1
|
Tao DL, Chen JM, Wu JP, Zhao SS, Qi BF, Yang X, Fan YY, Song JK, Zhao GH. Neospora caninum hijacks host PFKFB3-driven glycolysis to facilitate intracellular propagation of parasites. Vet Res 2025; 56:94. [PMID: 40307939 PMCID: PMC12042381 DOI: 10.1186/s13567-025-01524-w] [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: 12/16/2024] [Accepted: 02/13/2025] [Indexed: 05/02/2025] Open
Abstract
Infection with Neospora caninum leads to reproductive failure in ruminants, such as cattle and goats; however, no effective vaccines or treatments are currently available to control this infection. Carefully regulating the glycolysis of host cells is essential for the intracellular survival of pathogens. Nonetheless, the impact of N. caninum infection on host cell glycolysis and the effects and mechanisms of host cell glycolysis on the intracellular survival of this parasite remains unclear. In this study, the analysis of metabolomics and transcriptomics revealed that N. caninum infection increases the expression of glycolysis-related enzymes and lactate production in caprine endometrial epithelial cells (EECs). The study's findings demonstrate that the inhibition of host cell glycolysis using 2-DG or sodium oxamate (an LDH-A inhibitor) inhibits host cell glycolysis and the intracellular propagation of N. caninum tachyzoites. Moreover, the addition of lactate further promotes the replication of N. caninum tachyzoites both in vivo and in vitro. Further investigation found that N. caninum infection induces host cell glycolysis via up-regulating 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3) expression, while knockdown of PFKFB3 with small-interfering RNA or 3-PO significantly inhibits host cell glycolysis and the propagation of N. caninum tachyzoites both in vivo and in vitro. Additionally, a mechanistic study showed that N. caninum infection activates the JNK signalling pathway and inhibits the ubiquitination degradation of HIF-1α. Chromatin immunoprecipitation and dual-luciferase reporter assays revealed that N. caninum infection induces the expression of HIF-1α, which binds to the promoter region of pfkfb3. Our findings indicate that cellular glycolysis may serve as a potential therapeutic target for neosporosis, offering a novel insight for further investigating the intracellular survival mechanisms of N. caninum.
Collapse
Affiliation(s)
- De-Liang Tao
- Department of Parasitology, College of Veterinary Medicine, Northwest A&F University, Shaanxi, Yangling, China
| | - Jin-Ming Chen
- Department of Parasitology, College of Veterinary Medicine, Northwest A&F University, Shaanxi, Yangling, China
| | - Jiang-Ping Wu
- Department of Parasitology, College of Veterinary Medicine, Northwest A&F University, Shaanxi, Yangling, China
| | - Shan-Shan Zhao
- Department of Parasitology, College of Veterinary Medicine, Northwest A&F University, Shaanxi, Yangling, China
| | - Bu-Fan Qi
- Department of Parasitology, College of Veterinary Medicine, Northwest A&F University, Shaanxi, Yangling, China
| | - Xin Yang
- Department of Parasitology, College of Veterinary Medicine, Northwest A&F University, Shaanxi, Yangling, China
| | - Ying-Ying Fan
- Department of Parasitology, College of Veterinary Medicine, Northwest A&F University, Shaanxi, Yangling, China
| | - Jun-Ke Song
- Department of Parasitology, College of Veterinary Medicine, Northwest A&F University, Shaanxi, Yangling, China
| | - Guang-Hui Zhao
- Department of Parasitology, College of Veterinary Medicine, Northwest A&F University, Shaanxi, Yangling, China.
| |
Collapse
|
2
|
Kot K, Grabowska M, Tarnowski M, Kupnicka P, Tomasiak P, Kosik-Bogacka D, Łanocha-Arendarczyk N. Hypoxia and collagen deposition in the kidneys infected with Acanthamoeba sp. Sci Rep 2024; 14:28096. [PMID: 39543383 PMCID: PMC11564555 DOI: 10.1038/s41598-024-79848-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 11/12/2024] [Indexed: 11/17/2024] Open
Abstract
Acanthamoeba spp. are facultative, opportunistic pathogens that are found in diverse environments. In the hosts, they lead to multi-organ disease. Recent studies reported that they may induce changes in the kidneys of hosts. The aim of the study was to determine the influence of Acanthamoeba sp. on hypoxia and collagen deposition in the kidneys of immunocompetent and immunosuppressed mice infected with Acanthamoeba sp. The results strongly suggest that Acanthamoeba sp. induces hypoxia in mice with normal and reduced immune response by increasing gene and/or protein expression of HIF1α as well as HIF2α. Additionally, the activation of these factors is probably induced via NOX2/ROS. Hypoxia promotes vessel formation, and we found that angiogenesis occurs in the kidneys of mice infected with the parasite regardless of their immunological status. The proangiogenic factors released in hypoxic conditions cause modulation and inflammation in the kidney cells, which in turn leads to collagen deposition via TGF-β. This work reveals mechanisms occurring in the hosts infected with Acanthamoeba sp., highlights as well as supports the relevance of pathophysiology in the kidneys in hosts with systematic acanthamoebiasis.
Collapse
Affiliation(s)
- Karolina Kot
- Department of Biology, Parasitology, and Pharmaceutical Botany, Pomeranian Medical University in Szczecin, Szczecin, Poland.
| | - Marta Grabowska
- Department of Histology and Developmental Biology, Faculty of Health Sciences, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Maciej Tarnowski
- Department of Physiology in Health Sciences, Faculty of Health Sciences, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Patrycja Kupnicka
- Department of Biochemistry, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Patrycja Tomasiak
- Institute of Physical Culture Sciences, University of Szczecin, Szczecin, Poland
| | - Danuta Kosik-Bogacka
- Department of Biology, Parasitology, and Pharmaceutical Botany, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Natalia Łanocha-Arendarczyk
- Department of Biology, Parasitology, and Pharmaceutical Botany, Pomeranian Medical University in Szczecin, Szczecin, Poland
| |
Collapse
|
3
|
Baptista CG, Hosking S, Gas-Pascual E, Ciampossine L, Abel S, Hakimi MA, Jeffers V, Le Roch K, West CM, Blader IJ. The Toxoplasma gondii F-Box Protein L2 Functions as a Repressor of Stage Specific Gene Expression. PLoS Pathog 2024; 20:e1012269. [PMID: 38814984 PMCID: PMC11166348 DOI: 10.1371/journal.ppat.1012269] [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: 12/18/2023] [Revised: 06/11/2024] [Accepted: 05/17/2024] [Indexed: 06/01/2024] Open
Abstract
Toxoplasma gondii is a foodborne pathogen that can cause severe and life-threatening infections in fetuses and immunocompromised patients. Felids are its only definitive hosts, and a wide range of animals, including humans, serve as intermediate hosts. When the transmissible bradyzoite stage is orally ingested by felids, they transform into merozoites that expand asexually, ultimately generating millions of gametes for the parasite sexual cycle. However, bradyzoites in intermediate hosts differentiate exclusively to disease-causing tachyzoites, which rapidly disseminate throughout the host. Though tachyzoites are well-studied, the molecular mechanisms governing transitioning between developmental stages are poorly understood. Each parasite stage can be distinguished by a characteristic transcriptional signature, with one signature being repressed during the other stages. Switching between stages require substantial changes in the proteome, which is achieved in part by ubiquitination. F-box proteins mediate protein poly-ubiquitination by recruiting substrates to SKP1, Cullin-1, F-Box protein E3 ubiquitin ligase (SCF-E3) complexes. We have identified an F-box protein named Toxoplasma gondii F-Box Protein L2 (TgFBXL2), which localizes to distinct perinucleolar sites. TgFBXL2 is stably engaged in an SCF-E3 complex that is surprisingly also associated with a COP9 signalosome complex that negatively regulates SCF-E3 function. At the cellular level, TgFBXL2-depleted parasites are severely defective in centrosome replication and daughter cell development. Most remarkable, RNAseq data show that TgFBXL2 conditional depletion induces the expression of stage-specific genes including a large cohort of genes necessary for sexual commitment. Together, these data suggest that TgFBXL2 is a latent guardian of stage specific gene expression in Toxoplasma and poised to remove conflicting proteins in response to an unknown trigger of development.
Collapse
Affiliation(s)
- Carlos G. Baptista
- Department of Microbiology and Immunology, University at Buffalo School of Medicine, Buffalo, New York, United States of America
| | - Sarah Hosking
- Department of Microbiology and Immunology, University at Buffalo School of Medicine, Buffalo, New York, United States of America
| | - Elisabet Gas-Pascual
- Department of Biochemistry & Molecular Biology, Center for Tropical & Emerging Global Diseases, University of Georgia, Athens, Georgia United States of America
| | - Loic Ciampossine
- Department of Molecular, Cell, and Systems Biology, University of California Riverside, Riverside, California, United States of America
| | - Steven Abel
- Department of Molecular, Cell, and Systems Biology, University of California Riverside, Riverside, California, United States of America
| | - Mohamed-Ali Hakimi
- Host-Pathogen Interactions and Immunity to Infection, Institute for Advanced Biosciences (IAB), INSERM U1209, CNRS UMR 5309, Grenoble Alpes University, Grenoble, France
| | - Victoria Jeffers
- Department of Molecular, Cellular and Biomedical Sciences, University of New Hampshire, Durham, New Hampshire, United States of America
| | - Karine Le Roch
- Department of Molecular, Cell, and Systems Biology, University of California Riverside, Riverside, California, United States of America
| | - Christopher M. West
- Department of Biochemistry & Molecular Biology, Center for Tropical & Emerging Global Diseases, University of Georgia, Athens, Georgia United States of America
| | - Ira J. Blader
- Department of Microbiology and Immunology, University at Buffalo School of Medicine, Buffalo, New York, United States of America
| |
Collapse
|
4
|
Key M, Baptista CG, Bergmann A, Floyd K, Blader IJ, Dou Z. Toxoplasma gondii harbors a hypoxia-responsive coproporphyrinogen dehydrogenase-like protein. mSphere 2024; 9:e0009224. [PMID: 38411121 PMCID: PMC10964404 DOI: 10.1128/msphere.00092-24] [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: 02/05/2024] [Accepted: 02/06/2024] [Indexed: 02/28/2024] Open
Abstract
Toxoplasma gondii is an apicomplexan parasite that is the cause of toxoplasmosis, a potentially lethal disease for immunocompromised individuals. During in vivo infection, the parasites encounter various growth environments, such as hypoxia. Therefore, the metabolic enzymes in the parasites must adapt to such changes to fulfill their nutritional requirements. Toxoplasma can de novo biosynthesize some nutrients, such as heme. The parasites heavily rely on their own heme production for intracellular survival. Notably, the antepenultimate step within this pathway is facilitated by coproporphyrinogen III oxidase (CPOX), which employs oxygen to convert coproporphyrinogen III to protoporphyrinogen IX through oxidative decarboxylation. Conversely, some bacteria can accomplish this conversion independently of oxygen through coproporphyrinogen dehydrogenase (CPDH). Genome analysis found a CPDH ortholog in Toxoplasma. The mutant Toxoplasma lacking CPOX displays significantly reduced growth, implying that T. gondii CPDH (TgCPDH) potentially functions as an alternative enzyme to perform the same reaction as CPOX under low-oxygen conditions. In this study, we demonstrated that TgCPDH exhibits CPDH activity by complementing it in a heme synthesis-deficient Salmonella mutant. Additionally, we observed an increase in TgCPDH expression in Toxoplasma when it grew under hypoxic conditions. However, deleting TgCPDH in both wild-type and heme-deficient parasites did not alter their intracellular growth under both ambient and low-oxygen conditions. This research marks the first report of a CPDH-like protein in eukaryotic cells. Although TgCPDH responds to hypoxic conditions and possesses enzymatic activity, our findings revealed that it does not directly affect acute Toxoplasma infections in vitro and in vivo. IMPORTANCE Toxoplasma gondii is a ubiquitous parasite capable of infecting a wide range of warm-blooded hosts, including humans. During its life cycle, these parasites must adapt to varying environmental conditions, including situations with low-oxygen levels, such as intestine and spleen tissues. Our research, in conjunction with studies conducted by other laboratories, has revealed that Toxoplasma primarily relies on its own heme production during acute infections. Intriguingly, in addition to this classical heme biosynthetic pathway, the parasites encode a putative oxygen-independent coproporphyrinogen dehydrogenase (CPDH), suggesting its potential contribution to heme production under varying oxygen conditions, a feature typically observed in simpler organisms like bacteria. Notably, so far, CPDH has only been identified in some bacteria for heme biosynthesis. Our study discovered that Toxoplasma harbors a functional enzyme displaying CPDH activity, which alters its expression in the parasites when they face fluctuating oxygen levels in their surroundings.
Collapse
Affiliation(s)
- Melanie Key
- Department of Biological Sciences, Clemson University, Clemson, South Carolina, USA
- Eukaryotic Pathogens Innovation Center, Clemson University, Clemson, South Carolina, USA
| | - Carlos Gustavo Baptista
- Department of Microbiology and Immunology, University at Buffalo School of Medicine, The State University of New York, Buffalo, New York, USA
| | - Amy Bergmann
- Department of Biological Sciences, Clemson University, Clemson, South Carolina, USA
| | - Katherine Floyd
- Department of Biological Sciences, Clemson University, Clemson, South Carolina, USA
| | - Ira J. Blader
- Department of Microbiology and Immunology, University at Buffalo School of Medicine, The State University of New York, Buffalo, New York, USA
| | - Zhicheng Dou
- Department of Biological Sciences, Clemson University, Clemson, South Carolina, USA
- Eukaryotic Pathogens Innovation Center, Clemson University, Clemson, South Carolina, USA
| |
Collapse
|
5
|
Baptista CG, Hosking S, Gas-Pascual E, Ciampossine L, Abel S, Hakimi MA, Jeffers V, Le Roch K, West CM, Blader IJ. Toxoplasma gondii F-Box Protein L2 Silences Feline-Restricted Genes Necessary for Sexual Commitment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.18.572150. [PMID: 38187549 PMCID: PMC10769283 DOI: 10.1101/2023.12.18.572150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Toxoplasma gondii is a foodborne pathogen that can cause severe and life-threatening infections in fetuses and immunocompromised patients. Felids are its only definitive hosts, and a wide range of animals, including humans, serve as intermediate hosts. When the transmissible bradyzoite stage is orally ingested by felids, they transform into merozoites that expand asexually, ultimately generating millions of gametes for the parasite sexual cycle. However, bradyzoites in intermediate hosts differentiate exclusively to disease-causing tachyzoites, which rapidly disseminate throughout the host. Though tachyzoites are well-studied, the molecular mechanisms governing transitioning between developmental stages are poorly understood. Each parasite stage can be distinguished by a characteristic transcriptional signature, with one signature being repressed during the other stages. Switching between stages requires substantial changes in the proteome, which is achieved in part by ubiquitination. F-box proteins mediate protein poly-ubiquitination by recruiting substrates to SKP1, Cullin-1, F-Box protein E3 ubiquitin ligase (SCF-E3) complexes. We have identified an F-box protein named Toxoplasma gondii F-Box Protein L2 (TgFBXL2), which localizes to distinct nuclear sites. TgFBXL2 is stably engaged in an SCF-E3 complex that is surprisingly also associated with a COP9 signalosome complex that negatively regulates SCF-E3 function. At the cellular level, TgFBXL2-depleted parasites are severely defective in centrosome replication and daughter cell development. Most remarkable, RNA seq data show that TgFBXL2 conditional depletion induces the expression of genes necessary for sexual commitment. We suggest that TgFBXL2 is a latent guardian of sexual stage development in Toxoplasma and poised to remove conflicting proteins in response to an unknown trigger of sexual development.
Collapse
Affiliation(s)
- Carlos G. Baptista
- Department of Microbiology and Immunology, University at Buffalo School of Medicine, Buffalo, NY 14203 USA
| | - Sarah Hosking
- Department of Microbiology and Immunology, University at Buffalo School of Medicine, Buffalo, NY 14203 USA
| | - Elisabet Gas-Pascual
- Department of Biochemistry & Molecular Biology, Center for Tropical & Emerging Global Diseases, University of Georgia, Athens, GA 30602 USA
| | - Loic Ciampossine
- Department of Molecular, Cell, and Systems Biology, University of California Riverside, Riverside, CA, 92521USA
| | - Steven Abel
- Department of Molecular, Cell, and Systems Biology, University of California Riverside, Riverside, CA, 92521USA
| | - Mohamed-Ali Hakimi
- Host-Pathogen Interactions and Immunity to Infection, Institute for Advanced Biosciences (IAB), INSERM U1209, CNRS UMR 5309, Grenoble Alpes University, Grenoble, France
| | - Victoria Jeffers
- Department of Molecular, Cellular and Biomedical Sciences, University of New Hampshire, Durham, NH 03824 USA
| | - Karine Le Roch
- Department of Molecular, Cell, and Systems Biology, University of California Riverside, Riverside, CA, 92521USA
| | - Christopher M. West
- Department of Biochemistry & Molecular Biology, Center for Tropical & Emerging Global Diseases, University of Georgia, Athens, GA 30602 USA
| | - Ira J. Blader
- Department of Microbiology and Immunology, University at Buffalo School of Medicine, Buffalo, NY 14203 USA
| |
Collapse
|
6
|
Key M, Baptista CG, Bergmann A, Floyd K, Blader IJ, Dou Z. Toxoplasma gondii harbors a hypoxia-responsive coproporphyrinogen dehydrogenase-like protein. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.16.567449. [PMID: 38014006 PMCID: PMC10680763 DOI: 10.1101/2023.11.16.567449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Toxoplasma gondii is an apicomplexan parasite that is the cause of toxoplasmosis, a potentially lethal disease for immunocompromised individuals. During in vivo infection, the parasites encounter various growth environments, such as hypoxia. Therefore, the metabolic enzymes in the parasites must adapt to such changes to fulfill their nutritional requirements. Toxoplasma can de novo biosynthesize some nutrients, such as heme. The parasites heavily rely on their own heme production for intracellular survival. Notably, the antepenultimate step within this pathway is facilitated by coproporphyrinogen III oxidase (CPOX), which employs oxygen to convert coproporphyrinogen III to protoporphyrinogen IX through oxidative decarboxylation. Conversely, some bacteria can accomplish this conversion independently of oxygen through coproporphyrinogen dehydrogenase (CPDH). Genome analysis found a CPDH ortholog in Toxoplasma. The mutant Toxoplasma lacking CPOX displays significantly reduced growth, implying that TgCPDH potentially functions as an alternative enzyme to perform the same reaction as CPOX under low oxygen conditions. In this study, we demonstrated that TgCPDH exhibits coproporphyrinogen dehydrogenase activity by complementing it in a heme synthesis-deficient Salmonella mutant. Additionally, we observed an increase in TgCPDH expression in Toxoplasma when it grew under hypoxic conditions. However, deleting TgCPDH in both wildtype and heme-deficient parasites did not alter their intracellular growth under both ambient and low oxygen conditions. This research marks the first report of a coproporphyrinogen dehydrogenase-like protein in eukaryotic cells. Although TgCPDH responds to hypoxic conditions and possesses enzymatic activity, our findings suggest that it does not directly affect intracellular infection or the pathogenesis of Toxoplasma parasites.
Collapse
Affiliation(s)
- Melanie Key
- Department of Biological Sciences, Clemson University, Clemson, South Carolina, USA, 29634
- Eukaryotic Pathogens Innovation Center, Clemson University, Clemson, South Carolina, USA, 29634
| | - Carlos Gustavo Baptista
- Department of Microbiology and Immunology, University at Buffalo, The State University of New York, Buffalo, New York, USA 14260
| | - Amy Bergmann
- Department of Biological Sciences, Clemson University, Clemson, South Carolina, USA, 29634
| | - Katherine Floyd
- Department of Biological Sciences, Clemson University, Clemson, South Carolina, USA, 29634
| | - Ira J. Blader
- Department of Microbiology and Immunology, University at Buffalo, The State University of New York, Buffalo, New York, USA 14260
| | - Zhicheng Dou
- Department of Biological Sciences, Clemson University, Clemson, South Carolina, USA, 29634
- Eukaryotic Pathogens Innovation Center, Clemson University, Clemson, South Carolina, USA, 29634
| |
Collapse
|
7
|
Lis A, Baptista CG, Dahlgren K, Corvi MM, Blader IJ. Identification of Toxoplasma calcium-dependent protein kinase 3 as a stress-activated elongation factor 2 kinase. mSphere 2023; 8:e0015623. [PMID: 37272703 PMCID: PMC10449493 DOI: 10.1128/msphere.00156-23] [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] [Received: 03/24/2023] [Accepted: 04/19/2023] [Indexed: 06/06/2023] Open
Abstract
Toxoplasma gondii is an obligate intracellular parasite whose tachyzoite form causes disease via a lytic growth cycle. Its metabolic and cellular pathways are primarily designed to ensure parasite survival within a host cell. But during its lytic cycle, tachyzoites are exposed to the extracellular milieu and prolonged exposure requires activation of stress response pathways that include reprogramming the parasite proteome. Regulation of protein synthesis is therefore important for extracellular survival. We previously reported that in extracellularly stressed parasites, the elongation phase of protein synthesis is regulated by the Toxoplasma oxygen-sensing protein, PHYb. PHYb acts by promoting the activity of elongation factor eEF2, which is a GTPase that catalyzes the transfer of the peptidyl-tRNA from the A site to the P site of the ribosome. In the absence of PHYb, eEF2 is hyper-phosphorylated, which inhibits eEF2 from interacting with the ribosome. eEF2 kinases are atypical calcium-dependent kinases and BLAST analyses revealed the parasite kinase, CDPK3, as the most highly homologous to the Saccharomyces cerevisiae eEF2 kinase, RCK2. In parasites exposed to extracellular stress, loss of CDPK3 leads to decreased eEF2 phosphorylation and enhanced rates of elongation. Furthermore, co-immunoprecipitation studies revealed that CDPK3 and eEF2 interact in stressed parasites. Since CDPK3 and eEF2 normally localize to the plasma membrane and cytosol, respectively, we investigated how the two can interact. We report that under stress conditions, CDPK3 is not N-myristoylated likely leading to its cytoplasmic localization. In summary, we have identified a novel function for CDPK3 as the first protozoan extracellular stress-induced eEF2 kinase.IMPORTANCEAlthough it is an obligate intracellular parasite, Toxoplasma must be able to survive in the extracellular environment. Our previous work indicated that ensuring that elongation continues during protein synthesis is part of this stress response and that this is due to preventing phosphorylation of elongation factor 2. But the identity of the eEF2 kinase has remained unknown in Toxoplasma and other protozoan parasites. Here, we identify CDPK3 as the first protozoan eEF2 kinase and demonstrate that it is part of a stress response initiated when parasites are exposed to extracellular stress. We also demonstrate that CDPK3 engages eEF2 as a result of its relocalization from the plasma membrane to the cytosol.
Collapse
Affiliation(s)
- Agnieszka Lis
- Department of Microbiology and Immunology, SUNY at Buffalo School of Medicine, Buffalo, New York, USA
| | - Carlos Gustavo Baptista
- Department of Microbiology and Immunology, SUNY at Buffalo School of Medicine, Buffalo, New York, USA
| | - Kelsey Dahlgren
- Department of Microbiology and Immunology, SUNY at Buffalo School of Medicine, Buffalo, New York, USA
| | - Maria M. Corvi
- Laboratorio de Bioquímica y Biología Celular de Parásitos, Instituto Tecnológico de Chascomús (CONICET-UNSAM), Escuela de Bio y Nanotecnologías (UNSAM), Chascomús, Buenos Aires, Argentina
| | - Ira J. Blader
- Department of Microbiology and Immunology, SUNY at Buffalo School of Medicine, Buffalo, New York, USA
| |
Collapse
|
8
|
Akinsulie OC, Shahzad S, Ogunleye SC, Oladapo IP, Joshi M, Ugwu CE, Gbadegoye JO, Hassan FO, Adeleke R, Afolabi Akande Q, Adesola RO. Crosstalk between hypoxic cellular micro-environment and the immune system: a potential therapeutic target for infectious diseases. Front Immunol 2023; 14:1224102. [PMID: 37600803 PMCID: PMC10434535 DOI: 10.3389/fimmu.2023.1224102] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 06/26/2023] [Indexed: 08/22/2023] Open
Abstract
There are overwhelming reports on the promotional effect of hypoxia on the malignant behavior of various forms of cancer cells. This has been proposed and tested exhaustively in the light of cancer immunotherapy. However, there could be more interesting functions of a hypoxic cellular micro-environment than malignancy. There is a highly intricate crosstalk between hypoxia inducible factor (HIF), a transcriptional factor produced during hypoxia, and nuclear factor kappa B (NF-κB) which has been well characterized in various immune cell types. This important crosstalk shares common activating and inhibitory stimuli, regulators, and molecular targets. Impaired hydroxylase activity contributes to the activation of HIFs. Inflammatory ligands activate NF-κB activity, which leads to the expression of inflammatory and anti-apoptotic genes. The eventual sequelae of the interaction between these two molecular players in immune cells, either bolstering or abrogating functions, is largely cell-type dependent. Importantly, this holds promise for interesting therapeutic interventions against several infectious diseases, as some HIF agonists have helped prevent immune-related diseases. Hypoxia and inflammation are common features of infectious diseases. Here, we highlighted the role of this crosstalk in the light of functional immunity against infection and inflammation, with special focus on various innate and adaptive immune cells. Particularly, we discussed the bidirectional effects of this crosstalk in the regulation of immune responses by monocytes/macrophages, dendritic cells, neutrophils, B cells, and T cells. We believe an advanced understanding of the interplay between HIFs and NF-kB could reveal novel therapeutic targets for various infectious diseases with limited treatment options.
Collapse
Affiliation(s)
- Olalekan Chris Akinsulie
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
| | - Sammuel Shahzad
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
| | - Seto Charles Ogunleye
- College of Veterinary Medicine, Mississippi State University, Starkville, MS, United States
| | - Ifeoluwa Peace Oladapo
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
| | - Melina Joshi
- Center for Molecular Dynamics Nepal, Kathmandu, Nepal
| | - Charles Egede Ugwu
- Paul G. Allen School for Global Health, Washington State University, Pullman, WA, United States
| | - Joy Olaoluwa Gbadegoye
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Fasilat Oluwakemi Hassan
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Richard Adeleke
- College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Qudus Afolabi Akande
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, United States
| | | |
Collapse
|
9
|
DeMichele E, Sosnowski O, Buret AG, Allain T. Regulatory Functions of Hypoxia in Host-Parasite Interactions: A Focus on Enteric, Tissue, and Blood Protozoa. Microorganisms 2023; 11:1598. [PMID: 37375100 PMCID: PMC10303274 DOI: 10.3390/microorganisms11061598] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
Body tissues are subjected to various oxygenic gradients and fluctuations and hence can become transiently hypoxic. Hypoxia-inducible factor (HIF) is the master transcriptional regulator of the cellular hypoxic response and is capable of modulating cellular metabolism, immune responses, epithelial barrier integrity, and local microbiota. Recent reports have characterized the hypoxic response to various infections. However, little is known about the role of HIF activation in the context of protozoan parasitic infections. Growing evidence suggests that tissue and blood protozoa can activate HIF and subsequent HIF target genes in the host, helping or hindering their pathogenicity. In the gut, enteric protozoa are adapted to steep longitudinal and radial oxygen gradients to complete their life cycle, yet the role of HIF during these protozoan infections remains unclear. This review focuses on the hypoxic response to protozoa and its role in the pathophysiology of parasitic infections. We also discuss how hypoxia modulates host immune responses in the context of protozoan infections.
Collapse
Affiliation(s)
- Emily DeMichele
- Department of Biological Sciences, University of Calgary, Calgary, AB T2N 1N4, Canada; (E.D.); (O.S.); (A.G.B.)
- Inflammation Research Network, University of Calgary, Calgary, AB T2N 1N4, Canada
- Host-Parasite Interactions, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Olivia Sosnowski
- Department of Biological Sciences, University of Calgary, Calgary, AB T2N 1N4, Canada; (E.D.); (O.S.); (A.G.B.)
- Inflammation Research Network, University of Calgary, Calgary, AB T2N 1N4, Canada
- Host-Parasite Interactions, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Andre G. Buret
- Department of Biological Sciences, University of Calgary, Calgary, AB T2N 1N4, Canada; (E.D.); (O.S.); (A.G.B.)
- Inflammation Research Network, University of Calgary, Calgary, AB T2N 1N4, Canada
- Host-Parasite Interactions, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Thibault Allain
- Department of Biological Sciences, University of Calgary, Calgary, AB T2N 1N4, Canada; (E.D.); (O.S.); (A.G.B.)
- Inflammation Research Network, University of Calgary, Calgary, AB T2N 1N4, Canada
- Host-Parasite Interactions, University of Calgary, Calgary, AB T2N 1N4, Canada
| |
Collapse
|
10
|
Honfozo A, Houngue R, Vandeputte A, Dechavanne S, Nouatin O, Atindehou MC, Fanou LA, Massougbodji A, Dechavanne C, Brodin P, Tomavo S. An image-based high-content screening for compounds targeting Toxoplasma gondii repurposed inhibitors effective against the malaria parasite Plasmodium falciparum. Front Cell Infect Microbiol 2023; 13:1102551. [PMID: 36936758 PMCID: PMC10020723 DOI: 10.3389/fcimb.2023.1102551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 02/10/2023] [Indexed: 03/06/2023] Open
Abstract
Apicomplexa phylum includes numerous obligate intracellular protozoan parasites that are life threatening for humans and animals. In this context, Plasmodium falciparum and Toxoplasma gondii are of particular interest, as they are responsible for malaria and toxoplasmosis, respectively, for which efficient vaccines are presently lacking and therapies need to be improved. Apicomplexan parasites have a highly polarized morphology, with their apical end containing specific secretory organelles named rhoptries and micronemes, which depend on the unique receptor and transporter sortilin TgSORT for their biogenesis. In the present study, we took advantage of the subcellular polarity of the parasite to engineer a clonal transgenic Toxoplasma line that expresses simultaneously the green fluorescent protein TgSORT-GFP in the post-Golgi-endosome-like compartment and the red fluorescent protein rhoptry ROP1-mCherry near the apical end. We utilized this fluorescent transgenic T. gondii to develop a miniaturized image-based phenotype assay coupled to an automated image analysis. By applying this methodology to 1,120 compounds, we identified 12 that are capable of disrupting the T. gondii morphology and inhibiting intracellular replication. Analysis of the selected compounds confirmed that all 12 are kinase inhibitors and intramembrane pumps, with some exhibiting potent activity against Plasmodium falciparum. Our findings highlight the advantage of comparative and targeted phenotypic analysis involving two related parasite species as a means of identifying molecules with a conserved mode of action.
Collapse
Affiliation(s)
- Ariane Honfozo
- Université Paris Saclay, CNRS UMR 9198-CEA, Institute for Integrative Biology of the Cell (I2BC), Gif sur Yvette, France
| | - Rodrigue Houngue
- Université Paris Saclay, CNRS UMR 9198-CEA, Institute for Integrative Biology of the Cell (I2BC), Gif sur Yvette, France
| | - Alexandre Vandeputte
- Université de Lille, CNRS UMR 9017, INSERM U 1019, Institut Pasteur de Lille, US 41 − UAR 2014 − PLBS, CIIL − Center for Immunity and Infection of Lille, Lille, France
| | | | - Odilon Nouatin
- Institut de Recherche Clinique du Bénin, Abomey-Calavi, Benin
| | | | - Lucie Ayi Fanou
- Laboratoire de Biochimie et de Biologie Moléculaire, FAST/UAC, Cotonou, Benin
| | | | - Célia Dechavanne
- Université de Paris Cité, IRD, MERIT, Paris, France
- CERPAGE, Cotonou, Benin
| | - Priscille Brodin
- Université de Lille, CNRS UMR 9017, INSERM U 1019, Institut Pasteur de Lille, US 41 − UAR 2014 − PLBS, CIIL − Center for Immunity and Infection of Lille, Lille, France
| | - Stanislas Tomavo
- Université Paris Saclay, CNRS UMR 9198-CEA, Institute for Integrative Biology of the Cell (I2BC), Gif sur Yvette, France
- *Correspondence: Stanislas Tomavo,
| |
Collapse
|
11
|
Castello A, Fazio E, Alfonzetti T, Giunta RP, Salvaggio A, Ferlazzo AM, Cravana C, Bruschetta G, Medica P, Marino AMF. Assessment of selected molecular factors and 17-β estradiol dosage in response to Toxoplasma gondii infection in swine. Vet World 2022; 15:1641-1649. [PMID: 36185508 PMCID: PMC9394144 DOI: 10.14202/vetworld.2022.1641-1649] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 05/24/2022] [Indexed: 11/23/2022] Open
Abstract
Background and Aim: Toxoplasma gondii is a global zoonotic parasite infecting virtually all warm-blooded species, although a species-specific variability is evident referring to symptoms frame. Both the success of T. gondii and the outcome of infection depend on a delicate balance between host cellular pathways and the evasion or modulation strategies elicited by the parasite. The hormonal and molecular mechanisms involved in this delicate host-parasite balance are still unclear, especially when considering intermediate host species other than mouse. This study aimed to assess any correlation between T. gondii infection and selected molecular and hormonal factors involved in responses to infection in susceptible species such as swine. Moreover, blood counts and hematochemical assays (glucose, total cholesterol, and triglycerides dosage) were performed to evaluate the overall health condition of animals. Materials and Methods: Toxoplasmosis was diagnosed by enzyme-linked immunosorbent assay for antibodies determination and real-time polymerase chain reaction (RT-PCR) for T. gondii DNA detection. Target genes coding for key factors of cell responses to T. gondii infection were selected, and their transcription was assessed in various tissues by quantitative RT-PCR. 17-β estradiol concentrations were assessed by fluorimetric enzyme-linked immunoassay and the AIA-360 automated immunoassay analyzer. Blood count and hematochemical analyses were performed by a blood cell counter and a spectrophotometer, respectively. Results: The present research highlighted significant differences among infected and uninfected swine (control group) for both transcription profiles of some of the molecular factors considered and 17-β estradiol concentrations. Referring to the assessed hematological and biochemical parameters, no statistically significant differences were observed in infected swine compared to the control group. Conclusion: Our results contribute to the enrichment of data available about the subject and could be useful for a deeper knowledge of the interaction between this parasite and its hosts. However, more aspects are still unclear, such as the effective response of downstream molecules from the same pathways to the variation of factors observed in this study either assessing how the same factors respond to Toxoplasma gondii infection in other host speciesand further analyses should be performed on other host species.
Collapse
Affiliation(s)
- Annamaria Castello
- Department of Catania, Istituto Zooprofilattico Sperimentale Della Sicilia, Catania, Italy
| | - Esterina Fazio
- Department of Veterinary Sciences, University of Messina, Polo Universitario dell'Annunziata, Messina, Italy
| | - Tiziana Alfonzetti
- Department of Catania, Istituto Zooprofilattico Sperimentale Della Sicilia, Catania, Italy
| | - Renato Paolo Giunta
- Department of Catania, Istituto Zooprofilattico Sperimentale Della Sicilia, Catania, Italy
| | - Antonio Salvaggio
- Department of Catania, Istituto Zooprofilattico Sperimentale Della Sicilia, Catania, Italy
| | - Alida Maria Ferlazzo
- Department of Veterinary Sciences, University of Messina, Polo Universitario dell'Annunziata, Messina, Italy
| | - Cristina Cravana
- Department of Veterinary Sciences, University of Messina, Polo Universitario dell'Annunziata, Messina, Italy
| | - Giuseppe Bruschetta
- Department of Veterinary Sciences, University of Messina, Polo Universitario dell'Annunziata, Messina, Italy
| | - Pietro Medica
- Department of Veterinary Sciences, University of Messina, Polo Universitario dell'Annunziata, Messina, Italy
| | | |
Collapse
|
12
|
Chen M, Cheng T, Xu C, Pan M, Wu J, Wang T, Wu D, Yan G, Wang C, Shao J. Sodium houttuyfonate enhances the mono-therapy of fluconazole on oropharyngeal candidiasis (OPC) through HIF-1α/IL-17 axis by inhibiting cAMP mediated filamentation in Candida albicans-Candida glabrata dual biofilms. Virulence 2022; 13:428-443. [PMID: 35195502 PMCID: PMC8890385 DOI: 10.1080/21505594.2022.2035066] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Candida albicans and Candida glabrata are two common opportunistic fungi that can be co-isolated in oropharyngeal candidiasis (OPC). Hypha is a hallmark of the biofilm formation of C. albicans, indispensable for the attachment of C. glabrata, which is seldom in mycelial morphology. Increasing evidence reveals a hypoxic microenvironment in interior fungal biofilms, reminding of a fact that inflammation is usually accompanied by oxygen deprivation. As a result, it is assumed that the disaggregation of hypha-mediated hypoxia of biofilms might be a solution to alleviate OPC. Based on this hypothesis, sodium houttuyfonate (SH), a well-identified traditional herbal compound with antifungal activity, is used in combination with fluconazole (FLU), a well-informed synthesized antimycotics, to investigate their impact on filamentation in C. albicans and C. glabrata dual biofilms and the underlying mechanism of their combined treatment on OPC. The results show that compared with the single therapy, SH plus FLU can inhibit the hyphal growth in the mixed biofilms in vitro, decrease the fungal burden of oral tissues and internal organs, restore mucosal epithelial integrity and function, and reduce hypoxic microenvironment and inflammation in a mice OPC model. The possible mechanism of the combined therapy of SH plus FLU can be attributed to the regulation of HIF-1α/IL-17A axis through direct abrogation of the dual Candida biofilm formation. This study highlights the role of HIF-1α/IL-17A axis and the promising application of SH as a sensitizer of conventional antifungals in the treatment of OPC.
Collapse
Affiliation(s)
- Mengli Chen
- Laboratory of Infection and Immunity, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, Anhui P. R, China
| | - Ting Cheng
- Laboratory of Infection and Immunity, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, Anhui P. R, China
| | - Chen Xu
- Laboratory of Infection and Immunity, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, Anhui P. R, China
| | - Min Pan
- Laboratory of Infection and Immunity, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, Anhui P. R, China
| | - Jiadi Wu
- Department of Anatomy, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, P. R, China
| | - Tianming Wang
- Laboratory of Infection and Immunity, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, Anhui P. R, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, P. R, China
| | - Daqiang Wu
- Laboratory of Infection and Immunity, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, Anhui P. R, China.,Institute of Integrated Traditional Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, Anhui P. R, China.,Cas Center for Excellence in Molecular Cell Sciences, Ministry of Education Key Laboratory for Membrane-less Organelles & Cellular Dynamics, Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, P.r, China
| | - Guiming Yan
- Laboratory of Infection and Immunity, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, Anhui P. R, China.,Institute of Integrated Traditional Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, Anhui P. R, China
| | - Changzhong Wang
- Laboratory of Infection and Immunity, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, Anhui P. R, China.,Institute of Integrated Traditional Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, Anhui P. R, China
| | - Jing Shao
- Laboratory of Infection and Immunity, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, Anhui P. R, China.,Institute of Integrated Traditional Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, Anhui P. R, China
| |
Collapse
|
13
|
The anti-parasite action of imidazole derivatives likely involves oxidative stress but not HIF-1α signaling. Chem Biol Interact 2021; 349:109676. [PMID: 34592218 DOI: 10.1016/j.cbi.2021.109676] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 09/09/2021] [Accepted: 09/22/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND Therapeutic options for toxoplasmosis are limited. This fact underscores ongoing research efforts to identify and develop better therapy. Previously, we reported the anti-parasitic potential of a new series of derivatives of imidazole. OBJECTIVE In the current investigation, we attempted the investigation of the possible action mechanism of few promising anti-parasite imidazole derivatives namely C1 (bis-imidazole), C2 (phenyl-substituted 1H-imidazole) and C3 (thiophene-imidazole) METHODS: We evaluated if oxidative stress, hypoxia as well as metabolic reprogramming of host l-tryptophan pathway form part of the parasite growth inhibition by imidazoles. Anti-parasite assay was performed for imidazoles at concentrations ranging from 0 to 10 μM, while pyrimethamine was used as reference drug to validate assay. RESULTS Imidazole compounds restricted parasite growth dose-dependently. However, in the presence of an antioxidant (Trolox), l-tryptophan and/or CoCl2 (chemical inducer of hypoxia), the growth inhibitory efficacy of imidazoles was appreciably abolished. Further, imidazole treatment led to elevated level of reactive oxygen species, while reducing parasite mitochondrial membrane potential compared with control. In contrast, imidazole had no effect on host HIF-1α level suggesting its exclusion in the anti-parasite action. CONCLUSION Taken together, imidazole-based compounds might restrict parasite growth by causing oxidative stress. The findings provide new insight on the likely biochemical mechanisms of imidazoles as prospective anti-parasite therapy. Data gives new perspective that not only underscores the anti-parasite prospects of imidazoles, but implicates the host l-tryptophan pathway as a feasible treatment option for T. gondii infections.
Collapse
|
14
|
Bichiou H, Bouabid C, Rabhi I, Guizani-Tabbane L. Transcription Factors Interplay Orchestrates the Immune-Metabolic Response of Leishmania Infected Macrophages. Front Cell Infect Microbiol 2021; 11:660415. [PMID: 33898331 PMCID: PMC8058464 DOI: 10.3389/fcimb.2021.660415] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 03/22/2021] [Indexed: 12/24/2022] Open
Abstract
Leishmaniasis is a group of heterogenous diseases considered as an important public health problem in several countries. This neglected disease is caused by over 20 parasite species of the protozoa belonging to the Leishmania genus and is spread by the bite of a female phlebotomine sandfly. Depending on the parasite specie and the immune status of the patient, leishmaniasis can present a wide spectrum of clinical manifestations. As an obligate intracellular parasite, Leishmania colonize phagocytic cells, mainly the macrophages that orchestrate the host immune response and determine the fate of the infection. Once inside macrophages, Leishmania triggers different signaling pathways that regulate the immune and metabolic response of the host cells. Various transcription factors regulate such immune-metabolic responses and the associated leishmanicidal and inflammatory reaction against the invading parasite. In this review, we will highlight the most important transcription factors involved in these responses, their interactions and their impact on the establishment and the progression of the immune response along with their effect on the physiopathology of the disease.
Collapse
Affiliation(s)
- Haifa Bichiou
- Laboratory of Medical Parasitology, Biotechnology and Biomolecules (PMBB), Institut Pasteur de Tunis, Tunis, Tunisia.,Faculty of Sciences of Tunis, Université de Tunis El Manar, Tunis, Tunisia
| | - Cyrine Bouabid
- Laboratory of Medical Parasitology, Biotechnology and Biomolecules (PMBB), Institut Pasteur de Tunis, Tunis, Tunisia.,Faculty of Sciences of Tunis, Université de Tunis El Manar, Tunis, Tunisia
| | - Imen Rabhi
- Laboratory of Medical Parasitology, Biotechnology and Biomolecules (PMBB), Institut Pasteur de Tunis, Tunis, Tunisia.,Biotechnology Department, Higher Institute of Biotechnology at Sidi-Thabet (ISBST), Biotechpole Sidi-Thabet- University of Manouba, Tunis, Tunisia
| | - Lamia Guizani-Tabbane
- Laboratory of Medical Parasitology, Biotechnology and Biomolecules (PMBB), Institut Pasteur de Tunis, Tunis, Tunisia
| |
Collapse
|
15
|
Gossner A, Hassan MA. Transcriptional Analyses Identify Genes That Modulate Bovine Macrophage Response to Toxoplasma Infection and Immune Stimulation. Front Cell Infect Microbiol 2020; 10:437. [PMID: 33014886 PMCID: PMC7508302 DOI: 10.3389/fcimb.2020.00437] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 07/16/2020] [Indexed: 11/26/2022] Open
Abstract
The obligate intracellular parasite, Toxoplasma gondii, is highly prevalent among livestock species. Although cattle are generally resistant to Toxoplasma strains circulating in Europe and North America, the underlying mechanisms are largely unknown. Here, we report that bovine bone marrow-derived macrophage (BMDM) pre-stimulated with interferon gamma (IFNγ) restricts intracellular Toxoplasma growth independently of nitric oxide. While Toxoplasma promoted the expression of genes associated with alternative macrophage activation and lipid metabolism, IFNγ abrogated parasite-induced transcriptional responses and promoted the expression of genes linked to the classical macrophage activation phenotype. Additionally, several chemokines, including CCL22, that are linked to parasite-induced activation of the Wnt/β-catenin signaling were highly expressed in Toxoplasma-exposed naïve BMDMs. A chemical Wnt/β-catenin signaling pathway antagonist (IWR-1-endo) significantly reduced intracellular parasite burden in naïve BMDMs, suggesting that Toxoplasma activates this pathway to evade bovine macrophage anti-parasitic responses. Congruently, intracellular burden of a mutant Toxoplasma strain (RHΔASP5) that does not secrete dense granule proteins into the host cell, which is an essential requirement for parasite-induced activation of the Wnt/β-catenin pathway, was significantly reduced in naïve BMDMs. However, both the Wnt/β-catenin antagonist and RHASPΔ5 did not abolish parasite burden differences in naïve and IFNγ-stimulated BMDMs. Finally, we observed that parasites infecting IFNγ-stimulated BMDMs largely express genes associated with the slow dividing bradyzoite stage. Overall, this study provides novel insights into bovine macrophage transcriptional response to Toxoplasma. It establishes a foundation for a mechanistic analysis IFNγ-induced bovine anti-Toxoplasma responses and the counteracting Toxoplasma survival strategies.
Collapse
Affiliation(s)
- Anton Gossner
- Division of Infection and Immunity, The Roslin Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Musa A Hassan
- Division of Infection and Immunity, The Roslin Institute, The University of Edinburgh, Edinburgh, United Kingdom.,Centre for Tropical Livestock Genetics and Health, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
16
|
Repurposing bioenergetic modulators against protozoan parasites responsible for tropical diseases. INTERNATIONAL JOURNAL FOR PARASITOLOGY-DRUGS AND DRUG RESISTANCE 2020; 14:17-27. [PMID: 32829099 PMCID: PMC7452664 DOI: 10.1016/j.ijpddr.2020.07.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 07/17/2020] [Accepted: 07/20/2020] [Indexed: 12/30/2022]
Abstract
Malaria, leishmaniasis and trypanosomiasis are arthropod-borne, parasitic diseases that constitute a major global health problem. They are generally found in developing countries, where lack of access to preventive tools and treatment hinders their management. Because these parasites share an increased demand on glucose consumption with most cancer cells, six compounds used in anti-tumoral research were selected to be tested as antiparasitic agents in in vitro models of Leishmania infantum, Trypanosoma brucei, T. cruzi, and Plasmodium falciparum: dichloroacetic acid (DCA), 3-bromopyruvic acid (3BP), 2-deoxy-D-glucose (2DG), lonidamine (LND), metformin (MET), and sirolimus (SIR). No parasite-killing activity was found in L. infantum promastigotes, whereas DCA and 3BP reduced the burden of intra-macrophagic amastigotes. For T. brucei all selected compounds, but 2DG, decreased parasite survival. DCA, 2DG, LND and MET showed parasite-killing activity in T. cruzi. Finally, anti-plasmodial activity was found for DCA, 2DG, LND, MET and SIR. These results reinforce the hypothesis that drugs with proven efficacy in the treatment of cancer by interfering with ATP production, proliferation, and survival cell strategies might be useful in treating threatening parasitic diseases and provide new opportunities for their repurposing. Parasitic diseases are prevalent among the poorest of the poor. Some parasitic protists degrade glucose into CO2 even aerobically making this a target. Degrading glucose into CO2 (Warburg effect) is also characteristic for cancer cells. Repurposing cancer glycolysis blockers may provide cost-effective treatments for the poorest.
Collapse
|
17
|
Delling C, Daugschies A, Bangoura B, Dengler F. Cryptosporidium parvum alters glucose transport mechanisms in infected enterocytes. Parasitol Res 2019; 118:3429-3441. [PMID: 31667591 DOI: 10.1007/s00436-019-06471-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 09/24/2019] [Indexed: 11/25/2022]
Abstract
The parasite Cryptosporidium parvum Tyzzer 1912 destroys parts of the intestinal brush border membrane which is important for the uptake of nutrients like glucose. In this study, glucose transport mechanisms of the host cells (IPEC-J2 cells) infected by C. parvum were investigated. The mRNA expression levels of glucose transporters (GLUT) 1 and 2 and Na+-coupled glucose transporter (SGLT) 1 were compared in infected and uninfected cells over an infection time of 24-96 h by RT-qPCR. Furthermore, the protein expression of SGLT 1 and GLUT 2 was quantified in western blot studies. While the protein expression of SGLT 1 was not altered in infected cells, mRNA expression of SGLT 1 and GLUT 1 was significantly increased 24 h p. i. and decreased 96 h p. i. The mRNA expression of GLUT 2 was significantly decreased 24 h, 72 h, and 96 h p. i. and also correlated significantly with the infection dose at 72 h p. i. In contrast to that, the protein expression of GLUT 2 was significantly increased 48 h p. i., associated with a significantly higher intracellular glucose level in infected cells compared with control cells at that time point of infection. This points to an adaptation of the host cells' glucose uptake taking place in the acute phase of the infection. A better understanding of these molecular mechanisms following a C. parvum infection may probably lead to an improvement of therapy strategies in the future.
Collapse
Affiliation(s)
- Cora Delling
- Institute of Parasitology, Faculty of Veterinary Medicine, University of Leipzig, An den Tierkliniken 35, 04103, Leipzig, Germany.
| | - Arwid Daugschies
- Institute of Parasitology, Faculty of Veterinary Medicine, University of Leipzig, An den Tierkliniken 35, 04103, Leipzig, Germany
- Albrecht Daniel Thaer Institute, An den Tierkliniken 29, 04103, Leipzig, Germany
| | - Berit Bangoura
- Wyoming State Veterinary Laboratory, Department of Veterinary Sciences, University of Wyoming, 1174 Snowy Range Road, Laramie, WY, 82070, USA
| | - Franziska Dengler
- Institute of Veterinary Physiology, Faculty of Veterinary Medicine, University of Leipzig, An den Tierkliniken 7, 04103, Leipzig, Germany
| |
Collapse
|
18
|
Deng Y, Wu T, Zhai SQ, Li CH. Recent progress on anti-Toxoplasma drugs discovery: Design, synthesis and screening. Eur J Med Chem 2019; 183:111711. [PMID: 31585276 DOI: 10.1016/j.ejmech.2019.111711] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 08/27/2019] [Accepted: 09/16/2019] [Indexed: 01/30/2023]
Abstract
Toxoplasma gondii severely threaten the health of immunocompromised patients and pregnant women as this parasite can cause several disease, including brain and eye disease. Current treatment for toxoplasmosis commonly have high cytotoxic side effects on host and require long durations ranging from one week to more than one year. The regiments lack efficacy to eradicate T. gondii tissue cysts to cure chromic infection results in the needs for long treatment and relapsing disease. In addition, there has not been approved drugs for treating the pregnant women infected by T. gondii. Moreover, Toxoplasma vaccine researches face a wide variety of challenges. Developing high efficient and low toxic agents against T. gondii is urgent and important. Over the last decade, tremendous progress have been made in identifying and developing novel compounds for the treatment of toxoplasmosis. This review summarized and discussed recent advances between 2009 and 2019 in exploring effective agents against T. gondii from five aspects of drug discovery.
Collapse
Affiliation(s)
- Yu Deng
- Institute of Veterinary Sciences & Pharmaceuticals, Chongqing Academy of Animal Sciences, Rongchang, 402460, China
| | - Tao Wu
- Institute of Veterinary Sciences & Pharmaceuticals, Chongqing Academy of Animal Sciences, Rongchang, 402460, China
| | - Shao-Qin Zhai
- Institute of Veterinary Sciences & Pharmaceuticals, Chongqing Academy of Animal Sciences, Rongchang, 402460, China
| | - Cheng-Hong Li
- Institute of Veterinary Sciences & Pharmaceuticals, Chongqing Academy of Animal Sciences, Rongchang, 402460, China.
| |
Collapse
|
19
|
Baptista CG, Lis A, Deng B, Gas-Pascual E, Dittmar A, Sigurdson W, West CM, Blader IJ. Toxoplasma F-box protein 1 is required for daughter cell scaffold function during parasite replication. PLoS Pathog 2019; 15:e1007946. [PMID: 31348812 PMCID: PMC6685633 DOI: 10.1371/journal.ppat.1007946] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 08/07/2019] [Accepted: 06/27/2019] [Indexed: 01/06/2023] Open
Abstract
By binding to the adaptor protein SKP1 and serving as substrate receptors for the SKP1 Cullin, F-box E3 ubiquitin ligase complex, F-box proteins regulate critical cellular processes including cell cycle progression and membrane trafficking. While F-box proteins are conserved throughout eukaryotes and are well studied in yeast, plants, and animals, studies in parasitic protozoa are lagging. We have identified eighteen putative F-box proteins in the Toxoplasma genome of which four have predicted homologs in Plasmodium. Two of the conserved F-box proteins were demonstrated to be important for Toxoplasma fitness and here we focus on an F-box protein, named TgFBXO1, because it is the most highly expressed by replicative tachyzoites and was also identified in an interactome screen as a Toxoplasma SKP1 binding protein. TgFBXO1 interacts with Toxoplasma SKP1 confirming it as a bona fide F-box protein. In interphase parasites, TgFBXO1 is a component of the Inner Membrane Complex (IMC), which is an organelle that underlies the plasma membrane. Early during replication, TgFBXO1 localizes to the developing daughter cell scaffold, which is the site where the daughter cell IMC and microtubules form and extend from. TgFBXO1 localization to the daughter cell scaffold required centrosome duplication but before kinetochore separation was completed. Daughter cell scaffold localization required TgFBXO1 N-myristoylation and was dependent on the small molecular weight GTPase, TgRab11b. Finally, we demonstrate that TgFBXO1 is required for parasite growth due to its function as a daughter cell scaffold effector. TgFBXO1 is the first F-box protein to be studied in apicomplexan parasites and represents the first protein demonstrated to be important for daughter cell scaffold function.
Collapse
Affiliation(s)
- Carlos Gustavo Baptista
- Department of Microbiology and Immunology, University at Buffalo School of Medicine, Buffalo, New York, United States of America
| | - Agnieszka Lis
- Department of Microbiology and Immunology, University at Buffalo School of Medicine, Buffalo, New York, United States of America
| | - Bowen Deng
- Department of Biochemistry & Molecular Biology, University of Georgia, Athens, Georgia, United States of America
| | - Elisabet Gas-Pascual
- Department of Biochemistry & Molecular Biology, University of Georgia, Athens, Georgia, United States of America
| | - Ashley Dittmar
- Department of Microbiology and Immunology, University at Buffalo School of Medicine, Buffalo, New York, United States of America
| | - Wade Sigurdson
- Department of Physiology and Biophysics, University at Buffalo School of Medicine, Buffalo, New York, United States of America
| | - Christopher M. West
- Department of Biochemistry & Molecular Biology, University of Georgia, Athens, Georgia, United States of America
| | - Ira J. Blader
- Department of Microbiology and Immunology, University at Buffalo School of Medicine, Buffalo, New York, United States of America
| |
Collapse
|
20
|
The HIF-1α/LC3-II Axis Impacts Fungal Immunity in Human Macrophages. Infect Immun 2019; 87:IAI.00125-19. [PMID: 31036602 PMCID: PMC6589057 DOI: 10.1128/iai.00125-19] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 04/16/2019] [Indexed: 01/03/2023] Open
Abstract
The fungal pathogen Histoplasma capsulatum causes a spectrum of disease, ranging from local pulmonary infection to disseminated disease. The organism seeks residence in macrophages, which are permissive for its survival. Hypoxia-inducible factor 1α (HIF-1α), a principal regulator of innate immunity to pathogens, is necessary for macrophage-mediated immunity to H. capsulatum in mice. In the present study, we analyzed the effect of HIF-1α in human macrophages infected with this fungus. HIF-1α stabilization was detected in peripheral blood monocyte-derived macrophages at 2 to 24 h after infection with viable yeast cells. Further, host mitochondrial respiration and glycolysis were enhanced. In contrast, heat-killed yeasts induced early, but not later, stabilization of HIF-1α. Since the absence of HIF-1α is detrimental to host control of infection, we asked if large amounts of HIF-1α protein, exceeding those induced by H. capsulatum, altered macrophage responses to this pathogen. Exposure of infected macrophages to an HIF-1α stabilizer significantly reduced recovery of H. capsulatum from macrophages and produced a decrement in mitochondrial respiration and glycolysis compared to those of controls. We observed recruitment of the autophagy-related protein LC3-II to the phagosome, whereas enhancing HIF-1α reduced phagosomal decoration. This finding suggested that H. capsulatum exploited an autophagic process to survive. In support of this assertion, inhibition of autophagy activated macrophages to limit intracellular growth of H. capsulatum Thus, enhancement of HIF-1α creates a hostile environment for yeast cells in human macrophages by interrupting the ability of the pathogen to provoke host cell autophagy.
Collapse
|
21
|
Lima TS, Lodoen MB. Mechanisms of Human Innate Immune Evasion by Toxoplasma gondii. Front Cell Infect Microbiol 2019; 9:103. [PMID: 31041194 PMCID: PMC6476913 DOI: 10.3389/fcimb.2019.00103] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 03/25/2019] [Indexed: 12/16/2022] Open
Abstract
Toxoplasma gondii is an intracellular protozoan parasite of global importance that can remarkably infect, survive, and replicate in nearly all mammalian cells. Notably, 110 years after its discovery, Toxoplasmosis is still a neglected parasitic infection. Although most human infections with T. gondii are mild or asymptomatic, T. gondii infection can result in life-threatening disease in immunocompromised individuals and in the developing fetus due to congenital infection, underscoring the role of the host immune system in controlling the parasite. Recent evidence indicates that T. gondii elicits a robust innate immune response during infection. Interestingly, however, T. gondii has evolved strategies to successfully bypass or manipulate the immune system and establish a life-long infection in infected hosts. In particular, T. gondii manipulates host immunity through the control of host gene transcription and dysregulation of signaling pathways that result in modulation of cell adhesion and migration, secretion of immunoregulatory cytokines, production of microbicidal molecules, and apoptosis. Many of these host-pathogen interactions are governed by parasite effector proteins secreted from the apical secretory organelles, including the rhoptries and dense granules. Here, we review recent findings on mechanisms by which T. gondii evades host innate immunity, with a focus on parasite evasion of the human innate immune system.
Collapse
Affiliation(s)
- Tatiane S Lima
- Department of Molecular Biology and Biochemistry and the Institute for Immunology, University of California, Irvine, Irvine, CA, United States
| | - Melissa B Lodoen
- Department of Molecular Biology and Biochemistry and the Institute for Immunology, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
22
|
Florimond C, Cordonnier C, Taujale R, van der Wel H, Kannan N, West CM, Blader IJ. A Toxoplasma Prolyl Hydroxylase Mediates Oxygen Stress Responses by Regulating Translation Elongation. mBio 2019; 10:e00234-19. [PMID: 30914506 PMCID: PMC6437050 DOI: 10.1128/mbio.00234-19] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Accepted: 02/08/2019] [Indexed: 02/08/2023] Open
Abstract
As the protozoan parasite Toxoplasma gondii disseminates through its host, it responds to environmental changes by altering its gene expression, metabolism, and other processes. Oxygen is one variable environmental factor, and properly adapting to changes in oxygen levels is critical to prevent the accumulation of reactive oxygen species and other cytotoxic factors. Thus, oxygen-sensing proteins are important, and among these, 2-oxoglutarate-dependent prolyl hydroxylases are highly conserved throughout evolution. Toxoplasma expresses two such enzymes, TgPHYa, which regulates the SCF-ubiquitin ligase complex, and TgPHYb. To characterize TgPHYb, we created a Toxoplasma strain that conditionally expresses TgPHYb and report that TgPHYb is required for optimal parasite growth under normal growth conditions. However, exposing TgPHYb-depleted parasites to extracellular stress leads to severe decreases in parasite invasion, which is likely due to decreased abundance of parasite adhesins. Adhesin protein abundance is reduced in TgPHYb-depleted parasites as a result of inactivation of the protein synthesis elongation factor eEF2 that is accompanied by decreased rates of translational elongation. In contrast to most other oxygen-sensing proteins that mediate cellular responses to low O2, TgPHYb is specifically required for parasite growth and protein synthesis at high, but not low, O2 tensions as well as resistance to reactive oxygen species. In vivo, reduced TgPHYb expression leads to lower parasite burdens in oxygen-rich tissues. Taken together, these data identify TgPHYb as a sensor of high O2 levels, in contrast to TgPHYa, which supports the parasite at low O2IMPORTANCE Because oxygen plays a key role in the growth of many organisms, cells must know how much oxygen is available. O2-sensing proteins are therefore critical cellular factors, and prolyl hydroxylases are the best-studied type of O2-sensing proteins. In general, prolyl hydroxylases trigger cellular responses to decreased oxygen availability. But, how does a cell react to high levels of oxygen? Using the protozoan parasite Toxoplasma gondii, we discovered a prolyl hydroxylase that allows the parasite to grow at elevated oxygen levels and does so by regulating protein synthesis. Loss of this enzyme also reduces parasite burden in oxygen-rich tissues, indicating that sensing both high and low levels of oxygen impacts the growth and physiology of Toxoplasma.
Collapse
Affiliation(s)
- Celia Florimond
- Department of Microbiology and Immunology, University at Buffalo School of Medicine, Buffalo, New York, USA
| | - Charlotte Cordonnier
- Department of Microbiology and Immunology, University at Buffalo School of Medicine, Buffalo, New York, USA
| | - Rahil Taujale
- Institute of Bioinformatics, University of Georgia, Athens, Georgia, USA
| | - Hanke van der Wel
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
| | - Natarajan Kannan
- Institute of Bioinformatics, University of Georgia, Athens, Georgia, USA
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
| | - Christopher M West
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, USA
| | - Ira J Blader
- Department of Microbiology and Immunology, University at Buffalo School of Medicine, Buffalo, New York, USA
| |
Collapse
|
23
|
Lis A, Wiley M, Vaughan J, Gray PC, Blader IJ. The Activin Receptor, Activin-Like Kinase 4, Mediates Toxoplasma Gondii Activation of Hypoxia Inducible Factor-1. Front Cell Infect Microbiol 2019; 9:36. [PMID: 30891432 PMCID: PMC6411701 DOI: 10.3389/fcimb.2019.00036] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 02/04/2019] [Indexed: 12/11/2022] Open
Abstract
To grow and cause disease, intracellular pathogens modulate host cell processes. Identifying these processes as well as the mechanisms used by the pathogens to manipulate them is important for the development of more effective therapeutics. As an example, the intracellular parasite Toxoplasma gondii induces a wide variety of changes to its host cell, including altered membrane trafficking, cytoskeletal reorganization, and differential gene expression. Although several parasite molecules and their host targets have been identified that mediate- these changes, few are known to be required for parasite replication. One exception is the host cell transcription factor, hypoxia-inducible factor-1 (HIF-1), which is required for parasite replication in an oxygen-dependent manner. Toxoplasma activates HIF-1 by stabilizing the HIF-1α subunit, and this is dependent on the signaling from the Activin-Like Kinase (ALK) receptor superfamily. Here, we demonstrate that specific overexpression of the ALK family member, ALK4, increased HIF-1 activity in Toxoplasma-infected cells, and this increase required ALK4 kinase activity. Moreover, Toxoplasma stimulated ALK4 to dimerize with its co-receptor, ActRII, and also increased ALK4 kinase activity, thereby demonstrating that Toxoplasma activates the ALK4 receptor. ALK4 activation of HIF-1 was independent of canonical SMAD signaling but rather was dependent on the non-canonical Rho GTPase and JNK MAP kinase signaling pathways. Finally, Toxoplasma increased rates of ALK4 ubiquitination and turnover. These data provide the first evidence indicating that ALK4 signaling is a target for a microbial pathogen to manipulate its host cell.
Collapse
Affiliation(s)
- Agnieszka Lis
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| | - Mandi Wiley
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | | | | | - Ira J Blader
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| |
Collapse
|
24
|
Adeyemi OS, Otohinoyi DA, Awakan OJ, Adeyanju AA. Cellular apoptosis of HFF cells by inorganic nanoparticles not susceptible to modulation by Toxoplasma gondii infection in vitro. Toxicol In Vitro 2018; 54:280-285. [PMID: 30359720 DOI: 10.1016/j.tiv.2018.10.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 10/17/2018] [Accepted: 10/21/2018] [Indexed: 12/20/2022]
Abstract
The interaction of nanoparticles with living cells is becoming one of the urgent areas of collaborative research in materials science and biology. Previously, we showed that nanoparticles have promising anti-Toxoplasma gondii properties. Meanwhile, Toxoplasma gondii has been shown to avert apoptosis in host cells whereas nanoparticles have been implicated for apoptotic tendency. Therefore, in the present study, we assessed the in vitro apoptotic properties of inorganic nanoparticles in the absence or presence of Toxoplasma infection and/or small molecules used as metabolic modulators. Results showed that inorganic nanoparticles dose-dependently caused cellular apoptosis. However, in the presence of infection by Toxoplasma gondii, nanoparticles-induced cellular apoptosis was not mitigated. Likewise, use of several small molecules (anti-metabolites) as metabolic modulators either mildly or nearly failed to abate cellular apoptosis by nanoparticles. Taken together, our findings do not only confirm the apoptotic potential of inorganic nanoparticles but show evidence that cellular apoptosis by inorganic nanoparticles of gold and silver might not be susceptible to modulation by Toxoplasma gondii infection. The findings are new and contribute to deepen our understanding of the cellular interaction of nanoparticles.
Collapse
Affiliation(s)
- Oluyomi Stephen Adeyemi
- Medicinal Biochemistry, Nanomedicine & Toxicology Laboratory, Department of Biochemistry, Landmark University, PMB 10017 Omu-Aran, Nigeria.
| | | | - Oluwakemi Josephine Awakan
- Medicinal Biochemistry, Nanomedicine & Toxicology Laboratory, Department of Biochemistry, Landmark University, PMB 10017 Omu-Aran, Nigeria
| | | |
Collapse
|
25
|
Kumar V, Kumar A, Das S, Kumar A, Abhishek K, Verma S, Mandal A, Singh RK, Das P. Leishmania donovani Activates Hypoxia Inducible Factor-1α and miR-210 for Survival in Macrophages by Downregulation of NF-κB Mediated Pro-inflammatory Immune Response. Front Microbiol 2018; 9:385. [PMID: 29568285 PMCID: PMC5852103 DOI: 10.3389/fmicb.2018.00385] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 02/20/2018] [Indexed: 12/13/2022] Open
Abstract
Micro RNAs (miRNAs) have emerged as a critical regulator of several biological processes in both animals and plants. They have also been associated with regulation of immune responses in many human diseases during recent years. Visceral leishmaniasis (VL) is the most severe form of leishmaniasis, which is characterized by impairment of both innate and adaptive immune responses. In the present study, we observed that Leishmania establishes hypoxic environment in host macrophages that induces the expression of hypoxia inducible factor-1α (HIF-1α) and miRNA-210. Further, the expression of miRNA-210 was found to be dependent on activation of HIF-1α expression. The HIF-1α silencing by siRNA resulted in significantly (p < 0.001) decreased expression of miR-210 in parasites infected macrophages. We also observed that in siHIF-1α or antagomir-210 treated L. donovani infected macrophages, the parasitic load and percentage infectivity were significantly (p < 0.001) decreased. Furthermore, we found that inhibition of miR-210 leads to activation of NF-κB subunit p50, and it forms heterodimer with p65 and translocates into the nucleus from the cytoplasm. This significantly (p < 0.05) induced the transcription of pro-inflammatory cytokines genes such as TNF-α and IL-12 in miRNA-210 inhibited macrophages compared to uninhibited macrophages whereas the level of IL-10, an anti-inflammatory cytokine, was found to be significantly decreased (p < 0.001). These findings suggested that L. donovani infection induces hypoxic environment inside the macrophages that activates HIF-1α. Further, HIF-1α upregulates miR-210, which eventually establishes a suitable environment for the survival of parasite inside the host macrophages by downregulating NF-κB mediated pro-inflammatory immune responses.
Collapse
Affiliation(s)
- Vinod Kumar
- Division of Molecular Biology, Rajendra Memorial Research Institute of Medical Sciences (ICMR), Patna, India
| | - Ajay Kumar
- Division of Molecular Biology, Rajendra Memorial Research Institute of Medical Sciences (ICMR), Patna, India
| | - Sushmita Das
- Department of Microbiology, All India Institute of Medical Sciences, Patna, India
| | - Ashish Kumar
- Division of Molecular Biology, Rajendra Memorial Research Institute of Medical Sciences (ICMR), Patna, India
| | - Kumar Abhishek
- Division of Molecular Biology, Rajendra Memorial Research Institute of Medical Sciences (ICMR), Patna, India
| | - Sudha Verma
- Division of Molecular Biology, Rajendra Memorial Research Institute of Medical Sciences (ICMR), Patna, India
| | - Abhishek Mandal
- Division of Molecular Biology, Rajendra Memorial Research Institute of Medical Sciences (ICMR), Patna, India
| | - Rakesh K Singh
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Pradeep Das
- Division of Molecular Biology, Rajendra Memorial Research Institute of Medical Sciences (ICMR), Patna, India
| |
Collapse
|
26
|
Chakraborty S, Roy S, Mistry HU, Murthy S, George N, Bhandari V, Sharma P. Potential Sabotage of Host Cell Physiology by Apicomplexan Parasites for Their Survival Benefits. Front Immunol 2017; 8:1261. [PMID: 29081773 PMCID: PMC5645534 DOI: 10.3389/fimmu.2017.01261] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 09/21/2017] [Indexed: 12/26/2022] Open
Abstract
Plasmodium, Toxoplasma, Cryptosporidium, Babesia, and Theileria are the major apicomplexan parasites affecting humans or animals worldwide. These pathogens represent an excellent example of host manipulators who can overturn host signaling pathways for their survival. They infect different types of host cells and take charge of the host machinery to gain nutrients and prevent itself from host attack. The mechanisms by which these pathogens modulate the host signaling pathways are well studied for Plasmodium, Toxoplasma, Cryptosporidium, and Theileria, except for limited studies on Babesia. Theileria is a unique pathogen taking into account the way it modulates host cell transformation, resulting in its clonal expansion. These parasites majorly modulate similar host signaling pathways, however, the disease outcome and effect is different among them. In this review, we discuss the approaches of these apicomplexan to manipulate the host–parasite clearance pathways during infection, invasion, survival, and egress.
Collapse
Affiliation(s)
| | - Sonti Roy
- National Institute of Animal Biotechnology (NIAB-DBT), Hyderabad, India
| | - Hiral Uday Mistry
- National Institute of Animal Biotechnology (NIAB-DBT), Hyderabad, India
| | - Shweta Murthy
- National Institute of Animal Biotechnology (NIAB-DBT), Hyderabad, India
| | - Neena George
- National Institute of Animal Biotechnology (NIAB-DBT), Hyderabad, India
| | | | - Paresh Sharma
- National Institute of Animal Biotechnology (NIAB-DBT), Hyderabad, India
| |
Collapse
|
27
|
Adeyemi OS, Murata Y, Sugi T, Han Y, Kato K. Modulation of host HIF-1α activity and the tryptophan pathway contributes to the anti- Toxoplasma gondii potential of nanoparticles. Biochem Biophys Rep 2017; 11:84-92. [PMID: 28955772 PMCID: PMC5614707 DOI: 10.1016/j.bbrep.2017.07.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 07/04/2017] [Accepted: 07/04/2017] [Indexed: 01/24/2023] Open
Abstract
Background Toxoplasmosis constitutes a large global burden that is further exacerbated by the shortcomings of available therapeutic options, thus underscoring the urgent need for better anti-Toxoplasma gondii therapy or strategies. Recently, we showed that the anti-parasitic action of inorganic nanoparticles (NPs) could, in part, be due to changes in redox status as well as in the parasite mitochondrial membrane potential. Methods In the present study, we explored the in vitro mode of action of the anti-T. gondii effect of NPs by evaluating the contributions of host cellular processes, including the tryptophan pathway and hypoxia-inducing factor activity. NPs, at concentrations ranging from 0.01 to 200 µg/ml were screened for anti-parasitic activity. Sulfadiazine and/or pyrimethamine served as positive controls. Results We found that interplay among multiple host cellular processes, including HIF-1α activity, indoleamine 2,3-dioxygenase activity, and to a larger extent the tryptophan pathway, contribute to the anti-parasitic action of NPs. Conclusion To our knowledge, this is the first study to demonstrate an effect of NPs on the tryptophan and/or kynurenine pathway. General significance Our findings deepen our understanding of the mechanism of action of NPs and suggest that modulation of the host nutrient pool may represent a viable approach to the development of new and effective anti-parasitic agents. L-tryptophan relieved parasite growth restriction by nanoparticles. Nanoparticles modulate host HIF-1α and IDO activity while mildly activating kynurenine pathway.
Collapse
Affiliation(s)
- Oluyomi Stephen Adeyemi
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido 080-8555, Japan.,Medicinal Biochemistry and Toxicology Laboratory, Department of Biological Sciences, Landmark University, PMB 1004, Ipetu Road, Omu-Aran 370102, Nigeria
| | - Yuho Murata
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido 080-8555, Japan
| | - Tatsuki Sugi
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido 080-8555, Japan
| | - Yongmei Han
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido 080-8555, Japan
| | - Kentaro Kato
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido 080-8555, Japan
| |
Collapse
|
28
|
Molecular characterization of mudskipper (Boleophthalmus pectinirostris) hypoxia-inducible factor-1α (HIF-1α) and analysis of its function in monocytes/macrophages. PLoS One 2017; 12:e0177960. [PMID: 28542591 PMCID: PMC5443510 DOI: 10.1371/journal.pone.0177960] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 05/05/2017] [Indexed: 01/13/2023] Open
Abstract
Hypoxia-inducible factor-1α (HIF-1α) plays a critical role in immune and inflammatory responses and is important in controlling a variety of processes in monocytes and macrophages. However, very little information is available about the functions of HIF-1α in fish monocytes/macrophages (MO/MФ). In this study, the cDNA sequence of the mudskipper (Boleophthalmus pectinirostris) HIF-1α gene (BpHIF-1α) was determined. Sequence comparison and phylogenetic tree analysis showed that BpHIF-1α is clustered in the fish HIF-1α tree. Constitutive expression of BpHIF-1α mRNA was detected by real-time quantitative PCR in all tested tissues, and the expression was found to be dramatically increased in the skin, liver, spleen, and kidney after Edwardsiella tarda infection. In addition, hypoxia and infection induced the expression of the BpHIF-1α transcript and protein in MO/MФ, respectively. Hypoxia caused an increase in phagocytic and bactericidal capacity of mudskipper MO/MФ in a BpHIF-1α-dependent manner. BpHIF-1α induced an anti-inflammatory status in MO/MФ upon E. tarda infection and hypoxia. Therefore, BpHIF-1α may play a predominant role in the modulation of mudskipper MO/MФ function in the innate immune system.
Collapse
|
29
|
Abstract
Early electron microscopy studies revealed the elaborate cellular features that define the unique adaptations of apicomplexan parasites. Among these were bulbous rhoptry (ROP) organelles and small, dense granules (GRAs), both of which are secreted during invasion of host cells. These early morphological studies were followed by the exploration of the cellular contents of these secretory organelles, revealing them to be comprised of highly divergent protein families with few conserved domains or predicted functions. In parallel, studies on host-pathogen interactions identified many host signaling pathways that were mysteriously altered by infection. It was only with the advent of forward and reverse genetic strategies that the connections between individual parasite effectors and the specific host pathways that they targeted finally became clear. The current repertoire of parasite effectors includes ROP kinases and pseudokinases that are secreted during invasion and that block host immune pathways. Similarly, many secretory GRA proteins alter host gene expression by activating host transcription factors, through modification of chromatin, or by inducing small noncoding RNAs. These effectors highlight novel mechanisms by which T. gondii has learned to harness host signaling to favor intracellular survival and will guide future studies designed to uncover the additional complexity of this intricate host-pathogen interaction.
Collapse
|
30
|
Hu X, Binns D, Reese ML. The coccidian parasites Toxoplasma and Neospora dysregulate mammalian lipid droplet biogenesis. J Biol Chem 2017; 292:11009-11020. [PMID: 28487365 DOI: 10.1074/jbc.m116.768176] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 05/05/2017] [Indexed: 11/06/2022] Open
Abstract
Upon infection, the intracellular parasite Toxoplasma gondii co-opts critical functions of its host cell to avoid immune clearance and gain access to nutritional resources. One route by which Toxoplasma co-opts its host cell is through hijacking host organelles, many of which have roles in immunomodulation. Here we demonstrate that Toxoplasma infection results in increased biogenesis of host lipid droplets through rewiring of multiple components of host neutral lipid metabolism. These metabolic changes cause increased responsiveness of host cells to free fatty acid, leading to a radical increase in the esterification of free fatty acids into triacylglycerol. We identified c-Jun kinase and mammalian target of rapamycin (mTOR) as components of two distinct host signaling pathways that modulate the parasite-induced lipid droplet accumulation. We also found that, unlike many host processes dysregulated during Toxoplasma infection, the induction of lipid droplet generation is conserved not only during infection with genetically diverse Toxoplasma strains but also with Neospora caninum, which is closely related to Toxoplasma but has a restricted host range and uses different effector proteins to alter host signaling. Finally, by showing that a Toxoplasma strain deficient in exporting a specific class of effectors is unable to induce lipid droplet accumulation, we demonstrate that the parasite plays an active role in this process. These results indicate that, despite their different host ranges, Toxoplasma and Neospora use a conserved mechanism to co-opt these host organelles, which suggests that lipid droplets play a critical role at the coccidian host-pathogen interface.
Collapse
Affiliation(s)
- Xiaoyu Hu
- From the Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9041
| | - Derk Binns
- From the Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9041
| | - Michael L Reese
- From the Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9041
| |
Collapse
|
31
|
Devraj G, Beerlage C, Brüne B, Kempf VAJ. Hypoxia and HIF-1 activation in bacterial infections. Microbes Infect 2016; 19:144-156. [PMID: 27903434 DOI: 10.1016/j.micinf.2016.11.003] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 11/14/2016] [Accepted: 11/21/2016] [Indexed: 12/22/2022]
Abstract
For most of the living beings, oxygen is one of the essential elements required to sustain life. Deprivation of oxygen causes tissue hypoxia and this severely affects host cell and organ functions. Tissue hypoxia is a prominent microenvironmental condition occurring in infections and there is a body of evidence that hypoxia and inflammation are interconnected with each other. The primary key factor mediating the mammalian hypoxic response is hypoxia inducible factor (HIF)-1, which regulates oxygen homeostasis on cellular, tissue and organism level. Recent studies show that HIF-1 plays a central role in angiogenesis, cancer and cardiovascular disease but also in bacterial infections. Activation of HIF-1 depends on the nature of the pathogen and the characteristics of infections in certain hosts. Up to date, it is not completely clear whether the phenomenon of HIF-1 activation in infections has a protective or detrimental effect on the host. In this review, we give an overview of whether and how hypoxia and HIF-1 affect the course of infections.
Collapse
Affiliation(s)
- Gayatri Devraj
- Institute of Medical Microbiology and Infection Control, Goethe-University, Paul-Ehrlich-Str. 40, D-60596 Frankfurt am Main, Germany
| | - Christiane Beerlage
- Institute of Medical Microbiology and Infection Control, Goethe-University, Paul-Ehrlich-Str. 40, D-60596 Frankfurt am Main, Germany
| | - Bernhard Brüne
- Institute of Biochemistry I - Pathobiochemistry, Goethe-University, Theodor-Stern-Kai 7, D-60590 Frankfurt am Main, Germany
| | - Volkhard A J Kempf
- Institute of Medical Microbiology and Infection Control, Goethe-University, Paul-Ehrlich-Str. 40, D-60596 Frankfurt am Main, Germany.
| |
Collapse
|
32
|
Charpentier T, Hammami A, Stäger S. Hypoxia inducible factor 1α: A critical factor for the immune response to pathogens and Leishmania. Cell Immunol 2016; 309:42-49. [DOI: 10.1016/j.cellimm.2016.06.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 06/15/2016] [Accepted: 06/21/2016] [Indexed: 12/17/2022]
|
33
|
Friedrich D, Fecher RA, Rupp J, Deepe GS. Impact of HIF-1α and hypoxia on fungal growth characteristics and fungal immunity. Microbes Infect 2016; 19:204-209. [PMID: 27810563 DOI: 10.1016/j.micinf.2016.10.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 10/20/2016] [Accepted: 10/24/2016] [Indexed: 12/28/2022]
Abstract
Human pathogenic fungi are highly adaptable to a changing environment. The ability to adjust to low oxygen conditions is crucial for colonization and infection of the host. Recently, the impact of mammalian hypoxia-inducible factor-1α (HIF-1α) on fungal immunity has emerged. In this review, the role of hypoxia and HIF-1α in fungal infections is discussed regarding the innate immune response.
Collapse
Affiliation(s)
- Dirk Friedrich
- Department of Infectious Diseases and Microbiology, University of Lübeck, 23538 Lübeck, Germany.
| | - Roger A Fecher
- Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45220, USA
| | - Jan Rupp
- Department of Infectious Diseases and Microbiology, University of Lübeck, 23538 Lübeck, Germany
| | - George S Deepe
- Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; Medical Service, Veterans Affairs Hospital, Cincinnati, OH 45220, USA
| |
Collapse
|
34
|
Schatz V, Strüssmann Y, Mahnke A, Schley G, Waldner M, Ritter U, Wild J, Willam C, Dehne N, Brüne B, McNiff JM, Colegio OR, Bogdan C, Jantsch J. Myeloid Cell-Derived HIF-1α Promotes Control of Leishmania major. THE JOURNAL OF IMMUNOLOGY 2016; 197:4034-4041. [PMID: 27798163 DOI: 10.4049/jimmunol.1601080] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 09/15/2016] [Indexed: 12/30/2022]
Abstract
Hypoxia-inducible factor-1α (HIF-1α), which accumulates in mammalian host organisms during infection, supports the defense against microbial pathogens. However, whether and to what extent HIF-1α expressed by myeloid cells contributes to the innate immune response against Leishmania major parasites is unknown. We observed that Leishmania-infected humans and L. major-infected C57BL/6 mice exhibited substantial amounts of HIF-1α in acute cutaneous lesions. In vitro, HIF-1α was required for leishmanicidal activity and high-level NO production by IFN-γ/LPS-activated macrophages. Mice deficient for HIF-1α in their myeloid cell compartment had a more severe clinical course of infection and increased parasite burden in the skin lesions compared with wild-type controls. These findings were paralleled by reduced expression of type 2 NO synthase by lesional CD11b+ cells. Together, these data illustrate that HIF-1α is required for optimal innate leishmanicidal immune responses and, thereby, contributes to the cure of cutaneous leishmaniasis.
Collapse
Affiliation(s)
- Valentin Schatz
- Institute of Clinical Microbiology and Hygiene, University Hospital of Regensburg, University of Regensburg, 93053 Regensburg, Germany
| | - Yannic Strüssmann
- Institute of Clinical Microbiology and Hygiene, University Hospital of Regensburg, University of Regensburg, 93053 Regensburg, Germany
| | - Alexander Mahnke
- Mikrobiologisches Institut, Klinische Mikrobiologie, Immunologie, und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Gunnar Schley
- Medizinische Klinik 4, Nephrologie und Hypertensiologie, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Maximilian Waldner
- Medizinische Klinik 1, Gastroenterologie, Pneumologie und Endokrinologie, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Uwe Ritter
- Institute of Immunology, University of Regensburg, 93053 Regensburg, Germany
| | - Jens Wild
- Institute of Clinical Microbiology and Hygiene, University Hospital of Regensburg, University of Regensburg, 93053 Regensburg, Germany
| | - Carsten Willam
- Medizinische Klinik 4, Nephrologie und Hypertensiologie, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Nathalie Dehne
- Institute of Biochemistry I, Goethe-University Frankfurt, 60590 Frankfurt, Germany; and
| | - Bernhard Brüne
- Institute of Biochemistry I, Goethe-University Frankfurt, 60590 Frankfurt, Germany; and
| | - Jennifer M McNiff
- Department of Dermatology, Yale University School of Medicine, New Haven, CT 06510
| | - Oscar R Colegio
- Department of Dermatology, Yale University School of Medicine, New Haven, CT 06510
| | - Christian Bogdan
- Mikrobiologisches Institut, Klinische Mikrobiologie, Immunologie, und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Jonathan Jantsch
- Institute of Clinical Microbiology and Hygiene, University Hospital of Regensburg, University of Regensburg, 93053 Regensburg, Germany;
| |
Collapse
|
35
|
Fecher RA, Horwath MC, Friedrich D, Rupp J, Deepe GS. Inverse Correlation between IL-10 and HIF-1α in Macrophages Infected with Histoplasma capsulatum. THE JOURNAL OF IMMUNOLOGY 2016; 197:565-79. [PMID: 27271565 DOI: 10.4049/jimmunol.1600342] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 05/13/2016] [Indexed: 01/28/2023]
Abstract
Hypoxia-inducible factor (HIF)-1α is a transcription factor that regulates metabolic and immune response genes in the setting of low oxygen tension and inflammation. We investigated the function of HIF-1α in the host response to Histoplasma capsulatum because granulomas induced by this pathogenic fungus develop hypoxic microenvironments during the early adaptive immune response. In this study, we demonstrated that myeloid HIF-1α-deficient mice exhibited elevated fungal burden during the innate immune response (prior to 7 d postinfection) as well as decreased survival in response to a sublethal inoculum of H. capsulatum The absence of myeloid HIF-1α did not alter immune cell recruitment to the lungs of infected animals but was associated with an elevation of the anti-inflammatory cytokine IL-10. Treatment with mAb to IL-10 restored protective immunity to the mutant mice. Macrophages (Mϕs) constituted most IL-10-producing cells. Deletion of HIF-1α in neutrophils or dendritic cells did not alter fungal burden, thus implicating Mϕs as the pivotal cell in host resistance. HIF-1α was stabilized in Mϕs following infection. Increased activity of the transcription factor CREB in HIF-1α-deficient Mϕs drove IL-10 production in response to H. capsulatum IL-10 inhibited Mϕ control of fungal growth in response to the activating cytokine IFN-γ. Thus, we identified a critical function for Mϕ HIF-1α in tempering IL-10 production following infection. We established that transcriptional regulation of IL-10 by HIF-1α and CREB is critical for activation of Mϕs by IFN-γ and effective handling of H. capsulatum.
Collapse
Affiliation(s)
- Roger A Fecher
- Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati, OH 45267; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45220
| | - Michael C Horwath
- Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati, OH 45267; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45220
| | - Dirk Friedrich
- Department of Infectious Diseases and Microbiology, University of Lübeck, 23538 Lübeck, Germany; and
| | - Jan Rupp
- Department of Infectious Diseases and Microbiology, University of Lübeck, 23538 Lübeck, Germany; and
| | - George S Deepe
- Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati, OH 45267; Medical Service, Veterans Affairs Hospital, Cincinnati, OH 45220
| |
Collapse
|
36
|
Drug Repurposing Screening Identifies Novel Compounds That Effectively Inhibit Toxoplasma gondii Growth. mSphere 2016; 1:mSphere00042-15. [PMID: 27303726 PMCID: PMC4894684 DOI: 10.1128/msphere.00042-15] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 02/11/2016] [Indexed: 01/16/2023] Open
Abstract
The urgent need to develop new antimicrobial therapies has spawned the development of repurposing screens in which well-studied drugs and other types of compounds are tested for potential off-label uses. As a proof-of-principle screen to identify compounds effective against Toxoplasma gondii, we screened a collection of 1,120 compounds for the ability to significantly reduce Toxoplasma replication. A total of 94 compounds blocked parasite replication with 50% inhibitory concentrations of <5 µM. A significant number of these compounds are established inhibitors of dopamine or estrogen signaling. Follow-up experiments with the dopamine receptor inhibitor pimozide revealed that the drug impacted both parasite invasion and replication but did so independently of inhibition of dopamine or other neurotransmitter receptor signaling. Tamoxifen, which is an established inhibitor of the estrogen receptor, also reduced parasite invasion and replication. Even though Toxoplasma can activate the estrogen receptor, tamoxifen inhibits parasite growth independently of this transcription factor. Tamoxifen is also a potent inducer of autophagy, and we find that the drug stimulates recruitment of the autophagy marker light chain 3-green fluorescent protein onto the membrane of the vacuolar compartment in which the parasite resides and replicates. In contrast to other antiparasitic drugs, including pimozide, tamoxifen treatment of infected cells leads to a time-dependent elimination of intracellular parasites. Taken together, these data suggest that tamoxifen restricts Toxoplasma growth by inducing xenophagy or autophagic destruction of this obligate intracellular parasite. IMPORTANCE There is an urgent need to develop new therapies to treat microbial infections, and the repurposing of well-characterized compounds is emerging as one approach to achieving this goal. Using the protozoan parasite Toxoplasma gondii, we screened a library of 1,120 compounds and identified several compounds with significant antiparasitic activities. Among these were pimozide and tamoxifen, which are well-characterized drugs prescribed to treat patients with psychiatric disorders and breast cancer, respectively. The mechanisms by which these compounds target these disorders are known, but we show here that these drugs kill Toxoplasma through novel pathways, highlighting the potential utility of off-target effects in the treatment of infectious diseases.
Collapse
|
37
|
McMurtrey C, Trolle T, Sansom T, Remesh SG, Kaever T, Bardet W, Jackson K, McLeod R, Sette A, Nielsen M, Zajonc DM, Blader IJ, Peters B, Hildebrand W. Toxoplasma gondii peptide ligands open the gate of the HLA class I binding groove. eLife 2016; 5. [PMID: 26824387 PMCID: PMC4775218 DOI: 10.7554/elife.12556] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 01/28/2016] [Indexed: 01/10/2023] Open
Abstract
HLA class I presentation of pathogen-derived peptide ligands is essential for CD8+ T-cell recognition of Toxoplasma gondii infected cells. Currently, little data exist pertaining to peptides that are presented after T. gondii infection. Herein we purify HLA-A*02:01 complexes from T. gondii infected cells and characterize the peptide ligands using LCMS. We identify 195 T. gondii encoded ligands originating from both secreted and cytoplasmic proteins. Surprisingly, T. gondii ligands are significantly longer than uninfected host ligands, and these longer pathogen-derived peptides maintain a canonical N-terminal binding core yet exhibit a C-terminal extension of 1–30 amino acids. Structural analysis demonstrates that binding of extended peptides opens the HLA class I F’ pocket, allowing the C-terminal extension to protrude through one end of the binding groove. In summary, we demonstrate that unrealized structural flexibility makes MHC class I receptive to parasite-derived ligands that exhibit unique C-terminal peptide extensions. DOI:http://dx.doi.org/10.7554/eLife.12556.001 Toxoplasma gondii is a parasite that can infect most warm-blooded animals and cause a disease called toxoplasmosis. In humans, toxoplasmosis generally does not cause any noticeable symptoms, but it can cause serious problems in pregnant women and individuals with weakened immune systems. T. gondii is one of many parasites that hide within human cells in an attempt to avoid detection by the immune system. However, proteins called Human Leukocyte Antigens, or HLAs, can reveal hidden parasites by carrying small sections of them from the inside the infected cell to the cell’s surface. The immune system can then recognize the fragments as foreign and attack the parasite. HLAs typically pick up parasite fragments of a certain length, which enables the immune system to recognize that what is being displayed is a piece of parasite. By purifying HLAs from cells that have been infected by T. gondii, McMurtrey et al. have now learned more about which fragments of the parasite are displayed to the immune system. This analysis revealed that the parasite somehow manipulates the HLAs to carry parasite fragments that are considerably longer than can be explained with our current knowledge of how HLAs work. By using a technique called X-ray crystallography, McMurtrey et al. also show that the structure of the HLA assumes a previously unseen configuration when interacting with fragments of T. gondii. In the future, it will be important to understand how infected cells give rise to unusual structural configurations of HLAs and to unravel how these structures affect the immune system’s ability to fight infections. DOI:http://dx.doi.org/10.7554/eLife.12556.002
Collapse
Affiliation(s)
- Curtis McMurtrey
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, United States.,Pure MHC LLC, Austin, United States
| | - Thomas Trolle
- Center for Biological Sequence Analysis, Technical University of Denmark, Kongens Lyngby, Denmark.,La Jolla Institute for Allergy and Immunology, La Jolla, United States
| | - Tiffany Sansom
- Department of Microbiology and Immunology, University at Buffalo School of Medicine, Buffalo, United States
| | - Soumya G Remesh
- La Jolla Institute for Allergy and Immunology, La Jolla, United States
| | - Thomas Kaever
- La Jolla Institute for Allergy and Immunology, La Jolla, United States
| | - Wilfried Bardet
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, United States
| | - Kenneth Jackson
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, United States
| | - Rima McLeod
- University of Chicago, Chicago, United States
| | - Alessandro Sette
- La Jolla Institute for Allergy and Immunology, La Jolla, United States
| | - Morten Nielsen
- Center for Biological Sequence Analysis, Technical University of Denmark, Kongens Lyngby, Denmark.,Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín, Buenos Aires, Argentina
| | - Dirk M Zajonc
- La Jolla Institute for Allergy and Immunology, La Jolla, United States
| | - Ira J Blader
- Department of Microbiology and Immunology, University at Buffalo School of Medicine, Buffalo, United States
| | - Bjoern Peters
- La Jolla Institute for Allergy and Immunology, La Jolla, United States
| | - William Hildebrand
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, United States.,Pure MHC LLC, Austin, United States
| |
Collapse
|
38
|
Abstract
UNLABELLED During infections with the protozoan parasite Toxoplasma gondii, gamma-aminobutyric acid (GABA) is utilized as a carbon source for parasite metabolism and also to facilitate parasite dissemination by stimulating dendritic-cell motility. The best-recognized function for GABA, however, is its role in the nervous system as an inhibitory neurotransmitter that regulates the flow and timing of excitatory neurotransmission. When this pathway is altered, seizures develop. Human toxoplasmosis patients suffer from seizures, suggesting that Toxoplasma interferes with GABA signaling in the brain. Here, we show that while excitatory glutamatergic presynaptic proteins appeared normal, infection with type II ME49 Toxoplasma tissue cysts led to global changes in the distribution of glutamic acid decarboxylase 67 (GAD67), a key enzyme that catalyzes GABA synthesis in the brain. Alterations in GAD67 staining were not due to decreased expression but rather to a change from GAD67 clustering at presynaptic termini to a more diffuse localization throughout the neuropil. Consistent with a loss of GAD67 from the synaptic terminals, Toxoplasma-infected mice develop spontaneous seizures and are more susceptible to drugs that induce seizures by antagonizing GABA receptors. Interestingly, GABAergic protein mislocalization and the response to seizure-inducing drugs were observed in mice infected with type II ME49 but not type III CEP strain parasites, indicating a role for a polymorphic parasite factor(s) in regulating GABAergic synapses. Taken together, these data support a model in which seizures and other neurological complications seen in Toxoplasma-infected individuals are due, at least in part, to changes in GABAergic signaling. IMPORTANCE Infections of the central nervous system can cause seizures. While inflammation in the brain has been proposed to initiate the onset of the seizures, relatively little is known about how inflammation impacts the structure and function of the neurons. Here we used a parasite called Toxoplasma gondii that infects the brain and showed that seizures arise due to a defect in signaling of GABA, which is the neurotransmitter primarily responsible for preventing the onset of seizures.
Collapse
|
39
|
Abstract
Toxoplasmosis is the clinical and pathological consequence of acute infection with the obligate intracellular apicomplexan parasite Toxoplasma gondii. Symptoms result from tissue destruction that accompanies lytic parasite growth. This review updates current understanding of the host cell invasion, parasite replication, and eventual egress that constitute the lytic cycle, as well as the ways T. gondii manipulates host cells to ensure its survival. Since the publication of a previous iteration of this review 15 years ago, important advances have been made in our molecular understanding of parasite growth and mechanisms of host cell egress, and knowledge of the parasite's manipulation of the host has rapidly progressed. Here we cover molecular advances and current conceptual frameworks that include each of these topics, with an eye to what may be known 15 years from now.
Collapse
Affiliation(s)
- Ira J Blader
- Department of Microbiology and Immunology, University at Buffalo, Buffalo, New York 14127;
| | - Bradley I Coleman
- Department of Biology, Boston College, Chestnut Hill, Massachusetts 02467; , ,
| | - Chun-Ti Chen
- Department of Biology, Boston College, Chestnut Hill, Massachusetts 02467; , ,
| | - Marc-Jan Gubbels
- Department of Biology, Boston College, Chestnut Hill, Massachusetts 02467; , ,
| |
Collapse
|
40
|
Cheeseman K, Weitzman JB. Host–parasite interactions: an intimate epigenetic relationship. Cell Microbiol 2015; 17:1121-32. [DOI: 10.1111/cmi.12471] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 05/28/2015] [Accepted: 06/01/2015] [Indexed: 12/13/2022]
Affiliation(s)
- Kevin Cheeseman
- Sorbonne Paris Cité Epigenetics and Cell Fate UMR 7216 CNRS Université Paris Diderot Paris France
| | - Jonathan B. Weitzman
- Sorbonne Paris Cité Epigenetics and Cell Fate UMR 7216 CNRS Université Paris Diderot Paris France
| |
Collapse
|
41
|
siRNA Screening Identifies the Host Hexokinase 2 (HK2) Gene as an Important Hypoxia-Inducible Transcription Factor 1 (HIF-1) Target Gene in Toxoplasma gondii-Infected Cells. mBio 2015; 6:e00462. [PMID: 26106078 PMCID: PMC4479703 DOI: 10.1128/mbio.00462-15] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Although it is established that oxygen availability regulates cellular metabolism and growth, little is known regarding how intracellular pathogens use host factors to grow at physiological oxygen levels. Therefore, large-scale human small interfering RNA screening was performed to identify host genes important for growth of the intracellular protozoan parasite Toxoplasma gondii at tissue oxygen tensions. Among the genes identified by this screen, we focused on the hexokinase 2 (HK2) gene because its expression is regulated by hypoxia-inducible transcription factor 1 (HIF-1), which is important for Toxoplasma growth. Toxoplasma increases host HK2 transcript and protein levels in a HIF-1-dependent manner. In addition, parasite growth at 3% oxygen is restored in HIF-1-deficient cells transfected with HK2 expression plasmids. Both HIF-1 activation and HK2 expression were accompanied by increases in host glycolytic flux, suggesting that enhanced HK2 expression in parasite-infected cells is functionally significant. Parasite dependence on host HK2 and HIF-1 expression is not restricted to transformed cell lines, as both are required for parasite growth in nontransformed C2C12 myoblasts and HK2 is upregulated in vivo following infection. While HK2 is normally associated with the cytoplasmic face of the outer mitochondrial membrane at physiological O2 levels, HK2 relocalizes to the host cytoplasm following infection, a process that is required for parasite growth at 3% oxygen. Taken together, our findings show that HIF-1-dependent expression and relocalization of HK2 represent a novel mechanism by which Toxoplasma establishes its replicative niche at tissue oxygen tensions. Little is known regarding how the host cell contributes to the survival of the intracellular parasite Toxoplasma gondii at oxygen levels that mimic those found in tissues. Our previous work showed that Toxoplasma activates the expression of an oxygen-regulated transcription factor that is required for growth. Here, we report that Toxoplasma regulates the abundance and activity of a key host metabolic enzyme, hexokinase 2, by activating HIF-1 and by promoting dissociation of hexokinase 2 from the mitochondrial membrane. Collectively, our data reveal HIF-1/hexokinase 2 as a novel target for an intracellular pathogen that acts by reprograming the host cell’s metabolism to create an environment conducive for parasite replication at physiological oxygen levels.
Collapse
|
42
|
West CM, Blader IJ. Oxygen sensing by protozoans: how they catch their breath. Curr Opin Microbiol 2015; 26:41-7. [PMID: 25988702 DOI: 10.1016/j.mib.2015.04.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Accepted: 04/24/2015] [Indexed: 01/09/2023]
Abstract
Cells must know the local levels of available oxygen and either adapt accordingly or relocate to more favorable environments. Prolyl 4-hydroxylases (P4Hs) are emerging as universal cellular oxygen sensors. In animals, these oxygen sensors respond to decreased oxygen availability by up-regulating hypoxia-inducible transcription factors. In protozoa, the P4Hs appear to activate E3-SCF ubiquitin ligase complexes via a glycosylation-dependent mechanism, potentially to turn over their proteomes. Intracellular parasites are impacted by both types of oxygen-sensing pathways. Since parasites are exposed to diverse oxygen tensions during their life cycles, this review identifies emerging oxygen-sensing mechanisms and discusses how these mechanisms probably contribute to the regulation of unicellular eukaryotes.
Collapse
Affiliation(s)
- Christopher M West
- Department of Biochemistry & Molecular Biology, Oklahoma Center for Medical Glycobiology, University of Oklahoma Health Sciences Center, 975 NE 10th St., BRC 417, Oklahoma City, OK 73104, USA.
| | - Ira J Blader
- Department of Microbiology and Immunology, University at Buffalo School of Medicine, 347 Biomedical Research Building, 3435 Main Street, Buffalo, NY 14214, USA
| |
Collapse
|
43
|
Metheni M, Lombès A, Bouillaud F, Batteux F, Langsley G. HIF-1α induction, proliferation and glycolysis of Theileria-infected leukocytes. Cell Microbiol 2015; 17:467-72. [PMID: 25620534 DOI: 10.1111/cmi.12421] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 01/20/2015] [Accepted: 01/22/2015] [Indexed: 01/30/2023]
Abstract
Within 2 h of infection by Theileria annulata sporozoites, bovine macrophages display a two- to fourfold increase in transcription of hypoxia inducible factor (HIF-1α). Twenty hours post-invasion sporozoites develop into multi-nucleated macroschizonts that transform the infected macrophage into an immortalized, permanently proliferating, hyper-invasive and disease-causing leukaemia-like cell. Once immortalized Theileria-infected leukocytes can be propagated as cell lines and even though cultivated under normoxic conditions, both infected B cells and macrophages display sustained activation of HIF-1α. Attenuated macrophages used as live vaccines against tropical theileriosis also display HIF-1α activation even though they have lost their tumorigenic phenotype. Here, we review data that ascribes HIF-1α activation to the proliferation status of the infected leukocyte and discuss the possibility that Theileria may have lost its ability to render its host macrophage virulent due to continuous parasite replication in a high Reactive Oxygen Species (ROS) environment. We propose a model where uninfected macrophages have low levels of H2 O2 output, whereas virulent-infected macrophages produce high amounts of H2 O2 . Further increase in H2 O2 output leads to dampening of infected macrophage virulence, a characteristic of disease-resistant macrophages. At the same time exposure to H2 O2 sustains HIF-1α that induces the switch from mitochondrial oxidative phosphorylation to Warburg glycolysis, a metabolic shift that underpins uncontrolled infected macrophage proliferation. We propose that as macroschizonts develop into merozoites and infected macrophage proliferation arrests, HIF-1α levels will decrease and glycolysis will switch back from Warburg to oxidative glycolysis. As Theileria infection transforms its host leukocyte into an aggressive leukaemic-like cell, we propose that manipulating ROS levels, HIF-1α induction and oxidative over Warburg glycolysis could contribute to improved disease control. Finally, as excess amounts of H2 O2 drive virulent Theileria-infected macrophages towards attenuation it highlights how infection-induced pathology and redox balance are intimately linked.
Collapse
Affiliation(s)
- Mehdi Metheni
- Inserm U1016, Cnrs UMR8104, Cochin Institute, Paris, France; Laboratoire de Biologie Cellulaire Comparative des Apicomplexes, Faculté de Médicine, Université Paris Descartes - Sorbonne Paris Cité, France
| | | | | | | | | |
Collapse
|
44
|
Bhandari T, Nizet V. Hypoxia-Inducible Factor (HIF) as a Pharmacological Target for Prevention and Treatment of Infectious Diseases. Infect Dis Ther 2014; 3:159-74. [PMID: 25134687 PMCID: PMC4269623 DOI: 10.1007/s40121-014-0030-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2014] [Indexed: 02/07/2023] Open
Abstract
In the present era of ever-increasing antibiotic resistance and increasingly complex and immunosuppressed patient populations, physicians and scientists are seeking novel approaches to battle difficult infectious disease conditions. Development of a serious infection implies a failure of innate immune capabilities in the patient, and one may consider whether pharmacological strategies exist to correct and enhance innate immune cell function. Hypoxia-inducible factor-1 (HIF-1), the central regulator of the cellular response to hypoxic stress, has recently been recognized to control the activation state and key microbicidal functions of immune cells. HIF-1 boosting drugs are in clinical development for anemia and other indications, and could be repositioned as infectious disease therapeutics. With equal attention to opportunities and complexities, we review our current understanding of HIF-1 regulation of microbial host-pathogen interactions with an eye toward future drug development.
Collapse
Affiliation(s)
- Tamara Bhandari
- Center for Immunity, Infection and Inflammation, Department of Pediatrics and Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, USA
| | - Victor Nizet
- Center for Immunity, Infection and Inflammation, Department of Pediatrics and Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, USA.
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, USA.
- Center for Immunity, Infection and Inflammation, Medical Sciences Research 4113, University of California, San Diego, 9500 Gilman Drive, MC 0760, La Jolla, CA, 92093-0760, USA.
| |
Collapse
|
45
|
Toxoplasma gondii development of its replicative niche: in its host cell and beyond. EUKARYOTIC CELL 2014; 13:965-76. [PMID: 24951442 DOI: 10.1128/ec.00081-14] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Intracellular pathogens can replicate efficiently only after they manipulate and modify their host cells to create an environment conducive to replication. While diverse cellular pathways are targeted by different pathogens, metabolism, membrane and cytoskeletal architecture formation, and cell death are the three primary cellular processes that are modified by infections. Toxoplasma gondii is an obligate intracellular protozoan that infects ∼30% of the world's population and causes severe and life-threatening disease in developing fetuses, in immune-comprised patients, and in certain otherwise healthy individuals who are primarily found in South America. The high prevalence of Toxoplasma in humans is in large part a result of its ability to modulate these three host cell processes. Here, we highlight recent work defining the mechanisms by which Toxoplasma interacts with these processes. In addition, we hypothesize why some processes are modified not only in the infected host cell but also in neighboring uninfected cells.
Collapse
|
46
|
Brown KM, Suvorova E, Farrell A, McLain A, Dittmar A, Wiley GB, Marth G, Gaffney PM, Gubbels MJ, White M, Blader IJ. Forward genetic screening identifies a small molecule that blocks Toxoplasma gondii growth by inhibiting both host- and parasite-encoded kinases. PLoS Pathog 2014; 10:e1004180. [PMID: 24945800 PMCID: PMC4055737 DOI: 10.1371/journal.ppat.1004180] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 04/28/2014] [Indexed: 02/02/2023] Open
Abstract
The simultaneous targeting of host and pathogen processes represents an untapped approach for the treatment of intracellular infections. Hypoxia-inducible factor-1 (HIF-1) is a host cell transcription factor that is activated by and required for the growth of the intracellular protozoan parasite Toxoplasma gondii at physiological oxygen levels. Parasite activation of HIF-1 is blocked by inhibiting the family of closely related Activin-Like Kinase (ALK) host cell receptors ALK4, ALK5, and ALK7, which was determined in part by use of an ALK4,5,7 inhibitor named SB505124. Besides inhibiting HIF-1 activation, SB505124 also potently blocks parasite replication under normoxic conditions. To determine whether SB505124 inhibition of parasite growth was exclusively due to inhibition of ALK4,5,7 or because the drug inhibited a second kinase, SB505124-resistant parasites were isolated by chemical mutagenesis. Whole-genome sequencing of these mutants revealed mutations in the Toxoplasma MAP kinase, TgMAPK1. Allelic replacement of mutant TgMAPK1 alleles into wild-type parasites was sufficient to confer SB505124 resistance. SB505124 independently impacts TgMAPK1 and ALK4,5,7 signaling since drug resistant parasites could not activate HIF-1 in the presence of SB505124 or grow in HIF-1 deficient cells. In addition, TgMAPK1 kinase activity is inhibited by SB505124. Finally, mice treated with SB505124 had significantly lower tissue burdens following Toxoplasma infection. These data therefore identify SB505124 as a novel small molecule inhibitor that acts by inhibiting two distinct targets, host HIF-1 and TgMAPK1. Understanding how a compound blocks growth of an intracellular pathogen is important not only for developing these compounds into drugs that can be prescribed to patients, but also because these data will likely provide novel insight into the biology of these pathogens. Forward genetic screens are one established approach towards defining these mechanisms. But performing these screens with intracellular parasites has been limited not only because of technical limitations but also because the compounds may have off-target effects in either the host or parasite. Here, we report the first compound that kills a pathogen by simultaneously inhibiting distinct host- and parasite-encoded targets. Because developing drug resistance simultaneously to two targets is less likely, this work may highlight a new approach to antimicrobial drug discovery.
Collapse
Affiliation(s)
- Kevin M Brown
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Elena Suvorova
- Departments of Molecular Medicine & Global Health, University of South Florida, Tampa, Florida, United States of America
| | - Andrew Farrell
- Department of Biology, Boston College, Chestnut Hill, Massachusetts, United States of America
| | - Aaron McLain
- Department of Microbiology and Immunology, University at Buffalo, Buffalo, New York, United States of America
| | - Ashley Dittmar
- Department of Microbiology and Immunology, University at Buffalo, Buffalo, New York, United States of America
| | - Graham B Wiley
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States of America
| | - Gabor Marth
- Department of Biology, Boston College, Chestnut Hill, Massachusetts, United States of America
| | - Patrick M Gaffney
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States of America
| | - Marc Jan Gubbels
- Department of Biology, Boston College, Chestnut Hill, Massachusetts, United States of America
| | - Michael White
- Departments of Molecular Medicine & Global Health, University of South Florida, Tampa, Florida, United States of America
| | - Ira J Blader
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America; Department of Microbiology and Immunology, University at Buffalo, Buffalo, New York, United States of America
| |
Collapse
|
47
|
Metheni M, Echebli N, Chaussepied M, Ransy C, Chéreau C, Jensen K, Glass E, Batteux F, Bouillaud F, Langsley G. The level of H₂O₂ type oxidative stress regulates virulence of Theileria-transformed leukocytes. Cell Microbiol 2013; 16:269-79. [PMID: 24112286 PMCID: PMC3906831 DOI: 10.1111/cmi.12218] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Revised: 08/30/2013] [Accepted: 09/20/2013] [Indexed: 02/07/2023]
Abstract
Theileria annulata infects predominantly macrophages, and to a lesser extent B cells, and causes a widespread disease of cattle called tropical theileriosis. Disease-causing infected macrophages are aggressively invasive, but this virulence trait can be attenuated by long-term culture. Attenuated macrophages are used as live vaccines against tropical theileriosis and via their characterization one gains insights into what host cell trait is altered concomitant with loss of virulence. We established that sporozoite infection of monocytes rapidly induces hif1-α transcription and that constitutive induction of HIF-1α in transformed leukocytes is parasite-dependent. In both infectedmacrophages and B cells induction of HIF-1α activates transcription of its target genes that drive host cells to perform Warburg-like glycolysis. We propose that Theileria-infected leukocytes maintain a HIF-1α-driven transcriptional programme typical of Warburg glycolysis in order to reduce as much as possible host cell H2O2 type oxidative stress. However, in attenuated macrophages H2O2 production increases and HIF-1α levels consequently remained high, even though adhesion and aggressive invasiveness diminished. This indicates that Theileria infection generates a host leukocytes hypoxic response that if not properly controlled leads to loss of virulence.
Collapse
Affiliation(s)
- Mehdi Metheni
- Laboratoire de Biologie Cellulaire Comparative des Apicomplexes, Faculté de Médicine, Université Paris Descartes - Sorbonne Paris Cité, Paris, France; Inserm U1016, Cnrs UMR8104, Cochin Institute, Paris, 75014, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Theileria induces oxidative stress and HIF1α activation that are essential for host leukocyte transformation. Oncogene 2013; 33:1809-17. [PMID: 23665677 DOI: 10.1038/onc.2013.134] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Revised: 02/27/2013] [Accepted: 03/04/2013] [Indexed: 02/06/2023]
Abstract
Complex links between infection and cancer suggest that we still can learn much about tumorigenesis by studying how infectious agents hijack the host cell machinery. We studied the effects of an intracellular parasite called Theileria that infects bovine leukocytes and turns them into invasive cancer-like cells. We investigated the host cells pathways that are deregulated in infected leukocytes and might link infection and lymphoproliferative disease. We show that intracellular Theileria parasites drive a Warburg-like phenotype in infected host leukocytes, characterized by increased expression of metabolic regulators, increased glucose uptake and elevated lactate production, which were lost when the parasite was eliminated. The cohabitation of the parasites within the host cells leads to disruption of the redox balance (as measured by reduced/oxidized glutathione ratio) and elevated ROS (reactive oxygen species) levels, associated with chronic stabilization of the hypoxia-inducible factor 1 alpha (HIF1α). Inhibition of HIF1α (pharmacologically or genetically), or treatment with antioxidants, led to a marked reduction in expression of aerobic glycolytic genes and inhibited the transformed phenotype. These data show that stabilization of HIF1α, following increased ROS production, modulates host glucose metabolism and is critical for parasite-induced transformation. Our study expands knowledge about the molecular strategy used by the parasite Theileria to induce the transformed phenotypes of infected cells via reprogramming of glucose metabolism and redox signaling.
Collapse
|
49
|
Wu B, Teng H, Yang G, Wu L, Wang R. Hydrogen sulfide inhibits the translational expression of hypoxia-inducible factor-1α. Br J Pharmacol 2013; 167:1492-505. [PMID: 22831549 DOI: 10.1111/j.1476-5381.2012.02113.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND AND PURPOSE The accumulation of hypoxia-inducible factor-1α (HIF-1α) is under the influence of hydrogen sulfide (H(2) S), which regulates hypoxia responses. The regulation of HIF-1α accumulation by H(2) S has been shown, but the mechanisms for this effect are largely elusive and controversial. This study aimed at addressing the controversial mechanisms for and the functional importance of the interaction of H(2) S and HIF-1α protein. EXPERIMENTAL APPROACH HIF-1α protein levels and HIF-1α transcriptional activity were detected by Western blotting and luciferase assay. The mechanisms for H(2) S-regulated HIF-1α protein levels were determined using short interfering RNA transfection, co-immunoprecipitation and 7-methyl-GTP sepharose 4B pull-down assay. Angiogenic activity was evaluated using tube formation assay in EA.hy926 cells. KEY RESULTS The accumulation of HIF-1α protein under hypoxia (1% O(2) ) or hypoxia-mimetic conditions was reversed by sodium hydrosulfide (NaHS). This effect of NaHS was not altered after blocking the ubiquitin-proteasomal pathway for HIF-1α degradation; however, blockade of protein translation with cycloheximide abolished the effect of NaHS on the half-life of HIF-1α protein. Knockdown of eukaryotic translation initiation factor 2α (eIF2α) suppressed the effect of NaHS on HIF-1α protein accumulation under hypoxia. NaHS inhibited the expression of VEGF under hypoxia. It also decreased in vitro capillary tube formation and cell proliferation of EA.hy926 cells under hypoxia, but stimulated the tube formation under normoxia. CONCLUSIONS AND IMPLICATIONS H(2) S suppresses HIF-1α translation by enhancing eIF2α phosphorylation under hypoxia. The interaction of H(2) S and HIF-1α inhibits the angiogenic activity of vascular endothelial cells under hypoxia through the down-regulation of VEGF.
Collapse
Affiliation(s)
- Bo Wu
- Department of Pathophysiology, Harbin Medical University, Harbin, China Department of Biology, Lakehead University, Thunder Bay, ON, Canada
| | | | | | | | | |
Collapse
|
50
|
Singh AK, Mukhopadhyay C, Biswas S, Singh VK, Mukhopadhyay CK. Intracellular pathogen Leishmania donovani activates hypoxia inducible factor-1 by dual mechanism for survival advantage within macrophage. PLoS One 2012; 7:e38489. [PMID: 22701652 PMCID: PMC3373497 DOI: 10.1371/journal.pone.0038489] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2011] [Accepted: 05/07/2012] [Indexed: 11/19/2022] Open
Abstract
Recent evidence established a crucial role for mammalian oxygen sensing transcription factor hypoxia inducible factor-1 (HIF-1) in innate immunity against intracellular pathogens. In response to most of these pathogens host phagocytes increase transcription of HIF-1α, the regulatory component of HIF-1 to express various effector molecules against invaders. Leishmania donovani (LD), a protozoan parasite and the causative agent of fatal visceral leishmaniasis resides in macrophages within mammalian host. The mechanism of HIF-1 activation or its role in determining the fate of LD in infected macrophages is still not known. To determine that J774 macrophages were infected with LD and about four-fold increase in HIF-1 activity and HIF-1α expression were detected. A strong increase in HIF-1α expression and nuclear localization was also detected in LD-infected J774 cells, peritoneal macrophages and spleen derived macrophages of LD-infected BALB/c mice. A two-fold increase in HIF-1α mRNA was detected in LD-infected macrophages suggesting involvement of a transcriptional mechanism that was confirmed by promoter activity. We further revealed that LD also induced HIF-1α expression by depleting host cellular iron pool to affect prolyl hydroxylase activity resulting in to stabilization of HIF-1α. To determine the role of HIF-1 on intracellular LD, cells were transfected with HIF-1α siRNA to attenuate its expression and then infected with LD. Although, initial infection rate of LD in HIF-1α attenuated cells was not affected but intracellular growth of LD was significantly inhibited; while, over-expression of stabilized form of HIF-1α promoted intracellular growth of LD in host macrophage. Our results strongly suggest that LD activates HIF-1 by dual mechanism for its survival advantage within macrophage.
Collapse
Affiliation(s)
- Amit Kumar Singh
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Chaitali Mukhopadhyay
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Sudipta Biswas
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Vandana Kumari Singh
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Chinmay K. Mukhopadhyay
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
- * E-mail:
| |
Collapse
|