1
|
Manjunatha UH, Lakshminarayana SB, Jumani RS, Chao AT, Young JM, Gable JE, Knapp M, Hanna I, Galarneau JR, Cantwell J, Kulkarni U, Turner M, Lu P, Darrell KH, Watson LC, Chan K, Patra D, Mamo M, Luu C, Cuellar C, Shaul J, Xiao L, Chen YB, Carney SK, Lakshman J, Osborne CS, Zambriski JA, Aziz N, Sarko C, Diagana TT. Cryptosporidium PI(4)K inhibitor EDI048 is a gut-restricted parasiticidal agent to treat paediatric enteric cryptosporidiosis. Nat Microbiol 2024; 9:2817-2835. [PMID: 39379634 PMCID: PMC11522000 DOI: 10.1038/s41564-024-01810-x] [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: 01/17/2024] [Accepted: 08/15/2024] [Indexed: 10/10/2024]
Abstract
Diarrhoeal disease caused by Cryptosporidium is a major cause of morbidity and mortality in young and malnourished children from low- and middle-income countries, with no vaccine or effective treatment. Here we describe the discovery of EDI048, a Cryptosporidium PI(4)K inhibitor, designed to be active at the infection site in the gastrointestinal tract and undergo rapid metabolism in the liver. By using mutational analysis and crystal structure, we show that EDI048 binds to highly conserved amino acid residues in the ATP-binding site. EDI048 is orally efficacious in an immunocompromised mouse model despite negligible circulating concentrations, thus demonstrating that gastrointestinal exposure is necessary and sufficient for efficacy. In neonatal calves, a clinical model of cryptosporidiosis, EDI048 treatment resulted in rapid resolution of diarrhoea and significant reduction in faecal oocyst shedding. Safety and pharmacological studies demonstrated predictable metabolism and low systemic exposure of EDI048, providing a substantial safety margin required for a paediatric indication. EDI048 is a promising clinical candidate for the treatment of life-threatening paediatric cryptosporidiosis.
Collapse
Affiliation(s)
| | | | - Rajiv S Jumani
- Global Health, Biomedical Research, Novartis, Emeryville, CA, USA
| | - Alexander T Chao
- Global Health, Biomedical Research, Novartis, Emeryville, CA, USA
| | | | - Jonathan E Gable
- Global Health, Biomedical Research, Novartis, Emeryville, CA, USA
| | - Mark Knapp
- Biomedical Research, Novartis, Emeryville, CA, USA
| | - Imad Hanna
- Biomedical Research, Novartis, East Hanover, NJ, USA
| | | | | | | | | | - Peichao Lu
- Biomedical Research, Novartis, Emeryville, CA, USA
| | - Kristen H Darrell
- Global Health, Biomedical Research, Novartis, Emeryville, CA, USA
- Metagenomi, Inc., Emeryville, CA, USA
| | - Lucy C Watson
- Global Health, Biomedical Research, Novartis, Emeryville, CA, USA
| | - Katherine Chan
- Global Health, Biomedical Research, Novartis, Emeryville, CA, USA
| | - Debjani Patra
- Global Health, Biomedical Research, Novartis, Emeryville, CA, USA
| | | | | | | | - Jacob Shaul
- Biomedical Research, Novartis, Emeryville, CA, USA
- Absci Corporation, Vancouver, WA, USA
| | - Linda Xiao
- Global Health, Biomedical Research, Novartis, Emeryville, CA, USA
| | - Ying-Bo Chen
- Global Health, Biomedical Research, Novartis, Emeryville, CA, USA
| | - Shannon K Carney
- Department of Population Health Sciences, College of Veterinary Medicine, Center for One Health Research, Blacksburg, VA, USA
- Cornell University, College of Veterinary Medicine, Department of Population Medicine and Diagnostic Sciences, Ithaca, NY, USA
| | - Jay Lakshman
- Novartis Pharmaceutical Corporation, East Hanover, NJ, USA
| | - Colin S Osborne
- Global Health, Biomedical Research, Novartis, Emeryville, CA, USA
| | - Jennifer A Zambriski
- Department of Population Health Sciences, College of Veterinary Medicine, Center for One Health Research, Blacksburg, VA, USA
- Veterinarians for Global Solutions, Washington, DC, USA
| | - Natasha Aziz
- Global Health, Biomedical Research, Novartis, Emeryville, CA, USA
- Genentech Research and Early Development, South San Francisco, CA, USA
| | | | | |
Collapse
|
2
|
Caldwell N, Peet C, Miller P, Colon BL, Taylor MG, Cocco M, Dawson A, Lukac I, Teixeira JE, Robinson L, Frame L, Seizova S, Damerow S, Tamaki F, Post J, Riley J, Mutter N, Hanna JC, Ferguson L, Hu X, Tinti M, Forte B, Norcross NR, Campbell PS, Svensen N, Caldwell FC, Jansen C, Postis V, Read KD, Huston CD, Gilbert IH, Baragaña B, Pawlowic MC. Cryptosporidium lysyl-tRNA synthetase inhibitors define the interplay between solubility and permeability required to achieve efficacy. Sci Transl Med 2024; 16:eadm8631. [PMID: 39441903 DOI: 10.1126/scitranslmed.adm8631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 05/27/2024] [Accepted: 09/27/2024] [Indexed: 10/25/2024]
Abstract
Cryptosporidiosis is a diarrheal disease caused by infection with Cryptosporidium spp. parasites and is a leading cause of death in malnourished children worldwide. The only approved treatment, nitazoxanide, has limited efficacy in this at-risk patient population. Additional safe therapeutics are urgently required to tackle this unmet medical need. However, the development of anti-cryptosporidial drugs is hindered by a lack of understanding of the optimal compound properties required to treat this gastrointestinal infection. To address this knowledge gap, a diverse set of potent lysyl-tRNA synthetase inhibitors was profiled to identify optimal physicochemical and pharmacokinetic properties required for efficacy in a chronic mouse model of infection. The results from this comprehensive study illustrated the importance of balancing solubility and permeability to achieve efficacy in vivo. Our results establish in vitro criteria for solubility and permeability that are predictive of compound efficacy in vivo to guide the optimization of anti-cryptosporidial drugs. Two compounds from chemically distinct series (DDD489 and DDD508) were identified as demonstrating superior efficacy and prioritized for further evaluation. Both compounds achieved marked parasite reduction in immunocompromised mouse models and a disease-relevant calf model of infection. On the basis of these promising data, these compounds have been selected for progression to preclinical safety studies, expanding the portfolio of potential treatments for this neglected infectious disease.
Collapse
Affiliation(s)
- Nicola Caldwell
- Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Caroline Peet
- Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Peter Miller
- Department of Medicine, University of Vermont, Larner College of Medicine, Burlington, VT 05401, USA
| | - Beatrice L Colon
- Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Malcolm G Taylor
- Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Mattia Cocco
- Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Alice Dawson
- Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Iva Lukac
- Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Jose E Teixeira
- Department of Medicine, University of Vermont, Larner College of Medicine, Burlington, VT 05401, USA
| | - Lee Robinson
- Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Laura Frame
- Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Simona Seizova
- Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Sebastian Damerow
- Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Fabio Tamaki
- Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - John Post
- Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Jennifer Riley
- Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Nicole Mutter
- Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Jack C Hanna
- Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Liam Ferguson
- Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Xiao Hu
- Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Michele Tinti
- Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Barbara Forte
- Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Neil R Norcross
- Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Peter S Campbell
- Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Nina Svensen
- Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Flora C Caldwell
- Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Chimed Jansen
- Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Vincent Postis
- Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Kevin D Read
- Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Christopher D Huston
- Department of Medicine, University of Vermont, Larner College of Medicine, Burlington, VT 05401, USA
| | - Ian H Gilbert
- Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Beatriz Baragaña
- Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Mattie C Pawlowic
- Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| |
Collapse
|
3
|
He W, Cui H, Li N, Guo Y, Zeng S, Feng Y, Xiao L, Xu R. Involvement of INS15 in the development and pathogenicity of the zoonotic pathogen Cryptosporidium parvum. PLoS Negl Trop Dis 2024; 18:e0012569. [PMID: 39361715 PMCID: PMC11478815 DOI: 10.1371/journal.pntd.0012569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 10/15/2024] [Accepted: 09/24/2024] [Indexed: 10/05/2024] Open
Abstract
BACKGROUND Cryptosporidium parvum is a common protozoan pathogen responsible for moderate to severe diarrhea in humans and animals. The C. parvum genome contains 22 genes encoding insulinase-like M16 proteases (INS) with diverse structures and sequences, suggesting that members of the protein family may have distinct biological functions in the life cycle of parasites. Here, we investigated the role of INS15 and INS16, two proteases encoded by neighboring genes with high sequence identity, in the growth and development of C. parvum in vivo and in vitro. METHODOLOGY/PRINCIPAL FINDINGS INS15 and INS16 genes were tagged and knocked out using CRISPR/Cas9 technology in C. parvum IIdA20G1-HLJ isolate. The expression of INS15 and INS16 was determined by immunofluorescence analysis and immunoelectron microscopy. The effect of depletion of INS15 and INS16 on parasite growth and pathogenicity were assessed on HCT-8 cells and in interferon-γ knockout mice. Endogenous tagging showed that INS15 and INS16 expressed in the oocyst, trophozoite, meront and female gametes. INS15 also expressed in male gamonts, while INS16 was not detected in the male gamonts. Although depletion of the INS15 or INS16 gene affected late development of C. parvum in vitro, only depletion of INS15 significantly reduced parasite burden in infected mice. Mice infected with the INS15-depleted strain had reduced clinical signs, body weight, intestinal villus length to crypt height ratio, and survival time compared to infected with the tagging mutant. CONCLUSIONS/SIGNIFICANCE The results of this study indicate that INS15 is mainly involved in the late development of C. parvum. Depletion of this gene attenuates the pathogenicity of this important zoonotic parasite.
Collapse
Affiliation(s)
- Wei He
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
- School of Biology and Agriculture, Shaoguan University, Shaoguan, China
| | - Hao Cui
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
| | - Na Li
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
| | - Yaqiong Guo
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
| | - Songrong Zeng
- School of Biology and Agriculture, Shaoguan University, Shaoguan, China
| | - Yaoyu Feng
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
| | - Lihua Xiao
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
| | - Rui Xu
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
| |
Collapse
|
4
|
Tottey J, Etienne-Mesmin L, Chalançon S, Sausset A, Denis S, Mazal C, Blavignac C, Sallé G, Laurent F, Blanquet-Diot S, Lacroix-Lamandé S. Exploring the impact of digestive physicochemical parameters of adults and infants on the pathophysiology of Cryptosporidium parvum using the dynamic TIM-1 gastrointestinal model. Gut Pathog 2024; 16:55. [PMID: 39354600 PMCID: PMC11443851 DOI: 10.1186/s13099-024-00648-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 09/23/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUND Human cryptosporidiosis is distributed worldwide, and it is recognised as a leading cause of acute diarrhoea and death in infants in low- and middle-income countries. Besides immune status, the higher incidence and severity of this gastrointestinal disease in young children could also be attributed to the digestive environment. For instance, human gastrointestinal physiology undergoes significant changes with age, however the role this variability plays in Cryptosporidium parvum pathogenesis is not known. In this study, we analysed for the first time the impact of digestive physicochemical parameters on C. parvum infection in a human and age-dependent context using a dynamic in vitro gastrointestinal model. RESULTS Our results showed that the parasite excystation, releasing sporozoites from oocysts, occurs in the duodenum compartment after one hour of digestion in both child (from 6 months to 2 years) and adult experimental conditions. In the child small intestine, slightly less sporozoites were released from excystation compared to adult, however they exhibited a higher luciferase activity, suggesting a better physiological state. Sporozoites collected from the child jejunum compartment also showed a higher ability to invade human intestinal epithelial cells compared to the adult condition. Global analysis of the parasite transcriptome through RNA-sequencing demonstrated a more pronounced modulation in ileal effluents compared to gastric ones, albeit showing less susceptibility to age-related digestive condition. Further analysis of gene expression and enriched pathways showed that oocysts are highly active in protein synthesis in the stomach compartment, whereas sporozoites released in the ileum showed downregulation of glycolysis as well as strong modulation of genes potentially related to gliding motility and secreted effectors. CONCLUSIONS Digestion in a sophisticated in vitro gastrointestinal model revealed that invasive sporozoite stages are released in the small intestine, and are highly abundant and active in the ileum compartment, supporting reported C. parvum tissue tropism. Our comparative analysis suggests that physicochemical parameters encountered in the child digestive environment can influence the amount, physiological state and possibly invasiveness of sporozoites released in the small intestine, thus potentially contributing to the higher susceptibility of young individuals to cryptosporidiosis.
Collapse
Affiliation(s)
- Julie Tottey
- UMR 1282 ISP, Infectiologie et Santé Publique, INRAE, Université de Tours, Nouzilly, France.
| | - Lucie Etienne-Mesmin
- UMR 454 MEDIS, Microbiologie Environnement Digestif et Santé, Université Clermont Auvergne, INRAE, Clermont-Ferrand, France
| | - Sandrine Chalançon
- UMR 454 MEDIS, Microbiologie Environnement Digestif et Santé, Université Clermont Auvergne, INRAE, Clermont-Ferrand, France
| | - Alix Sausset
- UMR 1282 ISP, Infectiologie et Santé Publique, INRAE, Université de Tours, Nouzilly, France
| | - Sylvain Denis
- UMR 454 MEDIS, Microbiologie Environnement Digestif et Santé, Université Clermont Auvergne, INRAE, Clermont-Ferrand, France
| | - Carine Mazal
- UMR 454 MEDIS, Microbiologie Environnement Digestif et Santé, Université Clermont Auvergne, INRAE, Clermont-Ferrand, France
| | - Christelle Blavignac
- Centre Imagerie Cellulaire Santé, Université Clermont Auvergne, Clermont- Ferrand, France
| | - Guillaume Sallé
- UMR 1282 ISP, Infectiologie et Santé Publique, INRAE, Université de Tours, Nouzilly, France
| | - Fabrice Laurent
- UMR 1282 ISP, Infectiologie et Santé Publique, INRAE, Université de Tours, Nouzilly, France
| | - Stéphanie Blanquet-Diot
- UMR 454 MEDIS, Microbiologie Environnement Digestif et Santé, Université Clermont Auvergne, INRAE, Clermont-Ferrand, France
| | - Sonia Lacroix-Lamandé
- UMR 1282 ISP, Infectiologie et Santé Publique, INRAE, Université de Tours, Nouzilly, France
| |
Collapse
|
5
|
Ajiboye J, Teixeira JE, Gasonoo M, Mattice EB, Korwin-Mihavics B, Miller P, Cameron AC, Stebbins E, Campbell SD, Griggs DW, Spangenberg T, Meyers MJ, Huston CD. Identification of potent and orally efficacious phosphodiesterase inhibitors in Cryptosporidium parvum-infected immunocompromised male mice. Nat Commun 2024; 15:8272. [PMID: 39333545 PMCID: PMC11436873 DOI: 10.1038/s41467-024-52658-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 09/17/2024] [Indexed: 09/29/2024] Open
Abstract
Cryptosporidium parvum and C. hominis are parasites that cause life-threatening diarrhea in children and immunocompromised people. There is only one approved treatment that is modestly effective for children and ineffective for AIDS patients. Here, screening 278 compounds from the Merck KGaA, Darmstadt, Germany collection and accelerated follow-up enabled by prior investigation of the compounds identifies a series of pyrazolopyrimidine human phosphodiesterase (PDE)-V (hPDE-V) inhibitors with potent anticryptosporidial activity and efficacy following oral administration in C. parvum-infected male mice. The lead compounds affect parasite host cell egress, inhibit both C. parvum and C. hominis, work rapidly, and have minimal off-target effects in a safety screening panel. Interestingly, the hPDE-V inhibitors sildenafil and the 4-aminoquinoline compound 7a do not affect Cryptosporidium. C. parvum expresses one PDE (CpPDE1) continuously during asexual growth, the inhibited life stage. According to homology modeling and docking, the lead compounds interact with CpPDE1. Bulkier amino acids (Val900 and His884) in the CpPDE1 active site replace alanines in hPDE-V and block sildenafil binding. Supporting this, sildenafil kills a CRISPR-engineered Cryptosporidium CpPDE1 V900A mutant. The CpPDE1 mutation also alters parasite susceptibility to pyrazolopyrimidines. CpPDE1 is therefore a validated pyrazolopyrimidine molecular target to exploit for target-based optimization for improved anticryptosporidial development.
Collapse
Affiliation(s)
- Jubilee Ajiboye
- Department of Medicine, University of Vermont Larner College of Medicine, Room 202 Stafford Hall, 95 Carrigan Drive, Burlington, Vermont, USA
| | - José E Teixeira
- Department of Medicine, University of Vermont Larner College of Medicine, Room 202 Stafford Hall, 95 Carrigan Drive, Burlington, Vermont, USA
| | - Makafui Gasonoo
- Department of Chemistry, Saint Louis University, Room 206 Monsanto Hall, 3501 Laclede Avenue, Saint Louis, MO, USA
| | - Ethan B Mattice
- Department of Medicine, University of Vermont Larner College of Medicine, Room 202 Stafford Hall, 95 Carrigan Drive, Burlington, Vermont, USA
| | - Bethany Korwin-Mihavics
- Department of Medicine, University of Vermont Larner College of Medicine, Room 202 Stafford Hall, 95 Carrigan Drive, Burlington, Vermont, USA
| | - Peter Miller
- Department of Medicine, University of Vermont Larner College of Medicine, Room 202 Stafford Hall, 95 Carrigan Drive, Burlington, Vermont, USA
| | - Alexandra C Cameron
- Department of Medicine, University of Vermont Larner College of Medicine, Room 202 Stafford Hall, 95 Carrigan Drive, Burlington, Vermont, USA
| | - Erin Stebbins
- Department of Medicine, University of Vermont Larner College of Medicine, Room 202 Stafford Hall, 95 Carrigan Drive, Burlington, Vermont, USA
| | - Scott D Campbell
- Department of Molecular Microbiology and Immunology, Saint Louis University, Room 316 Doisy Research Center, 1100 South Grand Boulevard, Saint Louis, MO, USA
| | - David W Griggs
- Department of Molecular Microbiology and Immunology, Saint Louis University, Room 316 Doisy Research Center, 1100 South Grand Boulevard, Saint Louis, MO, USA
| | - Thomas Spangenberg
- Global Health R&D of Merck Healthcare, Ares Trading S.A. (a subsidiary of Merck KGaA, Darmstadt, Germany), Route de Crassier 1, Eysins, Switzerland
| | - Marvin J Meyers
- Department of Chemistry, Saint Louis University, Room 206 Monsanto Hall, 3501 Laclede Avenue, Saint Louis, MO, USA.
| | - Christopher D Huston
- Department of Medicine, University of Vermont Larner College of Medicine, Room 202 Stafford Hall, 95 Carrigan Drive, Burlington, Vermont, USA.
| |
Collapse
|
6
|
Ali DH, Gaji RY. TKL family kinases in human apicomplexan pathogens. Mol Biochem Parasitol 2024; 259:111628. [PMID: 38719028 PMCID: PMC11182715 DOI: 10.1016/j.molbiopara.2024.111628] [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: 01/18/2024] [Revised: 03/28/2024] [Accepted: 04/30/2024] [Indexed: 05/18/2024]
Abstract
Apicomplexan parasites are the primary causative agents of many human diseases, including malaria, toxoplasmosis, and cryptosporidiosis. These opportunistic pathogens undergo complex life cycles with multiple developmental stages, wherein many key steps are regulated by phosphorylation mechanisms. The genomes of apicomplexan pathogens contain protein kinases from different groups including tyrosine kinase-like (TKL) family proteins. Although information on the role of TKL kinases in apicomplexans is quite limited, recent studies have revealed the important role of this family of proteins in apicomplexan biology. TKL kinases in these protozoan pathogens show unique organization with many novel domains thus making them attractive candidates for drug development. In this mini review, we summarize the current understanding of the role of TKL kinases in human apicomplexan pathogens' (Toxoplasma gondii, Plasmodium falciparum and Cryptosporidium parvum) biology and pathogenesis.
Collapse
Affiliation(s)
- Dima Hajj Ali
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, USA
| | - Rajshekhar Y Gaji
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, USA.
| |
Collapse
|
7
|
Haase R, Puthenpurackal A, Maco B, Guérin A, Soldati-Favre D. γ-tubulin complex controls the nucleation of tubulin-based structures in Apicomplexa. Mol Biol Cell 2024; 35:ar121. [PMID: 39046777 PMCID: PMC11449391 DOI: 10.1091/mbc.e24-03-0100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/26/2024] [Accepted: 07/18/2024] [Indexed: 07/25/2024] Open
Abstract
Apicomplexan parasites rely on tubulin structures throughout their cell and life cycles, particularly in the polymerization of spindle microtubules to separate the replicated nucleus into daughter cells. Additionally, tubulin structures, including conoid and subpellicular microtubules, provide the necessary rigidity and structure for dissemination and host cell invasion. However, it is unclear whether these tubulin structures are nucleated via a highly conserved γ-tubulin complex or through a specific process unique to apicomplexans. This study demonstrates that Toxoplasma γ-tubulin is responsible for nucleating spindle microtubules, akin to higher eukaryotes, facilitating nucleus division in newly formed parasites. Interestingly, γ-tubulin colocalizes with nascent conoid and subpellicular microtubules during division, potentially nucleating these structures as well. Loss of γ-tubulin results in significant morphological defects due to impaired nucleus scission and the loss of conoid and subpellicular microtubule nucleation, crucial for parasite shape and rigidity. Additionally, the nucleation process of tubulin structures involves a concerted action of γ-tubulin and Gamma Tubulin Complex proteins (GCPs), recapitulating the localization and phenotype of γ-tubulin. This study also introduces new molecular markers for cytoskeletal structures and applies iterative expansion microscopy to reveal microtubule-based architecture in Cryptosporidium parvum sporozoites, further demonstrating the conserved localization and probable function of γ-tubulin in Cryptosporidium.
Collapse
Affiliation(s)
- Romuald Haase
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, CH-1221 Geneva 4, Switzerland
| | - Annet Puthenpurackal
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, CH-1221 Geneva 4, Switzerland
| | - Bohumil Maco
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, CH-1221 Geneva 4, Switzerland
| | - Amandine Guérin
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, CH-1221 Geneva 4, Switzerland
| | - Dominique Soldati-Favre
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, CH-1221 Geneva 4, Switzerland
| |
Collapse
|
8
|
Shaw S, Striepen B. Why is Parasite Sex Sexy? J Infect Dis 2024; 230:278-280. [PMID: 38813921 DOI: 10.1093/infdis/jiae277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 05/28/2024] [Indexed: 05/31/2024] Open
Affiliation(s)
- Sebastian Shaw
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 380 South University Avenue, Philadelphia, Pennsylvania 19104, USA
| | - Boris Striepen
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 380 South University Avenue, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
9
|
Wallbank BA, Pardy RD, Brodsky IE, Hunter CA, Striepen B. Cryptosporidium impacts epithelial turnover and is resistant to induced death of the host cell. mBio 2024; 15:e0172024. [PMID: 38995074 PMCID: PMC11323733 DOI: 10.1128/mbio.01720-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 06/13/2024] [Indexed: 07/13/2024] Open
Abstract
Infection with the apicomplexan parasite Cryptosporidium is a leading cause of diarrheal disease. Cryptosporidiosis is of particular importance in infants and shows a strong association with malnutrition, both as a risk factor and as a consequence. Cryptosporidium invades and replicates within the small intestine epithelial cells. This is a highly dynamic tissue that is developmentally stratified along the villus axis. New cells emerge from a stem cell niche in the crypt and differentiate into mature epithelial cells while moving toward the villus tip, where they are ultimately shed. Here, we studied the impact of Cryptosporidium infection on this dynamic architecture. Tracing DNA synthesis in pulse-chase experiments in vivo, we quantified the genesis and migration of epithelial cells along the villus. We found proliferation and epithelial migration to be elevated in response to Cryptosporidium infection. Infection also resulted in significant cell loss documented by imaging and molecular assays. Consistent with these observations, single-cell RNA sequencing of infected intestines showed a gain of young and a loss of mature cells. Interestingly, enhanced epithelial cell loss was not a function of enhanced apoptosis of infected cells. To the contrary, Cryptosporidium-infected cells were less likely to be apoptotic than bystanders, and experiments in tissue culture demonstrated that infection provided enhanced resistance to chemically induced apoptosis to the host but not bystander cells. Overall, this study suggests that Cryptosporidium may modulate cell apoptosis and documents pronounced changes in tissue homeostasis due to parasite infection, which may contribute to its long-term impact on the developmental and nutritional state of children. IMPORTANCE The intestine must balance its roles in digestion and nutrient absorption with the maintenance of an effective barrier to colonization and breach by numerous potential pathogens. An important component of this balance is its constant turnover, which is modulated by a gain of cells due to proliferation and loss due to death or extrusion. Here, we report that Cryptosporidium infection changes the dynamics of this process increasing both gain and loss of enterocytes speeding up the villus elevator. This leads to a much more immature epithelium and a reduction of the number of those cells typically found toward the villus apex best equipped to take up key nutrients including carbohydrates and lipids. These changes in the cellular architecture and physiology of the small intestine may be linked to the profound association between cryptosporidiosis and malnutrition.
Collapse
Affiliation(s)
- Bethan A. Wallbank
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ryan D. Pardy
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Igor E. Brodsky
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Christopher A. Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Boris Striepen
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
10
|
Nava MG, Szewczyk J, Arrington JV, Alam T, Vinayak S. The Cryptosporidium signaling kinase CDPK5 plays an important role in male gametogenesis and parasite virulence. Cell Rep 2024; 43:114263. [PMID: 38814783 PMCID: PMC11312397 DOI: 10.1016/j.celrep.2024.114263] [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: 10/03/2023] [Revised: 04/02/2024] [Accepted: 05/07/2024] [Indexed: 06/01/2024] Open
Abstract
The protozoan parasite Cryptosporidium is a leading cause of diarrhea in young children. The parasite's life cycle involves a coordinated and timely progression from asexual to sexual stages, leading to the formation of the transmissible oocyst. Underlying molecular signaling mechanisms orchestrating sexual development are not known. Here, we describe the function of a signaling kinase in Cryptosporidium male gametogenesis. We reveal the expression of Cryptosporidium parvum calcium-dependent protein kinase 5 (CDPK5) during male gamete development and its important role in the egress of mature gametes. Genetic ablation of this kinase results in viable parasites, indicating that this gene is dispensable for parasite survival. Interestingly, cdpk5 deletion decreases parasite virulence and impacts oocyst shedding in immunocompromised mice. Using phosphoproteomics, we identify possible CDPK5 substrates and biological processes regulated by this kinase. Collectively, these findings illuminate parasite cell biology by revealing a mechanism controlling male gamete production and a potential target to block disease transmission.
Collapse
Affiliation(s)
- Maria G Nava
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61802, USA
| | - Joanna Szewczyk
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61802, USA
| | - Justine V Arrington
- Proteomics Core Facility, Roy J. Carver Biotechnology Center, University of Illinois at Urbana-Champaign, Urbana, IL 61802, USA
| | - Tauqeer Alam
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61802, USA
| | - Sumiti Vinayak
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61802, USA.
| |
Collapse
|
11
|
Walzer KA, Tandel J, Byerly JH, Daniels AM, Gullicksrud JA, Whelan EC, Carro SD, Krespan E, Beiting DP, Striepen B. Transcriptional control of the Cryptosporidium life cycle. Nature 2024; 630:174-180. [PMID: 38811723 DOI: 10.1038/s41586-024-07466-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 04/25/2024] [Indexed: 05/31/2024]
Abstract
The parasite Cryptosporidium is a leading agent of diarrhoeal disease in young children, and a cause and consequence of chronic malnutrition1,2. There are no vaccines and only limited treatment options3. The parasite infects enterocytes, in which it engages in asexual and sexual replication4, both of which are essential to continued infection and transmission. However, their molecular mechanisms remain largely unclear5. Here we use single-cell RNA sequencing to reveal the gene expression programme of the entire Cryptosporidium parvum life cycle in culture and in infected animals. Diverging from the prevailing model6, we find support for only three intracellular stages: asexual type-I meronts, male gamonts and female gametes. We reveal a highly organized program for the assembly of components at each stage. Dissecting the underlying regulatory network, we identify the transcription factor Myb-M as the earliest determinant of male fate, in an organism that lacks genetic sex determination. Conditional expression of this factor overrides the developmental program and induces widespread maleness, while conditional deletion ablates male development. Both have a profound impact on the infection. A large set of stage-specific genes now provides the opportunity to understand, engineer and disrupt parasite sex and life cycle progression to advance the development of vaccines and treatments.
Collapse
Affiliation(s)
- Katelyn A Walzer
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jayesh Tandel
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jessica H Byerly
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Abigail M Daniels
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jodi A Gullicksrud
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Eoin C Whelan
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Stephen D Carro
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elise Krespan
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel P Beiting
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Boris Striepen
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
12
|
Kimball A, Funkhouser-Jones L, Huang W, Xu R, Witola WH, Sibley LD. Mendelian segregation and high recombination rates facilitate genetic analyses in Cryptosporidium parvum. PLoS Genet 2024; 20:e1011162. [PMID: 38885280 PMCID: PMC11213348 DOI: 10.1371/journal.pgen.1011162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 06/28/2024] [Accepted: 05/27/2024] [Indexed: 06/20/2024] Open
Abstract
Very little is known about the process of meiosis in the apicomplexan parasite Cryptosporidium despite the essentiality of sex in its life cycle. Most cell lines only support asexual growth of Cryptosporidium parvum (C. parvum), but stem cell derived intestinal epithelial cells grown under air-liquid interface (ALI) conditions support the sexual cycle. To examine chromosomal dynamics during meiosis in C. parvum, we generated two transgenic lines of parasites that were fluorescently tagged with mCherry or GFP on chromosomes 1 or 5, respectively. Infection of ALI cultures or Ifngr1-/- mice with mCherry and GFP parasites resulted in cross-fertilization and the formation of "yellow" oocysts, which contain 4 haploid sporozoites that are the product of meiosis. Recombinant oocysts from the F1 generation were purified and used to infect HCT-8 cultures, and phenotypes of the progeny were observed by microscopy. All possible phenotypes predicted by independent segregation were represented equally (~25%) in the population, indicating that C. parvum chromosomes exhibit a Mendelian inheritance pattern. The most common pattern observed from the outgrowth of single oocysts included all possible parental and recombinant phenotypes derived from a single meiotic event, suggesting a high rate of crossover. To estimate the frequency of crossover, additional loci on chromosomes 1 and 5 were tagged and used to monitor intrachromosomal crosses in Ifngr1-/- mice. Both chromosomes showed a high frequency of crossover compared to other apicomplexans with map distances (i.e., 1% recombination) of 3-12 kb. Overall, a high recombination rate may explain many unique characteristics observed in Cryptosporidium spp. such as high rates of speciation, wide variation in host range, and rapid evolution of host-specific virulence factors.
Collapse
Affiliation(s)
- Abigail Kimball
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Lisa Funkhouser-Jones
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Wanyi Huang
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Rui Xu
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - William H. Witola
- Department of Pathobiology, University of Illinois Urbana-Champaign College of Veterinary Medicine, Urbana, Illinois, United States of America
| | - L. David Sibley
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| |
Collapse
|
13
|
Pardy RD, Walzer KA, Wallbank BA, Byerly JH, O’Dea KM, Cohn IS, Haskins BE, Roncaioli JL, Smith EJ, Buenconsejo GY, Striepen B, Hunter CA. Analysis of intestinal epithelial cell responses to Cryptosporidium highlights the temporal effects of IFN-γ on parasite restriction. PLoS Pathog 2024; 20:e1011820. [PMID: 38718306 PMCID: PMC11078546 DOI: 10.1371/journal.ppat.1011820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 04/14/2024] [Indexed: 05/12/2024] Open
Abstract
The production of IFN-γ is crucial for control of multiple enteric infections, but its impact on intestinal epithelial cells (IEC) is not well understood. Cryptosporidium parasites exclusively infect epithelial cells and the ability of interferons to activate the transcription factor STAT1 in IEC is required for parasite clearance. Here, the use of single cell RNA sequencing to profile IEC during infection revealed an increased proportion of mid-villus enterocytes during infection and induction of IFN-γ-dependent gene signatures that was comparable between uninfected and infected cells. These analyses were complemented by in vivo studies, which demonstrated that IEC expression of the IFN-γ receptor was required for parasite control. Unexpectedly, treatment of Ifng-/- mice with IFN-γ showed the IEC response to this cytokine correlates with a delayed reduction in parasite burden but did not affect parasite development. These data sets provide insight into the impact of IFN-γ on IEC and suggest a model in which IFN-γ signalling to uninfected enterocytes is important for control of Cryptosporidium.
Collapse
Affiliation(s)
- Ryan D. Pardy
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Katelyn A. Walzer
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Bethan A. Wallbank
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jessica H. Byerly
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Keenan M. O’Dea
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Ian S. Cohn
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Breanne E. Haskins
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Justin L. Roncaioli
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Eleanor J. Smith
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Gracyn Y. Buenconsejo
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Boris Striepen
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Christopher A. Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
14
|
Liffner B, Absalon S. Expansion microscopy of apicomplexan parasites. Mol Microbiol 2024; 121:619-635. [PMID: 37571814 DOI: 10.1111/mmi.15135] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 07/15/2023] [Accepted: 07/20/2023] [Indexed: 08/13/2023]
Abstract
Apicomplexan parasites comprise significant pathogens of humans, livestock and wildlife, but also represent a diverse group of eukaryotes with interesting and unique cell biology. The study of cell biology in apicomplexan parasites is complicated by their small size, and historically this has required the application of cutting-edge microscopy techniques to investigate fundamental processes like mitosis or cell division in these organisms. Recently, a technique called expansion microscopy has been developed, which rather than increasing instrument resolution like most imaging modalities, physically expands a biological sample. In only a few years since its development, a derivative of expansion microscopy known as ultrastructure-expansion microscopy (U-ExM) has been widely adopted and proven extremely useful for studying cell biology of Apicomplexa. Here, we review the insights into apicomplexan cell biology that have been enabled through the use of U-ExM, with a specific focus on Plasmodium, Toxoplasma and Cryptosporidium. Further, we summarize emerging expansion microscopy modifications and modalities and forecast how these may influence the field of parasite cell biology in future.
Collapse
Affiliation(s)
- Benjamin Liffner
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Sabrina Absalon
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
15
|
Hasan MM, Mattice EB, Teixeira JE, Jumani RS, Stebbins EE, Klopfer CE, Franco SE, Love MS, McNamara CW, Huston CD. Cryptosporidium life cycle small molecule probing implicates translational repression and an Apetala 2 transcription factor in macrogamont differentiation. PLoS Pathog 2024; 20:e1011906. [PMID: 38669269 PMCID: PMC11078545 DOI: 10.1371/journal.ppat.1011906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 05/08/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
The apicomplexan parasite Cryptosporidium is a leading cause of childhood diarrhea in developing countries. Current treatment options are inadequate and multiple preclinical compounds are being actively pursued as potential drugs for cryptosporidiosis. Unlike most apicomplexans, Cryptosporidium spp. sequentially replicate asexually and then sexually within a single host to complete their lifecycles. Anti-cryptosporidial compounds are generally identified or tested through in vitro phenotypic assays that only assess the asexual stages. Therefore, compounds that specifically target the sexual stages remain unexplored. In this study, we leveraged the ReFRAME drug repurposing library against a newly devised multi-readout imaging assay to identify small-molecule compounds that modulate macrogamont differentiation and maturation. RNA-seq studies confirmed selective modulation of macrogamont differentiation for 10 identified compounds (9 inhibitors and 1 accelerator). The collective transcriptomic profiles of these compounds indicates that translational repression accompanies Cryptosporidium sexual differentiation, which we validated experimentally. Additionally, cross comparison of the RNA-seq data with promoter sequence analysis for stage-specific genes converged on a key role for an Apetala 2 (AP2) transcription factor (cgd2_3490) in differentiation into macrogamonts. Finally, drug annotation for the ReFRAME hits indicates that an elevated supply of energy equivalence in the host cell is critical for macrogamont formation.
Collapse
Affiliation(s)
- Muhammad M. Hasan
- Department of Medicine, University of Vermont Larner College of Medicine, Burlington, Vermont, United States of America
- Cell, Molecular, and Biomedical Sciences Graduate Program, University of Vermont, Burlington, Vermont, United States of America
| | - Ethan B. Mattice
- Department of Medicine, University of Vermont Larner College of Medicine, Burlington, Vermont, United States of America
- Cell, Molecular, and Biomedical Sciences Graduate Program, University of Vermont, Burlington, Vermont, United States of America
| | - José E. Teixeira
- Department of Medicine, University of Vermont Larner College of Medicine, Burlington, Vermont, United States of America
| | - Rajiv S. Jumani
- Department of Medicine, University of Vermont Larner College of Medicine, Burlington, Vermont, United States of America
- Cell, Molecular, and Biomedical Sciences Graduate Program, University of Vermont, Burlington, Vermont, United States of America
| | - Erin E. Stebbins
- Department of Medicine, University of Vermont Larner College of Medicine, Burlington, Vermont, United States of America
| | - Connor E. Klopfer
- Department of Medicine, University of Vermont Larner College of Medicine, Burlington, Vermont, United States of America
| | - Sebastian E. Franco
- Department of Medicine, University of Vermont Larner College of Medicine, Burlington, Vermont, United States of America
| | - Melissa S. Love
- Calibr at Scripps Research, San Diego, California, United States of America
| | - Case W. McNamara
- Calibr at Scripps Research, San Diego, California, United States of America
| | - Christopher D. Huston
- Department of Medicine, University of Vermont Larner College of Medicine, Burlington, Vermont, United States of America
- Cell, Molecular, and Biomedical Sciences Graduate Program, University of Vermont, Burlington, Vermont, United States of America
| |
Collapse
|
16
|
Agyabeng-Dadzie F, Xiao R, Kissinger JC. Cryptosporidium Genomics - Current Understanding, Advances, and Applications. CURRENT TROPICAL MEDICINE REPORTS 2024; 11:92-103. [PMID: 38813571 PMCID: PMC11130048 DOI: 10.1007/s40475-024-00318-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2024] [Indexed: 05/31/2024]
Abstract
Purpose of Review Here we highlight the significant contribution that genomics-based approaches have had on the field of Cryptosporidium research and the insights these approaches have generated into Cryptosporidium biology and transmission. Recent Findings There are advances in genomics, genetic manipulation, gene expression, and single-cell technologies. New and better genome sequences have revealed variable sub-telomeric gene families and genes under selection. RNA expression data now include single-cell and post-infection time points. These data have provided insights into the Cryptosporidium life cycle and host-pathogen interactions. Antisense and ncRNA transcripts are abundant. The critical role of the dsRNA virus is becoming apparent. Summary The community's ability to identify genomic targets in the abundant, yet still lacking, collection of genomic data, combined with their increased ability to assess function via gene knock-out, is revolutionizing the field. Advances in the detection of virulence genes, surveillance, population genomics, recombination studies, and epigenetics are upon us.
Collapse
Affiliation(s)
| | - Rui Xiao
- Institute of Bioinformatics, University of Georgia, Athens, GA 30602 USA
| | - Jessica C. Kissinger
- Department of Genetics, University of Georgia, Athens, GA 30602 USA
- Institute of Bioinformatics, University of Georgia, Athens, GA 30602 USA
- Center for Tropical and Emerging Global Diseases, University of Georgia, Coverdell Center, 107, 500 D.W. Brooks Drive, Athens, GA 30602 USA
| |
Collapse
|
17
|
Moss WJ, Brusini L, Kuehnel R, Brochet M, Brown KM. Apicomplexan phosphodiesterases in cyclic nucleotide turnover: conservation, function, and therapeutic potential. mBio 2024; 15:e0305623. [PMID: 38132724 PMCID: PMC10865986 DOI: 10.1128/mbio.03056-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023] Open
Abstract
Apicomplexa encompasses a large number of intracellular parasites infecting a wide range of animals. Cyclic nucleotide signaling is crucial for a variety of apicomplexan life stages and cellular processes. The cyclases and kinases that synthesize and respond to cyclic nucleotides (i.e., 3',5'-cyclic guanosine monophosphate and 3',5'-cyclic adenosine monophosphate) are highly conserved and essential throughout the parasite phylum. Growing evidence indicates that phosphodiesterases (PDEs) are also critical for regulating cyclic nucleotide signaling via cyclic nucleotide hydrolysis. Here, we discuss recent advances in apicomplexan PDE biology and opportunities for therapeutic interventions, with special emphasis on the major human apicomplexan parasite genera Plasmodium, Toxoplasma, Cryptosporidium, and Babesia. In particular, we show a highly flexible repertoire of apicomplexan PDEs associated with a wide range of cellular requirements across parasites and lifecycle stages. Despite this phylogenetic diversity, cellular requirements of apicomplexan PDEs for motility, host cell egress, or invasion are conserved. However, the molecular wiring of associated PDEs is extremely malleable suggesting that PDE diversity and redundancy are key for the optimization of cyclic nucleotide turnover to respond to the various environments encountered by each parasite and life stage. Understanding how apicomplexan PDEs are regulated and integrating multiple signaling systems into a unified response represent an untapped avenue for future exploration.
Collapse
Affiliation(s)
- William J. Moss
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Lorenzo Brusini
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Ronja Kuehnel
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Mathieu Brochet
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Kevin M. Brown
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| |
Collapse
|
18
|
Kimball A, Funkhouser-Jones L, Huang W, Xu R, Witola WH, Sibley LD. Mendelian segregation and high recombination rates facilitate genetic analyses in Cryptosporidium parvum. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.02.578536. [PMID: 38352509 PMCID: PMC10862819 DOI: 10.1101/2024.02.02.578536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
Very little is known about the process of meiosis in the apicomplexan parasite Cryptosporidium despite the essentiality of sex in its life cycle. Most cell lines only support asexual growth of Cryptosporidium parvum (C. parvum), but stem cell derived intestinal epithelial cells grown under air-liquid interface (ALI) conditions support the sexual cycle. To examine chromosomal dynamics during meiosis in C. parvum, we generated two transgenic lines of parasites that were fluorescently tagged with mCherry or GFP on chromosomes 1 or 5, respectively. Infection of ALI cultures or Ifngr1-/- mice with mCherry and GFP parasites produced "yellow" oocysts generated by cross-fertilization. Outcrossed oocysts from the F1 generation were purified and used to infect HCT-8 cultures, and phenotypes of the progeny were observed by microscopy. All possible phenotypes predicted by independent segregation were represented equally (~25%) in the population, indicating that C. parvum chromosomes exhibit a Mendelian inheritance pattern. Unexpectedly, the most common pattern observed from the outgrowth of single oocysts included all possible parental and recombinant phenotypes derived from a single meiotic event, suggesting a high rate of crossover. To estimate the frequency of crossover, additional loci on chromosomes 1 and 5 were tagged and used to monitor intrachromosomal crosses in Ifngr1-/- mice. Both chromosomes showed a high frequency of crossover compared to other apicomplexans with map distances (i.e., 1% recombination) of 3-12 kb. Overall, a high recombination rate may explain many unique characteristics observed in Cryptosporidium spp. such as high rates of speciation, wide variation in host range, and rapid evolution of host-specific virulence factors.
Collapse
Affiliation(s)
- Abigail Kimball
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Lisa Funkhouser-Jones
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Wanyi Huang
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Rui Xu
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - William H. Witola
- Department of Pathobiology, University of Illinois Urbana-Champaign College of Veterinary Medicine, Urbana, IL 61802, USA
| | - L. David Sibley
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
19
|
Pardy RD, Wallbank BA, Striepen B, Hunter CA. Immunity to Cryptosporidium: insights into principles of enteric responses to infection. Nat Rev Immunol 2024; 24:142-155. [PMID: 37697084 DOI: 10.1038/s41577-023-00932-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/28/2023] [Indexed: 09/13/2023]
Abstract
Cryptosporidium parasites replicate within intestinal epithelial cells and are an important cause of diarrhoeal disease in young children and in patients with primary and acquired defects in T cell function. This Review of immune-mediated control of Cryptosporidium highlights advances in understanding how intestinal epithelial cells detect this infection, the induction of innate resistance and the processes required for activation of T cell responses that promote parasite control. The development of a genetic tool set to modify Cryptosporidium combined with tractable mouse models provide new opportunities to understand the principles that govern the interface between intestinal epithelial cells and the immune system that mediate resistance to enteric pathogens.
Collapse
Affiliation(s)
- Ryan D Pardy
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bethan A Wallbank
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Boris Striepen
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Christopher A Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
20
|
Shaw S, Cohn IS, Baptista RP, Xia G, Melillo B, Agyabeng-Dadzie F, Kissinger JC, Striepen B. Genetic crosses within and between species of Cryptosporidium. Proc Natl Acad Sci U S A 2024; 121:e2313210120. [PMID: 38147547 PMCID: PMC10769859 DOI: 10.1073/pnas.2313210120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 11/12/2023] [Indexed: 12/28/2023] Open
Abstract
Parasites and their hosts are engaged in reciprocal coevolution that balances competing mechanisms of virulence, resistance, and evasion. This often leads to host specificity, but genomic reassortment between different strains can enable parasites to jump host barriers and conquer new niches. In the apicomplexan parasite Cryptosporidium, genetic exchange has been hypothesized to play a prominent role in adaptation to humans. The sexual lifecycle of the parasite provides a potential mechanism for such exchange; however, the boundaries of Cryptosporidium sex are currently undefined. To explore this experimentally, we established a model for genetic crosses. Drug resistance was engineered using a mutated phenylalanyl tRNA synthetase gene and marking strains with this and the previously used Neo transgene enabled selection of recombinant progeny. This is highly efficient, and genomic recombination is evident and can be continuously monitored in real time by drug resistance, flow cytometry, and PCR mapping. Using this approach, multiple loci can now be modified with ease. We demonstrate that essential genes can be ablated by crossing a Cre recombinase driver strain with floxed strains. We further find that genetic crosses are also feasible between species. Crossing Cryptosporidium parvum, a parasite of cattle and humans, and Cryptosporidium tyzzeri a mouse parasite resulted in progeny with a recombinant genome derived from both species that continues to vigorously replicate sexually. These experiments have important fundamental and translational implications for the evolution of Cryptosporidium and open the door to reverse- and forward-genetic analysis of parasite biology and host specificity.
Collapse
Affiliation(s)
- Sebastian Shaw
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Ian S. Cohn
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Rodrigo P. Baptista
- Department of Medicine, Houston Methodist Research Institute, Houston, TX77030
| | - Guoqin Xia
- Department of Chemistry, Scripps Research, La Jolla, CA92037
| | - Bruno Melillo
- Department of Chemistry, Scripps Research, La Jolla, CA92037
- Chemical Biology and Therapeutics Science Program, Broad Institute, Cambridge, MA02142
| | | | - Jessica C. Kissinger
- Department of Genetics, University of Georgia, Athens, GA30602
- Center for Tropical and Emerging Global Diseases and Institute of Bioinformatics, University of Georgia, Athens, GA30602
| | - Boris Striepen
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA19104
| |
Collapse
|
21
|
Gunasekera S, Clode PL, King B, Monis P, Thierry B, Carr JM, Chopra A, Watson M, O'Dea M, Hijjawi N, Ryan U. Comparison of in vitro growth characteristics of Cryptosporidium hominis (IdA15G1) and Cryptosporidium parvum (Iowa-IIaA17G2R1 and IIaA18G3R1). Parasitol Res 2023; 122:2891-2905. [PMID: 37776335 PMCID: PMC10667462 DOI: 10.1007/s00436-023-07979-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 09/14/2023] [Indexed: 10/02/2023]
Abstract
Cryptosporidium is a major cause of diarrhoeal disease and mortality in young children in resource-poor countries, for which no vaccines or adequate therapeutic options are available. Infection in humans is primarily caused by two species: C. hominis and C. parvum. Despite C. hominis being the dominant species infecting humans in most countries, very little is known about its growth characteristics and life cycle in vitro, given that the majority of our knowledge of the in vitro development of Cryptosporidium has been based on C. parvum. In the present study, the growth and development of two C. parvum isolates (subtypes Iowa-IIaA17G2R1 and IIaA18G3R1) and one C. hominis isolate (subtype IdA15G1) in HCT-8 cells were examined and compared at 24 h and 48 h using morphological data acquired with scanning electron microscopy. Our data indicated no significant differences in the proportion of meronts or merozoites between species or subtypes at either time-point. Sexual development was observed at the 48-h time-point across both species through observations of both microgamonts and macrogamonts, with a higher frequency of macrogamont observations in C. hominis (IdA15G1) cultures at 48-h post-infection compared to both C. parvum subtypes. This corresponded to differences in the proportion of trophozoites observed at the same time point. No differences in proportion of microgamonts were observed between the three subtypes, which were rarely observed across all cultures. In summary, our data indicate that asexual development of C. hominis is similar to that of C. parvum, while sexual development is accelerated in C. hominis. This study provides new insights into differences in the in vitro growth characteristics of C. hominis when compared to C. parvum, which will facilitate our understanding of the sexual development of both species.
Collapse
Affiliation(s)
- Samantha Gunasekera
- Harry Butler Institute, College of Environmental and Life Sciences, Murdoch University, Murdoch, Western Australia, 6150, Australia.
| | - Peta L Clode
- Centre for Microscopy, Characterisation, and Analysis and School of Biological Sciences, The University of Western Australia, Crawley, Western Australia, 6009, Australia
| | - Brendon King
- South Australian Water Corporation, Adelaide, South Australia, 5000, Australia
| | - Paul Monis
- South Australian Water Corporation, Adelaide, South Australia, 5000, Australia
| | - Benjamin Thierry
- Future Industries Institute, University of South Australia, Adelaide, South Australia, 5095, Australia
| | - Jillian M Carr
- College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Bedford Park, South Australia, 5042, Australia
| | - Abha Chopra
- Immunology and Infectious Diseases, Murdoch University, Murdoch, Western Australia, 6150, Australia
| | - Mark Watson
- Immunology and Infectious Diseases, Murdoch University, Murdoch, Western Australia, 6150, Australia
| | - Mark O'Dea
- Harry Butler Institute, College of Environmental and Life Sciences, Murdoch University, Murdoch, Western Australia, 6150, Australia
| | - Nawal Hijjawi
- Department of Medical Laboratory Sciences, Faculty of Applied Health Sciences, The Hashemite University, P.O. Box 150459, Zarqa, 13115, Jordan
| | - Una Ryan
- Harry Butler Institute, College of Environmental and Life Sciences, Murdoch University, Murdoch, Western Australia, 6150, Australia.
| |
Collapse
|
22
|
Hurle GR, Brainard J, Tyler KM. Microbiome diversity is a modifiable virulence factor for cryptosporidiosis. Virulence 2023; 14:2273004. [PMID: 37872759 PMCID: PMC10653618 DOI: 10.1080/21505594.2023.2273004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 10/15/2023] [Indexed: 10/25/2023] Open
Abstract
Cryptosporidium spp. infection causes significant disease in immunosuppressed individuals and children under the age of 5 years. The severity of the pathological presentation of cryptosporidiosis is a function of the host and parasite genotypes, host immune status, and the enteric environment or microbiome of the host. Cryptosporidiosis often presents with abdominal pain and severe diarrhoea and is associated with intestinal dysbiosis and inflammation. Our systematic analysis of the available literature revealed that bacterial diversity is reduced during infection in larger animal models, lending support to recent studies which indicate that the use of probiotics or the presence of a naturally diverse gut microbiome can prevent or minimise pathology caused by gastrointestinal pathogens. In summary, we present evidence that the presence of a diverse gut microbiome, natural or induced, reduces both symptomatic pathology and oocyst output.
Collapse
Affiliation(s)
| | - Julii Brainard
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Kevin. M. Tyler
- Norwich Medical School, University of East Anglia, Norwich, UK
| |
Collapse
|
23
|
Pardy RD, Walzer KA, Wallbank BA, Byerly JH, O’Dea KM, Cohn IS, Haskins BE, Roncaioli JL, Smith EJ, Buenconsejo GY, Striepen B, Hunter CA. Analysis of intestinal epithelial cell responses to Cryptosporidium highlights the temporal effects of IFN-γ on parasite restriction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.14.567008. [PMID: 38014210 PMCID: PMC10680692 DOI: 10.1101/2023.11.14.567008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
The production of IFN-γ is crucial for control of multiple enteric infections, but its impact on intestinal epithelial cells (IEC) is not well understood. Cryptosporidium parasites exclusively infect epithelial cells and the ability of interferons to activate the transcription factor STAT1 in IEC is required for parasite clearance. The use of single cell RNA sequencing to profile IEC during infection revealed induction of IFN-γ-dependent gene signatures that was comparable between uninfected and infected cells, and IEC expression of the IFN-γ receptor was required for parasite control. Unexpectedly, treatment of Ifng-/- mice with IFN-γ demonstrated the IEC response to this cytokine correlates with a delayed reduction in parasite burden but did not affect parasite development. These data sets provide insight into the impact of IFN-γ on IEC and suggest a model in which IFN-γ-mediated bystander activation of uninfected enterocytes is important for control of Cryptosporidium.
Collapse
Affiliation(s)
- Ryan D. Pardy
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Katelyn A. Walzer
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bethan A. Wallbank
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jessica H. Byerly
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Keenan M. O’Dea
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ian S. Cohn
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Breanne E. Haskins
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Justin L. Roncaioli
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Eleanor J. Smith
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gracyn Y. Buenconsejo
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Boris Striepen
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christopher A. Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
24
|
Korwin-Mihavics BR, Dews EA, di Genova BM, Huston CD. Organoid-based in vitro systems to model Cryptosporidium parvum infection in 2D and 3D. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.29.560165. [PMID: 37808810 PMCID: PMC10557739 DOI: 10.1101/2023.09.29.560165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Typical cancer cell-based culture systems cannot support the full life cycle of Cryptosporidium parvum, despite its monoxenous life cycle which is completed in the small intestine of a single host. There is a block to fertilization and zygote formation in vitro. In this paper, we adapted a 2D organoid derived monolayer system and a 3D inverted enteroid system for use in C. parvum culture. 3D inverted enteroids were successfully infected by C. parvum without the need for microinjection and supported subculture of C. parvum. Using the 2D organoid derived monolayer (ODM) system, the infection can be maintained for at least 3 weeks with new oocyst production throughout. Fertilization was confirmed based on successful mating of two strains of C. parvum. We demonstrated that the apparent block to fertilization in typical cell culture is overcome using ODMs.
Collapse
Affiliation(s)
- Bethany R Korwin-Mihavics
- Department of Medicine, University of Vermont, Burlington, Vermont, USA
- Microbiology and Molecular Genetics, University of Vermont, Burlington, Vermont, USA
- Cellular, Molecular, and Biomedical Sciences, University of Vermont, Burlington, Vermont, USA
| | - Emmett A Dews
- Department of Medicine, University of Vermont, Burlington, Vermont, USA
- Microbiology and Molecular Genetics, University of Vermont, Burlington, Vermont, USA
| | - Bruno Martorelli di Genova
- Microbiology and Molecular Genetics, University of Vermont, Burlington, Vermont, USA
- Cellular, Molecular, and Biomedical Sciences, University of Vermont, Burlington, Vermont, USA
| | - Christopher D Huston
- Department of Medicine, University of Vermont, Burlington, Vermont, USA
- Microbiology and Molecular Genetics, University of Vermont, Burlington, Vermont, USA
- Cellular, Molecular, and Biomedical Sciences, University of Vermont, Burlington, Vermont, USA
| |
Collapse
|
25
|
Shaw S, Cohn IS, Baptista RP, Xia G, Melillo B, Agyabeng-Dadzie F, Kissinger JC, Striepen B. Genetic crosses within and between species of Cryptosporidium. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.04.551960. [PMID: 37577700 PMCID: PMC10418217 DOI: 10.1101/2023.08.04.551960] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Parasites and their hosts are engaged in rapid coevolution that balances competing mechanisms of virulence, resistance, and evasion. This often leads to host specificity, but genomic reassortment between different strains can enable parasites to jump host barriers and conquer new niches. In the apicomplexan parasite Cryptosporidium genetic exchange has been hypothesized to play a prominent role in adaptation to humans. The sexual lifecycle of the parasite provides a potential mechanism for such exchange; however, the boundaries of Cryptosporidium sex are currently undefined. To explore this experimentally, we established a model for genetic crosses. Drug resistance was engineered using a mutated phenylalanyl tRNA synthetase gene and marking strains with this and the previously used Neo transgene enabled selection of recombinant progeny. This is highly efficient, and genomic recombination is evident and can be continuously monitored in real time by drug resistance, flow cytometry, and PCR mapping. Using this approach multiple loci can now be modified with ease. We demonstrate that essential genes can be ablated by crossing a Cre recombinase driver strain with floxed strains. We further find that genetic crosses are also feasible between species. Crossing C. parvum, a parasite of cattle and humans, and C. tyzzeri a mouse parasite resulted in progeny with a recombinant genome derived from both species that continues to vigorously replicate sexually. These experiments have important fundamental and translational implications for the evolution of Cryptosporidium and open the door to reverse- and forward- genetic analysis of parasite biology and host specificity.
Collapse
Affiliation(s)
- Sebastian Shaw
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA
| | - Ian S. Cohn
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA
| | | | - Guoqin Xia
- Department of Chemistry, Scripps Research, La Jolla, CA
| | - Bruno Melillo
- Department of Chemistry, Scripps Research, La Jolla, CA
- Chemical Biology and Therapeutics Science Program, Broad Institute, Cambridge, MA
| | | | - Jessica C. Kissinger
- Department of Genetics, University of Georgia, Athens, GA
- Center for Tropical and Emerging Global Diseases and Institute of Bioinformatics University of Georgia, Athens, GA
| | - Boris Striepen
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
26
|
Hanna JC, Corpas-Lopez V, Seizova S, Colon BL, Bacchetti R, Hall GMJ, Sands EM, Robinson L, Baragaña B, Wyllie S, Pawlowic MC. Mode of action studies confirm on-target engagement of lysyl-tRNA synthetase inhibitor and lead to new selection marker for Cryptosporidium. Front Cell Infect Microbiol 2023; 13:1236814. [PMID: 37600947 PMCID: PMC10436570 DOI: 10.3389/fcimb.2023.1236814] [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: 06/08/2023] [Accepted: 07/10/2023] [Indexed: 08/22/2023] Open
Abstract
Introduction Cryptosporidiosis is a leading cause of diarrheal-associated morbidity and mortality, predominantly affecting children under 5 years old in low-and-middle-income countries. There is no effective treatment and no vaccine. New therapeutics are emerging from drug discovery efforts. It is critical that mode of action studies are performed alongside drug discovery to ensure the best clinical outcomes. Unfortunately, technology to identify and validate drug targets for Cryptosporidium is severely lacking. Methods We used C. parvum lysyl-tRNA synthetase (CpKRS) and DDD01510706 as a target-compound pair to develop both chemical and genetic tools for mode of action studies for Cryptosporidium. We adapted thermal proteome profiling (TPP) for Cryptosporidium, an unbiased approach for target identification. Results Using TPP we identified the molecular target of DDD01510706 and confirm that it is CpKRS. Genetic tools confirm that CpKRS is expressed throughout the life cycle and that this target is essential for parasite survival. Parasites genetically modified to over-express CpKRS or parasites with a mutation at the compound-binding site are resistant to treatment with DDD01510706. We leveraged these mutations to generate a second drug selection marker for genetic modification of Cryptosporidium, KRSR. This second selection marker is interchangeable with the original selection marker, NeoR, and expands the range of reverse genetic approaches available to study parasite biology. Due to the sexual nature of the Cryptosporidium life cycle, parental strains containing different drug selection markers can be crossed in vivo. Discussion Selection with both drug markers produces highly efficient genetic crosses (>99% hybrid progeny), paving the way for forward genetics approaches in Cryptosporidium.
Collapse
Affiliation(s)
- Jack C. Hanna
- Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Victor Corpas-Lopez
- Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Simona Seizova
- Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Beatrice L. Colon
- Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Ross Bacchetti
- Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Grant M. J. Hall
- Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Emma M. Sands
- Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Lee Robinson
- Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Beatriz Baragaña
- Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dundee, United Kingdom
- Drug Discovery Unit, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Susan Wyllie
- Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Mattie C. Pawlowic
- Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
27
|
Tůmová L, Ježková J, Prediger J, Holubová N, Sak B, Konečný R, Květoňová D, Hlásková L, Rost M, McEvoy J, Xiao L, Santín M, Kváč M. Cryptosporidium mortiferum n. sp. (Apicomplexa: Cryptosporidiidae), the species causing lethal cryptosporidiosis in Eurasian red squirrels (Sciurus vulgaris). Parasit Vectors 2023; 16:235. [PMID: 37454101 DOI: 10.1186/s13071-023-05844-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 06/19/2023] [Indexed: 07/18/2023] Open
Abstract
BACKGROUND Cryptosporidium spp. are globally distributed parasites that infect epithelial cells in the microvillus border of the gastrointestinal tract of all classes of vertebrates. Cryptosporidium chipmunk genotype I is a common parasite in North American tree squirrels. It was introduced into Europe with eastern gray squirrels and poses an infection risk to native European squirrel species, for which infection is fatal. In this study, the biology and genetic variability of different isolates of chipmunk genotype I were investigated. METHODS The genetic diversity of Cryptosporidium chipmunk genotype I was analyzed by PCR/sequencing of the SSU rRNA, actin, HSP70, COWP, TRAP-C1 and gp60 genes. The biology of chipmunk genotype I, including oocyst size, localization of the life cycle stages and pathology, was examined by light and electron microscopy and histology. Infectivity to Eurasian red squirrels and eastern gray squirrels was verified experimentally. RESULTS Phylogenic analyses at studied genes revealed that chipmunk genotype I is genetically distinct from other Cryptosporidium spp. No detectable infection occurred in chickens and guinea pigs experimentally inoculated with chipmunk genotype I, while in laboratory mice, ferrets, gerbils, Eurasian red squirrels and eastern gray squirrels, oocyst shedding began between 4 and 11 days post infection. While infection in mice, gerbils, ferrets and eastern gray squirrels was asymptomatic or had mild clinical signs, Eurasian red squirrels developed severe cryptosporidiosis that resulted in host death. The rapid onset of clinical signs characterized by severe diarrhea, apathy, loss of appetite and subsequent death of the individual may explain the sporadic occurrence of this Cryptosporidium in field studies and its concurrent spread in the population of native European squirrels. Oocysts obtained from a naturally infected human, the original inoculum, were 5.64 × 5.37 μm and did not differ in size from oocysts obtained from experimentally infected hosts. Cryptosporidium chipmunk genotype I infection was localized exclusively in the cecum and anterior part of the colon. CONCLUSIONS Based on these differences in genetics, host specificity and pathogenicity, we propose the name Cryptosporidium mortiferum n. sp. for this parasite previously known as Cryptosporidium chipmunk genotype I.
Collapse
Affiliation(s)
- Lenka Tůmová
- Faculty of Agriculture and Technology, University of South Bohemia in České Budějovice, Studentská 1668, 37005, České Budějovice, Czech Republic
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branišovská 31, 370 05, České Budějovice, Czech Republic
| | - Jana Ježková
- Faculty of Agriculture and Technology, University of South Bohemia in České Budějovice, Studentská 1668, 37005, České Budějovice, Czech Republic
| | - Jitka Prediger
- Faculty of Agriculture and Technology, University of South Bohemia in České Budějovice, Studentská 1668, 37005, České Budějovice, Czech Republic
| | - Nikola Holubová
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branišovská 31, 370 05, České Budějovice, Czech Republic
| | - Bohumil Sak
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branišovská 31, 370 05, České Budějovice, Czech Republic
| | - Roman Konečný
- Faculty of Agriculture and Technology, University of South Bohemia in České Budějovice, Studentská 1668, 37005, České Budějovice, Czech Republic
| | - Dana Květoňová
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branišovská 31, 370 05, České Budějovice, Czech Republic
| | - Lenka Hlásková
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branišovská 31, 370 05, České Budějovice, Czech Republic
| | - Michael Rost
- Faculty of Agriculture and Technology, University of South Bohemia in České Budějovice, Studentská 1668, 37005, České Budějovice, Czech Republic
| | - John McEvoy
- Microbiological Sciences Department, North Dakota State University, 1523 Centennial Blvd, Van Es Hall, Fargo, ND, 58102, USA
| | - Lihua Xiao
- Center for Emerging and Zoonotic Diseases, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, Guangdong, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, Guangdong, China
| | - Monica Santín
- Environmental Microbial and Food Safety Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, US Department of Agriculture, Beltsville, MD, USA
| | - Martin Kváč
- Faculty of Agriculture and Technology, University of South Bohemia in České Budějovice, Studentská 1668, 37005, České Budějovice, Czech Republic.
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branišovská 31, 370 05, České Budějovice, Czech Republic.
| |
Collapse
|
28
|
Coppens I, Ward HD. Cryptosporidium secretome sheds new light on host-parasite interactions. Trends Parasitol 2023; 39:499-500. [PMID: 37150658 DOI: 10.1016/j.pt.2023.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 04/25/2023] [Indexed: 05/09/2023]
Abstract
Invasive Cryptosporidium sporozoites contain organelles that secrete unique proteins to facilitate invasion and remodeling of the infected cell. By identifying a novel secretory organelle, 'small granules', and defining the global content of all the secretory organelles, Guérin et al. set the stage to uncover molecular determinants of virulence at the host cell interface.
Collapse
Affiliation(s)
- Isabelle Coppens
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Honorine D Ward
- Division of Geographic Medicine and Infectious Diseases, Tufts Medical Center, Boston, MA, USA.
| |
Collapse
|
29
|
Oboh E, Teixeira JE, Schubert TJ, Maribona AS, Denman BN, Patel R, Huston CD, Meyers MJ. Structure-Activity relationships of replacements for the triazolopyridazine of Anti-Cryptosporidium lead SLU-2633. Bioorg Med Chem 2023; 86:117295. [PMID: 37148788 PMCID: PMC10201403 DOI: 10.1016/j.bmc.2023.117295] [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: 03/29/2023] [Revised: 04/18/2023] [Accepted: 04/21/2023] [Indexed: 05/08/2023]
Abstract
Cryptosporidiosis is a diarrheal disease particularly harmful to children and immunocompromised people. Infection is caused by the parasite Cryptosporidium and leads to dehydration, malnutrition, and death in severe cases. Nitazoxanide is the only FDA approved drug but is only modestly effective in children and ineffective in immunocompromised patients. To address this unmet medical need, we previously identified triazolopyridazine SLU-2633 as potent against Cryptosporidium parvum, with an EC50 of 0.17 µM. In the present study, we develop structure-activity relationships (SAR) for the replacement of the triazolopyridazine head group by exploring different heteroaryl groups with the aim of maintaining potency while reducing affinity for the hERG channel. 64 new analogs of SLU-2633 were synthesized and assayed for potency versus C. parvum. The most potent compound, 7,8-dihydro-[1,2,4]triazolo[4,3-b]pyridazine 17a, was found to have a Cp EC50 of 1.2 µM, 7-fold less potent than SLU-2633 but has an improved lipophilic efficiency (LipE) score. 17a was found to decrease inhibition in an hERG patch-clamp assay by about two-fold relative to SLU-2633 at 10 µM despite having similar inhibition in a [3H]-dofetilide competitive binding assay. While most other heterocycles were significantly less potent than the lead, some analogs such as azabenzothiazole 31b, have promising potency in the low micromolar range, similar to the drug nitazoxanide, and represent potential new leads for optimization. Overall, this work highlights the important role of the terminal heterocyclic head group and represents a significant extension of the understanding of the SAR for this class of anti-Cryptosporidium compounds.
Collapse
Affiliation(s)
- Edmund Oboh
- Department of Chemistry, School of Science and Engineering, Saint Louis University, Saint Louis, MO 63103, United States
| | - José E Teixeira
- Department of Medicine, University of Vermont Larner College of Medicine, Burlington, VT 05401, United States
| | - Tanner J Schubert
- Department of Chemistry, School of Science and Engineering, Saint Louis University, Saint Louis, MO 63103, United States
| | - Adriana S Maribona
- Department of Chemistry, School of Science and Engineering, Saint Louis University, Saint Louis, MO 63103, United States
| | - Brylon N Denman
- Department of Chemistry, School of Science and Engineering, Saint Louis University, Saint Louis, MO 63103, United States
| | - Radhika Patel
- Department of Chemistry, School of Science and Engineering, Saint Louis University, Saint Louis, MO 63103, United States
| | - Christopher D Huston
- Department of Medicine, University of Vermont Larner College of Medicine, Burlington, VT 05401, United States.
| | - Marvin J Meyers
- Department of Chemistry, School of Science and Engineering, Saint Louis University, Saint Louis, MO 63103, United States; Institute for Drug and Biotherapeutic Innovation, Saint Louis University, St. Louis, MO 63103, United States.
| |
Collapse
|
30
|
Tandel J, Walzer KA, Byerly JH, Pinkston B, Beiting DP, Striepen B. Genetic Ablation of a Female-Specific Apetala 2 Transcription Factor Blocks Oocyst Shedding in Cryptosporidium parvum. mBio 2023; 14:e0326122. [PMID: 36786597 PMCID: PMC10233709 DOI: 10.1128/mbio.03261-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 01/26/2023] [Indexed: 02/15/2023] Open
Abstract
The apicomplexan parasite Cryptosporidium is a leading global cause of diarrheal disease, and the infection poses a particularly grave threat to young children and those with weakened immune function. Infection occurs by ingestion of meiotic spores called oocysts, and transmission relies on fecal shedding of new oocysts. The entire life cycle thus occurs in a single host and features asexual as well as sexual forms of replication. Here, we identify and locus tag two Apetala 2-type (AP2) transcription factors and demonstrate that they are exclusively expressed in male and female gametes, respectively. To enable functional studies of essential genes in Cryptosporidium parvum, we develop and validate a small-molecule-inducible gene excision system, which we apply to the female factor AP2-F to achieve conditional gene knockout. Analyzing this mutant, we find the factor to be dispensable for asexual growth and early female fate determination in vitro but to be required for oocyst shedding in infected animals in vivo. Transcriptional analyses conducted in the presence or absence of AP2-F revealed that the factor controls the transcription of genes encoding crystalloid body proteins, which are exclusively expressed in female gametes. In C. parvum, the organelle is restricted to sporozoites, and its loss in other apicomplexan parasites leads to blocked transmission. Overall, our development of conditional gene ablation in C. parvum provides a robust method for genetic analysis in this parasite that enabled us to identify AP2-F as an essential regulator of transcription required for oocyst shedding and transmission. IMPORTANCE The parasite Cryptosporidium infects millions of people worldwide each year, leading to life-threatening diarrheal disease in young children and immunosuppressed individuals. There is no vaccine and only limited treatment. Transmission occurs via the fecal-oral route by an environmentally resilient spore-like oocyst. Infection takes place in the intestinal epithelium, where parasites initially propagate asexually before transitioning to male and female gametes, with sex leading to the formation of new oocysts. The essential role of sexual development for continuous infection and transmission makes it an attractive target for therapy and prevention. To study essential genes and potential drug targets across the life cycle, we established inducible gene excision for C. parvum. We determined that the female-specific transcription factor AP2-F is not required for asexual growth and early female development in vitro but is necessary for oocyst shedding in vivo. This work enhances the genetic tools available to study Cryptosporidium gene function.
Collapse
Affiliation(s)
- Jayesh Tandel
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, USA
| | - Katelyn A. Walzer
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jessica H. Byerly
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Brittain Pinkston
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, USA
| | - Daniel P. Beiting
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Boris Striepen
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
31
|
Warschkau D, Seeber F. Advances towards the complete in vitro life cycle of Toxoplasma gondii. Fac Rev 2023; 12:1. [PMID: 36846606 PMCID: PMC9944905 DOI: 10.12703/r/12-1] [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] [Indexed: 02/16/2023] Open
Abstract
The full life cycle of Toxoplasma gondii cannot be recapitulated in vitro, and access to certain stages, such as mature tissue cysts (bradyzoites) and oocysts (sporozoites), traditionally requires animal experiments. This has greatly hindered the study of the biology of these morphologically and metabolically distinct stages, which are essential for the infection of humans and animals. However, several breakthrough advances have been made in recent years towards obtaining these life stages in vitro, such as the discovery of several molecular factors that induce differentiation and commitment to the sexual cycle, and different culture methods that use, for example, myotubes and intestinal organoids to obtain mature bradyzoites and different sexual stages of the parasite. We review these novel tools and approaches, highlight their limitations and challenges, and discuss what research questions can already be answered with these models. We finally identify future routes for recapitulating the entire sexual cycle in vitro.
Collapse
Affiliation(s)
- David Warschkau
- FG16: Mycotic and Parasitic Agents and Mycobacteria, Robert Koch-Institut, Berlin, Germany
| | - Frank Seeber
- FG16: Mycotic and Parasitic Agents and Mycobacteria, Robert Koch-Institut, Berlin, Germany
| |
Collapse
|
32
|
Watanabe N, Bando H, Murakoshi F, Sakurai R, Kabir MHB, Fukuda Y, Kato K. The role of atypical MAP kinase 4 in the host interaction with Cryptosporidium parvum. Sci Rep 2023; 13:1096. [PMID: 36658270 PMCID: PMC9852575 DOI: 10.1038/s41598-023-28269-w] [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: 08/30/2022] [Accepted: 01/16/2023] [Indexed: 01/21/2023] Open
Abstract
Cryptosporidium parvum is an apicomplexan parasite that causes severe zoonotic diarrhea in humans and calves. Since there are no effective treatments or vaccines for infants or immunocompromised patients, it is important to understand the molecular mechanisms of the parasite-host interaction for novel drug discovery. Mitogen-activated protein kinase (MAP kinase) is a key host factor in interactions between host and various pathogens, including parasites. Although the function of conventional MAP kinases against parasite infection has been investigated, that of atypical MAP kinases remains largely unknown. Therefore, we focused on one of the atypical MAP kinases, MAPK4, and its effect on C. parvum infection in human intestinal cells. Here, we report that MAPK4-deficient intestinal cells showed a significant reduction in C. parvum infection. We also show that host MAPK4 has a role in host cell survival from C. parvum infection. In addition, we show that C. parvum requires host MAPK4 for its successful invasion and asexual reproduction. Taken together, our data suggest that MAPK4 is an important host factor contributing to C. parvum infection in human intestinal cells.
Collapse
Affiliation(s)
- Nina Watanabe
- Laboratory of Sustainable Animal Environment, Graduate School of Agricultural Science, Tohoku University, 232-3 Yomogida, Naruko-Onsen, Osaki, Miyagi, 989-6711, Japan
| | - Hironori Bando
- Laboratory of Sustainable Animal Environment, Graduate School of Agricultural Science, Tohoku University, 232-3 Yomogida, Naruko-Onsen, Osaki, Miyagi, 989-6711, Japan.,Department of Parasitology, Asahikawa Medical University, 2-1-1-1, Midorigaoka-Higashi, Asahikawa, Hokkaido, 078-8510, Japan
| | - Fumi Murakoshi
- Laboratory of Sustainable Animal Environment, Graduate School of Agricultural Science, Tohoku University, 232-3 Yomogida, Naruko-Onsen, Osaki, Miyagi, 989-6711, Japan.,Department of Infectious Diseases, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Riku Sakurai
- Laboratory of Sustainable Animal Environment, Graduate School of Agricultural Science, Tohoku University, 232-3 Yomogida, Naruko-Onsen, Osaki, Miyagi, 989-6711, Japan
| | - Mohammad Hazzaz Bin Kabir
- Laboratory of Sustainable Animal Environment, Graduate School of Agricultural Science, Tohoku University, 232-3 Yomogida, Naruko-Onsen, Osaki, Miyagi, 989-6711, Japan
| | - Yasuhiro Fukuda
- Laboratory of Sustainable Animal Environment, Graduate School of Agricultural Science, Tohoku University, 232-3 Yomogida, Naruko-Onsen, Osaki, Miyagi, 989-6711, Japan
| | - Kentaro Kato
- Laboratory of Sustainable Animal Environment, Graduate School of Agricultural Science, Tohoku University, 232-3 Yomogida, Naruko-Onsen, Osaki, Miyagi, 989-6711, Japan.
| |
Collapse
|
33
|
Khan SM, Witola WH. Past, current, and potential treatments for cryptosporidiosis in humans and farm animals: A comprehensive review. Front Cell Infect Microbiol 2023; 13:1115522. [PMID: 36761902 PMCID: PMC9902888 DOI: 10.3389/fcimb.2023.1115522] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 01/09/2023] [Indexed: 01/25/2023] Open
Abstract
The intracellular protozoan parasite of the genus Cryptosporidium is among the leading causes of waterborne diarrheal disease outbreaks throughout the world. The parasite is transmitted by ingestion of infective oocysts that are highly stable in the environment and resistant to almost all conventional disinfection methods and water treatments. Control of the parasite infection is exceedingly difficult due to the excretion of large numbers of oocysts in the feces of infected individuals that contaminate the environment and serve as a source of infection for susceptible hosts including humans and animals. Drug development against the parasite is challenging owing to its limited genetic tractability, absence of conventional drug targets, unique intracellular location within the host, and the paucity of robust cell culture platforms for continuous parasite propagation. Despite the high prevalence of the parasite, the only US Food and Drug Administration (FDA)-approved treatment of Cryptosporidium infections is nitazoxanide, which has shown moderate efficacy in immunocompetent patients. More importantly, no effective therapeutic drugs are available for treating severe, potentially life-threatening cryptosporidiosis in immunodeficient patients, young children, and neonatal livestock. Thus, safe, inexpensive, and efficacious drugs are urgently required to reduce the ever-increasing global cryptosporidiosis burden especially in low-resource countries. Several compounds have been tested for both in vitro and in vivo efficacy against the disease. However, to date, only a few experimental compounds have been subjected to clinical trials in natural hosts, and among those none have proven efficacious. This review provides an overview of the past and present anti-Cryptosporidium pharmacotherapy in humans and agricultural animals. Herein, we also highlight the progress made in the field over the last few years and discuss the different strategies employed for discovery and development of effective prospective treatments for cryptosporidiosis.
Collapse
|
34
|
Cohn IS, Henrickson SE, Striepen B, Hunter CA. Immunity to Cryptosporidium: Lessons from Acquired and Primary Immunodeficiencies. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:2261-2268. [PMID: 36469846 PMCID: PMC9731348 DOI: 10.4049/jimmunol.2200512] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 08/30/2022] [Indexed: 01/04/2023]
Abstract
Cryptosporidium is a ubiquitous protozoan parasite that infects gut epithelial cells and causes self-limited diarrhea in immunocompetent individuals. However, in immunocompromised hosts with global defects in T cell function, this infection can result in chronic, life-threatening disease. In addition, there is a subset of individuals with primary immunodeficiencies associated with increased risk for life-threatening cryptosporidiosis. These patients highlight MHC class II expression, CD40-CD40L interactions, NF-κB signaling, and IL-21 as key host factors required for resistance to this enteric pathogen. Understanding which immune deficiencies do (or do not) lead to increased risk for severe Cryptosporidium may reveal mechanisms of parasite restriction and aid in the identification of novel strategies to manage this common pathogen in immunocompetent and deficient hosts.
Collapse
Affiliation(s)
- Ian S. Cohn
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sarah E. Henrickson
- Institute for Immunology, University of Pennsylvania, Philadelphia, PA, USA
- Division of Allergy Immunology, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Boris Striepen
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christopher A. Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
35
|
The Marine Compound Tartrolon E Targets the Asexual and Early Sexual Stages of Cryptosporidium parvum. Microorganisms 2022; 10:microorganisms10112260. [PMID: 36422330 PMCID: PMC9693555 DOI: 10.3390/microorganisms10112260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/08/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022] Open
Abstract
New therapeutic agents for cryptosporidiosis are a critical medical need. The marine organic compound, tartrolon E (trtE), is highly effective against multiple apicomplexan parasites, including Cryptosporidium. Understanding the mechanism of action of trtE is required to advance in the drug development pipeline. Here, we validate using Nluc C. parvum parasites for the study of trtE and pinpoint the life stage targeted by trtE. Results show that trtE kills Nluc and wild type C. parvum with equal efficiency, confirming the use of the Nluc C. parvum to study this compound. Results revealed that trtE kills the parasite within an hour of treatment and while the compound has no effect on viability of sporozoites, trtE does inhibit establishment of infection. Targeting treatment at particular life cycle stages demonstrated that trtE is effective against asexual of the parasite but has reduced efficacy against mature sexual stages. Gene expression analysis shows that trtE inhibits the early sexual stage of the parasite. Results from these studies will aid the development of trtE as a therapeutic for cryptosporidiosis.
Collapse
|
36
|
Jex AR, Tonkin CJ, Ralph SA. 3, 2, 1, go! Cryptosporidium counts down to sex. PLoS Biol 2022; 20:e3001638. [PMID: 35552541 PMCID: PMC9129006 DOI: 10.1371/journal.pbio.3001638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 05/24/2022] [Indexed: 11/25/2022] Open
Abstract
Cryptosporidium is a leading cause of death from childhood diarrhoea, but its biology is poorly understood. This Primer explores a recent study in PLOS Biology that reveals hitherto unknown aspects of the parasite’s life cycle that may lead to improvements in ex vivo culture.
Collapse
Affiliation(s)
- Aaron R. Jex
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Christopher J. Tonkin
- Infectious Diseases and Immune Defence, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Stuart A. Ralph
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science & Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
- * E-mail:
| |
Collapse
|