aquaculture Science (Cefas), Weymouth, Dorset, United Kingdom
| | - Timothy W. Flegel
- Center of Excellence for Shrimp Molecular Biology and Biotechnology (Centex Shrimp), Faculty of Science, Mahidol University, Bangkok, Thailand
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani, Thailand
| | - Kallaya Sritunyalucksana
- Shrimp Pathogen Interaction Laboratory (SPI), National Center for Genetic Engineering and Biotechnology (BIOTEC), Bangkok, Thailand
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani, Thailand
| | - Ornchuma Itsathitphaisarn
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand
- Center of Excellence for Shrimp Molecular Biology and Biotechnology (Centex Shrimp), Faculty of Science, Mahidol University, Bangkok, Thailand
- * E-mail:
| |
Collapse
|
30
|
Sitjà-Bobadilla A, Estensoro I, Pérez-Sánchez J. Immunity to gastrointestinal microparasites of fish. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 64:187-201. [PMID: 26828391 DOI: 10.1016/j.dci.2016.01.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 01/25/2016] [Accepted: 01/25/2016] [Indexed: 06/05/2023]
Abstract
Fish intestinal parasites cause direct mortalities and also morbidity, poor growth, higher susceptibility to opportunistic pathogens and lower resistance to stress. This review is focused on microscopic parasites (Protozoa and Metazoa) that invade the gastrointestinal tract of fish. Intracellular parasites (mainly Microsporidia and Apicomplexa) evoke almost no host immune reaction while they are concealed in the cytoplasmic and nuclear compartments, and can even use fish cells (macrophages) as Trojan horses to spread in the host. Inflammatory reaction only appears when the parasite bursts infected cells. Immunity against extracellular parasites is depicted for the myxozoans Ceratonova shasta and Enteromyxum spp. The cellular and humoral innate responses and the production of antibodies are crucial for resolving some of these myxozoonoses, but an excessive inflammatory reaction (concerted by cytokines) can become a fatal pathophysiological consequence. The local immune response plays a key role, with numerous genes more strongly regulated in the intestine than at lymphohaematopoietic organs.
Collapse
Affiliation(s)
- Ariadna Sitjà-Bobadilla
- Fish Pathology Group, Institute of Aquaculture Torre de la Sal (IATS-CSIC), Consejo Superior de Investigaciones Científicas, Castellón, Spain.
| | - Itziar Estensoro
- Fish Pathology Group, Institute of Aquaculture Torre de la Sal (IATS-CSIC), Consejo Superior de Investigaciones Científicas, Castellón, Spain
| | - Jaume Pérez-Sánchez
- Nutrigenomics and Fish Growth Endocrinology Group, Institute of Aquaculture Torre de la Sal (IATS-CSIC), Consejo Superior de Investigaciones Científicas, Castellón, Spain
| |
Collapse
|
31
|
Microsporidia Intracellular Development Relies on Myc Interaction Network Transcription Factors in the Host. G3-GENES GENOMES GENETICS 2016; 6:2707-16. [PMID: 27402359 PMCID: PMC5015929 DOI: 10.1534/g3.116.029983] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Microsporidia are ubiquitous parasites that infect a wide range of animal hosts, and these fungal-related microbes undergo their entire replicative lifecycle inside of host cells. Despite being widespread in the environment and causing medical and agricultural harm, virtually nothing is known about the host factors important to facilitate their growth and development inside of host cells. Here, we perform a genetic screen to identify host transcription factors important for development of the microsporidian pathogen Nematocida parisii inside intestinal cells of its natural host, the nematode Caenorhabditis elegans. Through this screen, we identified the C. elegans Myc family of transcription factors as key host regulators of microsporidia growth and development. The Mad-like transcription factor MDL-1, and the Max-like transcription factors MXL-1 and MXL-2 promote pathogen levels, while the Myc-Mondo-like transcription factor MML-1 inhibits pathogen levels. We used epistasis analysis to show that MDL-1 and MXL-1, which are thought to function as a heterodimer, appear to be acting canonically. In contrast, MXL-2 and MML-1, which are also thought to function as a heterodimer, appear to be acting in separate pathways (noncanonically) in the context of pathogen infection. We also found that both MDL-1::GFP and MML-1::GFP are expressed in intestinal cells during infection. These findings provide novel insight into the host transcription factors that regulate microsporidia development.
Collapse
|
32
|
Evolution of the microtubular cytoskeleton (flagellar apparatus) in parasitic protists. Mol Biochem Parasitol 2016; 209:26-34. [DOI: 10.1016/j.molbiopara.2016.02.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Revised: 02/02/2016] [Accepted: 02/05/2016] [Indexed: 01/16/2023]
|
33
|
Luallen RJ, Reinke AW, Tong L, Botts MR, Félix MA, Troemel ER. Discovery of a Natural Microsporidian Pathogen with a Broad Tissue Tropism in Caenorhabditis elegans. PLoS Pathog 2016; 12:e1005724. [PMID: 27362540 PMCID: PMC4928854 DOI: 10.1371/journal.ppat.1005724] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 06/03/2016] [Indexed: 12/21/2022] Open
Abstract
Microbial pathogens often establish infection within particular niches of their host for replication. Determining how infection occurs preferentially in specific host tissues is a key aspect of understanding host-microbe interactions. Here, we describe the discovery of a natural microsporidian parasite of the nematode Caenorhabditis elegans that displays a unique tissue tropism compared to previously described parasites of this host. We characterize the life cycle of this new species, Nematocida displodere, including pathogen entry, intracellular replication, and exit. N. displodere can invade multiple host tissues, including the epidermis, muscle, neurons, and intestine of C. elegans. Despite robust invasion of the intestine very little replication occurs there, with the majority of replication occurring in the muscle and epidermis. This feature distinguishes N. displodere from two closely related microsporidian pathogens, N. parisii and N. sp. 1, which exclusively invade and replicate in the intestine. Comparison of the N. displodere genome with N. parisii and N. sp. 1 reveals that N. displodere is the earliest diverging species of the Nematocida genus. Over 10% of the proteins encoded by the N. displodere genome belong to a single species-specific family of RING-domain containing proteins of unknown function that may be mediating interactions with the host. Altogether, this system provides a powerful whole-animal model to investigate factors responsible for pathogen growth in different tissue niches. Pathogens evolve under selective pressure from host organisms to successfully invade and proliferate in different cells and tissues of the host for their own benefit. Microsporidia represent one of the most successful phyla of pathogens, with severely reduced genomes and loss of core cellular and metabolic pathways making them dependent on host cells for their own proliferation. We sampled around Paris, France, for wild nematodes infected with natural pathogens, and discovered a wild Caenorhabditis elegans that was infected with a new species of microsporidia. We characterize the life cycle of this new species, showing the pathogen enters via host feeding, replicates in multiple host tissues, and exits as new spores via a novel vulva bursting mechanism, leading us to name this species Nematocida displodere. Despite the capacity of N. displodere to invade multiple host tissues during infection, we found that the parasite showed very little replication in the intestine. This unique tissue specificity of N. displodere stands in stark contrast to its two closest-related species, Nematocida parisii and Nematocida sp. 1, which exclusively infect and proliferate in the intestine of C. elegans. We compared the genomes of these related species and found that N. displodere devotes over 10% of its genome to a single large gene family not found in any other species, and propose that their encoded proteins may be interacting with host factors during infection.
Collapse
Affiliation(s)
- Robert J. Luallen
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California San Diego (UCSD), La Jolla, California, United States of America
| | - Aaron W. Reinke
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California San Diego (UCSD), La Jolla, California, United States of America
| | - Linda Tong
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California San Diego (UCSD), La Jolla, California, United States of America
| | - Michael R. Botts
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California San Diego (UCSD), La Jolla, California, United States of America
| | - Marie-Anne Félix
- École Normale Supérieure, Institut de Biologie de l’ENS (IBENS), CNRS-INSERM, Paris, France
| | - Emily R. Troemel
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California San Diego (UCSD), La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
34
|
Exogenous gene can be integrated into Nosema bombycis genome by mediating with a non-transposon vector. Parasitol Res 2016; 115:3093-8. [PMID: 27083186 DOI: 10.1007/s00436-016-5064-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 04/08/2016] [Indexed: 01/25/2023]
Abstract
Nosema bombycis, a microsporidium, is a pathogen of pebrine disease of silkworms, and its genomic DNA sequences had been determined. Thus far, the research of gene functions of microsporidium including N. bombycis cannot be performed with gain/loss of function. In the present study, we targeted to construct transgenic N. bombycis. Therefore, hemocytes of the infected silkworm were transfected with a non-transposon vector pIZT/V5-His vector in vivo, and the blood, in which the hemocyte with green fluorescence could be observed, was added to the cultured BmN cells. Furthermore, normal BmN cells were infected with germinated N. bombycis, and the infected cells were transfected with pIZT/V5-His. Continuous fluorescence observations exposed that there were N. bombycis with green fluorescence in some N. bombycis-infected cells, and the extracted genome from the purified N. bombycis spore was used as templates. PCR amplification was carried out with a pair of primers for specifically amplifying the green fluorescence protein (GFP) gene; a specific product representing the gfp gene could be amplified. Expression of the GFP protein through Western blotting also demonstrated that the gfp gene was perfectly inserted into the genome of N. bombysis. These results illustrated that exogenous gene can be integrated into N. bombycis genome by mediating with a non-transposon vector. Our research not only offers a strategy for research on gene function of N. bombycis but also provides an important reference for constructing genetically modified microsporidium utilized for biocontrol of pests.
Collapse
|
35
|
Liu H, Li M, He X, Cai S, He X, Lu X. Transcriptome sequencing and characterization of ungerminated and germinated spores of Nosema bombycis. Acta Biochim Biophys Sin (Shanghai) 2016; 48:246-56. [PMID: 26837419 DOI: 10.1093/abbs/gmv140] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2015] [Accepted: 11/21/2015] [Indexed: 12/23/2022] Open
Abstract
Nosema bombycis is an obligate intracellular parasitic fungus that utilizes a distinctive mechanism to infect Bombyx mori. Germination, an indispensible process through which microsporidia infect the host cells, is regarded as a key developmental turning point for microsporidia from dormant state to reproduction state. Thus, elucidating the transcriptome changes before and after germination is crucial for parasite control. However, the molecular basis of germination of microsporidia remains unknown. To investigate this germination process, the transcriptome of N. bombycis ungerminated spores and germinated spores were sequenced and analyzed. More than 60 million high-quality transcript reads were generated from these two groups using RNA-Seq technology. After assembly, 2756 and 2690 unigenes were identified, respectively, and subsequently annotated based on known proteins. After analysis of differentially expressed genes, 66 genes were identified to be differentially expressed (P ≤ 0.05) between these two groups. A protein phosphatase-associated gene was first identified to be significantly up-regulated as determined by RNA-Seq and immunoblot analysis, indicating that dephosphorylation might potentially contribute to microsporidia germination. The DEGs that encode proteins involved in glycometabolism, spore wall proteins and ricin B lectin of N. bombycis were also analyzed. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses revealed genes responsible for some specific biological functions and processes. The datasets generated in this study provide a basic characterization of the transcriptome changes in N. bombycis during germination. The analysis of transcriptome data and identification of certain functional genes which are robust candidate genes related to germination will help to provide a deep understanding of spore germination and invasion.
Collapse
Affiliation(s)
- Han Liu
- Laboratory of Invertebrate Pathology, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Mingqian Li
- Tongde Hospital of Zhejiang Province, Hangzhou 310058, China
| | - Xinyi He
- Laboratory of Invertebrate Pathology, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Shunfeng Cai
- Laboratory of Invertebrate Pathology, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xiangkang He
- Laboratory of Invertebrate Pathology, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xingmeng Lu
- Laboratory of Invertebrate Pathology, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
36
|
Kinetoplastid Phylogenomics Reveals the Evolutionary Innovations Associated with the Origins of Parasitism. Curr Biol 2015; 26:161-172. [PMID: 26725202 PMCID: PMC4728078 DOI: 10.1016/j.cub.2015.11.055] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 11/17/2015] [Accepted: 11/19/2015] [Indexed: 01/01/2023]
Abstract
The evolution of parasitism is a recurrent event in the history of life and a core problem in evolutionary biology. Trypanosomatids are important parasites and include the human pathogens Trypanosoma brucei, Trypanosoma cruzi, and Leishmania spp., which in humans cause African trypanosomiasis, Chagas disease, and leishmaniasis, respectively. Genome comparison between trypanosomatids reveals that these parasites have evolved specialized cell-surface protein families, overlaid on a well-conserved cell template. Understanding how these features evolved and which ones are specifically associated with parasitism requires comparison with related non-parasites. We have produced genome sequences for Bodo saltans, the closest known non-parasitic relative of trypanosomatids, and a second bodonid, Trypanoplasma borreli. Here we show how genomic reduction and innovation contributed to the character of trypanosomatid genomes. We show that gene loss has “streamlined” trypanosomatid genomes, particularly with respect to macromolecular degradation and ion transport, but consistent with a widespread loss of functional redundancy, while adaptive radiations of gene families involved in membrane function provide the principal innovations in trypanosomatid evolution. Gene gain and loss continued during trypanosomatid diversification, resulting in the asymmetric assortment of ancestral characters such as peptidases between Trypanosoma and Leishmania, genomic differences that were subsequently amplified by lineage-specific innovations after divergence. Finally, we show how species-specific, cell-surface gene families (DGF-1 and PSA) with no apparent structural similarity are independent derivations of a common ancestral form, which we call “bodonin.” This new evidence defines the parasitic innovations of trypanosomatid genomes, revealing how a free-living phagotroph became adapted to exploiting hostile host environments. The Bodo saltans genome reveals evolutionary changes at the origin of parasitism Parasite genomes are streamlined, consistent with a loss of functional redundancy Expanded parasite transporter genes reflect a reorientation of membrane function Non-homologous, parasite cell-surface proteins evolved from a common ancestor
Collapse
|
37
|
The Prediction and Validation of Small CDSs Expand the Gene Repertoire of the Smallest Known Eukaryotic Genomes. PLoS One 2015; 10:e0139075. [PMID: 26421846 PMCID: PMC4589312 DOI: 10.1371/journal.pone.0139075] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 09/09/2015] [Indexed: 01/05/2023] Open
Abstract
The proper prediction of the gene catalogue of an organism is essential to obtain a representative snapshot of its overall lifestyle, especially when it is not amenable to culturing. Microsporidia are obligate intracellular, sometimes hard to culture, eukaryotic parasites known to infect members of every animal phylum. To date, sequencing and annotation of microsporidian genomes have revealed a poor gene complement with highly reduced gene sizes. In the present paper, we investigated whether such gene sizes may have induced biases for the methodologies used for genome annotation, with an emphasis on small coding sequence (CDS) gene prediction. Using better delineated intergenic regions from four Encephalitozoon genomes, we predicted de novo new small CDSs with sizes ranging from 78 to 255 bp (median 168) and corroborated these predictions by RACE-PCR experiments in Encephalitozoon cuniculi. Most of the newly found genes are present in other distantly related microsporidian species, suggesting their biological relevance. The present study provides a better framework for annotating microsporidian genomes and to train and evaluate new computational methods dedicated at detecting ultra-small genes in various organisms.
Collapse
|
38
|
Lallo MA, Vidoto Da Costa LF, Alvares-Saraiva AM, Rocha PRD, Spadacci-Morena DD, Konno FTDC, Suffredini IB. Culture and propagation of microsporidia of veterinary interest. J Vet Med Sci 2015; 78:171-6. [PMID: 26346746 PMCID: PMC4785104 DOI: 10.1292/jvms.15-0401] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Microsporidia are obligate intracellular mitochrondria-lacking pathogens that rely on host cells to grow and
multiply. Microsporidia, currently classified as fungi, are ubiquitous in nature and are found worldwide. They
infect a large number of mammals and are recognized as opportunistic infection agents in HIV-AIDS patients.
Its importance for veterinary medicine has been unveiled in recent years through the description of clinical
and subclinical forms of infection in domestic and wild animals. Domestic and wild birds may be infected by
the same human microsporidia, reinforcing their zoonotic potential. Microsporidiosis in fish is prevalent and
causes significant economic losses for fish farming. Some species of microsporidia have been propagated in
cell cultures, which may provide conditions for the development of diagnostic techniques, understanding of
pathogenesis and immune responses and for the discovery of potential therapies. Unfortunately, the cultivation
of these parasites is not fully standardized in most research laboratories, especially in the veterinary
field. The aim of this review is to relate the most important microsporidia of veterinary interest and
demonstrate how these pathogens can be grown and propagated in cell culture for diagnostic purposes or for
pathogenesis studies. Cultivation of microsporidia allowed the study of its life cycle, metabolism,
pathogenesis and diagnosis, and may also serve as a repository for these pathogens for molecular, biochemical,
antigenic and epidemiological studies.
Collapse
Affiliation(s)
- Maria Anete Lallo
- Environmental and Experimental Pathology, Paulista University, São Paulo, Brazil
| | | | | | | | | | | | | |
Collapse
|
39
|
Szumowski SC, Estes KA, Popovich JJ, Botts MR, Sek G, Troemel ER. Small GTPases promote actin coat formation on microsporidian pathogens traversing the apical membrane of Caenorhabditis elegans intestinal cells. Cell Microbiol 2015; 18:30-45. [PMID: 26147591 DOI: 10.1111/cmi.12481] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 06/01/2015] [Accepted: 06/12/2015] [Indexed: 12/16/2022]
Abstract
Many intracellular pathogens co-opt actin in host cells, but little is known about these interactions in vivo. We study the in vivo trafficking and exit of the microsporidian Nematocida parisii, which is an intracellular pathogen that infects intestinal cells of the nematode Caenorhabditis elegans. We recently demonstrated that N. parisii uses directional exocytosis to escape out of intestinal cells into the intestinal tract. Here, we show that an intestinal-specific isoform of C. elegans actin called ACT-5 forms coats around membrane compartments that contain single exocytosing spores, and that these coats appear to form after fusion with the apical membrane. We performed a genetic screen for host factors required for actin coat formation and identified small GTPases important for this process. Through analysis of animals defective in these factors, we found that actin coats are not required for pathogen exit although they may boost exocytic output. Later during infection, we find that ACT-5 also forms coats around membrane-bound vesicles that contain multiple spores. These vesicles are likely formed by clathrin-dependent compensatory endocytosis to retrieve membrane material that has been trafficked to the apical membrane as part of the exocytosis process. These findings provide insight into microsporidia interaction with host cells, and provide novel in vivo examples of the manner in which intracellular pathogens co-opt host actin during their life cycle.
Collapse
Affiliation(s)
- Suzannah C Szumowski
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California, La Jolla, CA, USA
| | - Kathleen A Estes
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California, La Jolla, CA, USA
| | - John J Popovich
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California, La Jolla, CA, USA
| | - Michael R Botts
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California, La Jolla, CA, USA
| | - Grace Sek
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California, La Jolla, CA, USA
| | - Emily R Troemel
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California, La Jolla, CA, USA
| |
Collapse
|
40
|
Luallen RJ, Bakowski MA, Troemel ER. Characterization of microsporidia-induced developmental arrest and a transmembrane leucine-rich repeat protein in Caenorhabditis elegans. PLoS One 2015; 10:e0124065. [PMID: 25874557 PMCID: PMC4395247 DOI: 10.1371/journal.pone.0124065] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 03/10/2015] [Indexed: 12/26/2022] Open
Abstract
Microsporidia comprise a highly diverged phylum of intracellular, eukaryotic pathogens, with some species able to cause life-threatening illnesses in immunocompromised patients. To better understand microsporidian infection in animals, we study infection of the genetic model organism Caenorhabditis elegans and a species of microsporidia, Nematocida parisii, which infects Caenorhabditis nematodes in the wild. We conducted a targeted RNAi screen for host C. elegans genes important for infection and growth of N. parisii, using nematode larval arrest as an assay for infection. Here, we present the results of this RNAi screen, and our analyses on one of the RNAi hits from the screen that was ultimately not corroborated by loss of function mutants. This hit was an RNAi clone against F56A8.3, a conserved gene that encodes a transmembrane protein containing leucine-rich repeats (LRRs), a domain found in numerous pathogen receptors from other systems. This RNAi clone caused C. elegans to be resistant to infection by N. parisii, leading to reduced larval arrest and lower pathogen load. Characterization of the endogenous F56A8.3 protein revealed that it is expressed in the intestine, localized to the membrane around lysosome-related organelles (LROs), and exists in two different protein isoforms in C. elegans. We used the CRISPR-Cas9 system to edit the F56A8.3 locus and created both a frameshift mutant resulting in a truncated protein and a complete knockout mutant. Neither of these mutants was able to recapitulate the infection phenotypes of the RNAi clone, indicating that the RNAi-mediated phenotypes are due to an off-target effect of the RNAi clone. Nevertheless, this study describes microsporidia-induced developmental arrest in C. elegans, presents results from an RNAi screen for host genes important for microsporidian infection, and characterizes aspects of the conserved F56A8.3 gene and its protein product.
Collapse
Affiliation(s)
- Robert J. Luallen
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California San Diego, La Jolla, California, United States of America
| | - Malina A. Bakowski
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California San Diego, La Jolla, California, United States of America
| | - Emily R. Troemel
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California San Diego, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
41
|
Vilcinskas A, Schmidtberg H, Estoup A, Tayeh A, Facon B, Vogel H. Evolutionary ecology of microsporidia associated with the invasive ladybird Harmonia axyridis. INSECT SCIENCE 2015; 22:313-324. [PMID: 25131382 DOI: 10.1111/1744-7917.12159] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 07/07/2014] [Indexed: 06/03/2023]
Abstract
Invasive species are characterized by the rapid growth and spread of their populations after establishing a foothold in new habitats, and there are now many examples of such species negatively affecting biodiversity and the economy. It is unclear why some species can become successful invaders, whereas most (even if closely related) remain noninvasive. We previously proposed a hypothesis that parasites associated with invading species can promote their invasive success if they are harmless toward the invaders but harmful to their competitors and/or predators in the newly colonized habitat. Here we discuss whether microsporidia that have recently been discovered in the invasive ladybird Harmonia axyridis contribute to its invasive success. We show that all H. axyridis beetles sourced from diverse collection sites all over the world carry abundant microsporidia. This suggests that both native and invasive H. axyridis populations are associated with these tolerated parasites, which were likely to have existed in native populations before expansion rather than being acquired in newly colonized areas. We describe the pathogenesis of the microsporidia during different developmental stages of H. axyridis and we address the possibility that the predation of its infected eggs and larvae by competing native ladybird species may lead to their infection and ultimately to their decline. Finally, we discuss our initial hypothesis: microsporidia that are tolerated by an invasive vector insect can be active against susceptible native competitors and/or predator species.
Collapse
Affiliation(s)
- Andreas Vilcinskas
- Institute of Phytopathology and Applied Zoology, Justus-Liebig-University of Giessen, Heinrich-Buff-Ring 26-32, Giessen, D-35392, Germany
| | - Henrike Schmidtberg
- Fraunhofer Institute for Molecular Biology and Applied Ecology, Winchester Strasse 2, Giessen, D-35394, Germany
| | | | | | | | - Heiko Vogel
- Max Planck Institute for Chemical Ecology, Hans-Knöll-Strasse 8, D-07745, Jena, Germany
| |
Collapse
|
42
|
Balla KM, Andersen EC, Kruglyak L, Troemel ER. A wild C. elegans strain has enhanced epithelial immunity to a natural microsporidian parasite. PLoS Pathog 2015; 11:e1004583. [PMID: 25680197 PMCID: PMC4334554 DOI: 10.1371/journal.ppat.1004583] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 11/18/2014] [Indexed: 12/27/2022] Open
Abstract
Microbial pathogens impose selective pressures on their hosts, and combatting these pathogens is fundamental to the propagation of a species. Innate immunity is an ancient system that provides the foundation for pathogen resistance, with epithelial cells in humans increasingly appreciated to play key roles in innate defense. Here, we show that the nematode C. elegans displays genetic variation in epithelial immunity against intestinal infection by its natural pathogen, Nematocida parisii. This pathogen belongs to the microsporidia phylum, which comprises a large phylum of over 1400 species of fungal-related parasites that can infect all animals, including humans, but are poorly understood. Strikingly, we find that a wild C. elegans strain from Hawaii is able to clear intracellular infection by N. parisii, with this ability restricted to young larval animals. Notably, infection of older larvae does not impair progeny production, while infection of younger larvae does. The early-life immunity of Hawaiian larvae enables them to produce more progeny later in life, providing a selective advantage in a laboratory setting—in the presence of parasite it is able to out-compete a susceptible strain in just a few generations. We show that enhanced immunity is dominant to susceptibility, and we use quantitative trait locus mapping to identify four genomic loci associated with resistance. Furthermore, we generate near-isogenic strains to directly demonstrate that two of these loci influence resistance. Thus, our findings show that early-life immunity of C. elegans against microsporidia is a complex trait that enables the host to produce more progeny later in life, likely improving its evolutionary success. Infectious diseases caused by microbes create some of the strongest forces in evolution, by killing their hosts, and impairing their ability to produce progeny. Microsporidia are very common microbes that cause disease in all animals, including roundworms, insects, fish and people. We investigated microsporidia infection in the roundworm C. elegans, and found that strains from diverse parts of the world have differing levels of resistance against infection. Interestingly, a C. elegans strain from Hawaii can clear infection but only during the earliest stage of life. This resistance appears to be evolutionarily important, because it is during this early stage of life when infection can greatly reduce the number of progeny produced by the host. Consistent with this idea, if the Hawaiian strain is infected when young, it will ultimately produce more progeny than a susceptible strain of C. elegans. We find that this early life resistance of Hawaiian animals is due to a combination of genetic regions, which together provide enhanced immunity against a natural pathogen, thus enabling this strain to have more offspring.
Collapse
Affiliation(s)
- Keir M. Balla
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, California, United States of America
| | - Erik C. Andersen
- Department of Molecular Biosciences, Northwestern University, Evanston, Illinois, United States of America
| | - Leonid Kruglyak
- Department of Human Genetics, Department of Biological Chemistry, and Howard Hughes Medical Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Emily R. Troemel
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
43
|
Natsopoulou ME, McMahon DP, Doublet V, Bryden J, Paxton RJ. Interspecific competition in honeybee intracellular gut parasites is asymmetric and favours the spread of an emerging infectious disease. Proc Biol Sci 2015; 282:20141896. [PMID: 25429014 PMCID: PMC4262169 DOI: 10.1098/rspb.2014.1896] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 10/24/2014] [Indexed: 02/03/2023] Open
Abstract
There is increasing appreciation that hosts in natural populations are subject to infection by multiple parasite species. Yet the epidemiological and ecological processes determining the outcome of mixed infections are poorly understood. Here, we use two intracellular gut parasites (Microsporidia), one exotic and one co-evolved in the western honeybee (Apis mellifera), in an experiment in which either one or both parasites were administered either simultaneously or sequentially. We provide clear evidence of within-host competition; order of infection was an important determinant of the competitive outcome between parasites, with the first parasite significantly inhibiting the growth of the second, regardless of species. However, the strength of this 'priority effect' was highly asymmetric, with the exotic Nosema ceranae exhibiting stronger inhibition of Nosema apis than vice versa. Our results reveal an unusual asymmetry in parasite competition that is dependent on order of infection. When incorporated into a mathematical model of disease prevalence, we find asymmetric competition to be an important predictor of the patterns of parasite prevalence found in nature. Our findings demonstrate the wider significance of complex multi-host-multi-parasite interactions as drivers of host-pathogen community structure.
Collapse
Affiliation(s)
- Myrsini E Natsopoulou
- Institut für Biologie, Martin-Luther-Universität Halle-Wittenberg, Hoher Weg 8, Halle (Saale) 06120, Germany
| | - Dino P McMahon
- Institut für Biologie, Martin-Luther-Universität Halle-Wittenberg, Hoher Weg 8, Halle (Saale) 06120, Germany School of Biological Sciences, MBC, Queen's University Belfast, Belfast BT9 7BL, UK Institute of Biology, Freie Universität Berlin, Schwendenerstr. 1, Berlin 14195, Germany Department for Materials and Environment, BAM Federal Institute for Materials Research and Testing, Unter den Eichen 87, Berlin 12205, Germany
| | - Vincent Doublet
- Institut für Biologie, Martin-Luther-Universität Halle-Wittenberg, Hoher Weg 8, Halle (Saale) 06120, Germany German Centre for Integrative Biodiversity Research (iDiv) Halle-Jena-Leipzig, Deutscher Platz 5e, Leipzig 04103, Germany
| | - John Bryden
- School of Biological Sciences, Royal Holloway, University of London, Egham, Surrey TW20 0EX, UK
| | - Robert J Paxton
- Institut für Biologie, Martin-Luther-Universität Halle-Wittenberg, Hoher Weg 8, Halle (Saale) 06120, Germany School of Biological Sciences, MBC, Queen's University Belfast, Belfast BT9 7BL, UK German Centre for Integrative Biodiversity Research (iDiv) Halle-Jena-Leipzig, Deutscher Platz 5e, Leipzig 04103, Germany
| |
Collapse
|
44
|
Early responses of silkworm midgut to microsporidium infection – A Digital Gene Expression analysis. J Invertebr Pathol 2015; 124:6-14. [DOI: 10.1016/j.jip.2014.10.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2014] [Revised: 10/04/2014] [Accepted: 10/07/2014] [Indexed: 02/03/2023]
|
45
|
Vidau C, Panek J, Texier C, Biron DG, Belzunces LP, Le Gall M, Broussard C, Delbac F, El Alaoui H. Differential proteomic analysis of midguts from Nosema ceranae-infected honeybees reveals manipulation of key host functions. J Invertebr Pathol 2014; 121:89-96. [DOI: 10.1016/j.jip.2014.07.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Revised: 06/20/2014] [Accepted: 07/08/2014] [Indexed: 01/14/2023]
|
46
|
Panek J, El Alaoui H, Mone A, Urbach S, Demettre E, Texier C, Brun C, Zanzoni A, Peyretaillade E, Parisot N, Lerat E, Peyret P, Delbac F, Biron DG. Hijacking of host cellular functions by an intracellular parasite, the microsporidian Anncaliia algerae. PLoS One 2014; 9:e100791. [PMID: 24967735 PMCID: PMC4072689 DOI: 10.1371/journal.pone.0100791] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 05/29/2014] [Indexed: 11/18/2022] Open
Abstract
Intracellular pathogens including bacteria, viruses and protozoa hijack host cell functions to access nutrients and to bypass cellular defenses and immune responses. These strategies have been acquired through selective pressure and allowed pathogens to reach an appropriate cellular niche for their survival and growth. To get new insights on how parasites hijack host cellular functions, we developed a SILAC (Stable Isotope Labeling by Amino Acids in Cell culture) quantitative proteomics workflow. Our study focused on deciphering the cross-talk in a host-parasite association, involving human foreskin fibroblasts (HFF) and the microsporidia Anncaliia algerae, a fungus related parasite with an obligate intracellular lifestyle and a strong host dependency. The host-parasite cross-talk was analyzed at five post-infection times 1, 6, 12 and 24 hours post-infection (hpi) and 8 days post-infection (dpi). A significant up-regulation of four interferon-induced proteins with tetratricopeptide repeats IFIT1, IFIT2, IFIT3 and MX1 was observed at 8 dpi suggesting a type 1 interferon (IFN) host response. Quantitative alteration of host proteins involved in biological functions such as signaling (STAT1, Ras) and reduction of the translation activity (EIF3) confirmed a host type 1 IFN response. Interestingly, the SILAC approach also allowed the detection of 148 A. algerae proteins during the kinetics of infection. Among these proteins many are involved in parasite proliferation, and an over-representation of putative secreted effectors proteins was observed. Finally our survey also suggests that A. algerae could use a transposable element as a lure strategy to escape the host innate immune system.
Collapse
Affiliation(s)
- Johan Panek
- Clermont Université, Université Blaise Pascal, Laboratoire Microorganismes: Génome et Environnement, Clermont-Ferrand, France
- CNRS, UMR 6023, LMGE, Aubière, France
| | - Hicham El Alaoui
- Clermont Université, Université Blaise Pascal, Laboratoire Microorganismes: Génome et Environnement, Clermont-Ferrand, France
- CNRS, UMR 6023, LMGE, Aubière, France
- * E-mail: (HEA); (DGB)
| | - Anne Mone
- Clermont Université, Université Blaise Pascal, Laboratoire Microorganismes: Génome et Environnement, Clermont-Ferrand, France
- CNRS, UMR 6023, LMGE, Aubière, France
| | - Serge Urbach
- Functional Proteomics Platform. UMR CNRS 5203, Montpellier, France
| | - Edith Demettre
- Functional Proteomics Platform. UMS CNRS 3426, Montpellier, France
| | - Catherine Texier
- Clermont Université, Université Blaise Pascal, Laboratoire Microorganismes: Génome et Environnement, Clermont-Ferrand, France
- CNRS, UMR 6023, LMGE, Aubière, France
| | - Christine Brun
- INSERM, UMR1090 TAGC, Marseille, Marseille, France
- Aix-Marseille Université, UMR1090 TAGC, Marseille, France
- CNRS, Marseille, France
| | - Andreas Zanzoni
- INSERM, UMR1090 TAGC, Marseille, Marseille, France
- Aix-Marseille Université, UMR1090 TAGC, Marseille, France
| | - Eric Peyretaillade
- Clermont Université, Université d'Auvergne, I.U.T., UFR Pharmacie, Clermont-Ferrand, France
- Clermont Université, Université d'Auvergne, EA 4678, Conception, Ingénierie et Développement de l'Aliment et du Médicament, Clermont-Ferrand, France
| | - Nicolas Parisot
- Clermont Université, Université d'Auvergne, I.U.T., UFR Pharmacie, Clermont-Ferrand, France
- Clermont Université, Université d'Auvergne, EA 4678, Conception, Ingénierie et Développement de l'Aliment et du Médicament, Clermont-Ferrand, France
| | - Emmanuelle Lerat
- Université de Lyon, Université Lyon 1, CNRS, UMR5558, Laboratoire de Biométrie et Biologie Evolutive, Villeurbanne, France
| | - Pierre Peyret
- Clermont Université, Université d'Auvergne, I.U.T., UFR Pharmacie, Clermont-Ferrand, France
- Clermont Université, Université d'Auvergne, EA 4678, Conception, Ingénierie et Développement de l'Aliment et du Médicament, Clermont-Ferrand, France
| | - Frederic Delbac
- Clermont Université, Université Blaise Pascal, Laboratoire Microorganismes: Génome et Environnement, Clermont-Ferrand, France
- CNRS, UMR 6023, LMGE, Aubière, France
| | - David G. Biron
- Clermont Université, Université Blaise Pascal, Laboratoire Microorganismes: Génome et Environnement, Clermont-Ferrand, France
- CNRS, UMR 6023, LMGE, Aubière, France
- * E-mail: (HEA); (DGB)
| |
Collapse
|
47
|
Zhu F, Shen Z, Xu L, Guo X. Molecular characteristics of the alpha- and beta-tubulin genes of Nosema philosamiae. Folia Parasitol (Praha) 2014; 60:411-5. [PMID: 24471282 DOI: 10.14411/fp.2013.043] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Microsporidia are intracellular parasites of insects and other higher eukaryotes. The microsporidian Nosema philosamiae Talukdar, 1961 was isolated from the eri silkworm, Philosamia cynthia ricini Grote. In the present study, alpha- and beta-tubulin genes from N. philosamiae were characterized. The identity analysis of nucleotide and amino acid sequences indicated high similarity with species of Nosema Nägeli, 1857 sensu lato (nucleotide sequences, > or = 96.0%; amino acid sequences, > or = 99.0%). However, the tubulin genes of N. philosamiae share low sequence similarity with that of N. ceranae Fries, Feng, da Silva, Slemenda et Pieniazek, 1996 (strain BRL01) and a Nosema/Vairimorpha species. Phylogenies based on alpha-, beta- and combined alpha- plus beta-tubulin gene sequences showed that N. philosamiae, along with the true Nosema species, forms a separate clade with a high bootstrap value, with N. ceranae BRL01 forming a clade of its own. The results indicated that the alpha- and beta-tubulin sequences may be useful as a diagnostic tool to discriminate the true Nosema group from the Nosema/Vairimorpha group.
Collapse
Affiliation(s)
- Feng Zhu
- Jiangsu University of Science and Technology, Zhenjiang, Jiangsu, China
| | - Zhongyuan Shen
- Jiangsu University of Science and Technology, Zhenjiang, Jiangsu, China
| | - Li Xu
- Jiangsu University of Science and Technology, Zhenjiang, Jiangsu, China
| | - Xijie Guo
- Jiangsu University of Science and Technology, Zhenjiang, Jiangsu, China
| |
Collapse
|
48
|
Sapir A, Dillman AR, Connon SA, Grupe BM, Ingels J, Mundo-Ocampo M, Levin LA, Baldwin JG, Orphan VJ, Sternberg PW. Microsporidia-nematode associations in methane seeps reveal basal fungal parasitism in the deep sea. Front Microbiol 2014; 5:43. [PMID: 24575084 PMCID: PMC3918590 DOI: 10.3389/fmicb.2014.00043] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Accepted: 01/21/2014] [Indexed: 12/23/2022] Open
Abstract
The deep sea is Earth's largest habitat but little is known about the nature of deep-sea parasitism. In contrast to a few characterized cases of bacterial and protistan parasites, the existence and biological significance of deep-sea parasitic fungi is yet to be understood. Here we report the discovery of a fungus-related parasitic microsporidium, Nematocenator marisprofundi n. gen. n. sp. that infects benthic nematodes at methane seeps on the Pacific Ocean floor. This infection is species-specific and has been temporally and spatially stable over 2 years of sampling, indicating an ecologically consistent host-parasite interaction. A high distribution of spores in the reproductive tracts of infected males and females and their absence from host nematodes' intestines suggests a sexual transmission strategy in contrast to the fecal-oral transmission of most microsporidia. N. marisprofundi targets the host's body wall muscles causing cell lysis, and in severe infection even muscle filament degradation. Phylogenetic analyses placed N. marisprofundi in a novel and basal clade not closely related to any described microsporidia clade, suggesting either that microsporidia-nematode parasitism occurred early in microsporidia evolution or that host specialization occurred late in an ancient deep-sea microsporidian lineage. Our findings reveal that methane seeps support complex ecosystems involving interkingdom interactions between bacteria, nematodes, and parasitic fungi and that microsporidia parasitism exists also in the deep-sea biosphere.
Collapse
Affiliation(s)
- Amir Sapir
- Howard Hughes Medical Institute and Division of Biology, California Institute of Technology Pasadena, CA, USA
| | - Adler R Dillman
- Howard Hughes Medical Institute and Division of Biology, California Institute of Technology Pasadena, CA, USA
| | - Stephanie A Connon
- Division of Geological and Planetary Sciences, California Institute of Technology Pasadena, CA, USA
| | - Benjamin M Grupe
- Integrative Oceanography Division, Center for Marine Biodiversity and Conservation, Scripps Institution of Oceanography La Jolla, CA, USA
| | - Jeroen Ingels
- Marine Life Support Systems, Plymouth Marine Laboratory, Prospect Place UK
| | - Manuel Mundo-Ocampo
- Department of Agricultural Biotecnology, Nematology Laboratory, CIIDIR-IPN Unidad Sinaloa Sinaloa, Mexico
| | - Lisa A Levin
- Integrative Oceanography Division, Center for Marine Biodiversity and Conservation, Scripps Institution of Oceanography La Jolla, CA, USA
| | - James G Baldwin
- Department of Nematology, University of California Riverside, CA, USA
| | - Victoria J Orphan
- Division of Geological and Planetary Sciences, California Institute of Technology Pasadena, CA, USA
| | - Paul W Sternberg
- Howard Hughes Medical Institute and Division of Biology, California Institute of Technology Pasadena, CA, USA
| |
Collapse
|
49
|
Ghani IA, Dieng H, Abu Hassan ZA, Ramli N, Kermani N, Satho T, Ahmad H, Abang FB, Fukumitsu Y, Ahmad AH. Pathogenicity of a microsporidium isolate from the diamondback moth against Noctuid moths: characterization and implications for microbiological pest management. PLoS One 2013; 8:e81642. [PMID: 24349104 PMCID: PMC3859509 DOI: 10.1371/journal.pone.0081642] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2013] [Accepted: 10/15/2013] [Indexed: 12/31/2022] Open
Abstract
Background Due to problems with chemical control, there is increasing interest in the use of microsporidia for control of lepidopteran pests. However, there have been few studies to evaluate the susceptibility of exotic species to microsporidia from indigenous Lepidoptera. Methodology/Principal Findings We investigated some biological characteristics of the microsporidian parasite isolated from wild Plutella xylostella (PX) and evaluated its pathogenicity on the laboratory responses of sympatric invasive and resident noctuid moths. There were significant differences in spore size and morphology between PX and Spodoptera litura (SL) isolates. Spores of PX isolate were ovocylindrical, while those of SL were oval. PX spores were 1.05 times longer than those of SL, which in turn were 1.49 times wider than those of the PX. The timing of infection peaks was much shorter in SL and resulted in earlier larval death. There were no noticeable differences in amplicon size (two DNA fragments were each about 1200 base pairs in length). Phylogenetic analysis revealed that the small subunit (SSU) rRNA gene sequences of the two isolates shared a clade with Nosema/Vairimorpha sequences. The absence of octospores in infected spodopteran tissues suggested that PX and SL spores are closely related to Nosema plutellae and N. bombycis, respectively. Both SL and S. exigua (SE) exhibited susceptibility to the PX isolate infection, but showed different infection patterns. Tissular infection was more diverse in the former and resulted in much greater spore production and larval mortality. Microsporidium-infected larvae pupated among both infected and control larvae, but adult emergence occurred only in the second group. Conclusion/Significance The PX isolate infection prevented completion of development of most leafworm and beet armyworm larvae. The ability of the microsporidian isolate to severely infect and kill larvae of both native and introduced spodopterans makes it a valuable candidate for biocontrol against lepidopteran pests.
Collapse
Affiliation(s)
- Idris Abd Ghani
- Faculty of Science and Technology, Universiti Kebangsaan Malaysia, Bangi, Malaysia
| | - Hamady Dieng
- School of Biological Sciences, Universiti Sains Malaysia, Penang, Malaysia
- * E-mail:
| | | | - Norazsida Ramli
- Department of Biomedical Science, Islamic University of Malaysia, Pahang, Malaysia
| | - Nadia Kermani
- Faculty of Science and Technology, Universiti Kebangsaan Malaysia, Bangi, Malaysia
| | - Tomomitsu Satho
- Faculty of Pharmaceutical Sciences, Fukuoka University, Japan
| | - Hamdan Ahmad
- School of Biological Sciences, Universiti Sains Malaysia, Penang, Malaysia
| | - Fatimah Bt Abang
- Faculty of Resource Science and Technology, Universiti Malaysia Sarawak, Kuching, Malaysia
| | - Yuki Fukumitsu
- Faculty of Pharmaceutical Sciences, Fukuoka University, Japan
| | - Abu Hassan Ahmad
- School of Biological Sciences, Universiti Sains Malaysia, Penang, Malaysia
| |
Collapse
|
50
|
Szumowski SC, Estes KA, Troemel ER. Preparing a discreet escape: Microsporidia reorganize host cytoskeleton prior to non-lytic exit from C. elegans intestinal cells. WORM 2013; 1:207-11. [PMID: 24058850 PMCID: PMC3670220 DOI: 10.4161/worm.20501] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2012] [Accepted: 04/24/2012] [Indexed: 11/25/2022]
Abstract
Intracellular pathogens commonly invade and replicate inside of intestinal cells and exit from these cells is a crucial step in pathogen transmission. For convenience, studies of intracellular pathogens are often conducted using in vitro cell culture systems, which unfortunately lack important features of polarized, intact intestinal epithelial cells. The nematode C. elegans provides a tractable system to study intracellular pathogens in vivo, where features of differentiated epithelial cells are easily visualized. In a recent paper, we used C. elegans as a host organism to study the exit strategy of Nematocida parisii, a naturally occurring intracellular pathogen in the microsporidia phylum. We showed that N. parisii remodels the C. elegans host cytoskeleton, and then exits host cells in an actin-dependent, non-lytic fashion. These findings illuminate key details about the transmission of microsporidia, which are poorly understood but ubiquitous pathogens. More generally, these findings have implications for exit strategies used by other intracellular pathogens that also infect epithelial cells.
Collapse
Affiliation(s)
- Suzannah C Szumowski
- Division of Biological Sciences; Section of Cell and Developmental Biology; University of California San Diego; La Jolla, CA USA
| | | | | |
Collapse
|