1
|
Fraering J, Salnot V, Gautier EF, Ezinmegnon S, Argy N, Peoc'h K, Manceau H, Alao J, Guillonneau F, Migot-Nabias F, Bertin GI, Kamaliddin C. Infected erythrocytes and plasma proteomics reveal a specific protein signature of severe malaria. EMBO Mol Med 2024; 16:319-333. [PMID: 38297098 PMCID: PMC10897182 DOI: 10.1038/s44321-023-00010-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 11/18/2023] [Accepted: 11/22/2023] [Indexed: 02/02/2024] Open
Abstract
Cerebral malaria (CM), the most lethal complication of Plasmodium falciparum severe malaria (SM), remains fatal for 15-25% of affected children despite the availability of treatment. P. falciparum infects and multiplies in erythrocytes, contributing to anemia, parasite sequestration, and inflammation. An unbiased proteomic assessment of infected erythrocytes and plasma samples from 24 Beninese children was performed to study the complex mechanisms underlying CM. A significant down-regulation of proteins from the ubiquitin-proteasome pathway and an up-regulation of the erythroid precursor marker transferrin receptor protein 1 (TFRC) were associated with infected erythrocytes from CM patients. At the plasma level, the samples clustered according to clinical presentation. Significantly, increased levels of the 20S proteasome components were associated with SM. Targeted quantification assays confirmed these findings on a larger cohort (n = 340). These findings suggest that parasites causing CM preferentially infect reticulocytes or erythroblasts and alter their maturation. Importantly, the host plasma proteome serves as a specific signature of SM and presents a remarkable opportunity for developing innovative diagnostic and prognostic biomarkers.
Collapse
Affiliation(s)
- Jeremy Fraering
- UMR261 MERIT, Université Paris Cité, IRD, F-75006, Paris, France
- Plateforme Proteom'IC, Institut Cochin, Université Paris Cité, INSERM U-1016, CNRS UMR8104, Paris, France
| | - Virginie Salnot
- Plateforme Proteom'IC, Institut Cochin, Université Paris Cité, INSERM U-1016, CNRS UMR8104, Paris, France
| | - Emilie-Fleur Gautier
- Plateforme Proteom'IC, Institut Cochin, Université Paris Cité, INSERM U-1016, CNRS UMR8104, Paris, France
- Institut Imagine-INSERM U1163, Hôpital Necker, Université Paris Cité, F-75015, Paris, France
- Laboratoire d'Excellence GR-Ex, F-75015, Paris, France
| | - Sem Ezinmegnon
- Groupe de Recherche Action en Santé, Ouagadougou, Burkina Faso
| | - Nicolas Argy
- UMR261 MERIT, Université Paris Cité, IRD, F-75006, Paris, France
- Laboratoire de parasitologie, Hôpital Bichat-Claude Bernard, APHP, Paris, France
| | - Katell Peoc'h
- Laboratoire d'Excellence GR-Ex, F-75015, Paris, France
- Biochimie Métabolique et Cellulaire, Hôpital Bichat-Claude Bernard, APHP, Paris, France
- Centre de Recherche sur l'Inflammation, UFR de Médecine Xavier Bichat, Université Paris Cité, INSERM UMR1149, Paris, France
| | - Hana Manceau
- Laboratoire d'Excellence GR-Ex, F-75015, Paris, France
- Biochimie Métabolique et Cellulaire, Hôpital Bichat-Claude Bernard, APHP, Paris, France
- Département de Biochimie, Hôpital Universitaire Beaujon, APHP, Clichy, France
| | - Jules Alao
- Service de Pédiatrie, Centre Hospitalier Universitaire de la Mère et de l'Enfant-Lagune de Cotonou, Cotonou, Benin
| | - François Guillonneau
- Plateforme Proteom'IC, Institut Cochin, Université Paris Cité, INSERM U-1016, CNRS UMR8104, Paris, France
- Unité OncoProtéomique, Institut de Cancérologie de l'Ouest, F-49055, Angers, France
- Université d'Angers, Inserm UMR 1307, CNRS UMR 6075, Nantes Université, CRCI2NA, F-49000, Angers, France
| | | | - Gwladys I Bertin
- UMR261 MERIT, Université Paris Cité, IRD, F-75006, Paris, France.
| | - Claire Kamaliddin
- UMR261 MERIT, Université Paris Cité, IRD, F-75006, Paris, France.
- Cumming School of Medicine, The University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
2
|
Zhong M, Zhou B. Plasmodium yoelii iron transporter PyDMT1 interacts with host ferritin and is required in full activity for malarial pathogenesis. BMC Biol 2023; 21:279. [PMID: 38049852 PMCID: PMC10696721 DOI: 10.1186/s12915-023-01776-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 11/22/2023] [Indexed: 12/06/2023] Open
Abstract
BACKGROUND The rapid reproduction of malaria parasites requires proper iron uptake. However, the process of iron absorption by parasites is rarely studied. Divalent metal transporter (DMT1) is a critical iron transporter responsible for uptaking iron. A homolog of human DMT1 exists in the malaria parasite genome, which in Plasmodium yoelii is hereafter named PyDMT1. RESULTS PyDMT1 knockout appears to be lethal. Surprisingly, despite dwelling in an iron-rich environment, the parasite cannot afford to lose even partial expression of PyDMT1; PyDMT1 hypomorphs were associated with severe growth defects and quick loss of pathogenicity. Iron supplementation could completely suppress the defect of the PyDMT1 hypomorph during in vitro culturing. Genetic manipulation through host ferritin (Fth1) knockout to increase intracellular iron levels enforced significant growth inhibition in vivo on the normal parasites but not the mutant. In vitro culturing with isolated ferritin knockout mouse erythrocytes completely rescued PyDMT1-hypomorph parasites. CONCLUSION A critical iron requirement of malaria parasites at the blood stage as mediated by this newly identified iron importer PyDMT1, and the iron homeostasis in malarial parasites is finely tuned. Tipping the iron balance between the parasite and host will efficiently kill the pathogenicity of the parasite. Lastly, PyDMT1 hypomorph parasites were less sensitive to the action of artemisinin.
Collapse
Affiliation(s)
- Mengjiao Zhong
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Bing Zhou
- Faculty of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
3
|
Koonyosying P, Srichairatanakool S, Tiwananthagorn S, Sthitmatee N. Inhibitory effects on bovine babesial infection by iron chelator, 1-(N-acetyl-6-aminohexyl)- 3-hydroxy-2-methylpyridin-4-one (CM1), and antimalarial drugs. Vet Parasitol 2023; 324:110055. [PMID: 37931475 DOI: 10.1016/j.vetpar.2023.110055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 10/12/2023] [Accepted: 10/17/2023] [Indexed: 11/08/2023]
Abstract
BACKGROUND Babesiosis is an infectious disease caused by protozoa of the apicomplexan phylum, genus Babesia. It is a malaria-like parasitic disease that can be transmitted via tick bites. The apicomplexan phylum of eukaryotic microbial parasites has had detrimental impacts on human and veterinary medicine. There are only a few drugs currently available to treat this disease; however, parasitic strains that are resistant to these commercial drugs are increasing in numbers. Plasmodium and Babesia are closely related as they share similar biological features including mechanisms for host cell invasion and metabolism. Therefore, antimalarial drugs may be useful in the treatment of Babesia infections. In addition to antimalarials, iron chelators also inhibit parasite growth. In this study, we aimed to evaluate the in vitro inhibitory efficacy of iron chelator and different antimalarials in the treatment of Babesia bovis. METHODS Cytotoxicity of antimalarial drugs; pyrimethamine, artefenomel, chloroquine, primaquine, dihydroarthemisinine, and the iron chelator, 1-(N-acetyl-6-aminohexyl)- 3-hydroxy-2 methylpyridin-4-one (CM1), were evaluated against Madin Darby Bovine Kidney (MDBK) cells and compared to diminazene aceturate, which is the currently available drug for animal babesiosis using an MTT solution. Afterwards, an evaluation of the in vitro growth-inhibitory effects of antimalarial drug concentrations was performed and monitored using a flow cytometer. Half maximal inhibitory concentrations (IC50) of each antimalarial and iron chelator were determined and compared to the antibabesial drug, diminazine aceturate, by interpolation using a curve-fitting technique. Subsequently, the effect of the drug combination was assessed by constructing an isobologram. Values of the sum of fractional inhibitions at 50% inhibition were then estimated. RESULTS Results indicate that all drugs tested could safely inhibit babesia parasite growth, as high as 2500 μM were non-toxic to mammalian cells. Although no drugs inhibited B. bovis more effectively than diminazine aceturate in this experiment, in vitro growth inhibition results with IC50 values of pyrimethamine 6.25 ± 2.59 μM, artefenomel 2.56 ± 0.67 μM, chloroquine 2.14 ± 0.76 μM, primaquine 22.61 ± 6.72 μM, dihydroarthemisinine 4.65 ± 0.22 μM, 1-(N-acetyl-6-aminohexyl)- 3-hydroxy-2 methylpyridin-4-one (CM1) 9.73 ± 1.90 μM, and diminazine aceturate 0.42 ± 0.01 μM, confirm that all drugs could inhibit B. bovis and could be used as alternative treatments for bovine babesial infection. Furthermore, the efficacy of a combination of the iron chelator, CM1, in combination with artefenomel dihydroarthemisinin or chloroquine, and artefenomel in combination with the iron chelator, CM1, dihydroarthemisinin or chloroquine, exhibited synergism against B. bovis in vitro. CONCLUSION Our evaluation of the inhibitory efficacy of the iron chelator CM1, antimalarial drugs, and a combination of these drugs against B. bovis could be potentially useful in the development and discovery of a novel drug for the treatment of B. bovis in the future.
Collapse
Affiliation(s)
- Pongpisid Koonyosying
- Laboratory of Veterinary Vaccine and Biological Products, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand; Office of Research Administration, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Somdet Srichairatanakool
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Saruda Tiwananthagorn
- Laboratory of Veterinary Vaccine and Biological Products, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand
| | - Nattawooti Sthitmatee
- Laboratory of Veterinary Vaccine and Biological Products, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand.
| |
Collapse
|
4
|
Wideman SK, Frost JN, Richter FC, Naylor C, Lopes JM, Viveiros N, Teh MR, Preston AE, White N, Yusuf S, Draper SJ, Armitage AE, Duarte TL, Drakesmith H. Cellular iron governs the host response to malaria. PLoS Pathog 2023; 19:e1011679. [PMID: 37812650 PMCID: PMC10586691 DOI: 10.1371/journal.ppat.1011679] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/19/2023] [Accepted: 09/11/2023] [Indexed: 10/11/2023] Open
Abstract
Malaria and iron deficiency are major global health problems with extensive epidemiological overlap. Iron deficiency-induced anaemia can protect the host from malaria by limiting parasite growth. On the other hand, iron deficiency can significantly disrupt immune cell function. However, the impact of host cell iron scarcity beyond anaemia remains elusive in malaria. To address this, we employed a transgenic mouse model carrying a mutation in the transferrin receptor (TfrcY20H/Y20H), which limits the ability of cells to internalise iron from plasma. At homeostasis TfrcY20H/Y20H mice appear healthy and are not anaemic. However, TfrcY20H/Y20H mice infected with Plasmodium chabaudi chabaudi AS showed significantly higher peak parasitaemia and body weight loss. We found that TfrcY20H/Y20H mice displayed a similar trajectory of malaria-induced anaemia as wild-type mice, and elevated circulating iron did not increase peak parasitaemia. Instead, P. chabaudi infected TfrcY20H/Y20H mice had an impaired innate and adaptive immune response, marked by decreased cell proliferation and cytokine production. Moreover, we demonstrated that these immune cell impairments were cell-intrinsic, as ex vivo iron supplementation fully recovered CD4+ T cell and B cell function. Despite the inhibited immune response and increased parasitaemia, TfrcY20H/Y20H mice displayed mitigated liver damage, characterised by decreased parasite sequestration in the liver and an attenuated hepatic immune response. Together, these results show that host cell iron scarcity inhibits the immune response but prevents excessive hepatic tissue damage during malaria infection. These divergent effects shed light on the role of iron in the complex balance between protection and pathology in malaria.
Collapse
Affiliation(s)
- Sarah K. Wideman
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Joe N. Frost
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Felix C. Richter
- Kennedy Institute of Rheumatology, Roosevelt Drive, Oxford, United Kingdom
| | - Caitlin Naylor
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - José M. Lopes
- Faculty of Medicine (FMUP) and Institute of Molecular Pathology, Immunology (IPATIMUP), University of Porto, Porto, Portugal
- Instituto de Biologia Molecular e Celular & Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
| | - Nicole Viveiros
- Instituto de Biologia Molecular e Celular & Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
| | - Megan R. Teh
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Alexandra E. Preston
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Natasha White
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Shamsideen Yusuf
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Simon J. Draper
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, United Kingdom
| | - Andrew E. Armitage
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Tiago L. Duarte
- Faculty of Medicine (FMUP) and Institute of Molecular Pathology, Immunology (IPATIMUP), University of Porto, Porto, Portugal
- Instituto de Biologia Molecular e Celular & Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
| | - Hal Drakesmith
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| |
Collapse
|
5
|
Hider RC, Pourzand C, Ma Y, Cilibrizzi A. Optical Imaging Opportunities to Inspect the Nature of Cytosolic Iron Pools. Molecules 2023; 28:6467. [PMID: 37764245 PMCID: PMC10537325 DOI: 10.3390/molecules28186467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 08/31/2023] [Accepted: 09/02/2023] [Indexed: 09/29/2023] Open
Abstract
The chemical nature of intracellular labile iron pools (LIPs) is described. By virtue of the kinetic lability of these pools, it is suggested that the isolation of such species by chromatography methods will not be possible, but rather mass spectrometric techniques should be adopted. Iron-sensitive fluorescent probes, which have been developed for the detection and quantification of LIP, are described, including those specifically designed to monitor cytosolic, mitochondrial, and lysosomal LIPs. The potential of near-infrared (NIR) probes for in vivo monitoring of LIP is discussed.
Collapse
Affiliation(s)
- Robert Charles Hider
- Institute of Pharmaceutical Science, King’s College London, London SE1 9NH, UK
- Department of Life Sciences, University of Bath, Bath BA2 7AY, UK;
| | - Charareh Pourzand
- Department of Life Sciences, University of Bath, Bath BA2 7AY, UK;
- Centre for Therapeutic Innovation, University of Bath, Bath BA2 7AY, UK
- Centre for Bioengineering and Biomedical Technologies, University of Bath, Bath BA2 7AY, UK
| | - Yongmin Ma
- Institute of Advanced Studies, School of Pharmaceutical and Chemical Engineering, Taizhou University, 1139 Shifu Avenue, Taizhou 318000, China;
| | - Agostino Cilibrizzi
- Institute of Pharmaceutical Science, King’s College London, London SE1 9NH, UK
- Centre for Therapeutic Innovation, University of Bath, Bath BA2 7AY, UK
| |
Collapse
|
6
|
Bahati YL, Delanghe J, Balaluka GB, Vandepoele K, Cirhuza JC, Kishabongo AS, Philippé J. Ferroportin Q248H mutation was not found to be protective against malaria and anemia in children under 5 years living in South Kivu/Democratic Republic of Congo, an endemic area of Plasmodium infection. Heliyon 2022; 8:e10460. [PMID: 36060466 PMCID: PMC9437791 DOI: 10.1016/j.heliyon.2022.e10460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 02/21/2022] [Accepted: 08/22/2022] [Indexed: 10/31/2022] Open
Abstract
Background Materials and methods Results Conclusion Frequency of ferroportin Q248H is high in African population. Ferroportin has been described to protect red blood cells against oxidative stress and malaria infection. Ferroportin allele frequency was similar in anemic and non-anemic children. No difference according to ferroportin allele frequency was observed among children in endemic area of Plasmodium infection. Ferroportin Q248H mutation, G6PD deficiency and sickle cell anemia did not affect susceptibility to malaria in endemic area.
Collapse
|
7
|
Li S, Xu W, Wang H, Tang T, Ma J, Cui Z, Shi H, Qin T, Zhou H, Li L, Jiang T, Li C. Ferroptosis plays an essential role in the antimalarial mechanism of low-dose dihydroartemisinin. Biomed Pharmacother 2022; 148:112742. [PMID: 35228063 DOI: 10.1016/j.biopha.2022.112742] [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: 12/17/2021] [Revised: 02/15/2022] [Accepted: 02/17/2022] [Indexed: 11/15/2022] Open
Abstract
The activation of artemisinin and its derivatives (ARTs) to generate ROS and other free radicals is mainly heme- or ferrous iron-dependent. ARTs induce ferroptosis in tumor cells, although the involvement of ferroptosis in malaria remains unclear. We found that three typical inducers of ferroptosis (erastin, RSL3 and sorafenib) could effectively mimic DHA inhibition on the growth of blood-stage parasites, which exhibited synergistic or nearly additive interactions in vitro with DHA, while the combination of DHA with ferroptosis inhibitors (deferoxamine, liproxstatin-1) had an obvious antagonistic effect. DHA, similar to ferroptosis inducers, can simultaneously induce the accumulation of ferroptosis-associated cellular labile iron and lipid peroxide. However, deferoxamine and liproxstatin-1 reduced the increase in ferrous iron and lipid peroxide caused by DHA. These results suggested that ferroptosis might be an effective way to induce cell death in parasites and could be a primary mechanism by which DHA kills parasites, with almost 50% contribution at low concentrations. These results provide a new strategy for antimalarial drug screening and clinical medication guidance.
Collapse
Affiliation(s)
- Shuo Li
- Research Center of Artemisinin, China Academy of Chinese Medical Sciences, Beijing 100700, China; Tang Center for Herbal Medicine Research, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Wenhui Xu
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Huajing Wang
- Research Center of Artemisinin, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Tian Tang
- Research Center of Artemisinin, China Academy of Chinese Medical Sciences, Beijing 100700, China; Tang Center for Herbal Medicine Research, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Ji Ma
- Research Center of Artemisinin, China Academy of Chinese Medical Sciences, Beijing 100700, China; Tang Center for Herbal Medicine Research, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Zhao Cui
- Research Center of Artemisinin, China Academy of Chinese Medical Sciences, Beijing 100700, China; Tang Center for Herbal Medicine Research, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Hang Shi
- Research Center of Artemisinin, China Academy of Chinese Medical Sciences, Beijing 100700, China; Tang Center for Herbal Medicine Research, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Ting Qin
- Research Center of Artemisinin, China Academy of Chinese Medical Sciences, Beijing 100700, China; Tang Center for Herbal Medicine Research, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Hongying Zhou
- Research Center of Artemisinin, China Academy of Chinese Medical Sciences, Beijing 100700, China; Tang Center for Herbal Medicine Research, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Lanfang Li
- Tang Center for Herbal Medicine Research, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Tingliang Jiang
- Research Center of Artemisinin, China Academy of Chinese Medical Sciences, Beijing 100700, China; Tang Center for Herbal Medicine Research, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Canghai Li
- Tang Center for Herbal Medicine Research, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| |
Collapse
|
8
|
Siddiqui FA, Liang X, Cui L. Plasmodium falciparum resistance to ACTs: Emergence, mechanisms, and outlook. Int J Parasitol Drugs Drug Resist 2021; 16:102-118. [PMID: 34090067 PMCID: PMC8188179 DOI: 10.1016/j.ijpddr.2021.05.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 04/06/2021] [Accepted: 05/21/2021] [Indexed: 01/18/2023]
Abstract
Emergence and spread of resistance in Plasmodium falciparum to the frontline treatment artemisinin-based combination therapies (ACTs) in the epicenter of multidrug resistance of Southeast Asia threaten global malaria control and elimination. Artemisinin (ART) resistance (or tolerance) is defined clinically as delayed parasite clearance after treatment with an ART drug. The resistance phenotype is restricted to the early ring stage and can be measured in vitro using a ring-stage survival assay. ART resistance is associated with mutations in the propeller domain of the Kelch family protein K13. As a pro-drug, ART is activated primarily by heme, which is mainly derived from hemoglobin digestion in the food vacuole. Activated ARTs can react promiscuously with a wide range of cellular targets, disrupting cellular protein homeostasis. Consistent with this mode of action for ARTs, the molecular mechanisms of K13-mediated ART resistance involve reduced hemoglobin uptake/digestion and increased cellular stress response. Mutations in other genes such as AP-2μ (adaptor protein-2 μ subunit), UBP-1 (ubiquitin-binding protein-1), and Falcipain 2a that interfere with hemoglobin uptake and digestion also increase resistance to ARTs. ART resistance has facilitated the development of resistance to the partner drugs, resulting in rapidly declining ACT efficacies. The molecular markers for resistance to the partner drugs are mostly associated with point mutations in the two food vacuole membrane transporters PfCRT and PfMDR1, and amplification of pfmdr1 and the two aspartic protease genes plasmepsin 2 and 3. It has been observed that mutations in these genes can have opposing effects on sensitivities to different partner drugs, which serve as the principle for designing triple ACTs and drug rotation. Although clinical ACT resistance is restricted to Southeast Asia, surveillance for drug resistance using in vivo clinical efficacy, in vitro assays, and molecular approaches is required to prevent or slow down the spread of resistant parasites.
Collapse
Affiliation(s)
- Faiza Amber Siddiqui
- Department of Internal Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Xiaoying Liang
- Department of Internal Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Liwang Cui
- Department of Internal Medicine, University of South Florida, Tampa, FL, 33612, USA.
| |
Collapse
|
9
|
Camarena V, Huff TC, Wang G. Epigenomic regulation by labile iron. Free Radic Biol Med 2021; 170:44-49. [PMID: 33493555 PMCID: PMC8217092 DOI: 10.1016/j.freeradbiomed.2021.01.026] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/17/2020] [Accepted: 01/11/2021] [Indexed: 12/21/2022]
Abstract
Iron is an essential micronutrient metal for cellular functions but can generate highly reactive oxygen species resulting in oxidative damage. For these reasons its uptake and metabolism is highly regulated. A small but dynamic fraction of ferrous iron inside the cell, termed intracellular labile iron, is redox-reactive and ready to participate multiples reactions of intracellular enzymes. Due to its nature its determination and precise quantification has been a roadblock. However, recent progress in the development of intracellular labile iron probes are allowing the reevaluation of our current understanding and unmasking new functions. The role of intracellular labile iron in regulating the epigenome was recently discovered. This chapter examine how intracellular labile iron can modulate histone and DNA demethylation and how its pool can mediate a signaling pathway from cAMP serving as a sensor of the metabolic needs of the cells.
Collapse
Affiliation(s)
- Vladimir Camarena
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Tyler C Huff
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Gaofeng Wang
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
10
|
Sharma P, Tóth V, Hyland EM, Law CJ. Characterization of the substrate binding site of an iron detoxifying membrane transporter from Plasmodium falciparum. Malar J 2021; 20:295. [PMID: 34193175 PMCID: PMC8247066 DOI: 10.1186/s12936-021-03827-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 06/17/2021] [Indexed: 11/10/2022] Open
Abstract
Background Plasmodium species are entirely dependent upon their host as a source of essential iron. Although it is an indispensable micronutrient, oxidation of excess ferrous iron to the ferric state in the cell cytoplasm can produce reactive oxygen species that are cytotoxic. The malaria parasite must therefore carefully regulate the processes involved in iron acquisition and storage. A 273 amino acid membrane transporter that is a member of the vacuolar iron transporter (VIT) family and an orthologue of the yeast Ca2+-sensitive cross complementer (CCC1) protein plays a major role in cytosolic iron detoxification of Plasmodium species and functions in transport of ferrous iron ions into the endoplasmic reticulum for storage. While this transporter, termed PfVIT, is not critical for viability of the parasite evidence from studies of mice infected with VIT-deficient Plasmodium suggests it could still provide an efficient target for chemoprophylactic treatment of malaria. Individual amino acid residues that constitute the Fe2+ binding site of the protein were identified to better understand the structural basis of substrate recognition and binding by PfVIT. Methods Using the crystal structure of a recently published plant VIT as a template, a high-quality homology model of PfVIT was constructed to identify the amino acid composition of the transporter’s substrate binding site and to act as a guide for subsequent mutagenesis studies. To test the effect of mutation of the substrate binding-site residues on PfVIT function a yeast complementation assay assessed the ability of overexpressed, recombinant wild type and mutant PfVIT to rescue an iron-sensitive deletion strain (ccc1∆) of Saccharomyces cerevisiae yeast from the toxic effects of a high concentration of extracellular iron. Results The combined in silico and mutagenesis approach identified a methionine residue located within the cytoplasmic metal binding domain of the transporter as essential for PfVIT function and provided insight into the structural basis for the Fe2+-selectivity of the protein. Conclusion The structural model of the metal binding site of PfVIT opens the door for rational design of therapeutics to interfere with iron homeostasis within the malaria parasite. Supplementary Information The online version contains supplementary material available at 10.1186/s12936-021-03827-7.
Collapse
Affiliation(s)
- Pragya Sharma
- School of Biological Sciences, Queen's University Belfast, 19 Chlorine Gardens, Belfast, BT9 5DL, UK
| | - Veronika Tóth
- School of Biological Sciences, Queen's University Belfast, 19 Chlorine Gardens, Belfast, BT9 5DL, UK
| | - Edel M Hyland
- School of Biological Sciences, Queen's University Belfast, 19 Chlorine Gardens, Belfast, BT9 5DL, UK
| | - Christopher J Law
- School of Biological Sciences, Queen's University Belfast, 19 Chlorine Gardens, Belfast, BT9 5DL, UK.
| |
Collapse
|
11
|
Dighal A, Mukhopadhyay D, Sengupta R, Moulik S, Mukherjee S, Roy S, Chaudhuri SJ, Das NK, Chatterjee M. Iron trafficking in patients with Indian Post kala-azar dermal leishmaniasis. PLoS Negl Trop Dis 2020; 14:e0007991. [PMID: 32023254 PMCID: PMC7001907 DOI: 10.1371/journal.pntd.0007991] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 12/12/2019] [Indexed: 12/19/2022] Open
Abstract
Background During infections involving intracellular pathogens, iron performs a double-edged function by providing the pathogen with nutrients, but also boosts the host’s antimicrobial arsenal. Although the role of iron has been described in visceral leishmaniasis, information regarding its status in the dermal sequel, Post Kala-azar Dermal Leishmaniasis (PKDL) remains limited. Accordingly, this study aimed to establish the status of iron within monocytes/macrophages of PKDL cases. Methodology/Principal findings The intramonocytic labile iron pool (LIP), status of CD163 (hemoglobin-haptoglobin scavenging receptor) and CD71 (transferrin receptor, Tfr) were evaluated within CD14+ monocytes by flow cytometry, and soluble CD163 by ELISA. At the lesional sites, Fe3+ status was evaluated by Prussian blue staining, parasite load by qPCR, while the mRNA expression of Tfr (TfR1/CD71), CD163, divalent metal transporter-1 (DMT-1), Lipocalin-2 (Lcn-2), Heme-oxygenase-1 (HO-1), Ferritin, Natural resistance-associated macrophage protein (NRAMP-1) and Ferroportin (Fpn-1) was evaluated by droplet digital PCR. Circulating monocytes demonstrated elevated levels of CD71, CD163 and soluble CD163, which corroborated with an enhanced lesional mRNA expression of TfR, CD163, DMT1 and Lcn-2. Additionally, the LIP was raised along with an elevated mRNA expression of ferritin and HO-1, as also iron exporters NRAMP-1 and Fpn-1. Conclusions/Significance In monocytes/macrophages of PKDL cases, enhancement of the iron influx gateways (TfR, CD163, DMT-1 and Lcn-2) possibly accounted for the enhanced LIP. However, enhancement of the iron exporters (NRAMP-1 and Fpn-1) defied the classical Ferritinlow/Ferroportinhigh phenotype of alternatively activated macrophages. The creation of such a pro-parasitic environment suggests incorporation of chemotherapeutic strategies wherein the availability of iron to the parasite can be restricted. Post kala-azar dermal leishmaniasis (PKDL), a dermal sequel of Visceral Leishmaniasis (VL) is caused by the digenetic protozoan parasite Leishmania donovani. The parasite infects humans and replicates intracellularly within macrophages, cells normally engaged in protecting the host from pathogens. Iron plays a crucial role in microbes and mammalian cells, being needed by the former for its growth and survival, while the latter uses it for activation of the immune system by facilitating generation of reactive oxygen species. Therefore, the availability of iron needs to be tightly regulated to ensure its accessibility for core biological functions, and yet prevent its utilization by intracellular pathogens. Here we investigated the status of intra-macrophage iron along with expression of its transporters in patients with PKDL. Our study suggests that within monocytes/macrophages there is an enhanced entry of iron via the upregulation of CD71 and CD163 that translates into an enhanced labile iron pool and Ferritin. However, the concomitant increase in expression of iron exporters NRAMP-1 and Fpn-1 suggested the host’s attempt to deny the pathogen access to iron. This Ferritinhigh/Ferroportinhigh phenotype was in contrast to the conventional Ferritinlow/Ferroportinhigh phenotype present in alternatively activated M2 macrophages. Taken together, the control of iron homeostasis is one of the contributors in the host-pathogen interplay as it influences the course of an infectious disease by favouring either the mammalian host or the invading pathogen.
Collapse
MESH Headings
- Adolescent
- Adult
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Antigens, Differentiation, Myelomonocytic/genetics
- Antigens, Differentiation, Myelomonocytic/metabolism
- Cation Transport Proteins/genetics
- Cation Transport Proteins/metabolism
- Female
- Humans
- India
- Iron/metabolism
- Leishmania donovani/drug effects
- Leishmania donovani/physiology
- Leishmaniasis, Cutaneous/metabolism
- Leishmaniasis, Cutaneous/parasitology
- Lipocalin-2/genetics
- Lipocalin-2/metabolism
- Macrophages/metabolism
- Male
- Monocytes/metabolism
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Receptors, Transferrin/genetics
- Receptors, Transferrin/metabolism
- Young Adult
Collapse
Affiliation(s)
- Aishwarya Dighal
- Dept. of Pharmacology, Institute of Postgraduate Medical Education and Research, Kolkata, India
| | - Debanjan Mukhopadhyay
- Dept. of Pharmacology, Institute of Postgraduate Medical Education and Research, Kolkata, India
| | - Ritika Sengupta
- Dept. of Pharmacology, Institute of Postgraduate Medical Education and Research, Kolkata, India
| | - Srija Moulik
- Dept. of Pharmacology, Institute of Postgraduate Medical Education and Research, Kolkata, India
| | - Shibabrata Mukherjee
- Dept. of Pharmacology, Institute of Postgraduate Medical Education and Research, Kolkata, India
| | - Susmita Roy
- Dept. of Pharmacology, Institute of Postgraduate Medical Education and Research, Kolkata, India
| | | | - Nilay K. Das
- Dept of Dermatology, Bankura Sammilani Medical College, Bankura, India
| | - Mitali Chatterjee
- Dept. of Pharmacology, Institute of Postgraduate Medical Education and Research, Kolkata, India
- * E-mail:
| |
Collapse
|
12
|
Lu F, He XL, Richard C, Cao J. A brief history of artemisinin: Modes of action and mechanisms of resistance. Chin J Nat Med 2020; 17:331-336. [PMID: 31171267 DOI: 10.1016/s1875-5364(19)30038-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Indexed: 12/18/2022]
Abstract
The cornerstone of antimalarial treatment, artemisinin, has reduced malaria associated morbidity and mortality worldwide. However, Plasmodium falciparum parasites with reduced sensitivity to artemisinin have emerged, and this threatens malaria control and elimination efforts. In this minireview, we describe the initial development of artemisinin as an antimalarial drug, its use both historically and currently, and our current understanding of its mode of action and the mechanisms by which malaria parasites achieve resistance.
Collapse
Affiliation(s)
- Feng Lu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225009, China; Key Laboratory of National Health Commission on Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi 214064, China; Jiangsu Key laboratory of integrated traditional Chinese and Western Medicine for prevention and treatment of Senile Diseases, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Xin-Long He
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225009, China
| | - Culleton Richard
- Malaria Unit, Institute of Tropical Medicine, Nagasaki University, Sakamoto, Nagasaki, Japan.
| | - Jun Cao
- Key Laboratory of National Health Commission on Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi 214064, China; Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Public Health Research Center, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
13
|
Inhibitory Mechanisms of DHA/CQ on pH and Iron Homeostasis of Erythrocytic Stage Growth of Plasmodium Falciparum. Molecules 2019; 24:molecules24101941. [PMID: 31137574 PMCID: PMC6571875 DOI: 10.3390/molecules24101941] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 05/11/2019] [Accepted: 05/13/2019] [Indexed: 12/19/2022] Open
Abstract
Malaria is an infectious disease caused by Plasmodium group. The mechanisms of antimalarial drugs DHA/CQ are still unclear today. The inhibitory effects (IC50) of single treatments with DHA/CQ or V-ATPase inhibitor Baf-A1 or combination treatments by DHA/CQ combined with Baf-A1 on the growth of Plasmodium falciparum strain 3D7 was investigated. Intracellular cytoplasmic pH and labile iron pool (LIP) were labeled by pH probe BCECF, AM and iron probe calcein, AM, the fluorescence of the probes was measured by FCM. The effects of low doses of DHA (0.2 nM, 0.4 nM, 0.8 nM) on gene expression of V-ATPases (vapE, vapA, vapG) located in the membrane of DV were tested by RT-qPCR. DHA combined with Baf-A1 showed a synergism effect (CI = 0.524) on the parasite growth in the concentration of IC50. Intracellular pH and irons were effected significantly by different doses of DHA/Baf-A1. Intracellular pH was decreased by CQ combined with Baf-A1 in the concentration of IC50. Intracellular LIP was increased by DHA combined with Baf-A1 in the concentration of 20 IC50. The expression of gene vapA was down-regulated by all low doses of DHA (0.2/0.4/0.8 nM) significantly (p < 0.001) and the expression of vapG/vapE were up-regulated by 0.8 nM DHA significantly (p < 0.001). Interacting with ferrous irons, affecting the DV membrane proton pumping and acidic pH or cytoplasmic irons homeostasis may be the antimalarial mechanism of DHA while CQ showed an effect on cytoplasmic pH of parasite in vitro. Lastly, this article provides us preliminary results and a new idea for antimalarial drugs combination and new potential antimalarial combination therapies.
Collapse
|
14
|
Muriuki JM, Mentzer AJ, Kimita W, Ndungu FM, Macharia AW, Webb EL, Lule SA, Morovat A, Hill AVS, Bejon P, Elliott AM, Williams TN, Atkinson SH. Iron Status and Associated Malaria Risk Among African Children. Clin Infect Dis 2019; 68:1807-1814. [PMID: 30219845 PMCID: PMC6522755 DOI: 10.1093/cid/ciy791] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 09/10/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND It remains unclear whether improving iron status increases malaria risk, and few studies have looked at the effect of host iron status on subsequent malaria infection. We therefore aimed to determine whether a child's iron status influences their subsequent risk of malaria infection in sub-Saharan Africa. METHODS We assayed iron and inflammatory biomarkers from community-based cohorts of 1309 Kenyan and 1374 Ugandan children aged 0-7 years and conducted prospective surveillance for episodes of malaria. Poisson regression models were fitted to determine the effect of iron status on the incidence rate ratio (IRR) of malaria using longitudinal data covering a period of 6 months. Models were adjusted for age, sex, parasitemia, inflammation, and study site. RESULTS At baseline, the prevalence of iron deficiency (ID) was 36.9% and 34.6% in Kenyan and Ugandan children, respectively. ID anemia (IDA) affected 23.6% of Kenyan and 17.6% of Ugandan children. Malaria risk was lower in children with ID (IRR, 0.7; 95% confidence interval [CI], 0.6, 0.8; P < .001) and IDA (IRR, 0.7; 95% CI, 0.6, 0.9; P = .006). Low transferrin saturation (<10%) was similarly associated with lower malaria risk (IRR, 0.8; 95% CI, 0.6, 0.9; P = .016). However, variation in hepcidin, soluble transferrin receptors (sTfR), and hemoglobin/anemia was not associated with altered malaria risk. CONCLUSIONS ID appears to protect against malaria infection in African children when defined using ferritin and transferrin saturation, but not when defined by hepcidin, sTfR, or hemoglobin. Additional research is required to determine causality. CLINICAL TRIALS REGISTRATION ISRCTN32849447.
Collapse
Affiliation(s)
| | - Alexander J Mentzer
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford
| | - Wandia Kimita
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | | | | | - Emily L Webb
- London School of Hygiene and Tropical Medicine, United Kingdom
| | - Swaib A Lule
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Alireza Morovat
- Department of Clinical Biochemistry, Oxford University Hospitals
| | - Adrian V S Hill
- Centre for Clinical Vaccinology and Tropical Medicine and the Jenner Institute Laboratories, University of Oxford
| | - Philip Bejon
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford
| | - Alison M Elliott
- London School of Hygiene and Tropical Medicine, United Kingdom
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Thomas N Williams
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Department of Medicine, Imperial College, London
| | - Sarah H Atkinson
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford
- Department of Paediatrics, University of Oxford, United Kingdom
| |
Collapse
|
15
|
Nairz M, Dichtl S, Schroll A, Haschka D, Tymoszuk P, Theurl I, Weiss G. Iron and innate antimicrobial immunity-Depriving the pathogen, defending the host. J Trace Elem Med Biol 2018; 48:118-133. [PMID: 29773170 DOI: 10.1016/j.jtemb.2018.03.007] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 02/25/2018] [Accepted: 03/06/2018] [Indexed: 02/08/2023]
Abstract
The acute-phase response is triggered by the presence of infectious agents and danger signals which indicate hazards for the integrity of the mammalian body. One central feature of this response is the sequestration of iron into storage compartments including macrophages. This limits the availability of this essential nutrient for circulating pathogens, a host defence strategy known as 'nutritional immunity'. Iron metabolism and the immune response are intimately linked. In infections, the availability of iron affects both the efficacy of antimicrobial immune pathways and pathogen proliferation. However, host strategies to withhold iron from microbes vary according to the localization of pathogens: Infections with extracellular bacteria such as Staphylococcus aureus, Streptococcus, Klebsiella or Yersinia stimulate the expression of the iron-regulatory hormone hepcidin which targets the cellular iron-exporter ferroportin-1 causing its internalization and blockade of iron egress from absorptive enterocytes in the duodenum and iron-recycling macrophages. This mechanism disrupts both routes of iron delivery to the circulation, contributes to iron sequestration in the mononuclear phagocyte system and mediates the hypoferraemia of the acute phase response subsequently resulting in the development of anaemia of inflammation. When intracellular microbes are present, other strategies of microbial iron withdrawal are needed. For instance, in macrophages harbouring intracellular pathogens such as Chlamydia, Mycobacterium tuberculosis, Listeria monocytogenes or Salmonella Typhimurium, ferroportin-1-mediated iron export is turned on for the removal of iron from infected cells. This also leads to reduced iron availability for intra-macrophage pathogens which inhibits their growth and in parallel strengthens anti-microbial effector pathways of macrophages including the formation of inducible nitric oxide synthase and tumour necrosis factor. Iron plays a key role in infectious diseases both as modulator of the innate immune response and as nutrient for microbes. We need to gain a more comprehensive understanding of how the body can differentially respond to infection by extra- or intracellular pathogens. This knowledge may allow us to modulate mammalian iron homeostasis pharmaceutically and to target iron-acquisition systems of pathogens, thus enabling us to treat infections with novel strategies that act independent of established antimicrobials.
Collapse
Affiliation(s)
- Manfred Nairz
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Austria.
| | - Stefanie Dichtl
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Austria
| | - Andrea Schroll
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Austria
| | - David Haschka
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Austria
| | - Piotr Tymoszuk
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Austria
| | - Igor Theurl
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Austria
| | - Günter Weiss
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Austria
| |
Collapse
|
16
|
Parasite-Mediated Degradation of Synthetic Ozonide Antimalarials Impacts In Vitro Antimalarial Activity. Antimicrob Agents Chemother 2018; 62:AAC.01566-17. [PMID: 29263074 DOI: 10.1128/aac.01566-17] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 12/16/2017] [Indexed: 01/05/2023] Open
Abstract
The peroxide bond of the artemisinins inspired the development of a class of fully synthetic 1,2,4-trioxolane-based antimalarials, collectively known as the ozonides. Similar to the artemisinins, heme-mediated degradation of the ozonides generates highly reactive radical species that are thought to mediate parasite killing by damaging critical parasite biomolecules. We examined the relationship between parasite dependent degradation and antimalarial activity for two ozonides, OZ277 (arterolane) and OZ439 (artefenomel), using a combination of in vitro drug stability and pulsed-exposure activity assays. Our results showed that drug degradation is parasite stage dependent and positively correlates with parasite load. Increasing trophozoite-stage parasitemia leads to substantially higher rates of degradation for both OZ277 and OZ439, and this is associated with a reduction in in vitro antimalarial activity. Under conditions of very high parasitemia (∼90%), OZ277 and OZ439 were rapidly degraded and completely devoid of activity in trophozoite-stage parasite cultures exposed to a 3-h drug pulse. This study highlights the impact of increasing parasite load on ozonide stability and in vitro antimalarial activity and should be considered when investigating the antimalarial mode of action of the ozonide antimalarials under conditions of high parasitemia.
Collapse
|
17
|
Barffour MA, Schulze KJ, Coles CL, Chileshe J, Kalungwana N, Arguello M, Siamusantu W, Moss WJ, West KP, Palmer AC. High Iron Stores in the Low Malaria Season Increase Malaria Risk in the High Transmission Season in a Prospective Cohort of Rural Zambian Children. J Nutr 2017; 147:1531-1536. [PMID: 28701387 DOI: 10.3945/jn.117.250381] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 04/28/2017] [Accepted: 06/12/2017] [Indexed: 11/14/2022] Open
Abstract
Background: Higher iron stores, defined by serum ferritin (SF) concentration, may increase malaria risk.Objective: We evaluated the association between SF assessed during low malaria season and the risk of malaria during high malaria season, controlling for inflammation.Methods: Data for this prospective study were collected from children aged 4-8 y (n = 745) participating in a biofortified maize efficacy trial in rural Zambia. All malaria cases were treated at baseline (September 2012). We used baseline SF and malaria status indicated by positive microscopy at endline (March 2013) to define exposure and outcome, respectively. Iron status was defined as deficient (corrected or uncorrected SF <12 or <15 μg/L, depending on age <5 or ≥5 y, respectively), moderate (<75 μg/L, excluding deficient), or high (≥75 μg/L). We used a modified Poisson regression to model the risk of malaria in the high transmission seasons (endline) as a function of iron status assessed in the low malaria seasons (baseline).Results: We observed an age-dependent, positive dose-response association between ferritin in the low malaria season and malaria incidence during the high malaria season in younger children. In children aged <6 y (but not older children), we observed a relative increase in malaria risk in the moderate iron status [incidence rate ratio (IRR) with SF: 1.56; 95% CI: 0.64, 3.86; IRR with inflammation-corrected SF: 1.92; 95% CI: 0.75, 4.93] and high iron status (IRR with SF: 2.66; 95% CI: 1.10, 6.43; or IRR with corrected SF: 2.93; 95% CI: 1.17, 7.33) categories compared with the deficient iron status category. The relative increase in malaria risk for children with high iron status was statistically significant only among those with a concurrently normal serum soluble transferrin receptor concentration (<8.3 mg/L; IRR: 1.97; 95% CI: 1.20, 7.37).Conclusions: Iron adequacy in 4- to 8-y-old children in rural Zambia was associated with increased malaria risk. Our findings underscore the need to integrate iron interventions with malaria control programs. This trial was registered at clinicaltrials.gov as NCT01695148.
Collapse
Affiliation(s)
- Maxwell A Barffour
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Kerry J Schulze
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Christian L Coles
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | | | | | - Margia Arguello
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | | | - William J Moss
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Keith P West
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Amanda C Palmer
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD;
| |
Collapse
|
18
|
Mwangi MN, Prentice AM, Verhoef H. Safety and benefits of antenatal oral iron supplementation in low-income countries: a review. Br J Haematol 2017; 177:884-895. [PMID: 28272734 PMCID: PMC5485170 DOI: 10.1111/bjh.14584] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The World Health Organization recommends universal iron supplementation of 30-60 mg/day in pregnancy but coverage is low in most countries. Its efficacy is uncertain, however, and there has been a vigorous debate in the last decade about its safety, particularly in areas with a high burden of malaria and other infectious diseases. We reviewed the evidence on the safety and efficacy of antenatal iron supplementation in low-income countries. We found no evidence that daily supplementation at a dose of 60 mg leads to increased maternal Plasmodium infection risk. On the other hand, recent meta-analyses found that antenatal iron supplementation provides benefits for maternal health (severe anaemia at postpartum, blood transfusion). For neonates, there was a reduced prematurity risk, and only a small or no effect on birth weight. A recent trial showed, however, that benefits of antenatal iron supplementation on maternal and neonatal health vary by maternal iron status, with substantial benefits in iron-deficient women. The benefits of universal iron supplementation are likely to vary with the prevalence of iron deficiency. As a consequence, the balance between benefits and risks is probably more favourable in low-income countries than in high-income countries despite the higher exposure to infectious pathogens.
Collapse
Affiliation(s)
- Martin N Mwangi
- Division of Human Nutrition, Wageningen University, Wageningen, The Netherlands
- Nutrition and Health Department, School of Public Health and Community Development, Maseno University, Maseno, Kenya
| | - Andrew M Prentice
- MRC Unit The Gambia, Banjul, The Gambia
- MRC International Nutrition Group, London School of Hygiene and Tropical Medicine, London, UK
| | - Hans Verhoef
- Division of Human Nutrition, Wageningen University, Wageningen, The Netherlands
- MRC International Nutrition Group, London School of Hygiene and Tropical Medicine, London, UK
- Cell Biology and Immunology Group, Wageningen University, Wageningen, The Netherlands
| |
Collapse
|
19
|
Sun J, Li C, Wang S. Organism-like formation of Schistosoma hemozoin and its function suggest a mechanism for anti-malarial action of artemisinin. Sci Rep 2016; 6:34463. [PMID: 27694940 PMCID: PMC5046088 DOI: 10.1038/srep34463] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 09/14/2016] [Indexed: 01/07/2023] Open
Abstract
The current theories of antimalarial mechanism of artemisinin are inadequate to fully explain the observed effects. In our study, “organism-like” formation of Schistosoma hemozoin granules by attaching to and utilizing erythrocytes to form new ones was observed. This indicates that heme iron is transferred from erythrocytes to hemozoin granules during their formation. However, as a disposal product of heme detoxification, these granules are not completely expelled from the Schistosoma gut, but decomposed again between microvilli in the posterior portion of the gut to transfer iron to eggs. Based on the function of iron transport supported by our observation of the unique process of Schistosoma hemozoin formation, here we propose a new viewpoint of antimalarial mechanism of artemisinin, which emphasizes the final outcome, i.e., interference of iron utilization in parasites by artemisinin, instead of focusing on the mode of interaction between artemisinin and heme or hemozoin. This suggests that artemisinin and its endoperoxides derivatives likely hit the Achilles’ heel of hemozoin-producing and iron-dependent organisms.
Collapse
Affiliation(s)
- Jun Sun
- Institute for Infectious Diseases and Vaccine Development, Tongji University School of Medicine, 1239 Siping Road, Shanghai 200092, P.R. China
| | - Chen Li
- Institute for Infectious Diseases and Vaccine Development, Tongji University School of Medicine, 1239 Siping Road, Shanghai 200092, P.R. China
| | - Suwen Wang
- Institute for Infectious Diseases and Vaccine Development, Tongji University School of Medicine, 1239 Siping Road, Shanghai 200092, P.R. China
| |
Collapse
|
20
|
Tilley L, Straimer J, Gnädig NF, Ralph SA, Fidock DA. Artemisinin Action and Resistance in Plasmodium falciparum. Trends Parasitol 2016; 32:682-696. [PMID: 27289273 DOI: 10.1016/j.pt.2016.05.010] [Citation(s) in RCA: 222] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 05/10/2016] [Accepted: 05/13/2016] [Indexed: 12/16/2022]
Abstract
The worldwide use of artemisinin-based combination therapies (ACTs) has contributed in recent years to a substantial reduction in deaths resulting from Plasmodium falciparum malaria. Resistance to artemisinins, however, has emerged in Southeast Asia. Clinically, resistance is defined as a slower rate of parasite clearance in patients treated with an artemisinin derivative or an ACT. These slow clearance rates associate with enhanced survival rates of ring-stage parasites briefly exposed in vitro to dihydroartemisinin. We describe recent progress made in defining the molecular basis of artemisinin resistance, which has identified a primary role for the P. falciparum K13 protein. Using K13 mutations as molecular markers, epidemiological studies are now tracking the emergence and spread of artemisinin resistance. Mechanistic studies suggest potential ways to overcome resistance.
Collapse
Affiliation(s)
- Leann Tilley
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Melbourne, Australia.
| | - Judith Straimer
- Department of Microbiology and Immunology, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Nina F Gnädig
- Department of Microbiology and Immunology, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Stuart A Ralph
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Melbourne, Australia
| | - David A Fidock
- Department of Microbiology and Immunology, Columbia University College of Physicians and Surgeons, New York, NY, USA; Division of Infectious Diseases, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, USA.
| |
Collapse
|
21
|
Slavic K, Krishna S, Lahree A, Bouyer G, Hanson KK, Vera I, Pittman JK, Staines HM, Mota MM. A vacuolar iron-transporter homologue acts as a detoxifier in Plasmodium. Nat Commun 2016; 7:10403. [PMID: 26786069 PMCID: PMC4735874 DOI: 10.1038/ncomms10403] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2015] [Accepted: 12/07/2015] [Indexed: 01/03/2023] Open
Abstract
Iron is an essential micronutrient but is also highly toxic. In yeast and plant cells, a key detoxifying mechanism involves iron sequestration into intracellular storage compartments, mediated by members of the vacuolar iron-transporter (VIT) family of proteins. Here we study the VIT homologue from the malaria parasites Plasmodium falciparum (PfVIT) and Plasmodium berghei (PbVIT). PfVIT-mediated iron transport in a yeast heterologous expression system is saturable (Km ∼ 14.7 μM), and selective for Fe(2+) over other divalent cations. PbVIT-deficient P. berghei lines (Pbvit(-)) show a reduction in parasite load in both liver and blood stages of infection in mice. Moreover, Pbvit(-) parasites have higher levels of labile iron in blood stages and are more sensitive to increased iron levels in liver stages, when compared with wild-type parasites. Our data are consistent with Plasmodium VITs playing a major role in iron detoxification and, thus, normal development of malaria parasites in their mammalian host.
Collapse
Affiliation(s)
- Ksenija Slavic
- Instituto de Medicina Molecular, Faculdade de Medicina Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Sanjeev Krishna
- Institute for Infection & Immunity, St. George's, University of London, London SW17 0RE, UK
| | - Aparajita Lahree
- Instituto de Medicina Molecular, Faculdade de Medicina Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Guillaume Bouyer
- Institute for Infection & Immunity, St. George's, University of London, London SW17 0RE, UK
- Sorbonne Universités, UPMC Univ Paris 6, CNRS, UMR 8227, Comparative Physiology of Erythrocytes, Station Biologique de Roscoff, CS 90074, 29688 Roscoff, France
| | - Kirsten K. Hanson
- Instituto de Medicina Molecular, Faculdade de Medicina Universidade de Lisboa, 1649-028 Lisbon, Portugal
- Present address: University of Texas at San Antonio, Department of Biology and STCEID, San Antonio, Texas 78249, USA
| | - Iset Vera
- Instituto de Medicina Molecular, Faculdade de Medicina Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Jon K. Pittman
- Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | - Henry M. Staines
- Institute for Infection & Immunity, St. George's, University of London, London SW17 0RE, UK
| | - Maria M. Mota
- Instituto de Medicina Molecular, Faculdade de Medicina Universidade de Lisboa, 1649-028 Lisbon, Portugal
| |
Collapse
|
22
|
Xie SC, Dogovski C, Hanssen E, Chiu F, Yang T, Crespo MP, Stafford C, Batinovic S, Teguh S, Charman S, Klonis N, Tilley L. Haemoglobin degradation underpins the sensitivity of early ring stage Plasmodium falciparum to artemisinins. J Cell Sci 2015; 129:406-16. [PMID: 26675237 PMCID: PMC4732288 DOI: 10.1242/jcs.178830] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 12/02/2015] [Indexed: 11/30/2022] Open
Abstract
Current first-line artemisinin antimalarials are threatened by the emergence of resistant Plasmodium falciparum. Decreased sensitivity is evident in the initial (early ring) stage of intraerythrocytic development, meaning that it is crucial to understand the action of artemisinins at this stage. Here, we examined the roles of iron (Fe) ions and haem in artemisinin activation in early rings using Fe ion chelators and a specific haemoglobinase inhibitor (E64d). Quantitative modelling of the antagonism accounted for its complex dependence on the chemical features of the artemisinins and on the drug exposure time, and showed that almost all artemisinin activity in early rings (>80%) is due to haem-mediated activation. The surprising implication that haemoglobin uptake and digestion is active in early rings is supported by identification of active haemoglobinases (falcipains) at this stage. Genetic down-modulation of the expression of the two main cysteine protease haemoglobinases, falcipains 2 and 3, renders early ring stage parasites resistant to artemisinins. This confirms the important role of haemoglobin-degrading falcipains in artemisinin activation, and shows that changes in the rate of artemisinin activation could mediate high-level artemisinin resistance. Summary: Down-modulation of the expression of haemoglobin-degrading falcipains in P. falciparum renders early ring stage parasites resistant to artemisinins.
Collapse
Affiliation(s)
- Stanley C Xie
- Department of Biochemistry and Molecular Biology, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Con Dogovski
- Department of Biochemistry and Molecular Biology, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Eric Hanssen
- Department of Biochemistry and Molecular Biology, The University of Melbourne, Melbourne, Victoria 3010, Australia Advanced Microscopy Facility, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Francis Chiu
- Monash Institute of Pharmaceutical Sciences, Faculty of Pharmacy and Pharmaceutical Sciences, Melbourne, Victoria 3010, Australia
| | - Tuo Yang
- Department of Biochemistry and Molecular Biology, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Maria P Crespo
- Department of Microbiology, University of Valle, 13 #100-00, Cali, Valle del Cauca, Colombia Department of Biomedical Sciences, Santiago de Cali University, 25, Cali, Valle del Cauca, Colombia
| | - Che Stafford
- Walter+Eliza Hall Institute, Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Steven Batinovic
- Department of Biochemistry and Molecular Biology, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Silvia Teguh
- Department of Biochemistry and Molecular Biology, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Susan Charman
- Monash Institute of Pharmaceutical Sciences, Faculty of Pharmacy and Pharmaceutical Sciences, Melbourne, Victoria 3010, Australia
| | - Nectarios Klonis
- Department of Biochemistry and Molecular Biology, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Leann Tilley
- Department of Biochemistry and Molecular Biology, The University of Melbourne, Melbourne, Victoria 3010, Australia
| |
Collapse
|
23
|
Thipubon P, Uthaipibull C, Kamchonwongpaisan S, Tipsuwan W, Srichairatanakool S. Inhibitory effect of novel iron chelator, 1-(N-acetyl-6-aminohexyl)-3-hydroxy-2-methylpyridin-4-one (CM1) and green tea extract on growth of Plasmodium falciparum. Malar J 2015; 14:382. [PMID: 26424148 PMCID: PMC4590262 DOI: 10.1186/s12936-015-0910-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 09/18/2015] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Iron is an essential micronutrient required by all living organisms including malaria parasites (Plasmodium spp.) for many biochemical reactions, especially growth and multiplication processes. Therefore, malaria parasite needs to take up the iron from outside or/and inside the parasitized red blood cells (PRBC). Iron chelators are widely used for the treatment of thalassaemia-related iron overload and also inhibit parasite growth at levels that are non-toxic to mammalian cells. METHODS Inhibitory effect of 1-(N-acetyl-6-aminohexyl)-3-hydroxy-2-methylpyridin-4-one (CM1) and green tea extract (GTE) on the growth of malaria parasite Plasmodium falciparum was compared with standard chelators including desferrioxamine (DFO), deferiprone (DFP) and deferasirox (DFX). A flow cytometric technique was used to enumerate PRBC stained with SYBR Green I fluorescent dye. The labile iron pool (LIP) was assayed using the calcein-acetoxymethyl fluorescent method. RESULTS The IC50 values of DFO, GTE, CM1, DFX and DFP against P. falciparum were 14.09, 21.11, 35.14, 44.71 and 58.25 µM, respectively. Importantly, CM1 was more effective in reducing LIP levels in the P. falciparum culture than DFP (p < 0.05). CONCLUSIONS CM1 and GTE exhibit anti-malarial activity. They could interfere with uptake of exogenous iron or deplete the intracellular labile iron pool in malaria parasites, leading to inhibition of their growth.
Collapse
Affiliation(s)
- Phitsinee Thipubon
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, 110 Inthawaroros Street, Tambol Sriphum, Amphur Muang, Chiang Mai, 50200, Thailand.
| | - Chairat Uthaipibull
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathum Thani, 12102, Thailand.
| | - Sumalee Kamchonwongpaisan
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathum Thani, 12102, Thailand.
| | - Wachiraporn Tipsuwan
- Division of Biochemistry, School of Medical Science, University of Phayao, Phayao, 56000, Thailand.
| | - Somdet Srichairatanakool
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, 110 Inthawaroros Street, Tambol Sriphum, Amphur Muang, Chiang Mai, 50200, Thailand.
| |
Collapse
|
24
|
Erythrocytic Iron Deficiency Enhances Susceptibility to Plasmodium chabaudi Infection in Mice Carrying a Missense Mutation in Transferrin Receptor 1. Infect Immun 2015; 83:4322-34. [PMID: 26303393 DOI: 10.1128/iai.00926-15] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 08/20/2015] [Indexed: 01/26/2023] Open
Abstract
The treatment of iron deficiency in areas of high malaria transmission is complicated by evidence which suggests that iron deficiency anemia protects against malaria, while iron supplementation increases malaria risk. Iron deficiency anemia results in an array of pathologies, including reduced systemic iron bioavailability and abnormal erythrocyte physiology; however, the mechanisms by which these pathologies influence malaria infection are not well defined. In the present study, the response to malaria infection was examined in a mutant mouse line, Tfrc(MRI24910), identified during an N-ethyl-N-nitrosourea (ENU) screen. This line carries a missense mutation in the gene for transferrin receptor 1 (TFR1). Heterozygous mice exhibited reduced erythrocyte volume and density, a phenotype consistent with dietary iron deficiency anemia. However, unlike the case in dietary deficiency, the erythrocyte half-life, mean corpuscular hemoglobin concentration, and intraerythrocytic ferritin content were unchanged. Systemic iron bioavailability was also unchanged, indicating that this mutation results in erythrocytic iron deficiency without significantly altering overall iron homeostasis. When infected with the rodent malaria parasite Plasmodium chabaudi adami, mice displayed increased parasitemia and succumbed to infection more quickly than their wild-type littermates. Transfusion of fluorescently labeled erythrocytes into malaria parasite-infected mice demonstrated an erythrocyte-autonomous enhanced survival of parasites within mutant erythrocytes. Together, these results indicate that TFR1 deficiency alters erythrocyte physiology in a way that is similar to dietary iron deficiency anemia, albeit to a lesser degree, and that this promotes intraerythrocytic parasite survival and an increased susceptibility to malaria in mice. These findings may have implications for the management of iron deficiency in the context of malaria.
Collapse
|
25
|
Ma Y, Abbate V, Hider RC. Iron-sensitive fluorescent probes: monitoring intracellular iron pools. Metallomics 2015; 7:212-22. [DOI: 10.1039/c4mt00214h] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
26
|
Antimalarial activity of granzyme B and its targeted delivery by a granzyme B-single-chain Fv fusion protein. Antimicrob Agents Chemother 2014; 59:669-72. [PMID: 25313223 DOI: 10.1128/aac.04190-14] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
We present here the first evidence that granzyme B acts against Plasmodium falciparum (50% inhibitory concentration [IC50], 1,590 nM; 95% confidence interval [95% CI], 1,197 to 2,112 nM). We created a novel antimalarial fusion protein consisting of granzyme B fused to a merozoite surface protein 4 (MSP4)-specific single-chain Fv protein (scFv), which targets the enzyme to infected erythrocytes, with up to an 8-fold reduction in the IC50 (176 nM; 95% CI, 154 to 202 nM). This study confirms the therapeutic efficacies of recombinant antibody-mediated antimalarial immunotherapeutics based on granzyme B.
Collapse
|
27
|
Brittenham GM, Andersson M, Egli I, Foman JT, Zeder C, Westerman ME, Hurrell RF. Circulating non-transferrin-bound iron after oral administration of supplemental and fortification doses of iron to healthy women: a randomized study. Am J Clin Nutr 2014; 100:813-20. [PMID: 25057155 PMCID: PMC4135492 DOI: 10.3945/ajcn.113.081505] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND After the oral administration of iron, the production of circulating non-transferrin-bound iron may contribute to an increased risk of illness in malaria-endemic areas that lack effective medical services. OBJECTIVE In healthy women with a range of body iron stores, we aimed to determine effects on the production of circulating non-transferrin-bound iron resulting from the oral administration of 1) a supplemental dose of iron (60 mg) with water, 2) a supplemental dose of iron (60 mg) with a standard test meal, and 3) a fortification dose of iron (6 mg) with a standard test meal. DESIGN With the use of serum ferritin as the indicator, healthy women with replete iron stores (ferritin concentration >25 μg/L; n = 16) and reduced iron stores (ferritin concentration ≤25 μg/L; n = 16) were enrolled in a prospective, randomized, crossover study. After the oral administration of aqueous solutions of ferrous sulfate isotopically labeled with ⁵⁴Fe, ⁵⁷Fe, or ⁵⁸Fe, blood samples were collected for 8 h, and iron absorption was estimated by erythrocyte incorporation at 14 d. RESULTS At 4 h, serum non-transferrin-bound iron reached peaks with geometric mean (95% CI) concentrations of 0.81 μmol/L (0.56, 1.1 μmol/L) for 60 mg Fe with water and 0.26 μmol/L (0.15, 0.38 μmol/L) for 60 mg Fe with food but was at assay limits of detection (0.1 μmol Fe/L) for 6 mg Fe with food. For the 60 mg Fe without food, the area under the curve over 8 h for serum non-transferrin-bound iron was positively correlated with the amount of iron absorbed (R = 0.49, P < 0.01) and negatively correlated with serum ferritin (R = -0.39, P < 0.05). CONCLUSIONS In healthy women, the production of circulating non-transferrin-bound iron is determined by the rate and amount of iron absorbed. The highest concentrations of non-transferrin-bound iron resulted from the administration of supplemental doses of iron without food. Little or no circulating non-transferrin-bound iron resulted from the consumption of a meal with a fortification dose of iron.
Collapse
Affiliation(s)
- Gary M Brittenham
- From the Human Nutrition Laboratory, Institute of Food, Nutrition and Health, ETH Zürich, Zürich, Switzerland (IE, JTF, CZ, MA, and RFH); the Intrinsic LifeSciences LLC, La Jolla, CA (MEW); and the Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY (GMB)
| | - Maria Andersson
- From the Human Nutrition Laboratory, Institute of Food, Nutrition and Health, ETH Zürich, Zürich, Switzerland (IE, JTF, CZ, MA, and RFH); the Intrinsic LifeSciences LLC, La Jolla, CA (MEW); and the Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY (GMB)
| | - Ines Egli
- From the Human Nutrition Laboratory, Institute of Food, Nutrition and Health, ETH Zürich, Zürich, Switzerland (IE, JTF, CZ, MA, and RFH); the Intrinsic LifeSciences LLC, La Jolla, CA (MEW); and the Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY (GMB)
| | - Jasmin Tajeri Foman
- From the Human Nutrition Laboratory, Institute of Food, Nutrition and Health, ETH Zürich, Zürich, Switzerland (IE, JTF, CZ, MA, and RFH); the Intrinsic LifeSciences LLC, La Jolla, CA (MEW); and the Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY (GMB)
| | - Christophe Zeder
- From the Human Nutrition Laboratory, Institute of Food, Nutrition and Health, ETH Zürich, Zürich, Switzerland (IE, JTF, CZ, MA, and RFH); the Intrinsic LifeSciences LLC, La Jolla, CA (MEW); and the Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY (GMB)
| | - Mark E Westerman
- From the Human Nutrition Laboratory, Institute of Food, Nutrition and Health, ETH Zürich, Zürich, Switzerland (IE, JTF, CZ, MA, and RFH); the Intrinsic LifeSciences LLC, La Jolla, CA (MEW); and the Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY (GMB)
| | - Richard F Hurrell
- From the Human Nutrition Laboratory, Institute of Food, Nutrition and Health, ETH Zürich, Zürich, Switzerland (IE, JTF, CZ, MA, and RFH); the Intrinsic LifeSciences LLC, La Jolla, CA (MEW); and the Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY (GMB)
| |
Collapse
|
28
|
Host iron status and iron supplementation mediate susceptibility to erythrocytic stage Plasmodium falciparum. Nat Commun 2014; 5:4446. [PMID: 25059846 DOI: 10.1038/ncomms5446] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 06/18/2014] [Indexed: 11/09/2022] Open
Abstract
Iron deficiency and malaria have similar global distributions, and frequently co-exist in pregnant women and young children. Where both conditions are prevalent, iron supplementation is complicated by observations that iron deficiency anaemia protects against falciparum malaria, and that iron supplements increase susceptibility to clinically significant malaria, but the mechanisms remain obscure. Here, using an in vitro parasite culture system with erythrocytes from iron-deficient and replete human donors, we demonstrate that Plasmodium falciparum infects iron-deficient erythrocytes less efficiently. In addition, owing to merozoite preference for young erythrocytes, iron supplementation of iron-deficient individuals reverses the protective effects of iron deficiency. Our results provide experimental validation of field observations reporting protective effects of iron deficiency and harmful effects of iron administration on human malaria susceptibility. Because recovery from anaemia requires transient reticulocytosis, our findings imply that in malarious regions iron supplementation should be accompanied by effective measures to prevent falciparum malaria.
Collapse
|
29
|
Clark MA, Goheen MM, Spidale NA, Kasthuri RS, Fulford A, Cerami C. RBC barcoding allows for the study of erythrocyte population dynamics and P. falciparum merozoite invasion. PLoS One 2014; 9:e101041. [PMID: 24984000 PMCID: PMC4077748 DOI: 10.1371/journal.pone.0101041] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 06/02/2014] [Indexed: 11/30/2022] Open
Abstract
Plasmodium falciparum invasion of host erythrocytes is essential for the propagation of the blood stage of malaria infection. Additionally, the brief extracellular merozoite stage of P. falciparum represents one of the rare windows during which the parasite is directly exposed to the host immune response. Therefore, efficient invasion of the host erythrocyte is necessary not only for productive host erythrocyte infection, but also for evasion of the immune response. Host traits, such as hemoglobinopathies and differential expression of erythrocyte invasion ligands, can protect individuals from malaria by impeding parasite erythrocyte invasion. Here we combine RBC barcoding with flow cytometry to study P. falciparum invasion. This novel high-throughput method allows for the (i) direct comparison of P. falciparum invasion into different erythrocyte populations and (ii) assessment of the impact of changing erythrocyte population dynamics on P. falciparum invasion.
Collapse
Affiliation(s)
- Martha A Clark
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Morgan M Goheen
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Nicholas A Spidale
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Raj S Kasthuri
- Division of Hematology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Anthony Fulford
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Carla Cerami
- Medical Research Council International Nutrition Group, London School of Hygiene and Tropical Medicine, London, United Kingdom & Medical Research Council Keneba, The Gambia
| |
Collapse
|
30
|
Clark MA, Goheen MM, Cerami C. Influence of host iron status on Plasmodium falciparum infection. Front Pharmacol 2014; 5:84. [PMID: 24834053 PMCID: PMC4018558 DOI: 10.3389/fphar.2014.00084] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 04/04/2014] [Indexed: 01/25/2023] Open
Abstract
Iron deficiency affects one quarter of the world's population and causes significant morbidity, including detrimental effects on immune function and cognitive development. Accordingly, the World Health Organization (WHO) recommends routine iron supplementation in children and adults in areas with a high prevalence of iron deficiency. However, a large body of clinical and epidemiological evidence has accumulated which clearly demonstrates that host iron deficiency is protective against falciparum malaria and that host iron supplementation may increase the risk of malaria. Although many effective antimalarial treatments and preventive measures are available, malaria remains a significant public health problem, in part because the mechanisms of malaria pathogenesis remain obscured by the complexity of the relationships that exist between parasite virulence factors, host susceptibility traits, and the immune responses that modulate disease. Here we review (i) the clinical and epidemiological data that describes the relationship between host iron status and malaria infection and (ii) the current understanding of the biological basis for these clinical and epidemiological observations.
Collapse
Affiliation(s)
- Martha A. Clark
- Microbiology and Immunology, University of North CarolinaChapel Hill, NC, USA
| | - Morgan M. Goheen
- Microbiology and Immunology, University of North CarolinaChapel Hill, NC, USA
| | - Carla Cerami
- Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel HillChapel Hill, NC, USA
| |
Collapse
|
31
|
Sangaré L, van Eijk AM, ter Kuile FO, Walson J, Stergachis A. The association between malaria and iron status or supplementation in pregnancy: a systematic review and meta-analysis. PLoS One 2014; 9:e87743. [PMID: 24551064 PMCID: PMC3925104 DOI: 10.1371/journal.pone.0087743] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2013] [Accepted: 12/27/2013] [Indexed: 12/20/2022] Open
Abstract
Introduction Malaria prevention and iron supplementation are associated with improved maternal and infant outcomes. However, evidence from studies in children suggests iron may adversely modify the risk of malaria. We reviewed the evidence in pregnancy of the association between malaria and markers of iron status, iron supplementation or parenteral treatment. Methods and Findings We searched MEDLINE, EMBASE, the Cochrane Central Register of Controlled Trials, the Global Health Library, and the Malaria in Pregnancy library to identify studies that investigated the association between iron status, iron treatment or supplementation during pregnancy and malaria. Thirty one studies contributed to the analysis; 3 experimental and 28 observational studies. Iron supplementation was not associated with an increased risk of P. falciparum malaria during pregnancy or delivery in Africa (summary Relative Risk = 0.89, 95% Confidence Interval (CI) 0.66–1.20, I2 = 78.8%, 5 studies). One study in Asia reported an increased risk of P. vivax within 30 days of iron supplementation (e.g. adjusted Hazard Ratio = 1.75, 95% CI 1.14–2.70 for 1–15 days), but not after 60 days. Iron deficiency (based on ferritin and C-reactive protein) was associated with lower odds for malaria infection (summary Odds Ratio = 0.35, 0.24–0.51, I2 = 59.2%, 5 studies). With the exception of the acute phase protein ferritin, biomarkers of iron deficiency were generally not associated with malaria infection. Conclusions Iron supplementation was associated with a temporal increase in P vivax, but not with an increased risk of P. falciparum; however, data are insufficient to rule out the potential for an increased risk of P. falciparum. Iron deficiency was associated with a decreased malaria risk in pregnancy only when measured with ferritin. Until there is more evidence, it is prudent to provide iron in combination with malaria prevention during pregnancy.
Collapse
Affiliation(s)
- Laura Sangaré
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
- Department of Bioengineering, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| | - Anna Maria van Eijk
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Feiko O. ter Kuile
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Judd Walson
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
- Department of Epidemiology, University of Washington, Seattle, Washington, United States of America
- Department of Medicine and Department of Pediatrics, University of Washington, Seattle, Washington, United States of America
| | - Andy Stergachis
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
- Department of Epidemiology, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
32
|
A ratiometric fluorescent probe for iron(III) and its application for detection of iron(III) in human blood serum. Anal Chim Acta 2014; 812:145-51. [DOI: 10.1016/j.aca.2013.12.024] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Revised: 12/09/2013] [Accepted: 12/17/2013] [Indexed: 11/13/2022]
|
33
|
Shapiro HM, Apte SH, Chojnowski GM, Hänscheid T, Rebelo M, Grimberg BT. Cytometry in malaria--a practical replacement for microscopy? ACTA ACUST UNITED AC 2014; Chapter 11:11.20.1-11.20.23. [PMID: 23835802 DOI: 10.1002/0471142956.cy1120s65] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Malaria, caused by protozoan Plasmodium parasites, kills ~800,000 people each year. Exact figures are uncertain because presumptive diagnoses are often made without identifying parasites in patients' blood either by microscopy, using Giemsa's century-old stain, or by simpler tests that are ultimately dependent on microscopy for quality control. Microscopy itself relies on trained observers' ability to detect subtle morphological features of parasitized red blood cells, only a few of which may be present on a slide. Quantitative and objective flow cytometric measurements of cellular constituents such as DNA, RNA, and the malaria pigment hemozoin are now useful in research in malaria biology and pharmacology, and can provide more reliable identification of parasite species and developmental stages and better detection of low-density parasitemia than could microscopy. The same measurements can now be implemented in much smaller, simpler, cheaper imaging cytometers, potentially providing a more accurate and precise diagnostic modality.
Collapse
|
34
|
Klonis N, Creek DJ, Tilley L. Iron and heme metabolism in Plasmodium falciparum and the mechanism of action of artemisinins. Curr Opin Microbiol 2013; 16:722-7. [PMID: 23932203 DOI: 10.1016/j.mib.2013.07.005] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Accepted: 07/04/2013] [Indexed: 11/17/2022]
Abstract
During the asexual blood stage of its lifecycle, the malaria parasite Plasmodium falciparum grows and multiplies in the hemoglobin-rich environment of the human erythrocyte. Although the parasite has evolved unique strategies to survive in this environment, its interaction with iron represents an Achilles' heel that is exploited by many antimalarial drugs. Recent work has shed new light on how the parasite deals with hemoglobin breakdown products and on the role of iron as a mediator of the action of the antimalarial drug, artemisinin.
Collapse
Affiliation(s)
- Nectarios Klonis
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, and ARC Centre of Excellence for Coherent X-ray Science, 30 Flemington Road, University of Melbourne, Parkville, VIC 3010, Australia
| | | | | |
Collapse
|