1
|
Mohan V, Vinjamuri SR, Sahoo P, Hatwar A, S N S, Krishna U, P V V, Vijayan K. Apicomplexan Espionage: Orchestrated Miscommunication at the Host-Parasite Interface. ACS Infect Dis 2024. [PMID: 39499240 DOI: 10.1021/acsinfecdis.4c00526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2024]
Abstract
Intracellular parasites, including Toxoplasma and Plasmodium, are entirely reliant on the active scavenging of host-derived nutrients to fuel their replicative cycle, as they are confined within a specialized membrane-bound compartment, the parasitophorous vacuole (PV). Initial observations, based on the proximity of host vesicles to the parasitophorous vacuole membrane (PVM), suggested that parasites utilize host vesicles to obtain essential nutrients. However, mounting evidence has now unequivocally demonstrated that intracellular pathogens establish membrane contacts with host organelles, establishing control over host cellular machinery. These intimate interactions enable the parasites to gain unimpeded access to cytosolic resources critical for development while evading host immune responses. This review consolidates the latest advancements in understanding the molecular machinery driving these transkingdom contacts and their functional roles. Further investigation into these processes promises to revolutionize our understanding of organelle communication, with profound implications for identifying new therapeutic targets and strategies.
Collapse
Affiliation(s)
- Vaisak Mohan
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, Thiruvananthapuram, Kerala 695551, India
| | - Sandeep Reddy Vinjamuri
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, Thiruvananthapuram, Kerala 695551, India
| | - Parnika Sahoo
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, Thiruvananthapuram, Kerala 695551, India
| | - Abhinav Hatwar
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, Thiruvananthapuram, Kerala 695551, India
| | - Sandra S N
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, Thiruvananthapuram, Kerala 695551, India
| | - Usha Krishna
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, Thiruvananthapuram, Kerala 695551, India
| | - Vyshnav P V
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, Thiruvananthapuram, Kerala 695551, India
| | - Kamalakannan Vijayan
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, Thiruvananthapuram, Kerala 695551, India
| |
Collapse
|
2
|
Fraser M, Curtis B, Phillips P, Yates PA, Lam KS, Netzel O, van Dooren GG, Ingmundson A, Matuschewski K, McLeod MD, Maier AG. Harnessing cholesterol uptake of malaria parasites for therapeutic applications. EMBO Mol Med 2024; 16:1515-1532. [PMID: 38862600 PMCID: PMC11251039 DOI: 10.1038/s44321-024-00087-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 05/14/2024] [Accepted: 05/24/2024] [Indexed: 06/13/2024] Open
Abstract
Parasites, such as the malaria parasite P. falciparum, are critically dependent on host nutrients. Interference with nutrient uptake can lead to parasite death and, therefore, serve as a successful treatment strategy. P. falciparum parasites cannot synthesise cholesterol, and instead source this lipid from the host. Here, we tested whether cholesterol uptake pathways could be 'hijacked' for optimal drug delivery to the intracellular parasite. We found that fluorescent cholesterol analogues were delivered from the extracellular environment to the intracellular parasite. We investigated the uptake and inhibitory effects of conjugate compounds, where proven antimalarial drugs (primaquine and artesunate) were attached to steroids that mimic the structure of cholesterol. These conjugated antimalarial drugs improved the inhibitory effects against multiple parasite lifecycle stages, multiple parasite species, and drug-resistant parasites, whilst also lowering the toxicity to human host cells. Steroids with introduced peroxides also displayed antimalarial activity. These results provide a proof-of-concept that cholesterol mimics can be developed as a drug delivery system against apicomplexan parasites with the potential to improve drug efficacy, increase therapeutic index, and defeat drug resistance.
Collapse
Affiliation(s)
- Merryn Fraser
- Research School of Biology, The Australian National University, Canberra, 2601, Australia
- Department of Molecular Parasitology, Institute of Biology, Humboldt University, Berlin, 10115, Germany
- Harvard T. H. Chan School of Public Health, Harvard University, Boston, MA, 02115, USA
| | - Blake Curtis
- Research School of Chemistry, The Australian National University, Canberra, 2601, Australia
- Metabolism of Microbial Pathogens, Robert Koch Institute, Berlin, 13353, Germany
| | - Patrick Phillips
- Research School of Biology, The Australian National University, Canberra, 2601, Australia
| | - Patrick A Yates
- Research School of Chemistry, The Australian National University, Canberra, 2601, Australia
| | - Kwong Sum Lam
- Research School of Biology, The Australian National University, Canberra, 2601, Australia
| | - Otto Netzel
- Department of Molecular Parasitology, Institute of Biology, Humboldt University, Berlin, 10115, Germany
| | - Giel G van Dooren
- Research School of Biology, The Australian National University, Canberra, 2601, Australia
| | - Alyssa Ingmundson
- Department of Molecular Parasitology, Institute of Biology, Humboldt University, Berlin, 10115, Germany
| | - Kai Matuschewski
- Department of Molecular Parasitology, Institute of Biology, Humboldt University, Berlin, 10115, Germany
| | - Malcolm D McLeod
- Research School of Chemistry, The Australian National University, Canberra, 2601, Australia.
| | - Alexander G Maier
- Research School of Biology, The Australian National University, Canberra, 2601, Australia.
| |
Collapse
|
3
|
Mamudu CO, Tebamifor ME, Sule MO, Dokunmu TM, Ogunlana OO, Iheagwam FN. Apicoplast-Resident Processes: Exploiting the Chink in the Armour of Plasmodium falciparum Parasites. Adv Pharmacol Pharm Sci 2024; 2024:9940468. [PMID: 38765186 PMCID: PMC11101256 DOI: 10.1155/2024/9940468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 03/25/2024] [Accepted: 04/20/2024] [Indexed: 05/21/2024] Open
Abstract
The discovery of a relict plastid, also known as an apicoplast (apicomplexan plastid), that houses housekeeping processes and metabolic pathways critical to Plasmodium parasites' survival has prompted increased research on identifying potent inhibitors that can impinge on apicoplast-localised processes. The apicoplast is absent in humans, yet it is proposed to originate from the eukaryote's secondary endosymbiosis of a primary symbiont. This symbiotic relationship provides a favourable microenvironment for metabolic processes such as haem biosynthesis, Fe-S cluster synthesis, isoprenoid biosynthesis, fatty acid synthesis, and housekeeping processes such as DNA replication, transcription, and translation, distinct from analogous mammalian processes. Recent advancements in comprehending the biology of the apicoplast reveal it as a vulnerable organelle for malaria parasites, offering numerous potential targets for effective antimalarial therapies. We provide an overview of the metabolic processes occurring in the apicoplast and discuss the organelle as a viable antimalarial target in light of current advances in drug discovery. We further highlighted the relevance of these metabolic processes to Plasmodium falciparum during the different stages of the lifecycle.
Collapse
Affiliation(s)
- Collins Ojonugwa Mamudu
- Department of Biochemistry, Covenant University, Ota, Nigeria
- Covenant Applied Informatics and Communication Africa Centre of Excellence, Ota, Nigeria
| | - Mercy Eyitomi Tebamifor
- Department of Biochemistry, Covenant University, Ota, Nigeria
- Covenant Applied Informatics and Communication Africa Centre of Excellence, Ota, Nigeria
| | - Mary Ohunene Sule
- Confluence University of Science and Technology, Osara, Kogi, Nigeria
| | - Titilope Modupe Dokunmu
- Department of Biochemistry, Covenant University, Ota, Nigeria
- Covenant Applied Informatics and Communication Africa Centre of Excellence, Ota, Nigeria
| | - Olubanke Olujoke Ogunlana
- Department of Biochemistry, Covenant University, Ota, Nigeria
- Covenant Applied Informatics and Communication Africa Centre of Excellence, Ota, Nigeria
- Covenant University Public Health and Wellbeing Research Cluster, Covenant University, Ota, Nigeria
| | - Franklyn Nonso Iheagwam
- Department of Biochemistry, Covenant University, Ota, Nigeria
- Covenant University Public Health and Wellbeing Research Cluster, Covenant University, Ota, Nigeria
| |
Collapse
|
4
|
Traore M, Sangare H, Diabate O, Diawara A, Cissé C, Nashiru O, Li J, Shaffer J, Wélé M, Doumbia S, Chikowore T, Soremekun O, Fatumo S. Causal effect of severe and non-severe malaria on dyslipidemia in African Ancestry individuals: A Mendelian randomization study. Ann Hum Genet 2024. [PMID: 38488696 DOI: 10.1111/ahg.12555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 01/16/2024] [Accepted: 02/07/2024] [Indexed: 10/08/2024]
Abstract
BACKGROUND Dyslipidemia is becoming prevalent in Africa, where malaria is endemic. Observational studies have documented the long-term protective effect of malaria on dyslipidemia; however, these study designs are prone to confounding. Therefore, we used Mendelian randomization (MR, a method robust to confounders and reverse causation) to determine the causal effect of severe malaria (SM) and the recurrence of non-severe malaria (RNM) on lipid traits. METHOD We performed two-sample MR using genome wide association study (GWAS) summary statistics for recurrent non-severe malaria (RNM) from a Benin cohort (N = 775) and severe malaria from the MalariaGEN dataset (N = 17,000) and lipid traits from summary-level data of a meta-analyzed African lipid GWAS (MALG, N = 24,215) from the African Partnership for Chronic Disease Research (APCDR) (N = 13,612) and the Africa Wits-IN-DEPTH partnership for genomics studies (AWI-Gen) dataset (N = 10,603). RESULT No evidence of significant causal association was obtained between RNM and high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), total cholesterol and triglycerides. However, a notable association emerged between severe malarial anaemia (SMA) which is a subtype of severe malaria and reduced HDL-C levels, suggesting a potential subtype-specific effect. Nonetheless, we strongly believe that the small sample size likely affects our estimates, warranting cautious interpretation of these results. CONCLUSION Our findings challenge the hypothesis of a broad causal relationship between malaria (both severe and recurrent non-severe forms) and dyslipidemia. The isolated association with SMA highlights an intriguing area for future research. However, we believe that conducting larger studies to investigate the connection between malaria and dyslipidemia in Africa will enhance our ability to better address the burden posed by both diseases.
Collapse
Affiliation(s)
- Mariam Traore
- The African Computational Genomics (TACG) Research Group, Medical Research Council /Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
- African Center of Excellence in Bioinformatics, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Harouna Sangare
- African Center of Excellence in Bioinformatics, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
- Department of Mathematics and Informatics, Faculty of Sciences and Techniques (FST), University of Sciences, Techniques and Technologies of Bamako (USTTB), Bamako, Mali
| | - Oudou Diabate
- African Center of Excellence in Bioinformatics, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Abdoulaye Diawara
- African Center of Excellence in Bioinformatics, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Cheickna Cissé
- African Center of Excellence in Bioinformatics, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
- Faculty of Medicine and Odonto-stomatology, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Oyekanmi Nashiru
- H3Africa Bioinformatics Network (H3ABioNet) Node, Centre for Genomics Research and Innovation, NABDA/FMST, Abuja, Nigeria
| | - Jian Li
- School of Public Health and Tropical Medicine, Tulane University, New Orleans, USA
| | - Jeffrey Shaffer
- School of Public Health and Tropical Medicine, Tulane University, New Orleans, USA
| | - Mamadou Wélé
- African Center of Excellence in Bioinformatics, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
- Department of Biological Sciences, Faculty of Sciences and Techniques, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Seydou Doumbia
- African Center of Excellence in Bioinformatics, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
- Faculty of Medicine and Odonto-stomatology, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Tinashe Chikowore
- MRC/Wits Developmental Pathways for Health Research Unit, Department of Pediatrics, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Opeyemi Soremekun
- The African Computational Genomics (TACG) Research Group, Medical Research Council /Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
- Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Segun Fatumo
- The African Computational Genomics (TACG) Research Group, Medical Research Council /Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
- H3Africa Bioinformatics Network (H3ABioNet) Node, Centre for Genomics Research and Innovation, NABDA/FMST, Abuja, Nigeria
- Department of Non-communicable Disease Epidemiology (NCDE), London School of Hygiene and Tropical Medicine, London, UK
| |
Collapse
|
5
|
Tan YJ, Jin Y, Zhou J, Yang YF. Lipid droplets in pathogen infection and host immunity. Acta Pharmacol Sin 2024; 45:449-464. [PMID: 37993536 PMCID: PMC10834987 DOI: 10.1038/s41401-023-01189-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 10/30/2023] [Indexed: 11/24/2023] Open
Abstract
As the hub of cellular lipid metabolism, lipid droplets (LDs) have been linked to a variety of biological processes. During pathogen infection, the biogenesis, composition, and functions of LDs are tightly regulated. The accumulation of LDs has been described as a hallmark of pathogen infection and is thought to be driven by pathogens for their own benefit. Recent studies have revealed that LDs and their subsequent lipid mediators contribute to effective immunological responses to pathogen infection by promoting host stress tolerance and reducing toxicity. In this comprehensive review, we delve into the intricate roles of LDs in governing the replication and assembly of a wide spectrum of pathogens within host cells. We also discuss the regulatory function of LDs in host immunity and highlight the potential for targeting LDs for the diagnosis and treatment of infectious diseases.
Collapse
Affiliation(s)
- Yan-Jie Tan
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, 250014, China
| | - Yi Jin
- Research Center of Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, China
| | - Jun Zhou
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, 250014, China.
- State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Cell Ecosystem, College of Life Sciences, Nankai University, Tianjin, 300071, China.
| | - Yun-Fan Yang
- Department of Cell Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| |
Collapse
|
6
|
Kandalgaonkar MR, Yeoh BS, Joe B, Schmidt NW, Vijay-Kumar M, Saha P. Hypertension Increases Susceptibility to Experimental Malaria in Mice. FUNCTION 2024; 5:zqae009. [PMID: 38706961 PMCID: PMC11065114 DOI: 10.1093/function/zqae009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 02/17/2024] [Accepted: 02/19/2024] [Indexed: 05/07/2024] Open
Abstract
Global prevalence of hypertension is on the rise, burdening healthcare, especially in developing countries where infectious diseases, such as malaria, are also rampant. Whether hypertension could predispose or increase susceptibility to malaria, however, has not been extensively explored. Previously, we reported that hypertension is associated with abnormal red blood cell (RBC) physiology and anemia. Since RBC are target host cells for malarial parasite, Plasmodium, we hypothesized that hypertensive patients with abnormal RBC physiology are at greater risk or susceptibility to Plasmodium infection. To test this hypothesis, normotensive (BPN/3J) and hypertensive (BPH/2J) mice were characterized for their RBC physiology and subsequently infected with Plasmodium yoelii (P. yoelii), a murine-specific non-lethal strain. When compared to BPN mice, BPH mice displayed microcytic anemia with RBC highly resistant to osmotic hemolysis. Further, BPH RBC exhibited greater membrane rigidity and an altered lipid composition, as evidenced by higher levels of phospholipids and saturated fatty acid, such as stearate (C18:0), along with lower levels of polyunsaturated fatty acid like arachidonate (C20:4). Moreover, BPH mice had significantly greater circulating Ter119+ CD71+ reticulocytes, or immature RBC, prone to P. yoelii infection. Upon infection with P. yoelii, BPH mice experienced significant body weight loss accompanied by sustained parasitemia, indices of anemia, and substantial increase in systemic pro-inflammatory mediators, compared to BPN mice, indicating that BPH mice were incompetent to clear P. yoelii infection. Collectively, these data demonstrate that aberrant RBC physiology observed in hypertensive BPH mice contributes to an increased susceptibility to P. yoelii infection and malaria-associated pathology.
Collapse
Affiliation(s)
- Mrunmayee R Kandalgaonkar
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Beng San Yeoh
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Bina Joe
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Nathan W Schmidt
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Herman B. Wells Center for Pediatric Research, and Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Matam Vijay-Kumar
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Piu Saha
- Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| |
Collapse
|
7
|
Nunes DLM, Carvalho-Araujo MF, Silva-Cabral S, Rios T, Chagas-Lima AC, de Sousa G, Ramos I, Gomes SAO, Atella GC. Lipid metabolism dynamic in Triatomine Rhodnius prolixus during acute Trypanosoma rangeli infection. Acta Trop 2023; 248:107032. [PMID: 37838024 DOI: 10.1016/j.actatropica.2023.107032] [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: 05/24/2023] [Revised: 09/18/2023] [Accepted: 09/19/2023] [Indexed: 10/16/2023]
Abstract
During its life cycle, Trypanosoma rangeli invades the hemolymph of its invertebrate host and colonizes hemocytes and salivary glands. The parasite cannot synthesize some lipid classes, and during its cycle, it depends on the uptake of these molecules from its vertebrate and invertebrate hosts to meet growth and differentiation requirements. However, until now, knowledge on how the parasite affects the lipid physiology of individual insect organs has been largely unknown. Herein, the biochemical and molecular dynamics of triatomine R. prolixus lipid metabolism in response to acute T. rangeli infection were investigated. Biochemical and microscopic assays revealed the lipid droplet profile and the levels of the different identified lipid classes. In addition, a qRT‒PCR approach was used to determine the expression profile of 6 protein-coding genes involved in the R. prolixus lipid physiology. We observed that triacylglycerol (TAG), monoacylglycerol (MAG), phosphatidylethanolamine (PE) and phosphatidylcholine (PC) levels in the fat body decreased in infected insects. On the other hand, high levels of free fatty acids were observed in the hemolymph during infection. Analysis by confocal microscopy revealed a decrease in lipid droplets size from infected fat bodies, and investigations by scanning electron microscopy revealed a significant number of parasites adhered to the surface of the organ. T. rangeli infection upregulated the transcript levels of the protein-coding gene for the acetyl-CoA carboxylase, the first enzyme in the de novo fatty acid synthesis pathway, responsible for the production of malonyl-CoA. On the other hand, downregulation of lipophorin receptor was observed. In conclusion, this study reveals a new set of molecular events that occur within the vector in response to the challenge imposed by the parasite.
Collapse
Affiliation(s)
- Daiene Lopes Moreira Nunes
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil; Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Maria Fernanda Carvalho-Araujo
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil; Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Suellen Silva-Cabral
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil; Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Thamara Rios
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil; Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alessandra Catarina Chagas-Lima
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil; Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Géssica de Sousa
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil; Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Isabela Ramos
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil; Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Suzete A O Gomes
- Laboratório de Biologia de Insetos, Universidade Federal Fluminense, Niterói, RJ, Brazil
| | - Georgia C Atella
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil; Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
8
|
Watanabe N, Saito-Nakano Y, Kurisawa N, Otomo K, Suenaga K, Nakano K, Nozaki T. Fumagillin inhibits growth of the enteric protozoan parasite Entamoeba histolytica by covalently binding to and selectively inhibiting methionine aminopeptidase 2. Antimicrob Agents Chemother 2023; 67:e0056023. [PMID: 37874291 PMCID: PMC10648944 DOI: 10.1128/aac.00560-23] [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: 05/02/2023] [Accepted: 08/27/2023] [Indexed: 10/25/2023] Open
Abstract
Amebiasis is an important cause of morbidity and mortality worldwide, and caused by infection with the protozoan parasite Entamoeba histolytica. Metronidazole is currently the first-line drug despite adverse effects and concerns on the emergence of drug resistance. Fumagillin, a fungal metabolite from Aspergillus fumigatus, and its structurally related natural and synthetic compounds have been previously explored as potential anti-angiogenesis inhibitors for cancers, anti-microbial, and anti-obese compounds. Although fumagillin was used for human amebiasis in clinical trials in 1950s, the mode of action of fumagillin remains elusive until now. In this report, we showed that fumagillin covalently binds to methionine aminopeptidase 2 (MetAP2) and non-covalently but abundantly binds to patatin family phospholipase A (PLA). Susceptibility against fumagillin of the amebic strains in which expression of E. histolytica MetAP2 (EhMetAP2) gene was silenced increased compared to control strain. Conversely, overexpression of EhMetAP2 mutants that harbors amino acid substitutions responsible for resistance to ovalicin, a fumagillin analog, in human MetAP2, also resulted in decrease in fumagillin susceptibility. In contrast, neither gene silencing nor overexpression of E. histolytica PLA (EhPLA) affected fumagillin susceptibility. These data suggest that EhPLA is not essential and not the target of fumagillin for its amebicidal activity. Taken together, our data have demonstrated that EhMetAP2 is the primary target for amebicidal activity of fumagillin, and EhMetAP2 represents a rational explorable target for the development of alternative therapeutic agents against amebiasis.
Collapse
Affiliation(s)
- Natsuki Watanabe
- Department of Biomedical Chemistry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yumiko Saito-Nakano
- Department of Parasitology and Antimicrobial Resistance Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Naoaki Kurisawa
- Department of Chemistry, Faculty of Science and Technology, Keio University, Kanagawa, Japan
| | - Keisuke Otomo
- Department of Chemistry, Faculty of Science and Technology, Keio University, Kanagawa, Japan
| | - Kiyotake Suenaga
- Department of Chemistry, Faculty of Science and Technology, Keio University, Kanagawa, Japan
| | - Kentaro Nakano
- Degree Programs in Biology, Graduate School of Science and Technology, University of Tsukuba, Ibaraki, Japan
| | - Tomoyoshi Nozaki
- Department of Biomedical Chemistry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
9
|
Yang ASP, Dutta D, Kretzschmar K, Hendriks D, Puschhof J, Hu H, Boonekamp KE, van Waardenburg Y, Chuva de Sousa Lopes SM, van Gemert GJ, de Wilt JHW, Bousema T, Clevers H, Sauerwein RW. Development of Plasmodium falciparum liver-stages in hepatocytes derived from human fetal liver organoid cultures. Nat Commun 2023; 14:4631. [PMID: 37532704 PMCID: PMC10397232 DOI: 10.1038/s41467-023-40298-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 07/19/2023] [Indexed: 08/04/2023] Open
Abstract
Plasmodium falciparum (Pf) parasite development in liver represents the initial step of the life-cycle in the human host after a Pf-infected mosquito bite. While an attractive stage for life-cycle interruption, understanding of parasite-hepatocyte interaction is inadequate due to limitations of existing in vitro models. We explore the suitability of hepatocyte organoids (HepOrgs) for Pf-development and show that these cells permitted parasite invasion, differentiation and maturation of different Pf strains. Single-cell messenger RNA sequencing (scRNAseq) of Pf-infected HepOrg cells has identified 80 Pf-transcripts upregulated on day 5 post-infection. Transcriptional profile changes are found involving distinct metabolic pathways in hepatocytes with Scavenger Receptor B1 (SR-B1) transcripts highly upregulated. A novel functional involvement in schizont maturation is confirmed in fresh primary hepatocytes. Thus, HepOrgs provide a strong foundation for a versatile in vitro model for Pf liver-stages accommodating basic biological studies and accelerated clinical development of novel tools for malaria control.
Collapse
Affiliation(s)
- Annie S P Yang
- Radboud Center of Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands.
| | - Devanjali Dutta
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
- Merus, Utrecht, the Netherlands
| | - Kai Kretzschmar
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
- Mildred Scheel Early Career Centre (MSNZ) for Cancer Research Würzburg, University Hospital Würzburg, Würzburg, Germany
| | - Delilah Hendriks
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Jens Puschhof
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
- Microbiome and Cancer Devision, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Huili Hu
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
- The Research Center of Stem Cell and Regenerative Medicine, Department of Systems Biomedicine, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Kim E Boonekamp
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
- Signaling and Functional Genomics Devision, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Youri van Waardenburg
- Radboud Center of Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | | | - Geert-Jan van Gemert
- Radboud Center of Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Johannes H W de Wilt
- Department of Surgery, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Teun Bousema
- Radboud Center of Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Hans Clevers
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands.
- Oncode Institute, Utrecht, The Netherlands.
- Princess Maxima Center (PMC) for Pediatric Oncology, Utrecht, the Netherlands.
- Pharma, Research and Early Development (pRED) of F. Hoffmann-La Roche Ltd, Basel, Switzerland.
| | - Robert W Sauerwein
- Radboud Center of Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands.
- TropIQ Health Sciences, Nijmegen, the Netherlands.
| |
Collapse
|
10
|
Lahree A, Mello-Vieira J, Mota MM. The nutrient games - Plasmodium metabolism during hepatic development. Trends Parasitol 2023; 39:445-460. [PMID: 37061442 DOI: 10.1016/j.pt.2023.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 03/15/2023] [Accepted: 03/17/2023] [Indexed: 04/17/2023]
Abstract
Malaria is a febrile illness caused by species of the protozoan parasite Plasmodium and is characterized by recursive infections of erythrocytes, leading to clinical symptoms and pathology. In mammals, Plasmodium parasites undergo a compulsory intrahepatic development stage before infecting erythrocytes. Liver-stage parasites have a metabolic configuration to facilitate the replication of several thousand daughter parasites. Their metabolism is of interest to identify cellular pathways essential for liver infection, to kill the parasite before onset of the disease. In this review, we summarize the current knowledge on nutrient acquisition and biosynthesis by liver-stage parasites mostly generated in murine malaria models, gaps in knowledge, and challenges to create a holistic view of the development and deficiencies in this field.
Collapse
Affiliation(s)
- Aparajita Lahree
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - João Mello-Vieira
- Institute of Biochemistry II, School of Medicine, Goethe University Frankfurt, Frankfurt am Main, Germany; Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Maria M Mota
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
11
|
Nwonuma CO, Balogun EA, Gyebi GA. Evaluation of Antimalarial Activity of Ethanolic Extract of Annona muricata L.: An in vivo and an in silico Approach. J Evid Based Integr Med 2023; 28:2515690X231165104. [PMID: 37019435 PMCID: PMC10084581 DOI: 10.1177/2515690x231165104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2023] Open
Abstract
In Nigeria, Annona muricata L. has been used to treat a variety of ailments. The mechanism of the antimalarial activity of ethanolic leaf extract of Annona muricata (EEAML) was investigated using both an in vivo and an in silico approach. The experimental mice were divided into five groups: A-F. The mice in groups B-F were inoculated with Plasmodium berghei NK-65 and treated accordingly. Groups A and B are the negative and positive controls (infected and untreated), respectively. Group C received 10 mg/kg chloroquine (standard drug), whereas groups D-F received 100, 200, and 300 mg/kg body weight of the extract orally respectively. The mice were euthanized eight days after infection, and their liver and blood were collected and used in biochemical tests. Molecular docking was performed using the extract's HPLC compounds and Plasmodium falciparum proteins. In the suppressive, prophylactic, and curative tests, there was a significant decrease (p < 0.05) in parasitemia levels in groups treated with the extract compared to the positive control and standard drug. When compared to the positive control, there was a significant (p < 0.05) reduction in liver MDA, total cholesterol, and total triglyceride levels. The binding energies of luteolin and apigenin-pfprotein complexes were significantly (p < 0.05) higher compared to their respective references. The anti-plasmodial activity of the extract may result from its hypolipidemic effect, which deprives the parasite of essential lipid molecules needed for parasite growth, as well as from the inhibitory effects of apigenin and luteolin on specific proteins required for the Plasmodium metabolic pathway.
Collapse
Affiliation(s)
- Charles Obiora Nwonuma
- Department of Biochemistry, College of Pure and Applied Science, Landmark University, Omu-Aran, Kwara State, Nigeria
| | | | - Gideon Ampoma Gyebi
- Department of Biochemistry, Faculty of Science and Technology, Bingham University, Karu, Nigeria
| |
Collapse
|
12
|
Maier AG, van Ooij C. The role of cholesterol in invasion and growth of malaria parasites. Front Cell Infect Microbiol 2022; 12:984049. [PMID: 36189362 PMCID: PMC9522969 DOI: 10.3389/fcimb.2022.984049] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/15/2022] [Indexed: 11/24/2022] Open
Abstract
Malaria parasites are unicellular eukaryotic pathogens that develop through a complex lifecycle involving two hosts, an anopheline mosquito and a vertebrate host. Throughout this lifecycle, the parasite encounters widely differing conditions and survives in distinct ways, from an intracellular lifestyle in the vertebrate host to exclusively extracellular stages in the mosquito. Although the parasite relies on cholesterol for its growth, the parasite has an ambiguous relationship with cholesterol: cholesterol is required for invasion of host cells by the parasite, including hepatocytes and erythrocytes, and for the development of the parasites in those cells. However, the parasite is unable to produce cholesterol itself and appears to remove cholesterol actively from its own plasma membrane, thereby setting up a cholesterol gradient inside the infected host erythrocyte. Overall a picture emerges in which the parasite relies on host cholesterol and carefully controls its transport. Here, we describe the role of cholesterol at the different lifecycle stages of the parasites.
Collapse
Affiliation(s)
- Alexander G. Maier
- Research School of Biology, The Australian National University, Canberra ACT, Australia
- *Correspondence: Alexander G. Maier, ; Christiaan van Ooij,
| | - Christiaan van Ooij
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, London, United Kingdom
- *Correspondence: Alexander G. Maier, ; Christiaan van Ooij,
| |
Collapse
|
13
|
Vijayan K, Arang N, Wei L, Morrison R, Geiger R, Parks KR, Lewis AJ, Mast FD, Douglass AN, Kain HS, Aitchison JD, Johnson JS, Aderem A, Kaushansky A. A genome-wide CRISPR-Cas9 screen identifies CENPJ as a host regulator of altered microtubule organization during Plasmodium liver infection. Cell Chem Biol 2022; 29:1419-1433.e5. [PMID: 35738280 PMCID: PMC9481707 DOI: 10.1016/j.chembiol.2022.06.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 02/03/2022] [Accepted: 06/01/2022] [Indexed: 11/30/2022]
Abstract
Prior to initiating symptomatic malaria, a single Plasmodium sporozoite infects a hepatocyte and develops into thousands of merozoites, in part by scavenging host resources, likely delivered by vesicles. Here, we demonstrate that host microtubules (MTs) dynamically reorganize around the developing liver stage (LS) parasite to facilitate vesicular transport to the parasite. Using a genome-wide CRISPR-Cas9 screen, we identified host regulators of cytoskeleton organization, vesicle trafficking, and ER/Golgi stress that regulate LS development. Foci of γ-tubulin localized to the parasite periphery; depletion of centromere protein J (CENPJ), a novel regulator identified in the screen, exacerbated this re-localization and increased infection. We demonstrate that the Golgi acts as a non-centrosomal MT organizing center (ncMTOC) by positioning γ-tubulin and stimulating MT nucleation at parasite periphery. Together, these data support a model where the Plasmodium LS recruits host Golgi to form MT-mediated conduits along which host organelles are recruited to PVM and support parasite development.
Collapse
Affiliation(s)
- Kamalakannan Vijayan
- Center for Infectious Disease Research, Seattle, WA, USA; Seattle Children's Research Institute, Seattle, WA, USA
| | - Nadia Arang
- Center for Infectious Disease Research, Seattle, WA, USA
| | - Ling Wei
- Seattle Children's Research Institute, Seattle, WA, USA
| | - Robert Morrison
- Center for Infectious Disease Research, Seattle, WA, USA; Seattle Children's Research Institute, Seattle, WA, USA
| | - Rechel Geiger
- MSTP Program, University of Washington, Seattle, WA, USA
| | - K Rachael Parks
- Department of Global Health, University of Washington, Seattle, WA, USA
| | - Adam J Lewis
- Center for Infectious Disease Research, Seattle, WA, USA
| | - Fred D Mast
- Center for Infectious Disease Research, Seattle, WA, USA; Seattle Children's Research Institute, Seattle, WA, USA
| | - Alyse N Douglass
- Department of Global Health, University of Washington, Seattle, WA, USA
| | - Heather S Kain
- Center for Infectious Disease Research, Seattle, WA, USA
| | - John D Aitchison
- Center for Infectious Disease Research, Seattle, WA, USA; Seattle Children's Research Institute, Seattle, WA, USA; Department of Biochemistry, University of Washington, Seattle, WA, USA; Department of Pediatrics, University of Washington, Seattle, WA, USA
| | | | - Alan Aderem
- Center for Infectious Disease Research, Seattle, WA, USA; Seattle Children's Research Institute, Seattle, WA, USA; Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Alexis Kaushansky
- Center for Infectious Disease Research, Seattle, WA, USA; Seattle Children's Research Institute, Seattle, WA, USA; Department of Global Health, University of Washington, Seattle, WA, USA; Department of Pediatrics, University of Washington, Seattle, WA, USA; Brotman Baty Institute for Precision Medicine, Seattle, WA, USA.
| |
Collapse
|
14
|
Schroeder EA, Chirgwin ME, Derbyshire ER. Plasmodium's fight for survival: escaping elimination while acquiring nutrients. Trends Parasitol 2022; 38:544-557. [PMID: 35534377 PMCID: PMC9187605 DOI: 10.1016/j.pt.2022.04.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/10/2022] [Accepted: 04/10/2022] [Indexed: 01/08/2023]
Abstract
Plasmodium parasites extensively alter their host hepatocyte to evade host detection and support an unprecedented replication rate. Host cell manipulation includes association with the host early and late endomembrane systems, where Plasmodium accesses nutrients while suppressing cellular immune processes. Early endomembrane organelles provide an opportunity to sequester an abundance of lipids and proteins, but the association with late endomembrane organelles also risks autophagy-mediated elimination. While not all parasites survive, those that do benefit from a plethora of nutrients provided through this pathway. In this review, we discuss recent advances in our understanding of how Plasmodium parasites balance the need for host nutrients while avoiding elimination during the liver stage.
Collapse
Affiliation(s)
- Erin A Schroeder
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
| | | | - Emily R Derbyshire
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA; Department of Chemistry, Duke University, Durham, NC, USA.
| |
Collapse
|
15
|
Atella T, Bittencourt-Cunha PR, Araujo MFC, Silva-Cardoso L, Maya-Monteiro CM, Atella GC. Trypanosoma cruzi modulates lipid metabolism and highjacks phospholipids from the midgut of Rhodnius prolixus. Acta Trop 2022; 233:106552. [PMID: 35671784 DOI: 10.1016/j.actatropica.2022.106552] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 06/02/2022] [Accepted: 06/03/2022] [Indexed: 11/30/2022]
Abstract
Chagas disease is potentially life-threatening and caused by the protozoan parasite Trypanosoma cruzi. The parasite cannot synthesize some lipids and depends on the uptake of these lipids from its vertebrate and invertebrate hosts. To achieve this, T. cruzi may need to modify the physiology of the insect host for its own benefit. In this study, we investigated the interaction of T. cruzi (Y strain) with its insect vector Rhodnius prolixus and how it manipulates the vector lipid metabolism. We observed a physiological change in lipid flux in of infected insects. In the fat body of infected insects, triacylglycerol levels decreased by 80.6% and lipid storage droplet-1(LSD-1) mRNA levels were lower, when compared to controls. Lipid sequestration by infected midguts led to increased levels of 5' AMP-activated protein kinase (AMPK) phosphorylation and activation in the fat body, inhibiting the synthesis of fatty acids and stimulating their oxidation. This led to reduced lipid levels in the fat body of infected insets, despite the fact that T. cruzi does not colonize this tissue. There was a 3-fold increase, in lipid uptake and synthesis in the midgut of infected insects. Finally, our results suggest that the parasite modifies the lipid flux and metabolism of its vector R. prolixus through the increase in lipid delivery from the fat body to midgut that are then scavenge by T cruzi.
Collapse
Affiliation(s)
- T Atella
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, 343 Carlos Chagas Filho Avenue, Rio de Janeiro, RJ 21941902, Brazil; Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - P R Bittencourt-Cunha
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, 343 Carlos Chagas Filho Avenue, Rio de Janeiro, RJ 21941902, Brazil; Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - M F C Araujo
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, 343 Carlos Chagas Filho Avenue, Rio de Janeiro, RJ 21941902, Brazil; Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - L Silva-Cardoso
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, 343 Carlos Chagas Filho Avenue, Rio de Janeiro, RJ 21941902, Brazil; Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - C M Maya-Monteiro
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | - G C Atella
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, 343 Carlos Chagas Filho Avenue, Rio de Janeiro, RJ 21941902, Brazil; Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
16
|
Lahree A, Baptista SDJS, Marques S, Perschin V, Zuzarte-Luís V, Goel M, Choudhary HH, Mishra S, Stigloher C, Zerial M, Sundaramurthy V, Mota MM. Active APPL1 sequestration by Plasmodium favors liver-stage development. Cell Rep 2022; 39:110886. [PMID: 35649358 DOI: 10.1016/j.celrep.2022.110886] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 01/10/2022] [Accepted: 05/06/2022] [Indexed: 11/03/2022] Open
Abstract
Intracellular pathogens manipulate host cells to survive and thrive. Cellular sensing and signaling pathways are among the key host machineries deregulated to favor infection. In this study, we show that liver-stage Plasmodium parasites compete with the host to sequester a host endosomal-adaptor protein (APPL1) known to regulate signaling in response to endocytosis. The enrichment of APPL1 at the parasitophorous vacuole membrane (PVM) involves an atypical Plasmodium Rab5 isoform (Rab5b). Depletion of host APPL1 alters neither the infection nor parasite development; however, upon overexpression of a GTPase-deficient host Rab5 mutant (hRab5_Q79L), the parasites are smaller and their PVM is stripped of APPL1. Infection with the GTPase-deficient Plasmodium berghei Rab5b mutant (PbRab5b_Q91L) in this case rescues the PVM APPL1 signal and parasite size. In summary, we observe a robust correlation between the level of APPL1 retention at the PVM and parasite size during exoerythrocytic development.
Collapse
Affiliation(s)
- Aparajita Lahree
- Instituto de Medicina Molecular- João Lobo Antunes (iMM-JLA), Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal; Departamento de Bioengenharia, Instituto Superior Técnico, Av. Rovisco Pais 1, 1049-001 Lisboa, Portugal
| | - Sara de Jesus Santos Baptista
- Instituto de Medicina Molecular- João Lobo Antunes (iMM-JLA), Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| | - Sofia Marques
- Instituto de Medicina Molecular- João Lobo Antunes (iMM-JLA), Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| | - Veronika Perschin
- Imaging Core Facility, Biocenter, University of Würzburg, 97074 Würzburg, Germany
| | - Vanessa Zuzarte-Luís
- Instituto de Medicina Molecular- João Lobo Antunes (iMM-JLA), Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| | - Manisha Goel
- National Centre for Biological Sciences, Tata Institute of Fundamental Research (NCBS), Bellary Road, Bangalore 560065, Karnataka, India
| | - Hadi Hasan Choudhary
- CSIR-Central Drug Research Institute Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, Uttar Pradesh, India
| | - Satish Mishra
- CSIR-Central Drug Research Institute Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, Uttar Pradesh, India
| | - Christian Stigloher
- Imaging Core Facility, Biocenter, University of Würzburg, 97074 Würzburg, Germany
| | - Marino Zerial
- Max Planck Institute of Molecular Cell Biology and Genetics (MPI-CBG), Pfotenhauerstraße 108, 01307 Dresden, Germany
| | - Varadharajan Sundaramurthy
- National Centre for Biological Sciences, Tata Institute of Fundamental Research (NCBS), Bellary Road, Bangalore 560065, Karnataka, India
| | - Maria M Mota
- Instituto de Medicina Molecular- João Lobo Antunes (iMM-JLA), Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal.
| |
Collapse
|
17
|
Roy D, Anas M, Manhas A, Saha S, Kumar N, Panda G. Synthesis, biological evaluation, Structure − Activity relationship studies of quinoline-imidazole derivatives as potent antimalarial agents. Bioorg Chem 2022; 121:105671. [DOI: 10.1016/j.bioorg.2022.105671] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 01/24/2022] [Accepted: 02/07/2022] [Indexed: 12/22/2022]
|
18
|
Shunmugam S, Arnold CS, Dass S, Katris NJ, Botté CY. The flexibility of Apicomplexa parasites in lipid metabolism. PLoS Pathog 2022; 18:e1010313. [PMID: 35298557 PMCID: PMC8929637 DOI: 10.1371/journal.ppat.1010313] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Apicomplexa are obligate intracellular parasites responsible for major human infectious diseases such as toxoplasmosis and malaria, which pose social and economic burdens around the world. To survive and propagate, these parasites need to acquire a significant number of essential biomolecules from their hosts. Among these biomolecules, lipids are a key metabolite required for parasite membrane biogenesis, signaling events, and energy storage. Parasites can either scavenge lipids from their host or synthesize them de novo in a relict plastid, the apicoplast. During their complex life cycle (sexual/asexual/dormant), Apicomplexa infect a large variety of cells and their metabolic flexibility allows them to adapt to different host environments such as low/high fat content or low/high sugar levels. In this review, we discuss the role of lipids in Apicomplexa parasites and summarize recent findings on the metabolic mechanisms in host nutrient adaptation.
Collapse
Affiliation(s)
- Serena Shunmugam
- Apicolipid Team, Institute for Advanced Biosciences, CNRS UMR5309, Université Grenoble Alpes, INSERM U1209, Grenoble, France
| | - Christophe-Sébastien Arnold
- Apicolipid Team, Institute for Advanced Biosciences, CNRS UMR5309, Université Grenoble Alpes, INSERM U1209, Grenoble, France
| | - Sheena Dass
- Apicolipid Team, Institute for Advanced Biosciences, CNRS UMR5309, Université Grenoble Alpes, INSERM U1209, Grenoble, France
| | - Nicholas J. Katris
- Apicolipid Team, Institute for Advanced Biosciences, CNRS UMR5309, Université Grenoble Alpes, INSERM U1209, Grenoble, France
| | - Cyrille Y. Botté
- Apicolipid Team, Institute for Advanced Biosciences, CNRS UMR5309, Université Grenoble Alpes, INSERM U1209, Grenoble, France
| |
Collapse
|
19
|
Ahiya AI, Bhatnagar S, Morrisey JM, Beck JR, Vaidya AB. Dramatic Consequences of Reducing Erythrocyte Membrane Cholesterol on Plasmodium falciparum. Microbiol Spectr 2022; 10:e0015822. [PMID: 35196803 PMCID: PMC8865471 DOI: 10.1128/spectrum.00158-22] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 02/01/2022] [Indexed: 01/11/2023] Open
Abstract
Cholesterol is the most abundant lipid in the erythrocyte. During its blood-stage development, the malaria parasite establishes an active cholesterol gradient across the various membrane systems within the infected erythrocyte. Interestingly, some antimalarial compounds have recently been shown to disrupt cholesterol homeostasis in the intraerythrocytic stages of Plasmodium falciparum. These studies point to the importance of cholesterol for parasite growth. Previously, reduction of cholesterol from the erythrocyte membrane by treatment with methyl-β-cyclodextrin (MβCD) was shown to inhibit parasite invasion and growth. In addition, MβCD treatment of trophozoite-stage P. falciparum was shown to result in parasite expulsion from the host cell. We have revisited these phenomena by using live video microscopy, ultrastructural analysis, and response to antimalarial compounds. By using time-lapse video microscopy of fluorescently tagged parasites, we show that MβCD treatment for just 30 min causes dramatic expulsion of the trophozoite-stage parasites. This forceful expulsion occurs within 10 s. Remarkably, the plasma membrane of the host cell from which the parasite has been expelled does not appear to be compromised. The parasitophorous vacuolar membrane (PVM) continued to surround the extruded parasite, but the PVM appeared damaged. Treatment with antimalarial compounds targeting PfATP4 or PfNCR1 prevented MβCD-mediated extrusion of the parasites, pointing to a potential role of cholesterol dynamics underlying the expulsion phenomena. We also confirmed the essential role of erythrocyte plasma membrane cholesterol for invasion and growth of P. falciparum. This defect can be partially complemented by cholesterol and desmosterol but not with epicholesterol, revealing stereospecificity underlying cholesterol function. Overall, our studies advance previous observations and reveal unusual cell biological features underlying cholesterol depletion of the infected erythrocyte plasma membrane. IMPORTANCE Malaria remains a major challenge in much of the world. Symptoms of malaria are caused by the growth of parasites belonging to Plasmodium spp. inside the red blood cells (RBCs), leading to their destruction. The parasite depends upon its host for much of its nutritional needs. Cholesterol is a major lipid in the RBC plasma membrane, which is the only source of this lipid for malaria parasites. We have previously shown that certain new antimalarial compounds disrupt cholesterol homeostasis in P. falciparum. Here, we use live time-lapse video microscopy to show dramatic expulsion of the parasite from the host RBC when the cholesterol content of the RBC is reduced. Remarkably, this expulsion is inhibited by the antimalarials that disrupt lipid homeostasis. We also show stereospecificity of cholesterol in supporting parasite growth inside RBC. Overall, these results point to a critical role of cholesterol in the physiology of malaria parasites.
Collapse
Affiliation(s)
- Avantika I. Ahiya
- Center for Molecular Parasitology, Institute for Molecular Medicine and Infectious Disease, Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Suyash Bhatnagar
- Center for Molecular Parasitology, Institute for Molecular Medicine and Infectious Disease, Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Joanne M. Morrisey
- Center for Molecular Parasitology, Institute for Molecular Medicine and Infectious Disease, Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Josh R. Beck
- Department of Biomedical Sciences, Iowa State University, Ames, Iowa, USA
| | - Akhil B. Vaidya
- Center for Molecular Parasitology, Institute for Molecular Medicine and Infectious Disease, Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
20
|
Silva LMR, Velásquez ZD, López-Osorio S, Hermosilla C, Taubert A. Novel Insights Into Sterol Uptake and Intracellular Cholesterol Trafficking During Eimeria bovis Macromeront Formation. Front Cell Infect Microbiol 2022; 12:809606. [PMID: 35223543 PMCID: PMC8878908 DOI: 10.3389/fcimb.2022.809606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 01/18/2022] [Indexed: 11/24/2022] Open
Abstract
Apicomplexan parasites are considered as defective in cholesterol synthesis. Consequently, they need to scavenge cholesterol from the host cell by either enhancing the uptake of extracellular cholesterol sources or by upregulating host cellular de-novo biosynthesis. Given that Eimeria bovis macromeront formation in bovine lymphatic endothelial host cells in vivo is a highly cholesterol-demanding process, we here examined host parasite interactions based on host cellular uptake of different low-density lipoprotein (LDL) types, i.e., of non-modified (LDL), oxidized (oxLDL), and acetylated LDL (acLDL). Furthermore, the expression of lipoprotein-oxidized receptor 1 (LOX-1), which mediates acLDL and oxLDL internalization, was monitored throughout first merogony, in vitro and ex vivo. Moreover, the effects of inhibitors blocking exogenous sterol uptake or intracellular transport were studied during E. bovis macromeront formation in vitro. Hence, E. bovis-infected primary bovine umbilical vein endothelial cells (BUVEC) were treated with inhibitors of sterol uptake (ezetimibe, poly-C, poly-I, sucrose) and of intracellular sterol transport and release from endosomes (progesterone, U18666A). As a read-out system, the size and number of macromeronts as well as merozoite I production were estimated. Overall, the internalization of all LDL modifications (LDL, oxLDL, acLDL) was observed in E. bovis-infected BUVEC but to different extents. Supplementation with oxLDL and acLDL at lower concentrations (5 and 10 µg/ml, respectively) resulted in a slight increase of both macromeront numbers and size; however, at higher concentrations (25-50 µg/ml), merozoite I production was diminished. LOX-1 expression was enhanced in E. bovis-infected BUVEC, especially toward the end of merogony. As an interesting finding, ezetimibe treatments led to a highly significant blockage of macromeront development and merozoite I production confirming the relevance of sterol uptake for intracellular parasite development. Less prominent effects were induced by non-specific inhibition of LDL internalization via sucrose, poly-I, and poly-C. In addition, blockage of cholesterol transport via progesterone and U18666A treatments resulted in significant inhibition of parasite development. Overall, current data underline the relevance of exogenous sterol uptake and intracellular cholesterol transport for adequate E. bovis macromeront development, unfolding new perspectives for novel drug targets against E. bovis.
Collapse
Affiliation(s)
- Liliana M. R. Silva
- Institute of Parasitology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Giessen, Germany
| | - Zahady D. Velásquez
- Institute of Parasitology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Giessen, Germany
| | - Sara López-Osorio
- Institute of Parasitology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Giessen, Germany
- Veterinary Medicine School, CIBAV Investigation Group, University of Antioquia, Medellin, Colombia
| | - Carlos Hermosilla
- Institute of Parasitology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Giessen, Germany
| | - Anja Taubert
- Institute of Parasitology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
21
|
In Vivo Antimalarial Activity of Polyalthia longifolia (Annonaceae) Leaf Extract and Assessment of Its Formulated Oral Dosage Forms. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:6519346. [PMID: 34858510 PMCID: PMC8632403 DOI: 10.1155/2021/6519346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 11/06/2021] [Indexed: 11/18/2022]
Abstract
Plant medicine is commonly employed to treat malaria and other infections in Ghana. However, many of these phytomedicines have not been scientifically investigated to justify their use. This study therefore sought to investigate the antimalarial property of Polyalthia longifolia leaves and to formulate suitable dosage forms for ease of administration. A four-day antiplasmodial suppressive and curative study was conducted on ethanol extract of P. longifolia leaves (PLE) using Plasmodium berghei infected albino mice. Tablet and suspension dosage forms of PLE were formulated and evaluated for quality and stability. Statistically significant (P < 0.05) parasitaemia suppression (61.25%) and cure (58.78%) were achieved at a PLE dose of 100 mg/kg, and increases in hematological indices (P < 0.001) were also observed in the PLE-treated mice as compared to the untreated group. The tablets passed the tests for uniformity of weight, friability (<1%), hardness, disintegration (<15 minutes), and in vitro dissolution (>70% release in 45 minutes). The sedimentation volume, rheology, viscosity, and pH of the formulated suspension were within the official specifications. The dosage forms showed consistency in PLE content (85-105%) and no changes in physicochemical properties over the six months period of stability study. The in vivo antimalarial activity of PLE has been established and oral dosage forms that conformed to Pharmacopoeial standards are formulated for use in the management of malaria.
Collapse
|
22
|
Hajialiani F, Shahbazzadeh D, Maleki F, Elmi T, Tabatabaie F, Zamani Z. The Metabolomic Profiles of Sera of Mice Infected with Plasmodium berghei and Treated by Effective Fraction of Naja naja oxiana Using 1H Nuclear Magnetic Resonance Spectroscopy. Acta Parasitol 2021; 66:1517-1527. [PMID: 34357584 DOI: 10.1007/s11686-021-00456-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 07/08/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND The use of venom fractions from the Iranian cobra could be useful adjunct treatments of malaria with chloroquine. A metabolomic investigation with 1HNMR spectroscopy was conducted on an effective fraction tested earlier using Plasmodium berghei as an experimental murine model. PURPOSE We sought to ascertain both safety and anti-parasitic effects of experimental therapies. METHODS After purification of the venom fractions, 25 mice were infected, then treated for 4 days with 0.2 ml of 5 mg/kg, 2.5 mg/kg and 1 mg/kg of the effective fraction, chloroquine, and a drug vehicle. An ED50 was obtained using Giemsa staining and real-time PCR analysis. The toxicity tests inspecting both liver and kidney tissues were performed. RESULTS A clear inhibitory effect on parasitaemia was observed (with 75% inhibition with 5 mg/kg and 50% reduction when 2.5 mg/kg dosage used). ED50 obtained 2.5 mg/kg. The metabolomics were identified as differentiation of aminoacyl-t-RNA biosynthesis, valine, leucine, isoleucine biosynthesis and degradation pathways were observed. CONCLUSION Upon therapeutic effects of cobra venom fraction, further optimization of dose-dependent response of pharmacokinetics would be worthwhile for further exploration in adjunct experimental venom therapies.
Collapse
|
23
|
Evaluation of Antimalarial Potential of Extracts from Alstonia boonei and Carica papaya in Plasmodium berghei-Infected Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:2599191. [PMID: 34659429 PMCID: PMC8514918 DOI: 10.1155/2021/2599191] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/26/2021] [Accepted: 09/02/2021] [Indexed: 01/18/2023]
Abstract
Extracts of Alstonia boonei and Carica papaya are used in herbal medicine for the treatment of malaria. This work investigated the phytochemical, antioxidant, and antimalarial effects of hydromethanolic extracts of Alstonia boonei and Carica papaya. A four-day chemosuppressive test was conducted to assess the ability of the extracts to prevent establishment of infection. Three doses of the extracts were administered—100, 200, and 400 mg/kg bw—prior to Plasmodium berghei challenge. Change in body weight, parasitemia, packed cell volume (PCV), and mean survival time was determined. A three-day curative test was also carried out on Plasmodium berghei-infected mice to determine the effects of the plant extracts (200 mg/kg bw) on parasitemia and biochemical indices of liver and kidney functions, lipid metabolism, and oxidative stress. The study revealed that the extracts possessed phenolic compounds (34.13 ± 1.90 mg GAE/g for Alstonia boonei and 27.99 ± 1.46 mg GAE/g for Carica papaya) and flavonoids (19.47 ± 1.89 mg QE/g for Alstonia boonei and 18.24 ± 1.36 mg QE/g for Carica papaya). In vitro antioxidant activity measured as total antioxidant power, total reducing power, and DPPH radical scavenging activity showed that the extracts possessed higher antioxidant activity than the reference compounds. The outcome of the chemosuppressive test revealed that whereas Plasmodium berghei-infected mice had high parasitemia, decreased mean survival time, exhibited loss of weight, and had low PCV, treatment with the extracts reversed the effects in a concentration-dependent manner. Similarly, the curative test revealed that the extracts significantly suppressed parasitemia compared with the malaria negative control group. This was mirrored by reversal of indices of hepatic toxicity (AST, ALT, and ALP levels), nephropathy (urea and creatinine levels), oxidative stress (SOD, CAT, GPx, GSH, and lipid peroxides), and dyslipidemia (TC, HDL, and TG levels and HMG-CoA reductase activity) in infected but treated mice compared with negative control. Put together, the results of this study demonstrate that the extracts of Alstonia boonei and Carica papaya possess antimalarial properties and are able to ameliorate metabolic dysregulations that characterize Plasmodium berghei infection. The phytoconstituents in these extracts are believed to be responsible for the pharmacological activity reported in this study.
Collapse
|
24
|
Abstract
Host-directed therapy (HDT) is gaining traction as a strategy to combat infectious diseases caused by viruses and intracellular bacteria, but its implementation in the context of parasitic diseases has received less attention. Here, we provide a brief overview of this field and advocate HDT as a promising strategy for antimalarial intervention based on untapped targets. HDT provides a basis from which repurposed drugs could be rapidly deployed and is likely to strongly limit the emergence of resistance. This strategy can be applied to any intracellular pathogen and is particularly well placed in situations in which rapid identification of treatments is needed, such as emerging infections and pandemics, as starkly illustrated by the current COVID-19 crisis.
Collapse
|
25
|
Ressurreição M, van Ooij C. Lipid transport proteins in malaria, from Plasmodium parasites to their hosts. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:159047. [PMID: 34461309 DOI: 10.1016/j.bbalip.2021.159047] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 08/08/2021] [Accepted: 08/10/2021] [Indexed: 11/25/2022]
Abstract
Eukaryotic unicellular pathogens from the genus Plasmodium are the etiological agents of malaria, a disease that persists over a wide range of vertebrate species, including humans. During its dynamic lifecycle, survival in the different hosts depends on the parasite's ability to establish a suitable environmental milieu. To achieve this, specific host processes are exploited to support optimal growth, including extensive modifications to the infected host cell. These modifications include the formation of novel membranous structures, which are induced by the parasite. Consequently, to maintain a finely tuned and dynamic lipid environment, the organisation and distribution of lipids to different cell sites likely requires specialised lipid transfer proteins (LTPs). Indeed, several parasite and host-derived LTPs have been identified and shown to be essential at specific stages. Here we describe the roles of LTPs in parasite development and adaptation to its host including how the latest studies are profiting from the improved genetic, lipidomic and imaging toolkits available to study Plasmodium parasites. Lastly, a list of predicted Plasmodium LTPs is provided to encourage research in this field.
Collapse
Affiliation(s)
- Margarida Ressurreição
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom.
| | - Christiaan van Ooij
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom.
| |
Collapse
|
26
|
Dumoulin PC, Burleigh BA. Metabolic flexibility in Trypanosoma cruzi amastigotes: implications for persistence and drug sensitivity. Curr Opin Microbiol 2021; 63:244-249. [PMID: 34455305 DOI: 10.1016/j.mib.2021.07.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 07/19/2021] [Accepted: 07/24/2021] [Indexed: 12/18/2022]
Abstract
Throughout their life cycle, parasitic organisms experience a variety of environmental conditions. To ensure persistence and transmission, some protozoan parasites are capable of adjusting their replication or converting to distinct life cycle stages. Trypanosoma cruzi is a 'generalist' parasite that is competent to infect various insect (triatomine) vectors and mammalian hosts. Within the mammalian host, T. cruzi replicates intracellularly as amastigotes and can persist for the lifetime of the host. The persistence of the parasites in tissues can lead to the development of Chagas disease. Recent work has identified growth plasticity and metabolic flexibility as aspects of amastigote biology that are important determinants of persistence in varied growth conditions and under drug pressure. A better understanding of the link between amastigote and host/tissue metabolism will aid in the development of new drugs or therapies that can limit disease pathology.
Collapse
Affiliation(s)
- Peter C Dumoulin
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA 02115, United States.
| | - Barbara A Burleigh
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA 02115, United States
| |
Collapse
|
27
|
Vijayan K, Wei L, Glennon EKK, Mattocks C, Bourgeois N, Staker B, Kaushansky A. Host-targeted Interventions as an Exciting Opportunity to Combat Malaria. Chem Rev 2021; 121:10452-10468. [PMID: 34197083 DOI: 10.1021/acs.chemrev.1c00062] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Terminal and benign diseases alike in adults, children, pregnant women, and others are successfully treated by pharmacological inhibitors that target human enzymes. Despite extensive global efforts to fight malaria, the disease continues to be a massive worldwide health burden, and new interventional strategies are needed. Current drugs and vector control strategies have contributed to the reduction in malaria deaths over the past 10 years, but progress toward eradication has waned in recent years. Resistance to antimalarial drugs is a substantial and growing problem. Moreover, targeting dormant forms of the malaria parasite Plasmodium vivax is only possible with two approved drugs, which are both contraindicated for individuals with glucose-6-phosphate dehydrogenase deficiency and in pregnant women. Plasmodium parasites are obligate intracellular parasites and thus have specific and absolute requirements of their hosts. Growing evidence has described these host necessities, paving the way for opportunities to pharmacologically target host factors to eliminate Plasmodium infection. Here, we describe progress in malaria research and adjacent fields and discuss key challenges that remain in implementing host-directed therapy against malaria.
Collapse
Affiliation(s)
| | - Ling Wei
- Seattle Children's Research Institute, Seattle, Washington 98109, United States
| | | | - Christa Mattocks
- Department of Global Health, University of Washington, Seattle, Washington 98195, United States
| | - Natasha Bourgeois
- Seattle Children's Research Institute, Seattle, Washington 98109, United States.,Department of Global Health, University of Washington, Seattle, Washington 98195, United States
| | - Bart Staker
- Seattle Children's Research Institute, Seattle, Washington 98109, United States
| | - Alexis Kaushansky
- Seattle Children's Research Institute, Seattle, Washington 98109, United States.,Department of Global Health, University of Washington, Seattle, Washington 98195, United States.,Department of Pediatrics, University of Washington, Seattle, Washington 98105, United States.,Brotman Baty Institute for Precision Medicine, Seattle, Washington 98195, United States
| |
Collapse
|
28
|
Ezetimibe blocks Toxoplasma gondii-, Neospora caninum- and Besnoitia besnoiti-tachyzoite infectivity and replication in primary bovine endothelial host cells. Parasitology 2021; 148:1107-1115. [PMID: 34024307 PMCID: PMC8273898 DOI: 10.1017/s0031182021000822] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Coccidia are obligate apicomplexan parasites that affect humans and animals. In fast replicating species, in vitro merogony takes only 24–48 h. In this context, successful parasite proliferation requires nutrients and other building blocks. Coccidian parasites are auxotrophic for cholesterol, so they need to obtain this molecule from host cells. In humans, ezetimibe has been applied successfully as hypolipidaemic compound, since it reduces intestinal cholesterol absorption via blockage of Niemann−Pick C-1 like-1 protein (NPC1L1), a transmembrane protein expressed in enterocytes. To date, few data are available on its potential anti-parasitic effects in primary host cells infected with apicomplexan parasites of human and veterinary importance, such as Toxoplasma gondii, Neospora caninum and Besnoitia besnoiti. Current inhibition experiments show that ezetimibe effectively blocks T. gondii, B. besnoiti and N. caninum tachyzoite infectivity and replication in primary bovine endothelial host cells. Thus, 20 μm ezetimibe blocked parasite proliferation by 73.1−99.2%, via marked reduction of the number of tachyzoites per meront, confirmed by 3D-holotomographic analyses. The effects were parasitostatic since withdrawal of the compound led to parasite recovery with resumed proliferation. Ezetimibe-glucuronide, the in vivo most effective metabolite, failed to affect parasite proliferation in vitro, thereby suggesting that ezetimibe effects might be NPC1L1-independent.
Collapse
|
29
|
Hernández J, Gabrielli M, Costa J, Uttaro AD. Phagocytic and pinocytic uptake of cholesterol in Tetrahymena thermophila impact differently on gene regulation for sterol homeostasis. Sci Rep 2021; 11:9067. [PMID: 33907281 PMCID: PMC8079401 DOI: 10.1038/s41598-021-88737-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 04/15/2021] [Indexed: 01/02/2023] Open
Abstract
The ciliate Tetrahymena thermophila can either synthesize tetrahymanol or when available, assimilate and modify sterols from its diet. This metabolic shift is mainly driven by transcriptional regulation of genes for tetrahymanol synthesis (TS) and sterol bioconversion (SB). The mechanistic details of sterol uptake, intracellular trafficking and the associated gene expression changes are unknown. By following cholesterol incorporation over time in a conditional phagocytosis-deficient mutant, we found that although phagocytosis is the main sterol intake route, a secondary endocytic pathway exists. Different expression patterns for TS and SB genes were associated with these entry mechanisms. Squalene synthase was down-regulated by a massive cholesterol intake only attainable by phagocytosis-proficient cells, whereas C22-sterol desaturase required ten times less cholesterol and was up-regulated in both wild-type and mutant cells. These patterns are suggestive of at least two different signaling pathways. Sterol trafficking beyond phagosomes and esterification was impaired by the NPC1 inhibitor U18666A. NPC1 is a protein that mediates cholesterol export from late endosomes/lysosomes in mammalian cells. U18666A also produced a delay in the transcriptional response to cholesterol, suggesting that the regulatory signals are triggered between lysosomes and the endoplasmic reticulum. These findings could hint at partial conservation of sterol homeostasis between eukaryote lineages.
Collapse
Affiliation(s)
- Josefina Hernández
- Instituto de Biología Molecular y Celular de Rosario, CONICET, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, S2000FHQ, Ocampo y Esmeralda, Rosario, Argentina
| | - Matías Gabrielli
- Instituto de Biología Molecular y Celular de Rosario, CONICET, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, S2000FHQ, Ocampo y Esmeralda, Rosario, Argentina
| | - Joaquín Costa
- Instituto de Biología Molecular y Celular de Rosario, CONICET, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, S2000FHQ, Ocampo y Esmeralda, Rosario, Argentina
| | - Antonio D Uttaro
- Instituto de Biología Molecular y Celular de Rosario, CONICET, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, S2000FHQ, Ocampo y Esmeralda, Rosario, Argentina.
| |
Collapse
|
30
|
Talyuli OAC, Bottino-Rojas V, Polycarpo CR, Oliveira PL, Paiva-Silva GO. Non-immune Traits Triggered by Blood Intake Impact Vectorial Competence. Front Physiol 2021; 12:638033. [PMID: 33737885 PMCID: PMC7960658 DOI: 10.3389/fphys.2021.638033] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 02/08/2021] [Indexed: 11/13/2022] Open
Abstract
Blood-feeding arthropods are considered an enormous public health threat. They are vectors of a plethora of infectious agents that cause potentially fatal diseases like Malaria, Dengue fever, Leishmaniasis, and Lyme disease. These vectors shine due to their own physiological idiosyncrasies, but one biological aspect brings them all together: the requirement of blood intake for development and reproduction. It is through blood-feeding that they acquire pathogens and during blood digestion that they summon a collection of multisystemic events critical for vector competence. The literature is focused on how classical immune pathways (Toll, IMD, and JAK/Stat) are elicited throughout the course of vector infection. Still, they are not the sole determinants of host permissiveness. The dramatic changes that are the hallmark of the insect physiology after a blood meal intake are the landscape where a successful infection takes place. Dominant processes that occur in response to a blood meal are not canonical immunological traits yet are critical in establishing vector competence. These include hormonal circuitries and reproductive physiology, midgut permeability barriers, midgut homeostasis, energy metabolism, and proteolytic activity. On the other hand, the parasites themselves have a role in the outcome of these blood triggered physiological events, consistently using them in their favor. Here, to enlighten the knowledge on vector-pathogen interaction beyond the immune pathways, we will explore different aspects of the vector physiology, discussing how they give support to these long-dated host-parasite relationships.
Collapse
Affiliation(s)
- Octavio A C Talyuli
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vanessa Bottino-Rojas
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carla R Polycarpo
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Rio de Janeiro, Brazil
| | - Pedro L Oliveira
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Rio de Janeiro, Brazil
| | - Gabriela O Paiva-Silva
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Rio de Janeiro, Brazil
| |
Collapse
|
31
|
Oringanje C, Delacruz LR, Han Y, Luckhart S, Riehle MA. Overexpression of Activated AMPK in the Anopheles stephensi Midgut Impacts Mosquito Metabolism, Reproduction and Plasmodium Resistance. Genes (Basel) 2021; 12:genes12010119. [PMID: 33478058 PMCID: PMC7835765 DOI: 10.3390/genes12010119] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/12/2021] [Accepted: 01/13/2021] [Indexed: 12/12/2022] Open
Abstract
Mitochondrial integrity and homeostasis in the midgut are key factors controlling mosquito fitness and anti-pathogen resistance. Targeting genes that regulate mitochondrial dynamics represents a potential strategy for limiting mosquito-borne diseases. AMP-activated protein kinase (AMPK) is a key cellular energy sensor found in nearly all eukaryotic cells. When activated, AMPK inhibits anabolic pathways that consume ATP and activates catabolic processes that synthesize ATP. In this study, we overexpressed a truncated and constitutively active α-subunit of AMPK under the control of the midgut-specific carboxypeptidase promotor in the midgut of female Anopheles stephensi. As expected, AMPK overexpression in homozygous transgenic mosquitoes was associated with changes in nutrient storage and metabolism, decreasing glycogen levels at 24 h post-blood feeding when transgene expression was maximal, and concurrently increasing circulating trehalose at the same time point. When transgenic lines were challenged with Plasmodium falciparum, we observed a significant decrease in the prevalence and intensity of infection relative to wild type controls. Surprisingly, we did not observe a significant difference in the survival of adult mosquitoes fed either sugar only or both sugar and bloodmeals throughout adult life. This may be due to the limited period that the transgene was activated before homeostasis was restored. However, we did observe a significant decrease in egg production, suggesting that manipulation of AMPK activity in the mosquito midgut resulted in the re-allocation of resources away from egg production. In summary, this work identifies midgut AMPK activity as an important regulator of metabolism, reproduction, and innate immunity in An. stephensi, a highly invasive and important malaria vector species.
Collapse
Affiliation(s)
| | | | - Yunan Han
- Department of Health Sciences, ECPI University, Virginia Beach, VA 23462, USA;
| | - Shirley Luckhart
- Department of Entomology, Plant Pathology and Nematology, University of Idaho, Moscow, ID 83844, USA;
- Department of Biological Sciences, University of Idaho, Moscow, ID 83844, USA
| | - Michael A. Riehle
- Department of Entomology, University of Arizona, Tucson, AZ 85721, USA;
- Correspondence: ; Tel.: +1-520-626-8500
| |
Collapse
|
32
|
Zhang Y, Wang C, Jia H. Biogenesis and maintenance of the apicoplast in model apicomplexan parasites. Parasitol Int 2020; 81:102270. [PMID: 33321224 DOI: 10.1016/j.parint.2020.102270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 12/08/2020] [Accepted: 12/09/2020] [Indexed: 11/27/2022]
Abstract
The apicoplast is a non-photosynthetic relict plastid of Apicomplexa that evolved from a secondary symbiotic system. During its evolution, most of the genes derived from its alga ancestor were lost. Only genes involved in several valuable metabolic pathways, such as the synthesis of isoprenoid precursors, heme, and fatty acids, have been transferred to the host genome and retained to help these parasites adapt to a complex life cycle and various living environments. The biological function of an apicoplast is essential for most apicomplexan parasites. Considering their potential as drug targets, the metabolic functions of this symbiotic organelle have been intensively investigated through computational and biological means. Moreover, we know that not only organellar metabolic functions are linked with other organelles, but also their biogenesis processes have developed and evolved to tailor their biological functions and proper inheritance. Several distinct features have been found in the biogenesis process of apicoplasts. For example, the apicoplast borrows a dynamin-related protein (DrpA) from its host to implement organelle division. The autophagy system has also been repurposed for linking the apicoplast and centrosome during replication and the division process. However, many vital questions remain to be answered about how these parasites maintain and properly inherit this symbiotic organelle. Here we review our current knowledge about its biogenesis process and discuss several critical questions remaining to be answered in this field.
Collapse
Affiliation(s)
- Ying Zhang
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang Province 163319, PR China; State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Haping Street 678, Nangang District, Harbin 150069, PR China
| | - Chunren Wang
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang Province 163319, PR China
| | - Honglin Jia
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Haping Street 678, Nangang District, Harbin 150069, PR China.
| |
Collapse
|
33
|
O'Neal AJ, Butler LR, Rolandelli A, Gilk SD, Pedra JH. Lipid hijacking: a unifying theme in vector-borne diseases. eLife 2020; 9:61675. [PMID: 33118933 PMCID: PMC7595734 DOI: 10.7554/elife.61675] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/20/2020] [Indexed: 12/16/2022] Open
Abstract
Vector-borne illnesses comprise a significant portion of human maladies, representing 17% of global infections. Transmission of vector-borne pathogens to mammals primarily occurs by hematophagous arthropods. It is speculated that blood may provide a unique environment that aids in the replication and pathogenesis of these microbes. Lipids and their derivatives are one component enriched in blood and are essential for microbial survival. For instance, the malarial parasite Plasmodium falciparum and the Lyme disease spirochete Borrelia burgdorferi, among others, have been shown to scavenge and manipulate host lipids for structural support, metabolism, replication, immune evasion, and disease severity. In this Review, we will explore the importance of lipid hijacking for the growth and persistence of these microbes in both mammalian hosts and arthropod vectors.
Collapse
Affiliation(s)
- Anya J O'Neal
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, United States
| | - L Rainer Butler
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, United States
| | - Agustin Rolandelli
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, United States
| | - Stacey D Gilk
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, United States
| | - Joao Hf Pedra
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, United States
| |
Collapse
|
34
|
Usman MA, Ibrahim MA, Salman AA, Sallau AB. Depletion of cholesterol could be associated with modulation of progesterone but not other sex hormone levels during Plasmodium falciparum infection in humans: a cross-sectional study from Zaria, Nigeria. Parasitol Res 2020; 119:4143-4150. [PMID: 32951142 DOI: 10.1007/s00436-020-06826-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 07/21/2020] [Indexed: 10/23/2022]
Abstract
In order for Plasmodium falciparum to grow and survive in its host, membrane biogenesis, fueled by host cholesterol, is essential for these processes. Consistent with this essential role, more insights into the cholesterol pathway would enhance the current understanding of the pathophysiology of malaria infection. To explore its broader potential, we conducted a cross-sectional study and assayed for the serum levels of cholesterol, vitamin D, progesterone, testosterone, estradiol and bile acid in both P. falciparum-infected patients and apparently healthy sex-matched participants. Our results revealed that the levels of cholesterol, vitamin D, progesterone, testosterone and estradiol in P. falciparum-infected patients were significantly (p < 0.05) lower compared to those in control groups whereas the level of bile acid in P. falciparum-infected patients was significantly (p < 0.05) higher compared to that in control groups. Additionally, cholesterol and the metabolic products with the exception of bile acid had a significant (p < 0.05) association with the parasite density in P. falciparum-infected patients with moderate and high P. falciparum infections. Furthermore, all the metabolic products of cholesterol had an insignificant (p > 0.05) association with the cholesterol in P. falciparum-infected patients with the exception of progesterone which showed a significant (p < 0.05) association with cholesterol in the malaria-infected female patients. Data from the present study demonstrated that progesterone depletion in P. falciparum-infected female patients could be a consequence of P. falciparum-induced decrease in cholesterol.
Collapse
|
35
|
Kenthirapalan S, Tran PN, Kooij TWA, Ridgway MC, Rauch M, Brown SHJ, Mitchell TW, Matuschewski K, Maier AG. Distinct adaptations of a gametocyte ABC transporter to murine and human Plasmodium parasites and its incompatibility in cross-species complementation. Int J Parasitol 2020; 50:511-522. [PMID: 32445722 DOI: 10.1016/j.ijpara.2020.03.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 03/25/2020] [Accepted: 03/27/2020] [Indexed: 11/18/2022]
Abstract
Parasites of the genus Plasmodium infect a wide range of mammalian hosts including humans, primates, bats and arboreal rodents. A hallmark of Plasmodium spp. is the very narrow host range, indicative of matching parasite-host coevolution. Accordingly, their respective genomes harbour many unique genes and gene families that typically encode proteins involved in host cell recognition and remodelling. Whether and to what extent conserved proteins that are shared across Plasmodium spp. also exert distinct species-specific roles remains largely untested. Here, we present detailed functional profiling of the female gametocyte-specific ATP-binding cassette transporter gABCG2 in the murine parasite Plasmodium berghei and compare our findings with data from the orthologous gene in the human parasite Plasmodium falciparum. We show that P. berghei gABCG2 is female-specific and continues to be expressed in zygotes and ookinetes. In contrast to a distinct localization to Iipid-rich gametocyte-specific spots as observed in P. falciparum, the murine malaria parasite homolog is found at the parasite plasma membrane. Plasmodium berghei lacking gABCG2 displays fast asexual blood-stage replication and increased proportions of female gametocytes, consistent with the corresponding P. falciparum knock-out phenotype. Strikingly, cross-species replacement of gABCG2 in either the murine or the human parasite did not restore normal growth rates. The lack of successful complementation despite high conservation across Plasmodium spp. is an indicator of distinct adaptations and tight parasite-host coevolution. Hence, incompatibility of conserved genes in closely related Plasmodium spp. might be more common than previously anticipated.
Collapse
Affiliation(s)
| | - Phuong N Tran
- Research School of Biology, The Australian National University, Canberra, Australian Capital Territory 2601, Australia
| | - Taco W A Kooij
- Department of Medical Microbiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands
| | - Melanie C Ridgway
- Research School of Biology, The Australian National University, Canberra, Australian Capital Territory 2601, Australia
| | - Manuel Rauch
- Parasitology Unit, Max Planck Institute for Infection Biology, 10117 Berlin, Germany; Dept. of Molecular Parasitology, Institute of Biology, Humboldt University, 10115 Berlin, Germany
| | - Simon H J Brown
- School of Biological Sciences, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Todd W Mitchell
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia; School of Medicine, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Kai Matuschewski
- Parasitology Unit, Max Planck Institute for Infection Biology, 10117 Berlin, Germany; Dept. of Molecular Parasitology, Institute of Biology, Humboldt University, 10115 Berlin, Germany.
| | - Alexander G Maier
- Research School of Biology, The Australian National University, Canberra, Australian Capital Territory 2601, Australia.
| |
Collapse
|
36
|
Oluba OM. Erythrocyte Lipid and Antioxidant Changes in <i>Plasmodium falciparum</i>-infected Children Attending Mother and Child Hospital in Akure, Nigeria. Pak J Biol Sci 2020; 22:257-264. [PMID: 31930848 DOI: 10.3923/pjbs.2019.257.264] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND AND OBJECTIVE Understanding the molecular and cellular pathways activated in response to Plasmodium falciparum infection is crucial for the development of pharmacological intervention to malaria. The present study was designed to evaluate the lipid components and the oxidative status of erythrocyte obtained from children under 5 years infected with Plasmodium falciparum. MATERIALS AND METHODS Parasitemia was assessed prior and after treatment with antimalarial, erythrocyte lipid profile, levels of lipid peroxidation and antioxidant status (reduced glutathione, GSH; superoxide dismutase, SOD; catalase and glutathione peroxidase, GPx) were measured. RESULTS Results obtained showed that in Plasmodium infected erythrocyte, the total phospholipids, cholesterol and LDL-cholesterol concentrations were significantly elevated beyond the normal level. In addition, an upsurge in erythrocyte oxidant (lipid peroxide) status with a concomitant downregulation of the antioxidant status (GSH concentration and SOD, catalase and GPx activities) was observed in P. falciparum-infected children. However, following a three-day treatment with artemisinin combination drugs, there was a significant reduction in erythrocyte phospholipids, total cholesterol and LDL-cholesterol concentration as well as lipid peroxide levels. Significant augmentation in erythrocyte antioxidant status (GSH, SOD, catalase and GPx) were also observed after treatment with antimalarials. CONCLUSION This study demonstrated that erythrocyte lipids and oxidative status are usually altered in Plasmodium falciparum-infected children. Thus, monitoring erythrocyte lipid profile and oxidative status could offer a viable diagnostic strategy in early detection of malaria in children.
Collapse
|
37
|
Silva LMR, Lütjohann D, Hamid P, Velasquez ZD, Kerner K, Larrazabal C, Failing K, Hermosilla C, Taubert A. Besnoitia besnoiti infection alters both endogenous cholesterol de novo synthesis and exogenous LDL uptake in host endothelial cells. Sci Rep 2019; 9:6650. [PMID: 31040348 PMCID: PMC6491585 DOI: 10.1038/s41598-019-43153-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 04/12/2019] [Indexed: 02/07/2023] Open
Abstract
Besnoitia besnoiti, an apicomplexan parasite of cattle being considered as emergent in Europe, replicates fast in host endothelial cells during acute infection and is in considerable need for energy, lipids and other building blocks for offspring formation. Apicomplexa are generally considered as defective in cholesterol synthesis and have to scavenge cholesterol from their host cells for successful replication. Therefore, we here analysed the influence of B. besnoiti on host cellular endogenous cholesterol synthesis and on sterol uptake from exogenous sources. GC-MS-based profiling of cholesterol-related sterols revealed enhanced cholesterol synthesis rates in B. besnoiti-infected cells. Accordingly, lovastatin and zaragozic acid treatments diminished tachyzoite production. Moreover, increased lipid droplet contents and enhanced cholesterol esterification was detected and inhibition of the latter significantly blocked parasite proliferation. Furthermore, artificial increase of host cellular lipid droplet disposability boosted parasite proliferation. Interestingly, lectin-like oxidized low density lipoprotein receptor 1 expression was upregulated in infected endothelial hostcells, whilst low density lipoproteins (LDL) receptor was not affected by parasite infection. However, exogenous supplementations with non-modified and acetylated LDL both boosted B. besnoiti proliferation. Overall, current data show that B. besnoiti simultaneously exploits both, endogenous cholesterol biosynthesis and cholesterol uptake from exogenous sources, during asexual replication.
Collapse
Affiliation(s)
- Liliana M R Silva
- Institute of Parasitology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Schubertstr. 81, D-35392, Giessen, Germany.
| | - Dieter Lütjohann
- Institute for Clinical Chemistry and Clinical Pharmacology, University Clinics Bonn, Laboratory for Special Lipid Diagnostics/Center Internal Medicine/Building 26/UG 68, Sigmund-Freud-Str. 25, D-53127, Bonn, Germany
| | - Penny Hamid
- Institute of Parasitology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Schubertstr. 81, D-35392, Giessen, Germany.,Department of Parasitology, Faculty of Veterinary Medicine, Universitas Gadjah Mada, Jl. Fauna No. 2 Karangmalang, 55281, Yogyakarta, Indonesia
| | - Zahady D Velasquez
- Institute of Parasitology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Schubertstr. 81, D-35392, Giessen, Germany
| | - Katharina Kerner
- Institute for Hygiene and Infectious Diseases of Animals, Justus-Liebig-University, Giessen, Frankfurter Str. 85-89, D-35392, Germany
| | - Camilo Larrazabal
- Institute of Parasitology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Schubertstr. 81, D-35392, Giessen, Germany
| | - Klaus Failing
- Unit for Biomathematics and Data Processing, Faculty of Veterinary Medicine, Justus Liebig University Giessen, Frankfurter Str. 95, D-35392, Giessen, Germany
| | - Carlos Hermosilla
- Institute of Parasitology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Schubertstr. 81, D-35392, Giessen, Germany
| | - Anja Taubert
- Institute of Parasitology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Schubertstr. 81, D-35392, Giessen, Germany
| |
Collapse
|
38
|
Abstract
Parasites undergo complex life cycles that comprise a wide variety of cellular differentiation events in different host compartments and transmission across multiple hosts. As parasites depend on host resources, it is not surprising they have developed efficient mechanisms to sense alterations and adapt to the available resources in a wide range of environments. Here we provide an overview of the nutritional needs of different parasites throughout their diverse life stages and highlight recent insights into strategies that both hosts and parasites have developed to meet these nutritional requirements needed for defense, survival, and replication. These studies will provide the foundation for a systems-level understanding of host-parasite interactions, which will require the integration of molecular, epidemiologic, and mechanistic data and the application of interdisciplinary approaches to model parasite regulatory networks that are triggered by alterations in host resources.
Collapse
|
39
|
Oluba OM. Ganoderma terpenoid extract exhibited anti-plasmodial activity by a mechanism involving reduction in erythrocyte and hepatic lipids in Plasmodium berghei infected mice. Lipids Health Dis 2019; 18:12. [PMID: 30636645 PMCID: PMC6330487 DOI: 10.1186/s12944-018-0951-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 12/18/2018] [Indexed: 01/11/2023] Open
Abstract
Bioactive components of Ganoderma lucidum has recently gained intense research attention due to their acclaimed nutritional and medicinal properties. Thus, the terpenoid extract from the fruit bodies of G. lucidum (GT) was evaluated for activity against Plasmodium berghei in mice in two separate experiments. In addition, the effects of the extract on erythrocyte and hepatic lipids as well as liver HMG-CoA reductase activity before and after the treatments were also assessed. Mice with established infection were administered 100 and 250 mg/kg/day GT alone and in combination with chloroquine (CQ), in either case two separate controls designated: CQ (30 mg/kg chloroquine) and INF-CTR (1 mL DMSO) were also included. Treatment was administered orally for 12 days and parasitemia determined every three days. Percentage survival was significantly increased to 87% from 66% due to combination of GT100 with CQ compared to GT100 alone and to 75% from 62% when GT250 was administered with CQ compared to GT250 alone. Erythrocyte triglycerides, total cholesterol (TC), LDL and phospholipids contents were significantly lower in GT + CQ-treated mice compared to CQ alone and INF-CTR. Similarly, hepatic TC and phospholipid levels were significantly lower in the GT + CQ-treated mice compared to CQ alone and INF-CTR and HMG-CoA reductase activity in the liver was significantly inhibited due to administration of GT + CQ. Data from this study suggest that the anti-plasmodial action of GT could involve mechanisms associated with its hypolipidemic activity. It was also demonstrated that chloroquine, when administered in combination with GT, potentiates its curative effect in P. berghei-infected mice.
Collapse
Affiliation(s)
- Olarewaju M Oluba
- Department of Biological Sciences, Food Safety and Toxicology Research Unit, Environment and Technology Research Cluster, College of Science and Engineering, Landmark University, P.M.B, Omu-Aran, Kwara State, 1001, Nigeria.
| |
Collapse
|
40
|
Glennon EKK, Dankwa S, Smith JD, Kaushansky A. Opportunities for Host-targeted Therapies for Malaria. Trends Parasitol 2018; 34:843-860. [PMID: 30122551 PMCID: PMC6168423 DOI: 10.1016/j.pt.2018.07.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 07/20/2018] [Accepted: 07/23/2018] [Indexed: 12/19/2022]
Abstract
Despite the recent successes of artemisinin-based antimalarial drugs, many still die from severe malaria, and eradication efforts are hindered by the limited drugs currently available to target transmissible gametocyte parasites and liver-resident dormant Plasmodium vivax hypnozoites. Host-targeted therapy is a new direction for infectious disease drug development and aims to interfere with host molecules, pathways, or networks that are required for infection or that contribute to disease. Recent advances in our understanding of host pathways involved in parasite development and pathogenic mechanisms in severe malaria could facilitate the development of host-targeted interventions against Plasmodium infection and malaria disease. This review discusses new opportunities for host-targeted therapeutics for malaria and the potential to harness drug polypharmacology to simultaneously target multiple host pathways using a single drug intervention.
Collapse
Affiliation(s)
- Elizabeth K K Glennon
- Center for Infectious Disease Research, 307 Westlake Ave N Suite 500, Seattle, WA 98109, USA; Department of Global Health, University of Washington, Harris Hydraulics Laboratory, Box 357965, Seattle, WA 98195, USA; These authors made an equal contribution
| | - Selasi Dankwa
- Center for Infectious Disease Research, 307 Westlake Ave N Suite 500, Seattle, WA 98109, USA; These authors made an equal contribution
| | - Joseph D Smith
- Center for Infectious Disease Research, 307 Westlake Ave N Suite 500, Seattle, WA 98109, USA; Department of Global Health, University of Washington, Harris Hydraulics Laboratory, Box 357965, Seattle, WA 98195, USA
| | - Alexis Kaushansky
- Center for Infectious Disease Research, 307 Westlake Ave N Suite 500, Seattle, WA 98109, USA; Department of Global Health, University of Washington, Harris Hydraulics Laboratory, Box 357965, Seattle, WA 98195, USA.
| |
Collapse
|
41
|
Abdulkareem AO, Babamale AO, Owolusi LO, Busari SA, Olatunji LA. Anti-plasmodial activity of sodium acetate in Plasmodium berghei-infected mice. J Basic Clin Physiol Pharmacol 2018; 29:493-498. [PMID: 29634486 DOI: 10.1515/jbcpp-2017-0203] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 02/23/2018] [Indexed: 06/08/2023]
Abstract
Abstract
Background
Continuous increase in drug resistance has hindered the control of malaria infection and resulted in multi-drug-resistant parasite strains. This, therefore, intensifies the search for alternative treatments with no or less side effects. Several histone deacetylase inhibitors have been characterised to possess anti-malaria activity; however, their further development as anti-malaria agents has not recorded much success. The present study investigated the anti-plasmodial activity of sodium acetate in Plasmodium berghei-infected mice, aiming at finding a better alternative source of malaria chemotherapy.
Methods
Thirty female Swiss albino mice were randomly distributed into six groups. Groups A (uninfected control) and B (infected control) received only distilled water. Group C (artesunate control) were infected and treated orally with 4 mg/kg artesunate on the first day, and subsequently 2 mg/kg artesunate. Groups D, E and F were infected and orally treated with 50, 100 and 200 mg/kg sodium acetate, respectively.
Results
Sodium acetate significantly lowered parasitaemia (p<0.05) after 4 days post-treatment, and the parasite inhibition rate of 68.5% at 50 mg/kg compared favourably with the 73.3% rate of artesunate. Similarly, administration of 50 mg/kg sodium acetate improved serum total cholesterol relatively better than artesunate. Our results also revealed that sodium acetate does not interfere with liver function, as there was no significant difference (p>0.05) in the serum activities of aspartate aminotransferase and alanine aminotransferase in both infected treated and uninfected mice.
Conclusions
This study shows that sodium acetate may be a safe alternative source of anti-malaria drugs. Its effect on the serum total cholesterol also predicts its ability in correcting malaria-induced metabolic syndromes.
Collapse
Affiliation(s)
- Adam O Abdulkareem
- Animal Physiology Unit, Department of Zoology, University of Ilorin, Ilorin, Nigeria, Phone: +2348066528548
| | | | - Lucky O Owolusi
- Animal Physiology Unit, Department of Zoology, University of Ilorin, Ilorin, Nigeria
| | - Simbiat A Busari
- Animal Physiology Unit, Department of Zoology, University of Ilorin, Ilorin, Nigeria
| | - Lawrence A Olatunji
- Cardiometabolic Research Unit, Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| |
Collapse
|
42
|
Taubert A, Silva LMR, Velásquez ZD, Larrazabal C, Lütjohann D, Hermosilla C. Modulation of cholesterol-related sterols during Eimeria bovis macromeront formation and impact of selected oxysterols on parasite development. Mol Biochem Parasitol 2018; 223:1-12. [PMID: 29909067 DOI: 10.1016/j.molbiopara.2018.06.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 06/12/2018] [Accepted: 06/12/2018] [Indexed: 11/16/2022]
Abstract
Obligate intracellular apicomplexan parasites are considered as deficient in cholesterol biosynthesis and scavenge cholesterol from their host cell in a parasite-specific manner. Compared to fast proliferating apicomplexan species producing low numbers of merozoites per host cell, (e. g. Toxoplasma gondii), the macromeront-forming protozoa Eimeria bovis is in extraordinary need for cholesterol for offspring production (≥ 170,000 merozoites I/macromeront). Interestingly, optimized in vitro E. bovis merozoite I production occurs under low foetal calf serum (FCS, 1.2%) supplementation. To analyze the impact of extensive E. bovis proliferation on host cellular sterol metabolism we here compared the sterol profiles of E. bovis-infected primary endothelial host cells grown under optimized (1.2% FCS) and non-optimized (10% FCS) cell culture conditions. Therefore, several sterols indicating endogenous de novo cholesterol synthesis, cholesterol conversion and sterol uptake (phytosterols) were analyzed via GC-MS-based approaches. Overall, significantly enhanced levels of phytosterols were detected in both FCS conditions indicating infection-triggered sterol uptake from extracellular sources as a major pathway of sterol acquisition. Interestingly, a simultaneous induction of endogenous cholesterol synthesis based on increased levels of distinct cholesterol precursors was only observed in case of optimized parasite proliferation indicating a parasite proliferation-dependent effect. Considering side-chain oxysterols, 25 hydroxycholesterol levels were selectively found increased in E. bovis-infected host cells, while 24 hydroxycholesterol and 27 hydroxycholesterol contents were not significantly altered by infection. Exogenous treatments with 25 hydroxycholesterol, 27 hydroxycholesterol, and 7 ketocholesterol revealed significant adverse effects on E. bovis intracellular development. Thus, the number and size of developing macromeronts and merozoite I production was significantly reduced indicating that these oxysterols bear direct or indirect antiparasitic properties. Overall, the current data indicate parasite-driven changes in the host cellular sterol profile reflecting the huge demand of E. bovis for cholesterol during macromeront formation and its versatility in the acquisition of cholesterol sources.
Collapse
Affiliation(s)
- A Taubert
- Institute of Parasitology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Schubertstr. 81, 35392 Giessen, Germany.
| | - L M R Silva
- Institute of Parasitology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Schubertstr. 81, 35392 Giessen, Germany.
| | - Z D Velásquez
- Institute of Parasitology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Schubertstr. 81, 35392 Giessen, Germany.
| | - C Larrazabal
- Institute of Parasitology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Schubertstr. 81, 35392 Giessen, Germany.
| | - D Lütjohann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Clinics of Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany.
| | - C Hermosilla
- Institute of Parasitology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Schubertstr. 81, 35392 Giessen, Germany.
| |
Collapse
|
43
|
Vallochi AL, Teixeira L, Oliveira KDS, Maya-Monteiro CM, Bozza PT. Lipid Droplet, a Key Player in Host-Parasite Interactions. Front Immunol 2018; 9:1022. [PMID: 29875768 PMCID: PMC5974170 DOI: 10.3389/fimmu.2018.01022] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 04/24/2018] [Indexed: 12/18/2022] Open
Abstract
Lipid droplets (lipid bodies, LDs) are dynamic organelles that have important roles in regulating lipid metabolism, energy homeostasis, cell signaling, membrane trafficking, and inflammation. LD biogenesis, composition, and functions are highly regulated and may vary according to the stimuli, cell type, activation state, and inflammatory environment. Increased cytoplasmic LDs are frequently observed in leukocytes and other cells in a number of infectious diseases. Accumulating evidence reveals LDs participation in fundamental mechanisms of host-pathogen interactions, including cell signaling and immunity. LDs are sources of eicosanoid production, and may participate in different aspects of innate signaling and antigen presentation. In addition, intracellular pathogens evolved mechanisms to subvert host metabolism and may use host LDs, as ways of immune evasion and nutrients source. Here, we review mechanisms of LDs biogenesis and their contributions to the infection progress, and discuss the latest discoveries on mechanisms and pathways involving LDs roles as regulators of the immune response to protozoan infection.
Collapse
Affiliation(s)
- Adriana Lima Vallochi
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | | | | | | | - Patricia T. Bozza
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| |
Collapse
|
44
|
Posfai D, Sylvester K, Reddy A, Ganley JG, Wirth J, Cullen QE, Dave T, Kato N, Dave SS, Derbyshire ER. Plasmodium parasite exploits host aquaporin-3 during liver stage malaria infection. PLoS Pathog 2018; 14:e1007057. [PMID: 29775485 PMCID: PMC5979039 DOI: 10.1371/journal.ppat.1007057] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 05/31/2018] [Accepted: 04/25/2018] [Indexed: 12/22/2022] Open
Abstract
Within the liver a single Plasmodium parasite transforms into thousands of blood-infective forms to cause malaria. Here, we use RNA-sequencing to identify host genes that are upregulated upon Plasmodium berghei infection of hepatocytes with the hypothesis that host pathways are hijacked to benefit parasite development. We found that expression of aquaporin-3 (AQP3), a water and glycerol channel, is significantly induced in Plasmodium-infected hepatocytes compared to uninfected cells. This aquaglyceroporin localizes to the parasitophorous vacuole membrane, the compartmental interface between the host and pathogen, with a temporal pattern that correlates with the parasite’s expansion in the liver. Depletion or elimination of host AQP3 expression significantly reduces P. berghei parasite burden during the liver stage and chemical disruption by a known AQP3 inhibitor, auphen, reduces P. falciparum asexual blood stage and P. berghei liver stage parasite load. Further use of this inhibitor as a chemical probe suggests that AQP3-mediated nutrient transport is an important function for parasite development. This study reveals a previously unknown potential route for host-dependent nutrient acquisition by Plasmodium which was discovered by mapping the transcriptional changes that occur in hepatocytes throughout P. berghei infection. The dataset reported may be leveraged to identify additional host factors that are essential for Plasmodium liver stage infection and highlights Plasmodium’s dependence on host factors within hepatocytes. Plasmodium parasites undergo an obligatory morphogenesis and replication within the liver before they invade red blood cells and cause malaria. The liver stage is clinically silent but essential for the Plasmodium parasite to complete its life cycle. During this time, the parasite relies on the host cell to support a massive replication event, yet host factors that are critical to this expansion are largely unknown. We identify human aquaporin-3 (AQP3), a water and glycerol channel, as essential for the proper development of the parasite within the liver cell. AQP3 localizes to the parasitophorous vacuole membrane, the interface between the host cytoplasm and the parasite, possibly aiding in the nutritional uptake for the parasite. Genetic disruption or treatment with the AQP3 inhibitor auphen, reduces parasite load in liver and blood cells.
Collapse
Affiliation(s)
- Dora Posfai
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, North Carolina, United States of America
| | - Kayla Sylvester
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, North Carolina, United States of America
| | - Anupama Reddy
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Jack G. Ganley
- Department of Chemistry, Duke University, Durham, North Carolina, United States of America
| | - Johannes Wirth
- Department of Chemistry, Duke University, Durham, North Carolina, United States of America
| | - Quinlan E. Cullen
- Department of Chemistry, Duke University, Durham, North Carolina, United States of America
| | - Tushar Dave
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Nobutaka Kato
- The Broad Institute, 7 Cambridge Center, Cambridge, Massachusetts, United States of America
| | - Sandeep S. Dave
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Emily R. Derbyshire
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, North Carolina, United States of America
- Department of Chemistry, Duke University, Durham, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
45
|
Rakovac Tisdall A, Crowley VEF, Crook MA. Undetectable high-density lipoprotein cholesterol in acute malaria. J Clin Lipidol 2018; 12:822-825. [DOI: 10.1016/j.jacl.2018.02.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 02/20/2018] [Accepted: 02/28/2018] [Indexed: 01/20/2023]
|
46
|
An evaluation of lipid metabolism in the insect trypanosomatid Herpetomonas muscarum uncovers a pathway for the uptake of extracellular insect lipoproteins. Parasitol Int 2018; 67:97-106. [DOI: 10.1016/j.parint.2017.10.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Revised: 10/28/2017] [Accepted: 10/31/2017] [Indexed: 11/18/2022]
|
47
|
Arama C, Diarra I, Kouriba B, Sirois F, Fedoryak O, Thera MA, Coulibaly D, Lyke KE, Plowe CV, Chrétien M, Doumbo OK, Mbikay M. Malaria severity: Possible influence of the E670G PCSK9 polymorphism: A preliminary case-control study in Malian children. PLoS One 2018; 13:e0192850. [PMID: 29447211 PMCID: PMC5813955 DOI: 10.1371/journal.pone.0192850] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 01/31/2018] [Indexed: 11/19/2022] Open
Abstract
Aim Proprotein Convertase Subtilisin/Kexin Type 9 (PCSK9) is a hepatic secretory protein which promotes the degradation of low-density lipoprotein receptors leading to reduced hepatic uptake of plasma cholesterol. Non-synonymous single-nucleotide polymorphisms in its gene have been linked to hypo- or hyper- cholesterolemia, depending on whether they decrease or increase PCSK9 activity, respectively. Since the proliferation and the infectivity of Plasmodium spp. partially depend on cholesterol from the host, we hypothesize that these PCSK9 genetic polymorphisms could influence the course of malaria infection in individuals who carry them. Here we examined the frequency distribution of one dominant (C679X) and two recessive (A443T, I474V) hypocholesterolemic polymorphisms as well as that of one recessive hypercholesterolemic polymorphism (E670G) among healthy and malaria-infected Malian children. Methods Dried blood spots were collected in Bandiagara, Mali, from 752 age, residence and ethnicity-matched children: 253 healthy controls, 246 uncomplicated malaria patients and 253 severe malaria patients. Their genomic DNA was extracted and genotyped for the above PCSK9 polymorphisms using Taqman assays. Associations of genotype distributions and allele frequencies with malaria were evaluated. Results The minor allele frequency of the A443T, I474V, E670G, and C679X polymorphisms in the study population sample was 0.12, 0.20, 0.26, and 0.02, respectively. For each polymorphism, the genotype distribution among the three health conditions was statistically insignificant, but for the hypercholesterolemic E670G polymorphism, a trend towards association of the minor allele with malaria severity was observed (P = 0.035). The association proved to be stronger when allele frequencies between healthy controls and severe malaria cases were compared (Odd Ratio: 1.34; 95% Confidence Intervals: 1.04–1.83); P = 0.031). Conclusions Carriers of the minor allele of the E670G PCSK9 polymorphism might be more susceptible to severe malaria. Further investigation of the cholesterol regulating function of PCSK9 in the pathophysiology of malaria is needed.
Collapse
Affiliation(s)
- Charles Arama
- Malaria Research and Training Center, International Centers for Excellence in Research, Université des Sciences, des Techniques et des Technologies de Bamako, Bamako, Mali
| | - Issa Diarra
- Malaria Research and Training Center, International Centers for Excellence in Research, Université des Sciences, des Techniques et des Technologies de Bamako, Bamako, Mali
| | - Bourèma Kouriba
- Malaria Research and Training Center, International Centers for Excellence in Research, Université des Sciences, des Techniques et des Technologies de Bamako, Bamako, Mali
| | - Francine Sirois
- Laboratoire de protéolyse fonctionnelle, Institut de recherches cliniques de Montréal, Montréal, Québec, Canada
| | - Olesya Fedoryak
- Laboratoire de protéolyse fonctionnelle, Institut de recherches cliniques de Montréal, Montréal, Québec, Canada
| | - Mahamadou A. Thera
- Malaria Research and Training Center, International Centers for Excellence in Research, Université des Sciences, des Techniques et des Technologies de Bamako, Bamako, Mali
| | - Drissa Coulibaly
- Malaria Research and Training Center, International Centers for Excellence in Research, Université des Sciences, des Techniques et des Technologies de Bamako, Bamako, Mali
| | - Kirsten E. Lyke
- Center for Vaccine Development and Division of Malaria Research, Institute for Global Health, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Christopher V. Plowe
- Center for Vaccine Development and Division of Malaria Research, Institute for Global Health, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Michel Chrétien
- Laboratoire de protéolyse fonctionnelle, Institut de recherches cliniques de Montréal, Montréal, Québec, Canada
- Chronic Disease Program, Ottawa Hospital Research Hospital, Ottawa, Ontario, Canada
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Ogobara K. Doumbo
- Malaria Research and Training Center, International Centers for Excellence in Research, Université des Sciences, des Techniques et des Technologies de Bamako, Bamako, Mali
- * E-mail: (MM); (OKD)
| | - Majambu Mbikay
- Laboratoire de protéolyse fonctionnelle, Institut de recherches cliniques de Montréal, Montréal, Québec, Canada
- Chronic Disease Program, Ottawa Hospital Research Hospital, Ottawa, Ontario, Canada
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- * E-mail: (MM); (OKD)
| |
Collapse
|
48
|
Cariaco Y, Lima WR, Sousa R, Nascimento LAC, Briceño MP, Fotoran WL, Wunderlich G, Dos Santos JL, Silva NM. Ethanolic extract of the fungus Trichoderma stromaticum decreases inflammation and ameliorates experimental cerebral malaria in C57BL/6 mice. Sci Rep 2018; 8:1547. [PMID: 29367729 PMCID: PMC5784021 DOI: 10.1038/s41598-018-19840-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 01/05/2018] [Indexed: 01/31/2023] Open
Abstract
Increased resistance to the first-line treatment against P. falciparum malaria, artemisinin-based combination therapies, has been reported. Here, we tested the effect of crude ethanolic extract of the fungus Trichoderma stromaticum (Ext-Ts) on the growth of P. falciparum NF54 in infected human red blood cells (ihRBCs) and its anti-malarial and anti-inflammatory properties in a mouse model of experimental cerebral malaria. For this purpose, ihRBCs were treated with Ext-Ts and analysed for parasitaemia; C57BL/6 mice were infected with P. berghei ANKA (PbA), treated daily with Ext-Ts, and clinical, biochemical, histological and immunological features of the disease were monitored. It was observed that Ext-Ts presented a dose-dependent ability to control P. falciparum in ihRBCs. In addition, it was demonstrated that Ext-Ts treatment of PbA-infected mice was able to increase survival, prevent neurological signs and decrease parasitaemia at the beginning of infection. These effects were associated with systemically decreased levels of lipids and IFN-γ, ICAM-1, VCAM-1 and CCR5 cerebral expression, preserving blood brain barrier integrity and attenuating the inflammatory lesions in the brain, liver and lungs. These results suggest that Ext-Ts could be a source of immunomodulatory and antimalarial compounds that could improve the treatment of cerebral malaria.
Collapse
Affiliation(s)
- Yusmaris Cariaco
- Laboratory of Immunopathology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, 38400-902, Minas Gerais, Brazil
| | - Wânia Rezende Lima
- Institute of Exact and Natural Sciences, Federal University of Mato Grosso, Rondonópolis, 78735-901, Mato Grosso, Brazil
| | - Romulo Sousa
- Laboratory of Immunopathology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, 38400-902, Minas Gerais, Brazil
| | - Layane Alencar Costa Nascimento
- Laboratory of Immunopathology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, 38400-902, Minas Gerais, Brazil
| | - Marisol Pallete Briceño
- Laboratory of Immunopathology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, 38400-902, Minas Gerais, Brazil
| | | | - Gerhard Wunderlich
- Department of Parasitology, University of São Paulo, São Paulo, 05508-900, Brazil
| | | | - Neide Maria Silva
- Laboratory of Immunopathology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, 38400-902, Minas Gerais, Brazil.
| |
Collapse
|
49
|
Aquaglyceroporin PbAQP is required for efficient progression through the liver stage of Plasmodium infection. Sci Rep 2018; 8:655. [PMID: 29330527 PMCID: PMC5766620 DOI: 10.1038/s41598-017-18987-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 12/19/2017] [Indexed: 12/04/2022] Open
Abstract
The discovery of aquaglyceroporins (AQP) has highlighted a new mechanism of membrane solute transport that may hold therapeutic potential for controlling parasitic infections, including malaria. Plasmodium parasites express a single AQP at the plasma membrane that functions as a channel for water, nutrients and waste into and out cells. We previously demonstrated that Plasmodium berghei targeted for PbAQP deletion are deficient in glycerol import and less virulent than wild-type parasites during the blood developmental stage. Here, we have examined the contribution of PbAQP to the infectivity of P. berghei in the liver. PbAQP is expressed in the sporozoite mosquito stage and is detected at low levels in intrahepatic parasites at the onset of hepatocyte infection. As the parasites progress to late hepatic stages, PbAQP transcription increases and PbAQP localizes to the plasma membrane of hepatic merozoites. Compared to wild-type parasites, PbAQP-null sporozoites exhibit a delay in blood stage infection due to slower replication in hepatocytes, resulting in retardation of merosome production. Furthermore, PbAQP disruption results in a significant reduction in erythrocyte infectivity by hepatocyte-derived merozoites. Hepatic merozoites incorporate exogenous glycerol into glycerophospholipids and PbAQP-null merozoites contain less phosphatidylcholine than wild-type merozoites, underlining the contribution of Plasmodium AQP to phospholipid syntheses.
Collapse
|
50
|
Moreira D, Estaquier J, Cordeiro-da-Silva A, Silvestre R. Metabolic Crosstalk Between Host and Parasitic Pathogens. EXPERIENTIA SUPPLEMENTUM (2012) 2018; 109:421-458. [PMID: 30535608 DOI: 10.1007/978-3-319-74932-7_12] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
A complex network that embraces parasite-host intrinsic factors and the microenvironment regulated the interaction between a parasite and its host. Nutritional pressures exerted by both elements of this duet thus dictate this host-parasite niche. To survive and proliferate inside a host and a harsh nutritional environment, the parasites modulate different nutrient sensing pathways to subvert host metabolic pathways. Such mechanism is able to change the flux of distinct nutrients/metabolites diverting them to be used by the parasites. Apart from this nutritional strategy, the scavenging of nutrients, particularly host fatty acids, constitutes a critical mechanism to fulfil parasite nutritional requirements, ultimately defining the host metabolic landscape. The host metabolic alterations that result from host-parasite metabolic coupling can certainly be considered important targets to improve diagnosis and also for the development of future therapies. Metabolism is in fact considered a key element within this complex interaction, its modulation being crucial to dictate the final infection outcome.
Collapse
Affiliation(s)
- Diana Moreira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
- i3S-Instituto de Investigacão e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- Departamento de Ciências Bioloógicas, Faculdade de Farmaácia, Universidade do Porto, Porto, Portugal
| | - Jérôme Estaquier
- CNRS FR 3636, Université Paris Descartes, Paris, France
- Centre de Recherche du CHU de Québec, Université Laval, Québec, Canada
| | - Anabela Cordeiro-da-Silva
- i3S-Instituto de Investigacão e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- Departamento de Ciências Bioloógicas, Faculdade de Farmaácia, Universidade do Porto, Porto, Portugal
| | - Ricardo Silvestre
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|