1
|
Blackwell AM, Jami-Alahmadi Y, Nasamu AS, Kudo S, Senoo A, Slam C, Tsumoto K, Wohlschlegel JA, Manuel Martinez Caaveiro J, Goldberg DE, Sigala PA. Malaria parasites require a divergent heme oxygenase for apicoplast gene expression and biogenesis. eLife 2024; 13:RP100256. [PMID: 39660822 PMCID: PMC11634067 DOI: 10.7554/elife.100256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2024] Open
Abstract
Malaria parasites have evolved unusual metabolic adaptations that specialize them for growth within heme-rich human erythrocytes. During blood-stage infection, Plasmodium falciparum parasites internalize and digest abundant host hemoglobin within the digestive vacuole. This massive catabolic process generates copious free heme, most of which is biomineralized into inert hemozoin. Parasites also express a divergent heme oxygenase (HO)-like protein (PfHO) that lacks key active-site residues and has lost canonical HO activity. The cellular role of this unusual protein that underpins its retention by parasites has been unknown. To unravel PfHO function, we first determined a 2.8 Å-resolution X-ray structure that revealed a highly α-helical fold indicative of distant HO homology. Localization studies unveiled PfHO targeting to the apicoplast organelle, where it is imported and undergoes N-terminal processing but retains most of the electropositive transit peptide. We observed that conditional knockdown of PfHO was lethal to parasites, which died from defective apicoplast biogenesis and impaired isoprenoid-precursor synthesis. Complementation and molecular-interaction studies revealed an essential role for the electropositive N-terminus of PfHO, which selectively associates with the apicoplast genome and enzymes involved in nucleic acid metabolism and gene expression. PfHO knockdown resulted in a specific deficiency in levels of apicoplast-encoded RNA but not DNA. These studies reveal an essential function for PfHO in apicoplast maintenance and suggest that Plasmodium repurposed the conserved HO scaffold from its canonical heme-degrading function in the ancestral chloroplast to fulfill a critical adaptive role in organelle gene expression.
Collapse
Affiliation(s)
- Amanda Mixon Blackwell
- Department of Biochemistry, University of Utah School of MedicineSalt Lake CityUnited States
| | - Yasaman Jami-Alahmadi
- Department of Biological Chemistry, University of California, Los AngelesLos AngelesUnited States
| | - Armiyaw S Nasamu
- Departments of Medicine and Molecular Microbiology, Washington University School of MedicineSt. LouisUnited States
| | - Shota Kudo
- Department of Chemistry & Biotechnology, The University of TokyoTokyoJapan
| | - Akinobu Senoo
- Department of Protein Drug Discovery, Graduate School of Pharmaceutical Sciences, Kyushu UniversityFukuokaJapan
| | - Celine Slam
- Department of Biochemistry, University of Utah School of MedicineSalt Lake CityUnited States
| | - Kouhei Tsumoto
- Department of Chemistry & Biotechnology, The University of TokyoTokyoJapan
- Department of Bioengineering, University of TokyoTokyoJapan
| | - James A Wohlschlegel
- Department of Biological Chemistry, University of California, Los AngelesLos AngelesUnited States
| | | | - Daniel E Goldberg
- Departments of Medicine and Molecular Microbiology, Washington University School of MedicineSt. LouisUnited States
| | - Paul A Sigala
- Department of Biochemistry, University of Utah School of MedicineSalt Lake CityUnited States
- Departments of Medicine and Molecular Microbiology, Washington University School of MedicineSt. LouisUnited States
| |
Collapse
|
2
|
Loveridge KM, Sigala PA. Identification of a divalent metal transporter required for cellular iron metabolism in malaria parasites. Proc Natl Acad Sci U S A 2024; 121:e2411631121. [PMID: 39467134 PMCID: PMC11551425 DOI: 10.1073/pnas.2411631121] [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: 06/12/2024] [Accepted: 09/23/2024] [Indexed: 10/30/2024] Open
Abstract
Plasmodium falciparum malaria parasites invade and multiply inside red blood cells (RBCs), the most iron-rich compartment in humans. Like all cells, P. falciparum requires nutritional iron to support essential metabolic pathways, but the critical mechanisms of iron acquisition and trafficking during RBC infection have remained obscure. Parasites internalize and liberate massive amounts of heme during large-scale digestion of RBC hemoglobin within an acidic food vacuole (FV) but lack a heme oxygenase to release porphyrin-bound iron. Although most FV heme is sequestered into inert hemozoin crystals, prior studies indicate that trace heme escapes biomineralization and is susceptible to nonenzymatic degradation within the oxidizing FV environment to release labile iron. Parasites retain a homolog of divalent metal transporter 1 (DMT1), a known mammalian iron transporter, but its role in P. falciparum iron acquisition has not been tested. Our phylogenetic studies indicate that P. falciparum DMT1 (PfDMT1) retains conserved molecular features critical for metal transport. We localized this protein to the FV membrane and defined its orientation in an export-competent topology. Conditional knockdown of PfDMT1 expression is lethal to parasites, which display broad cellular defects in iron-dependent functions, including impaired apicoplast biogenesis and mitochondrial polarization. Parasites are selectively rescued from partial PfDMT1 knockdown by supplementation with exogenous iron, but not other metals. These results support a cellular paradigm whereby PfDMT1 is the molecular gatekeeper to essential iron acquisition by blood-stage malaria parasites and suggest that therapeutic targeting of PfDMT1 may be a potent antimalarial strategy.
Collapse
Affiliation(s)
- Kade M. Loveridge
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT84112
| | - Paul A. Sigala
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT84112
| |
Collapse
|
3
|
Blackwell AM, Jami-Alahmadi Y, Nasamu AS, Kudo S, Senoo A, Slam C, Tsumoto K, Wohlschlegel JA, Caaveiro JMM, Goldberg DE, Sigala PA. Malaria parasites require a divergent heme oxygenase for apicoplast gene expression and biogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.30.596652. [PMID: 38853871 PMCID: PMC11160694 DOI: 10.1101/2024.05.30.596652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Malaria parasites have evolved unusual metabolic adaptations that specialize them for growth within heme-rich human erythrocytes. During blood-stage infection, Plasmodium falciparum parasites internalize and digest abundant host hemoglobin within the digestive vacuole. This massive catabolic process generates copious free heme, most of which is biomineralized into inert hemozoin. Parasites also express a divergent heme oxygenase (HO)-like protein (PfHO) that lacks key active-site residues and has lost canonical HO activity. The cellular role of this unusual protein that underpins its retention by parasites has been unknown. To unravel PfHO function, we first determined a 2.8 Å-resolution X-ray structure that revealed a highly α-helical fold indicative of distant HO homology. Localization studies unveiled PfHO targeting to the apicoplast organelle, where it is imported and undergoes N-terminal processing but retains most of the electropositive transit peptide. We observed that conditional knockdown of PfHO was lethal to parasites, which died from defective apicoplast biogenesis and impaired isoprenoid-precursor synthesis. Complementation and molecular-interaction studies revealed an essential role for the electropositive N-terminus of PfHO, which selectively associates with the apicoplast genome and enzymes involved in nucleic acid metabolism and gene expression. PfHO knockdown resulted in a specific deficiency in levels of apicoplast-encoded RNA but not DNA. These studies reveal an essential function for PfHO in apicoplast maintenance and suggest that Plasmodium repurposed the conserved HO scaffold from its canonical heme-degrading function in the ancestral chloroplast to fulfill a critical adaptive role in organelle gene expression.
Collapse
Affiliation(s)
| | | | - Armiyaw S. Nasamu
- Departments of Medicine and Molecular Microbiology, Washington University School of Medicine, St. Louis, MO
| | - Shota Kudo
- Department of Chemistry & Biotechnology, The University of Tokyo, Tokyo, Japan
| | - Akinobu Senoo
- Department of Protein Drug Discovery, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Celine Slam
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT
| | - Kouhei Tsumoto
- Department of Chemistry & Biotechnology, The University of Tokyo, Tokyo, Japan
- Department of Bioengineering, University of Tokyo, Tokyo, Japan
| | | | - Jose M. M. Caaveiro
- Department of Chemistry & Biotechnology, The University of Tokyo, Tokyo, Japan
| | - Daniel E. Goldberg
- Departments of Medicine and Molecular Microbiology, Washington University School of Medicine, St. Louis, MO
| | - Paul A. Sigala
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT
- Departments of Medicine and Molecular Microbiology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
4
|
Bombaça ACS, Caminha MA, Barbosa JMC, Pedra-Rezende Y, Ennes-Vidal V, Brunoro GVF, Archanjo BS, d'Avila CM, Valente RH, Menna-Barreto RFS. Heme metabolism in Strigomonas culicis: Implications of H 2O 2 resistance induction and symbiont elimination. J Biol Chem 2024; 300:107692. [PMID: 39159809 DOI: 10.1016/j.jbc.2024.107692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 08/07/2024] [Accepted: 08/09/2024] [Indexed: 08/21/2024] Open
Abstract
Monoxenous trypanosomatid Strigomonas culicis harbors an endosymbiotic bacterium, which enables the protozoa to survive without heme supplementation. The impact of H2O2 resistance and symbiont elimination on intracellular heme and Fe2+ availability was analyzed through a comparison of WT strain with both WT H2O2-resistant (WTR) and aposymbiotic (Apo) protozoa. The relative quantification of the heme biosynthetic pathway through label-free parallel reaction monitoring targeted mass spectrometry revealed that H2O2 resistance does not influence the abundance of tryptic peptides. However, the Apo strain showed increased coproporphyrinogen III oxidase and ferrochelatase levels. A putative ferrous iron transporter, homologous to LIT1 and TcIT from Leishmania major and Trypanosoma cruzi, was identified for the first time. Label-free parallel reaction monitoring targeted mass spectrometry also showed that S. culicis Iron Transporter (ScIT) increased 1.6- and 16.4-fold in WTR and Apo strains compared to WT. Accordingly, antibody-mediated blockage of ScIT decreased by 28.0% and 40.0% intracellular Fe2+concentration in both WTR and Apo strains, whereas no effect was detected in WT. In a heme-depleted medium, adding 10 μM hemin decreased ScIT transcript levels in Apo, whereas 10 μM PPIX, the substrate of ferrochelatase, increased intracellular Fe2+ concentration and ferric iron reduction. Overall, the data suggest mechanisms dependent on de novo heme synthesis (and its substrates) in the Apo strain to overcome reduced heme availability. Given the importance of heme and Fe2+ as cofactors in metabolic pathways, including oxidative phosphorylation and antioxidant systems, this study provides novel mechanistic insights associated with H2O2 resistance in S. culicis.
Collapse
Affiliation(s)
- Ana Cristina Souza Bombaça
- Laboratório de Biologia Celular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil; Laboratório de Doenças Parasitárias, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Marcelle Almeida Caminha
- Laboratório de Biologia Celular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil; Laboratório de Toxinologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro Brazil
| | | | - Yasmin Pedra-Rezende
- Laboratório de Biologia Celular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil; Laboratório de Toxinologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro Brazil
| | - Vitor Ennes-Vidal
- Laboratório de Doenças Parasitárias, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | | | - Bráulio Soares Archanjo
- Divisão de Metrologia de Materiais, Instituto Nacional de Metrologia, Qualidade e Tecnologia (Inmetro), Duque de Caxias, Brazil
| | - Claudia Masini d'Avila
- Laboratório de Doenças Parasitárias, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Richard Hemmi Valente
- Laboratório de Toxinologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro Brazil
| | | |
Collapse
|
5
|
Loveridge KM, Sigala PA. Identification of a divalent metal transporter required for cellular iron metabolism in malaria parasites. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.10.587216. [PMID: 38798484 PMCID: PMC11118319 DOI: 10.1101/2024.05.10.587216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Plasmodium falciparum malaria parasites invade and multiply inside red blood cells (RBCs), the most iron-rich compartment in humans. Like all cells, P. falciparum requires nutritional iron to support essential metabolic pathways, but the critical mechanisms of iron acquisition and trafficking during RBC infection have remained obscure. Parasites internalize and liberate massive amounts of heme during large-scale digestion of RBC hemoglobin within an acidic food vacuole (FV) but lack a heme oxygenase to release porphyrin-bound iron. Although most FV heme is sequestered into inert hemozoin crystals, prior studies indicate that trace heme escapes biomineralization and is susceptible to non-enzymatic degradation within the oxidizing FV environment to release labile iron. Parasites retain a homolog of divalent metal transporter 1 (DMT1), a known mammalian iron transporter, but its role in P. falciparum iron acquisition has not been tested. Our phylogenetic studies indicate that P. falciparum DMT1 (PfDMT1) retains conserved molecular features critical for metal transport. We localized this protein to the FV membrane and defined its orientation in an export-competent topology. Conditional knockdown of PfDMT1 expression is lethal to parasites, which display broad cellular defects in iron-dependent functions, including impaired apicoplast biogenesis and mitochondrial polarization. Parasites are selectively rescued from partial PfDMT1 knockdown by supplementation with exogenous iron, but not other metals. These results support a cellular paradigm whereby PfDMT1 is the molecular gatekeeper to essential iron acquisition by blood-stage malaria parasites and suggest that therapeutic targeting of PfDMT1 may be a potent antimalarial strategy.
Collapse
Affiliation(s)
- Kade M. Loveridge
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Paul A. Sigala
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, United States
| |
Collapse
|
6
|
Thiele PJ, Mela-Lopez R, Blandin SA, Klug D. Let it glow: genetically encoded fluorescent reporters in Plasmodium. Malar J 2024; 23:114. [PMID: 38643106 PMCID: PMC11032601 DOI: 10.1186/s12936-024-04936-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 04/06/2024] [Indexed: 04/22/2024] Open
Abstract
The use of fluorescent proteins (FPs) in Plasmodium parasites has been key to understand the biology of this obligate intracellular protozoon. FPs like the green fluorescent protein (GFP) enabled to explore protein localization, promoter activity as well as dynamic processes like protein export and endocytosis. Furthermore, FP biosensors have provided detailed information on physiological parameters at the subcellular level, and fluorescent reporter lines greatly extended the malariology toolbox. Still, in order to achieve optimal results, it is crucial to know exactly the properties of the FP of choice and the genetic scenario in which it will be used. This review highlights advantages and disadvantages of available landing sites and promoters that have been successfully applied for the ectopic expression of FPs in Plasmodium berghei and Plasmodium falciparum. Furthermore, the properties of newly developed FPs beyond DsRed and EGFP, in the visualization of cells and cellular structures as well as in the sensing of small molecules are discussed.
Collapse
Affiliation(s)
- Pia J Thiele
- Inserm, CNRS, Université de Strasbourg, UPR9022/U1257, Mosquito Immune Responses (MIR), IBMC, F-67000, Strasbourg, France
| | - Raquel Mela-Lopez
- Inserm, CNRS, Université de Strasbourg, UPR9022/U1257, Mosquito Immune Responses (MIR), IBMC, F-67000, Strasbourg, France
| | - Stéphanie A Blandin
- Inserm, CNRS, Université de Strasbourg, UPR9022/U1257, Mosquito Immune Responses (MIR), IBMC, F-67000, Strasbourg, France
| | - Dennis Klug
- Inserm, CNRS, Université de Strasbourg, UPR9022/U1257, Mosquito Immune Responses (MIR), IBMC, F-67000, Strasbourg, France.
- Institute of Physiology and Pathophysiology, Department of Molecular Cell Physiology, Philipps University Marburg, 35037, Marburg, Germany.
| |
Collapse
|
7
|
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
|
8
|
Singh R, Singh R, Srihari V, Makde RD. In Vitro Investigation Unveiling New Insights into the Antimalarial Mechanism of Chloroquine: Role in Perturbing Nucleation Events during Heme to β-Hematin Transformation. ACS Infect Dis 2023; 9:1647-1657. [PMID: 37471056 DOI: 10.1021/acsinfecdis.3c00278] [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] [Indexed: 07/21/2023]
Abstract
Malaria parasites generate toxic heme during hemoglobin digestion, which is neutralized by crystallizing into inert hemozoin (β-hematin). Chloroquine blocks this detoxification process, resulting in heme-mediated toxicity in malaria parasites. However, the exact mechanism of chloroquine's action remains unknown. This study investigates the impact of chloroquine on the transformation of heme into β-hematin. The results show that chloroquine does not completely halt the transformation process but rather slows it down. Additionally, chloroquine complexation with free heme does not affect substrate availability or inhibit β-hematin formation. Scanning electron microscopy (SEM) and X-ray powder diffraction (XRD) studies indicate that the size of β-hematin crystal particles and crystallites increases in the presence of chloroquine, suggesting that chloroquine does not impede crystal growth. These findings suggest that chloroquine delays hemozoin production by perturbing the nucleation events of crystals and/or the stability of crystal nuclei. Thus, contrary to prevailing beliefs, this study provides a new perspective on the working mechanism of chloroquine.
Collapse
Affiliation(s)
- Rahul Singh
- Beamline Development and Application Section, Bhabha Atomic Research Centre, Mumbai 400085, India
- Homi Bhabha National Institute, Mumbai 400085, India
| | - Rashmi Singh
- Laser & Functional Materials Division, Raja Ramanna Centre for Advanced Technology, Indore 452013, India
| | - Velaga Srihari
- High Pressure and Synchrotron Radiation Physics Division, Bhabha Atomic Research Centre, Mumbai 40008, Maharashtra, India
| | - Ravindra D Makde
- Beamline Development and Application Section, Bhabha Atomic Research Centre, Mumbai 400085, India
- Homi Bhabha National Institute, Mumbai 400085, India
| |
Collapse
|
9
|
Baptista V, Silva M, Ferreira GM, Calçada C, Minas G, Veiga MI, Catarino SO. Optical Spectrophotometry as a Promising Method for Quantification and Stage Differentiation of Plasmodium falciparum Parasites. ACS Infect Dis 2023; 9:140-149. [PMID: 36490289 DOI: 10.1021/acsinfecdis.2c00484] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Malaria is one of the most life-threatening infectious diseases worldwide, claiming half a million lives yearly. Prompt and accurate diagnosis is crucial for disease control and elimination. Currently used diagnostic methods require blood sampling and fail to detect low-level infections. At the symptomatic stage of infection, the parasites feed on red blood cells' (RBCs) hemoglobin, forming inert crystals, the hemozoin, in the process. Thus, along with parasite maturation inside the RBCs, the hemoglobin and hemozoin proportion is inversely related, and they generate specific optical spectra, according to their concentration. Herein, to address the issues of finger prick sampling and the lack of sensitivity of the parasitological test, we explored the optical features of Plasmodium falciparum-infected RBCs through absorbance and reflectance spectrophotometric characterization, aiming for their detection. This is the first work fully characterizing the spectrophotometric properties of P. falciparum-infected RBCs by using only 16 specific wavelengths within the visible optical spectra and two different post-processing algorithms. With such an innovative methodology, low-level infections can be detected and quantified, and early- and late-stage development can be clearly distinguished, not only improving the current detection limits but also proving the successful applicability of spectrophotometry for competitive and accurate malaria diagnosis.
Collapse
Affiliation(s)
- Vitória Baptista
- Microelectromechanical Systems Research Unit (CMEMS-UMinho), School of Engineering, University of Minho, Campus de Azurém, 4800-058 Guimarães, Portugal.,LABBELS─Associate Laboratory, 4800-058 Braga/Guimarães, Portugal.,Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal.,ICVS/3B's─PT Government Associate Laboratory, 4806-909 Guimarães, Braga/, Portugal
| | - Miguel Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal.,ICVS/3B's─PT Government Associate Laboratory, 4806-909 Guimarães, Braga/, Portugal
| | - Gabriel M Ferreira
- Microelectromechanical Systems Research Unit (CMEMS-UMinho), School of Engineering, University of Minho, Campus de Azurém, 4800-058 Guimarães, Portugal.,LABBELS─Associate Laboratory, 4800-058 Braga/Guimarães, Portugal
| | - Carla Calçada
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal.,ICVS/3B's─PT Government Associate Laboratory, 4806-909 Guimarães, Braga/, Portugal
| | - Graça Minas
- Microelectromechanical Systems Research Unit (CMEMS-UMinho), School of Engineering, University of Minho, Campus de Azurém, 4800-058 Guimarães, Portugal.,LABBELS─Associate Laboratory, 4800-058 Braga/Guimarães, Portugal
| | - Maria Isabel Veiga
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal.,ICVS/3B's─PT Government Associate Laboratory, 4806-909 Guimarães, Braga/, Portugal
| | - Susana O Catarino
- Microelectromechanical Systems Research Unit (CMEMS-UMinho), School of Engineering, University of Minho, Campus de Azurém, 4800-058 Guimarães, Portugal.,LABBELS─Associate Laboratory, 4800-058 Braga/Guimarães, Portugal
| |
Collapse
|
10
|
Kongklad G, Chitaree R, Taechalertpaisarn T, Panvisavas N, Nuntawong N. Discriminant Analysis PCA-LDA Assisted Surface-Enhanced Raman Spectroscopy for Direct Identification of Malaria-Infected Red Blood Cells. Methods Protoc 2022; 5:mps5030049. [PMID: 35736550 PMCID: PMC9231316 DOI: 10.3390/mps5030049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 05/31/2022] [Accepted: 06/07/2022] [Indexed: 12/13/2022] Open
Abstract
Various methods for detecting malaria have been developed in recent years, each with its own set of advantages. These methods include microscopic, antigen-based, and molecular-based analysis of blood samples. This study aimed to develop a new, alternative procedure for clinical use by using a large data set of surface-enhanced Raman spectra to distinguish normal and infected red blood cells. PCA-LDA algorithms were used to produce models for separating P. falciparum (3D7)-infected red blood cells and normal red blood cells based on their Raman spectra. Both average normalized spectra and spectral imaging were considered. However, these initial spectra could hardly differentiate normal cells from the infected cells. Then, discrimination analysis was applied to assist in the classification and visualization of the different spectral data sets. The results showed a clear separation in the PCA-LDA coordinate. A blind test was also carried out to evaluate the efficiency of the PCA-LDA separation model and achieved a prediction accuracy of up to 80%. Considering that the PCA-LDA separation accuracy will improve when a larger set of training data is incorporated into the existing database, the proposed method could be highly effective for the identification of malaria-infected red blood cells.
Collapse
Affiliation(s)
- Gunganist Kongklad
- Department of Physics, Faculty of Science, Mahidol University, Bangkok 10400, Thailand;
| | - Ratchapak Chitaree
- Department of Physics, Faculty of Science, Mahidol University, Bangkok 10400, Thailand;
- Correspondence:
| | - Tana Taechalertpaisarn
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand;
| | - Nathinee Panvisavas
- Department of Plant, Faculty of Science, Mahidol University, Bangkok 10400, Thailand;
| | - Noppadon Nuntawong
- National Electronics and Computer Technology Center (NECTEC), 112 Thailand Science Park, Pathum Thani 12120, Thailand;
| |
Collapse
|
11
|
Zhu P, Zhou B. The Antagonizing Role of Heme in the Antimalarial Function of Artemisinin: Elevating Intracellular Free Heme Negatively Impacts Artemisinin Activity in Plasmodium falciparum. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27061755. [PMID: 35335120 PMCID: PMC8949904 DOI: 10.3390/molecules27061755] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 03/02/2022] [Accepted: 03/04/2022] [Indexed: 11/25/2022]
Abstract
The rich source of heme within malarial parasites has been considered to underly the action specificity of artemisinin. We reasoned that increasing intraparasitic free heme levels might further sensitize the parasites to artemisinin. Various means, such as modulating heme synthesis, degradation, polymerization, or hemoglobin digestion, were tried to boost intracellular heme levels, and under several scenarios, free heme levels were significantly augmented. Interestingly, all results arrived at the same conclusion, i.e., elevating heme acted in a strongly negative way, impacting the antimalarial action of artemisinin, but exerted no effect on several other antimalarial drugs. Suppression of the elevated free heme level by introducing heme oxygenase expression effectively restored artemisinin potency. Consistently, zinc protoporphyrin IX/zinc mesoporphyrin, as analogues of heme, drastically increased free heme levels and, concomitantly, the EC50 values of artemisinin. We were unable to effectively mitigate free heme levels, possibly due to an unknown compensating heme uptake pathway, as evidenced by our observation of efficient uptake of a fluorescent heme homologue by the parasite. Our results thus indicate the existence of an effective and mutually compensating heme homeostasis network in the parasites, including an uncharacterized heme uptake pathway, to maintain a certain level of free heme and that augmentation of the free heme level negatively impacts the antimalarial action of artemisinin. Importance: It is commonly believed that heme is critical in activating the antimalarial action of artemisinins. In this work, we show that elevating free heme levels in the malarial parasites surprisingly negatively impacts the action of artemisinin. We tried to boost free heme levels with various means, such as by modulating heme synthesis, heme polymerization, hemoglobin degradation and using heme analogues. Whenever we saw elevation of free heme levels, reduction in artemisinin potency was also observed. The homeostasis of heme appears to be complex, as there exists an unidentified heme uptake pathway in the parasites, nullifying our attempts to effectively reduce intraparasitic free heme levels. Our results thus indicate that too much heme is not good for the antimalarial action of artemisinins. This research can help us better understand the biological properties of this mysterious drug.
Collapse
Affiliation(s)
- Pan Zhu
- State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China;
| | - Bing Zhou
- State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China;
- Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Correspondence:
| |
Collapse
|
12
|
Baptista V, Peng WK, Minas G, Veiga MI, Catarino SO. Review of Microdevices for Hemozoin-Based Malaria Detection. BIOSENSORS 2022; 12:bios12020110. [PMID: 35200370 PMCID: PMC8870200 DOI: 10.3390/bios12020110] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 02/04/2022] [Accepted: 02/06/2022] [Indexed: 05/21/2023]
Abstract
Despite being preventable and treatable, malaria still puts almost half of the world's population at risk. Thus, prompt, accurate and sensitive malaria diagnosis is crucial for disease control and elimination. Optical microscopy and immuno-rapid tests are the standard malaria diagnostic methods in the field. However, these are time-consuming and fail to detect low-level parasitemia. Biosensors and lab-on-a-chip devices, as reported to different applications, usually offer high sensitivity, specificity, and ease of use at the point of care. Thus, these can be explored as an alternative for malaria diagnosis. Alongside malaria infection inside the human red blood cells, parasites consume host hemoglobin generating the hemozoin crystal as a by-product. Hemozoin is produced in all parasite species either in symptomatic and asymptomatic individuals. Furthermore, hemozoin crystals are produced as the parasites invade the red blood cells and their content relates to disease progression. Hemozoin is, therefore, a unique indicator of infection, being used as a malaria biomarker. Herein, the so-far developed biosensors and lab-on-a-chip devices aiming for malaria detection by targeting hemozoin as a biomarker are reviewed and discussed to fulfil all the medical demands for malaria management towards elimination.
Collapse
Affiliation(s)
- Vitória Baptista
- Microelectromechanical Systems Research Unit (CMEMS-UMinho), School of Engineering, Campus de Azurém, University of Minho, 4800-058 Guimarães, Portugal; (G.M.); (S.O.C.)
- LABBELS-Associate Laboratory, Braga/Guimarães, 4806-909 Guimarães, Portugal
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal;
- ICVS/3B’s-PT Government Associate Laboratory, Braga/Guimarães, 4806-909 Guimarães, Portugal
- Correspondence:
| | - Weng Kung Peng
- Songshan Lake Materials Laboratory, Building A1, University Innovation Park, Dongguan 523808, China;
| | - Graça Minas
- Microelectromechanical Systems Research Unit (CMEMS-UMinho), School of Engineering, Campus de Azurém, University of Minho, 4800-058 Guimarães, Portugal; (G.M.); (S.O.C.)
- LABBELS-Associate Laboratory, Braga/Guimarães, 4806-909 Guimarães, Portugal
| | - Maria Isabel Veiga
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal;
- ICVS/3B’s-PT Government Associate Laboratory, Braga/Guimarães, 4806-909 Guimarães, Portugal
| | - Susana O. Catarino
- Microelectromechanical Systems Research Unit (CMEMS-UMinho), School of Engineering, Campus de Azurém, University of Minho, 4800-058 Guimarães, Portugal; (G.M.); (S.O.C.)
- LABBELS-Associate Laboratory, Braga/Guimarães, 4806-909 Guimarães, Portugal
| |
Collapse
|
13
|
Sloan MA, Aghabi D, Harding CR. Orchestrating a heist: uptake and storage of metals by apicomplexan parasites. MICROBIOLOGY (READING, ENGLAND) 2021; 167. [PMID: 34898419 PMCID: PMC7612242 DOI: 10.1099/mic.0.001114] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Megan A Sloan
- Wellcome Centre for Integrative Parasitology, Institute for Infection, Immunity and Inflammation, University of Glasgow, UK
| | - Dana Aghabi
- Wellcome Centre for Integrative Parasitology, Institute for Infection, Immunity and Inflammation, University of Glasgow, UK
| | | |
Collapse
|
14
|
Baptista V, Costa MS, Calçada C, Silva M, Gil JP, Veiga MI, Catarino SO. The Future in Sensing Technologies for Malaria Surveillance: A Review of Hemozoin-Based Diagnosis. ACS Sens 2021; 6:3898-3911. [PMID: 34735120 DOI: 10.1021/acssensors.1c01750] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Early and effective malaria diagnosis is vital to control the disease spread and to prevent the emergence of severe cases and death. Currently, malaria diagnosis relies on optical microscopy and immuno-rapid tests; however, these require a drop of blood, are time-consuming, or are not specific and sensitive enough for reliable detection of low-level parasitaemia. Thus, there is an urge for simpler, prompt, and accurate alternative diagnostic methods. Particularly, hemozoin has been increasingly recognized as an attractive biomarker for malaria detection. As the disease proliferates, parasites digest host hemoglobin, in the process releasing toxic haem that is detoxified into an insoluble crystal, the hemozoin, which accumulates along with infection progression. Given its magnetic, optical, and acoustic unique features, hemozoin has been explored for new label-free diagnostic methods. Thereby, herein, we review the hemozoin-based malaria detection methods and critically discuss their challenges and potential for the development of an ideal diagnostic device.
Collapse
Affiliation(s)
- Vitória Baptista
- Microelectromechanical Systems Research Unit (CMEMS-UMinho), School of Engineering, University of Minho, Campus de Azurém, 4800-058 Guimarães, Portugal
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- ICVS/3B’s − PT Government Associate Laboratory, 4805-017 Braga/Guimarães, Portugal
| | - Mariana S. Costa
- Microelectromechanical Systems Research Unit (CMEMS-UMinho), School of Engineering, University of Minho, Campus de Azurém, 4800-058 Guimarães, Portugal
| | - Carla Calçada
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- ICVS/3B’s − PT Government Associate Laboratory, 4805-017 Braga/Guimarães, Portugal
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal
| | - Miguel Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- ICVS/3B’s − PT Government Associate Laboratory, 4805-017 Braga/Guimarães, Portugal
| | - José Pedro Gil
- Stockholm Malaria Center, Department of Microbiology and Tumour Cell Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Maria Isabel Veiga
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- ICVS/3B’s − PT Government Associate Laboratory, 4805-017 Braga/Guimarães, Portugal
| | - Susana O. Catarino
- Microelectromechanical Systems Research Unit (CMEMS-UMinho), School of Engineering, University of Minho, Campus de Azurém, 4800-058 Guimarães, Portugal
| |
Collapse
|
15
|
de Villiers KA, Egan TJ. Heme Detoxification in the Malaria Parasite: A Target for Antimalarial Drug Development. Acc Chem Res 2021; 54:2649-2659. [PMID: 33982570 DOI: 10.1021/acs.accounts.1c00154] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Over the last century, malaria deaths have decreased by more than 85%. Nonetheless, there were 405 000 deaths in 2018, mostly resulting from Plasmodium falciparum infection. In the 21st century, much of the advance has arisen from the deployment of insecticide-treated bed nets and artemisinin combination therapy. However, over the past few decades parasites with a delayed artemisinin clearance phenotype have appeared in Southeast Asia, threatening further gains. The effort to find new drugs is thus urgent. A prominent process in blood stage malaria parasites, which we contend remains a viable drug target, is hemozoin formation. This crystalline material consisting of heme can be readily seen when parasites are viewed microscopically. The process of its formation in the parasite, however, is still not fully understood.In early work, we recognized hemozoin formation as a biomineralization process. We have subsequently investigated the kinetics of synthetic hemozoin (β-hematin) crystallization catalyzed at lipid-aqueous interfaces under biomimetic conditions. This led us to the use of neutral detergent-based high-throughput screening (HTS) for inhibitors of β-hematin formation. A good hit rate against malaria parasites was obtained. Simultaneously, we developed a pyridine-based assay which proved successful in measuring the concentrations of hematin not converted to β-hematin.The pyridine assay was adapted to determine the effects of chloroquine and other clinical antimalarials on hemozoin formation in the cell. This permitted the determination of the dose-dependent amounts of exchangeable heme and hemozoin in P. falciparum for the first time. These studies have shown that hemozoin inhibitors cause a dose-dependent increase in exchangeable heme, correlated with decreased parasite survival. Electron spectroscopic imaging (ESI) showed a relocation of heme iron into the parasite cytoplasm, while electron microscopy provided evidence of the disruption of hemozoin crystals. This cellular assay was subsequently extended to top-ranked hits from a wide range of scaffolds found by HTS. Intriguingly, the amounts of exchangeable heme at the parasite growth IC50 values of these scaffolds showed substantial variation. The amount of exchangeable heme was found to be correlated with the amount of inhibitor accumulated in the parasitized red blood cell. This suggests that heme-inhibitor complexes, rather than free heme, lead to parasite death. This was supported by ESI using a Br-containing compound which showed the colocalization of Fe and Br as well as by confocal Raman microscopy which confirmed the presence of a complex in the parasite. Current evidence indicates that inhibitors block hemozoin formation by surface adsorption. Indeed, we have successfully introduced molecular docking with hemozoin to find new inhibitors. It follows that the resulting increase in free heme leads to the formation of the parasiticidal heme-inhibitor complex. We have reported crystal structures of heme-drug complexes for several aryl methanol antimalarials in nonaqueous media. These form coordination complexes but most other inhibitors interact noncovalently, and the determination of their structures remains a major challenge.It is our view that key future developments will include improved assays to measure cellular heme levels, better in silico approaches for predicting β-hematin inhibition, and a concerted effort to determine the structure and properties of heme-inhibitor complexes.
Collapse
Affiliation(s)
- Katherine A. de Villiers
- Department of Chemistry and Polymer Science, Stellenbosch University, Private Bag, Matieland 7600, South Africa
| | - Timothy J. Egan
- Department of Chemistry, University of Cape Town, Private Bag, Rondebosch 7701, South Africa
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, Cape Town 7945, South Africa
| |
Collapse
|
16
|
Nairz M, Weiss G. Iron in infection and immunity. Mol Aspects Med 2020; 75:100864. [PMID: 32461004 DOI: 10.1016/j.mam.2020.100864] [Citation(s) in RCA: 177] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 04/25/2020] [Accepted: 05/05/2020] [Indexed: 12/12/2022]
Abstract
Iron is an essential micronutrient for virtually all living cells. In infectious diseases, both invading pathogens and mammalian cells including those of the immune system require iron to sustain their function, metabolism and proliferation. On the one hand, microbial iron uptake is linked to the virulence of most human pathogens. On the other hand, the sequestration of iron from bacteria and other microorganisms is an efficient strategy of host defense in line with the principles of 'nutritional immunity'. In an acute infection, host-driven iron withdrawal inhibits the growth of pathogens. Chronic immune activation due to persistent infection, autoimmune disease or malignancy however, sequesters iron not only from infectious agents, autoreactive lymphocytes and neoplastic cells but also from erythroid progenitors. This is one of the key mechanisms which collectively result in the anemia of chronic inflammation. In this review, we highlight the most important interconnections between iron metabolism and immunity, focusing on host defense against relevant infections and on the clinical consequences of anemia of inflammation.
Collapse
Affiliation(s)
- Manfred Nairz
- 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; Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, Austria.
| |
Collapse
|
17
|
Plasmodium falciparum Knockout for the GPCR-Like PfSR25 Receptor Displays Greater Susceptibility to 1,2,3-Triazole Compounds That Block Malaria Parasite Development. Biomolecules 2020; 10:biom10081197. [PMID: 32824696 PMCID: PMC7465636 DOI: 10.3390/biom10081197] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 08/13/2020] [Accepted: 08/15/2020] [Indexed: 01/05/2023] Open
Abstract
The search for new compounds with antimalarial activity is urgent, as resistance to ones in the classical drug, has already been described in more than one continent. Compounds derived from 1,2,3-triazoles are effective against parasites and bacteria. Here, we evaluated the potential antimalarial activity against the human malaria parasite Plasmodium falciparum in a culture of fifty-four triazole compounds derived from 1H-and 2H-1,2,3-triazole. We identified thirty-one compounds with potential antimalarial activity at concentrations in the micromolar order (µM) and IC50 values ranging from 2.80 µM (9) to 29.27 µM (21). Then, we selected some of these compounds to perform the same tests on the PfSR25- strain (knockout for P. falciparum G-protein coupled receptor-like, SR25). Our experiences with the PfSR25- strain showed that both compounds with higher antimalarial activity for the 3D7 strain and those with less activity resulted in lower IC50 values for the knockout strain. The cytotoxicity of the compounds was evaluated in human renal embryonic cells (HEK 293), using MTT assays. This demonstrated that the compounds with the highest activity (9, 13, 19, 22, 24, 29), showed no toxicity at the tested concentrations.
Collapse
|
18
|
Kloehn J, Harding CR, Soldati-Favre D. Supply and demand-heme synthesis, salvage and utilization by Apicomplexa. FEBS J 2020; 288:382-404. [PMID: 32530125 DOI: 10.1111/febs.15445] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 05/23/2020] [Accepted: 06/05/2020] [Indexed: 01/05/2023]
Abstract
The Apicomplexa phylum groups important human and animal pathogens that cause severe diseases, encompassing malaria, toxoplasmosis, and cryptosporidiosis. In common with most organisms, apicomplexans rely on heme as cofactor for several enzymes, including cytochromes of the electron transport chain. This heme derives from de novo synthesis and/or the development of uptake mechanisms to scavenge heme from their host. Recent studies have revealed that heme synthesis is essential for Toxoplasma gondii tachyzoites, as well as for the mosquito and liver stages of Plasmodium spp. In contrast, the erythrocytic stages of the malaria parasites rely on scavenging heme from the host red blood cell. The unusual heme synthesis pathway in Apicomplexa spans three cellular compartments and comprises enzymes of distinct ancestral origin, providing promising drug targets. Remarkably given the requirement for heme, T. gondii can tolerate the loss of several heme synthesis enzymes at a high fitness cost, while the ferrochelatase is essential for survival. These findings indicate that T. gondii is capable of salvaging heme precursors from its host. Furthermore, heme is implicated in the activation of the key antimalarial drug artemisinin. Recent findings established that a reduction in heme availability corresponds to decreased sensitivity to artemisinin in T. gondii and Plasmodium falciparum, providing insights into the possible development of combination therapies to tackle apicomplexan parasites. This review describes the microeconomics of heme in Apicomplexa, from supply, either from de novo synthesis or scavenging, to demand by metabolic pathways, including the electron transport chain.
Collapse
Affiliation(s)
- Joachim Kloehn
- Department of Microbiology and Molecular Medicine, CMU, University of Geneva, Switzerland
| | - Clare R Harding
- Wellcome Centre for Molecular Parasitology, Institute of Infection, Immunity & Inflammation, University of Glasgow, UK
| | | |
Collapse
|
19
|
Mach J, Sutak R. Iron in parasitic protists – from uptake to storage and where we can interfere. Metallomics 2020; 12:1335-1347. [DOI: 10.1039/d0mt00125b] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A comprehensive review of iron metabolism in parasitic protists and its potential use as a drug target.
Collapse
Affiliation(s)
- Jan Mach
- Department of Parasitology
- Faculty of Science - BIOCEV
- Charles University
- Vestec u Prahy
- Czech Republic
| | - Robert Sutak
- Department of Parasitology
- Faculty of Science - BIOCEV
- Charles University
- Vestec u Prahy
- Czech Republic
| |
Collapse
|
20
|
Ramos S, Carlos AR, Sundaram B, Jeney V, Ribeiro A, Gozzelino R, Bank C, Gjini E, Braza F, Martins R, Ademolue TW, Blankenhaus B, Gouveia Z, Faísca P, Trujillo D, Cardoso S, Rebelo S, Del Barrio L, Zarjou A, Bolisetty S, Agarwal A, Soares MP. Renal control of disease tolerance to malaria. Proc Natl Acad Sci U S A 2019; 116:5681-5686. [PMID: 30833408 PMCID: PMC6431151 DOI: 10.1073/pnas.1822024116] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Malaria, the disease caused by Plasmodium spp. infection, remains a major global cause of morbidity and mortality. Host protection from malaria relies on immune-driven resistance mechanisms that kill Plasmodium However, these mechanisms are not sufficient per se to avoid the development of severe forms of disease. This is accomplished instead via the establishment of disease tolerance to malaria, a defense strategy that does not target Plasmodium directly. Here we demonstrate that the establishment of disease tolerance to malaria relies on a tissue damage-control mechanism that operates specifically in renal proximal tubule epithelial cells (RPTEC). This protective response relies on the induction of heme oxygenase-1 (HMOX1; HO-1) and ferritin H chain (FTH) via a mechanism that involves the transcription-factor nuclear-factor E2-related factor-2 (NRF2). As it accumulates in plasma and urine during the blood stage of Plasmodium infection, labile heme is detoxified in RPTEC by HO-1 and FTH, preventing the development of acute kidney injury, a clinical hallmark of severe malaria.
Collapse
Affiliation(s)
- Susana Ramos
- Instituto Gulbenkian de Ciência, 2780-156 Oeiras, Portugal
| | | | | | - Viktoria Jeney
- Faculty of Medicine, Department of Internal Medicine, University of Debrecen, H-4032, Debrecen, Hungary
| | - Ana Ribeiro
- Instituto Gulbenkian de Ciência, 2780-156 Oeiras, Portugal
| | | | - Claudia Bank
- Instituto Gulbenkian de Ciência, 2780-156 Oeiras, Portugal
| | - Erida Gjini
- Instituto Gulbenkian de Ciência, 2780-156 Oeiras, Portugal
| | - Faouzi Braza
- Instituto Gulbenkian de Ciência, 2780-156 Oeiras, Portugal
| | - Rui Martins
- Instituto Gulbenkian de Ciência, 2780-156 Oeiras, Portugal
| | | | | | - Zélia Gouveia
- Instituto Gulbenkian de Ciência, 2780-156 Oeiras, Portugal
| | - Pedro Faísca
- Instituto Gulbenkian de Ciência, 2780-156 Oeiras, Portugal
| | - Damian Trujillo
- Department of Microbiology and Immunology, Stanford University, Stanford, CA 94305-5124
| | - Sílvia Cardoso
- Instituto Gulbenkian de Ciência, 2780-156 Oeiras, Portugal
| | - Sofia Rebelo
- Instituto Gulbenkian de Ciência, 2780-156 Oeiras, Portugal
| | | | - Abolfazl Zarjou
- Department of Medicine, Division of Nephrology, University of Alabama, Birmingham, AL 35294
| | - Subhashini Bolisetty
- Department of Medicine, Division of Nephrology, University of Alabama, Birmingham, AL 35294
| | - Anupam Agarwal
- Department of Medicine, Division of Nephrology, University of Alabama, Birmingham, AL 35294
- Birmingham Veterans Administration Medical Center, Birmingham, AL 35294
| | | |
Collapse
|
21
|
Markwalter C, Kantor AG, Moore CP, Richardson KA, Wright DW. Inorganic Complexes and Metal-Based Nanomaterials for Infectious Disease Diagnostics. Chem Rev 2019; 119:1456-1518. [PMID: 30511833 PMCID: PMC6348445 DOI: 10.1021/acs.chemrev.8b00136] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Indexed: 12/12/2022]
Abstract
Infectious diseases claim millions of lives each year. Robust and accurate diagnostics are essential tools for identifying those who are at risk and in need of treatment in low-resource settings. Inorganic complexes and metal-based nanomaterials continue to drive the development of diagnostic platforms and strategies that enable infectious disease detection in low-resource settings. In this review, we highlight works from the past 20 years in which inorganic chemistry and nanotechnology were implemented in each of the core components that make up a diagnostic test. First, we present how inorganic biomarkers and their properties are leveraged for infectious disease detection. In the following section, we detail metal-based technologies that have been employed for sample preparation and biomarker isolation from sample matrices. We then describe how inorganic- and nanomaterial-based probes have been utilized in point-of-care diagnostics for signal generation. The following section discusses instrumentation for signal readout in resource-limited settings. Next, we highlight the detection of nucleic acids at the point of care as an emerging application of inorganic chemistry. Lastly, we consider the challenges that remain for translation of the aforementioned diagnostic platforms to low-resource settings.
Collapse
Affiliation(s)
| | | | | | | | - David W. Wright
- Department of Chemistry, Vanderbilt
University, Nashville, Tennessee 37235, United States
| |
Collapse
|
22
|
Lechuga GC, Pereira MCS, Bourguignon SC. Heme metabolism as a therapeutic target against protozoan parasites. J Drug Target 2018; 27:767-779. [DOI: 10.1080/1061186x.2018.1536982] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Guilherme Curty Lechuga
- Laboratório de Interação celular e molecular, Departamento de Biologia Celular e Molecular, Universidade Federal Fluminense, Rua Outeiro São João Batista, Rio de Janeiro, Brazil
- Fundação Oswaldo Cruz, Laboratório de Ultraestrutura Celular, Rio de Janeiro, Brazil
- Instituto de Biologia, Programa de Pós-graduação em Ciências e Biotecnologia (PPBI), Universidade Federal Fluminense, Rio de Janeiro, Brazil
| | - Mirian C. S. Pereira
- Fundação Oswaldo Cruz, Laboratório de Ultraestrutura Celular, Rio de Janeiro, Brazil
| | - Saulo C. Bourguignon
- Laboratório de Interação celular e molecular, Departamento de Biologia Celular e Molecular, Universidade Federal Fluminense, Rua Outeiro São João Batista, Rio de Janeiro, Brazil
- Instituto de Biologia, Programa de Pós-graduação em Ciências e Biotecnologia (PPBI), Universidade Federal Fluminense, Rio de Janeiro, Brazil
| |
Collapse
|
23
|
The Asp99-Arg188 salt bridge of the Pseudomonas aeruginosa HemO is critical in allowing conformational flexibility during catalysis. J Biol Inorg Chem 2018; 23:1057-1070. [PMID: 30194537 DOI: 10.1007/s00775-018-1609-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 08/23/2018] [Indexed: 12/17/2022]
Abstract
The P. aeruginosa iron-regulated heme oxygenase (HemO) is required within the host for the utilization of heme as an iron source. As iron is essential for survival and virulence, HemO represents a novel antimicrobial target. We recently characterized small molecule inhibitors that bind to an allosteric site distant from the heme pocket, and further proposed binding at this site disrupts a nearby salt bridge between D99 and R188. Herein, through a combination of site-directed mutagenesis and hydrogen-deuterium exchange mass spectrometry (HDX-MS), we determined that the disruption of the D99-R188 salt bridge leads to significant decrease in conformational flexibility within the distal and proximal helices that form the heme-binding site. The RR spectra of the resting state Fe(III) and reduced Fe(II)-deoxy heme-HemO D99A, R188A and D99/R188A complexes are virtually identical to those of wild-type HemO, indicating no significant change in the heme environment. Furthermore, mutation of D99 or R188 leads to a modest decrease in the stability of the Fe(II)-O2 heme complex. Despite this slight difference in Fe(II)-O2 stability, we observe complete loss of enzymatic activity. We conclude the loss of activity is a result of decreased conformational flexibility in helices previously shown to be critical in accommodating variation in the distal ligand and the resulting chemical intermediates generated during catalysis. Furthermore, this newly identified allosteric binding site on HemO represents a novel alternative drug-design strategy to that of competitive inhibition at the active site or via direct coordination of ligands to the heme iron.
Collapse
|
24
|
Zhang DL, Wu J, Shah BN, Greutélaers KC, Ghosh MC, Ollivierre H, Su XZ, Thuma PE, Bedu-Addo G, Mockenhaupt FP, Gordeuk VR, Rouault TA. Erythrocytic ferroportin reduces intracellular iron accumulation, hemolysis, and malaria risk. Science 2018; 359:1520-1523. [PMID: 29599243 DOI: 10.1126/science.aal2022] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 08/15/2017] [Accepted: 01/25/2018] [Indexed: 12/17/2022]
Abstract
Malaria parasites invade red blood cells (RBCs), consume copious amounts of hemoglobin, and severely disrupt iron regulation in humans. Anemia often accompanies malaria disease; however, iron supplementation therapy inexplicably exacerbates malarial infections. Here we found that the iron exporter ferroportin (FPN) was highly abundant in RBCs, and iron supplementation suppressed its activity. Conditional deletion of the Fpn gene in erythroid cells resulted in accumulation of excess intracellular iron, cellular damage, hemolysis, and increased fatality in malaria-infected mice. In humans, a prevalent FPN mutation, Q248H (glutamine to histidine at position 248), prevented hepcidin-induced degradation of FPN and protected against severe malaria disease. FPN Q248H appears to have been positively selected in African populations in response to the impact of malaria disease. Thus, FPN protects RBCs against oxidative stress and malaria infection.
Collapse
Affiliation(s)
- De-Liang Zhang
- Section on Human Iron Metabolism, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jian Wu
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Binal N Shah
- Sickle Cell Center, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Katja C Greutélaers
- Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Tropical Medicine and International Health, Berlin 13353, Germany
| | - Manik C Ghosh
- Section on Human Iron Metabolism, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Hayden Ollivierre
- Section on Human Iron Metabolism, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Xin-Zhuan Su
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - George Bedu-Addo
- Komfo Anokye Teaching Hospital, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Frank P Mockenhaupt
- Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Tropical Medicine and International Health, Berlin 13353, Germany
| | - Victor R Gordeuk
- Sickle Cell Center, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Tracey A Rouault
- Section on Human Iron Metabolism, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
25
|
Lechuga GC, Borges JC, Calvet CM, de Araújo HP, Zuma AA, do Nascimento SB, Motta MCM, Bernardino AMR, Pereira MCDS, Bourguignon SC. Interactions between 4-aminoquinoline and heme: Promising mechanism against Trypanosoma cruzi. Int J Parasitol Drugs Drug Resist 2016; 6:154-164. [PMID: 27490082 PMCID: PMC4971285 DOI: 10.1016/j.ijpddr.2016.07.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 06/25/2016] [Accepted: 07/12/2016] [Indexed: 02/07/2023]
Abstract
Chagas disease is a neglected tropical disease caused by the flagellated protozoan Trypanosoma cruzi. The current drugs used to treat this disease have limited efficacy and produce severe side effects. Quinolines, nitrogen heterocycle compounds that form complexes with heme, have a broad spectrum of antiprotozoal activity and are a promising class of new compounds for Chagas disease chemotherapy. In this study, we evaluated the activity of a series of 4-arylaminoquinoline-3-carbonitrile derivatives against all forms of Trypanosoma cruzi in vitro. Compound 1g showed promising activity against epimastigote forms when combined with hemin (IC50<1 μM), with better performance than benznidazole, the reference drug. This compound also inhibited the viability of trypomastigotes and intracellular amastigotes. The potency of 1g in combination with heme was enhanced against epimastigotes and trypomastigotes, suggesting a similar mechanism of action that occurs in Plasmodium spp. The addition of hemin to the culture medium increased trypanocidal activity of analog 1g without changing the cytotoxicity of the host cell, reaching an IC50 of 11.7 μM for trypomastigotes. The mechanism of action was demonstrated by the interaction of compound 1g with hemin in solution and prevention of heme peroxidation. Compound 1g and heme treatment induced alterations of the mitochondrion-kinetoplast complex in epimastigotes and trypomastigotes and also, accumulation of electron-dense deposits in amastigotes as visualized by transmission electron microscopy. The trypanocidal activity of 4-aminoquinolines and the elucidation of the mechanism involving interaction with heme is a neglected field of research, given the parasite's lack of heme biosynthetic pathway and the importance of this cofactor for parasite survival and growth. The results of this study can improve and guide rational drug development and combination treatment strategies.
Collapse
Affiliation(s)
- Guilherme Curty Lechuga
- Laboratório de Interação celular e molecular, Departamento de Biologia Celular e Molecular, Universidade Federal Fluminense, Rua Outeiro São João Batista, 24020-141, Niterói, Rio de Janeiro, Brazil
| | - Júlio Cesar Borges
- Departamento de Química Orgânica, Universidade Federal Fluminense, Rua Outeiro São João Batista, 24020-141, Niterói, Rio de Janeiro, Brazil; Instituto Federal de Educação, Ciência e Tecnologia do Rio de Janeiro, Campus Nilópolis, 26530-060, RJ, Brazil
| | - Claudia Magalhães Calvet
- Laboratório de Ultraestrutura Celular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Avenida Brasil 4365, 21040-360, Rio de Janeiro, RJ, Brazil
| | - Humberto Pinheiro de Araújo
- Departamento de Imunologia, Instituto Nacional de Controle de Qualidade em Saúde, Fundação Oswaldo Cruz, Avenida Brasil 4365, 21040-360, Rio de Janeiro, RJ, Brazil
| | - Aline Araujo Zuma
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho, 373-bloco G. Cidade Universitária, Ilha do Fundão, Rio de Janeiro, RJ, Brazil
| | - Samara Braga do Nascimento
- Laboratório de Interação celular e molecular, Departamento de Biologia Celular e Molecular, Universidade Federal Fluminense, Rua Outeiro São João Batista, 24020-141, Niterói, Rio de Janeiro, Brazil
| | - Maria Cristina Machado Motta
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho, 373-bloco G. Cidade Universitária, Ilha do Fundão, Rio de Janeiro, RJ, Brazil
| | | | - Mirian Claudia de Souza Pereira
- Laboratório de Ultraestrutura Celular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Avenida Brasil 4365, 21040-360, Rio de Janeiro, RJ, Brazil.
| | - Saulo Cabral Bourguignon
- Laboratório de Interação celular e molecular, Departamento de Biologia Celular e Molecular, Universidade Federal Fluminense, Rua Outeiro São João Batista, 24020-141, Niterói, Rio de Janeiro, Brazil.
| |
Collapse
|
26
|
Exported Epoxide Hydrolases Modulate Erythrocyte Vasoactive Lipids during Plasmodium falciparum Infection. mBio 2016; 7:mBio.01538-16. [PMID: 27795395 PMCID: PMC5082902 DOI: 10.1128/mbio.01538-16] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Erythrocytes are reservoirs of important epoxide-containing lipid signaling molecules, including epoxyeicosatrienoic acids (EETs). EETs function as vasodilators and anti-inflammatory modulators in the bloodstream. Bioactive EETs are hydrolyzed to less active diols (dihydroxyeicosatrienoic acids) by epoxide hydrolases (EHs). The malaria parasite Plasmodium falciparum infects host red blood cells (RBCs) and exports hundreds of proteins into the RBC compartment. In this study, we show that two parasite epoxide hydrolases, P. falciparum epoxide hydrolases 1 (PfEH1) and 2 (PfEH2), both with noncanonical serine nucleophiles, are exported to the periphery of infected RBCs. PfEH1 and PfEH2 were successfully expressed in Escherichia coli, and they hydrolyzed physiologically relevant erythrocyte EETs. Mutations in active site residues of PfEH1 ablated the ability of the enzyme to hydrolyze an epoxide substrate. Overexpression of PfEH1 or PfEH2 in parasite-infected RBCs resulted in a significant alteration in the epoxide fatty acids stored in RBC phospholipids. We hypothesize that the parasite disruption of epoxide-containing signaling lipids leads to perturbed vascular function, creating favorable conditions for binding and sequestration of infected RBCs to the microvascular endothelium. The malaria parasite exports hundreds of proteins into the erythrocyte compartment. However, for most of these proteins, their physiological function is unknown. In this study, we investigate two “hypothetical” proteins of the α/β-hydrolase fold family that share sequence similarity with epoxide hydrolases (EHs)—enzymes that destroy bioactive epoxides. Altering EH expression in parasite-infected erythrocytes resulted in a significant change in the epoxide fatty acids stored in the host cell. We propose that these EH enzymes may help the parasite to manipulate host blood vessel opening and inflame the vessel walls as they pass through the circulation system. Understanding how the malaria parasite interacts with its host RBCs will aid in our ability to combat this deadly disease.
Collapse
|
27
|
Sigala PA, Morante K, Tsumoto K, Caaveiro JMM, Goldberg DE. In-Cell Enzymology To Probe His-Heme Ligation in Heme Oxygenase Catalysis. Biochemistry 2016; 55:4836-49. [PMID: 27490825 DOI: 10.1021/acs.biochem.6b00562] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Heme oxygenase (HO) is a ubiquitous enzyme with key roles in inflammation, cell signaling, heme disposal, and iron acquisition. HO catalyzes the oxidative conversion of heme to biliverdin (BV) using a conserved histidine to coordinate the iron atom of bound heme. This His-heme interaction has been regarded as being essential for enzyme activity, because His-to-Ala mutants fail to convert heme to biliverdin in vitro. We probed a panel of proximal His mutants of cyanobacterial, human, and plant HO enzymes using a live-cell activity assay based on heterologous co-expression in Escherichia coli of each HO mutant and a fluorescent biliverdin biosensor. In contrast to in vitro studies with purified proteins, we observed that multiple HO mutants retained significant activity within the intracellular environment of bacteria. X-ray crystallographic structures of human HO1 H25R with bound heme and additional functional studies suggest that HO mutant activity inside these cells does not involve heme ligation by a proximal amino acid. Our study reveals unexpected plasticity in the active site binding interactions with heme that can support HO activity within cells, suggests important contributions by the surrounding active site environment to HO catalysis, and can guide efforts to understand the evolution and divergence of HO function.
Collapse
Affiliation(s)
- Paul A Sigala
- Departments of Medicine and Molecular Microbiology, Washington University School of Medicine , St. Louis, Missouri 63110, United States
| | - Koldo Morante
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo , Bunkyo-ku, Tokyo 113-8654, Japan
| | - Kouhei Tsumoto
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo , Bunkyo-ku, Tokyo 113-8654, Japan.,Medical Proteomics Laboratory, Institute of Medical Science, The University of Tokyo , Minato-ku, Tokyo 108-8639, Japan
| | - Jose M M Caaveiro
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo , Bunkyo-ku, Tokyo 113-8654, Japan
| | - Daniel E Goldberg
- Departments of Medicine and Molecular Microbiology, Washington University School of Medicine , St. Louis, Missouri 63110, United States
| |
Collapse
|
28
|
Alves E, Maluf FV, Bueno VB, Guido RVC, Oliva G, Singh M, Scarpelli P, Costa F, Sartorello R, Catalani LH, Brady D, Tewari R, Garcia CRS. Biliverdin targets enolase and eukaryotic initiation factor 2 (eIF2α) to reduce the growth of intraerythrocytic development of the malaria parasite Plasmodium falciparum. Sci Rep 2016; 6:22093. [PMID: 26915471 PMCID: PMC4768138 DOI: 10.1038/srep22093] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 02/08/2016] [Indexed: 01/09/2023] Open
Abstract
In mammals, haem degradation to biliverdin (BV) through the action of haem oxygenase (HO) is a critical step in haem metabolism. The malaria parasite converts haem into the chemically inert haemozoin to avoid toxicity. We discovered that the knock-out of HO in P. berghei is lethal; therefore, we investigated the function of biliverdin (BV) and haem in the parasite. Addition of external BV and haem to P. falciparum-infected red blood cell (RBC) cultures delays the progression of parasite development. The search for a BV molecular target within the parasites identified P. falciparum enolase (Pf enolase) as the strongest candidate. Isothermal titration calorimetry using recombinant full-length Plasmodium enolase suggested one binding site for BV. Kinetic assays revealed that BV is a non-competitive inhibitor. We employed molecular modelling studies to predict the new binding site as well as the binding mode of BV to P. falciparum enolase. Furthermore, addition of BV and haem targets the phosphorylation of Plasmodium falciparum eIF2α factor, an eukaryotic initiation factor phosphorylated by eIF2α kinases under stress conditions. We propose that BV targets enolase to reduce parasite glycolysis rates and changes the eIF2α phosphorylation pattern as a molecular mechanism for its action.
Collapse
Affiliation(s)
- Eduardo Alves
- Núcleo de Pesquisa em Sinalização Celular Patógeno-Hospedeiro (NUSCEP), Departamento de Fisiologia, Instituto de Biociências, Universidade de São Paulo, Brasil.,Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Brasil
| | - Fernando V Maluf
- Centro de Pesquisa e Inovação em Biodiversidade e Fármacos, Instituto de Física de São Carlos, Universidade de São Paulo, Brasil
| | - Vânia B Bueno
- Departamento de Química Fundamental, Instituto de Química, Universidade de São Paulo, Brasil
| | - Rafael V C Guido
- Centro de Pesquisa e Inovação em Biodiversidade e Fármacos, Instituto de Física de São Carlos, Universidade de São Paulo, Brasil
| | - Glaucius Oliva
- Centro de Pesquisa e Inovação em Biodiversidade e Fármacos, Instituto de Física de São Carlos, Universidade de São Paulo, Brasil
| | - Maneesh Singh
- Núcleo de Pesquisa em Sinalização Celular Patógeno-Hospedeiro (NUSCEP), Departamento de Fisiologia, Instituto de Biociências, Universidade de São Paulo, Brasil.,Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Brasil
| | - Pedro Scarpelli
- Núcleo de Pesquisa em Sinalização Celular Patógeno-Hospedeiro (NUSCEP), Departamento de Fisiologia, Instituto de Biociências, Universidade de São Paulo, Brasil.,Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Brasil
| | - Fahyme Costa
- Núcleo de Pesquisa em Sinalização Celular Patógeno-Hospedeiro (NUSCEP), Departamento de Fisiologia, Instituto de Biociências, Universidade de São Paulo, Brasil.,Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, Brasil
| | - Robson Sartorello
- Núcleo de Pesquisa em Sinalização Celular Patógeno-Hospedeiro (NUSCEP), Departamento de Fisiologia, Instituto de Biociências, Universidade de São Paulo, Brasil
| | - Luiz H Catalani
- Departamento de Química Fundamental, Instituto de Química, Universidade de São Paulo, Brasil
| | - Declan Brady
- School of Life Sciences, University of Nottingham, UK
| | - Rita Tewari
- School of Life Sciences, University of Nottingham, UK
| | - Celia R S Garcia
- Núcleo de Pesquisa em Sinalização Celular Patógeno-Hospedeiro (NUSCEP), Departamento de Fisiologia, Instituto de Biociências, Universidade de São Paulo, Brasil
| |
Collapse
|
29
|
Zíková A, Hampl V, Paris Z, Týč J, Lukeš J. Aerobic mitochondria of parasitic protists: Diverse genomes and complex functions. Mol Biochem Parasitol 2016; 209:46-57. [PMID: 26906976 DOI: 10.1016/j.molbiopara.2016.02.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 02/16/2016] [Accepted: 02/17/2016] [Indexed: 02/08/2023]
Abstract
In this review the main features of the mitochondria of aerobic parasitic protists are discussed. While the best characterized organelles are by far those of kinetoplastid flagellates and Plasmodium, we also consider amoebae Naegleria and Acanthamoeba, a ciliate Ichthyophthirius and related lineages. The simplistic view of the mitochondrion as just a power house of the cell has already been abandoned in multicellular organisms and available data indicate that this also does not apply for protists. We discuss in more details the following mitochondrial features: genomes, post-transcriptional processing, translation, biogenesis of iron-sulfur complexes, heme metabolism and the electron transport chain. Substantial differences in all these core mitochondrial features between lineages are compatible with the view that aerobic protists harbor organelles that are more complex and flexible than previously appreciated.
Collapse
Affiliation(s)
- Alena Zíková
- Institute of Parasitology, Biology Centre, České Budějovice (Budweis), Czech Republic; University of South Bohemia, Faculty of Science, České Budějovice (Budweis), Czech Republic.
| | - Vladimír Hampl
- Charles University in Prague, Faculty of Science, Prague, Czech Republic
| | - Zdeněk Paris
- Institute of Parasitology, Biology Centre, České Budějovice (Budweis), Czech Republic
| | - Jiří Týč
- Institute of Parasitology, Biology Centre, České Budějovice (Budweis), Czech Republic
| | - Julius Lukeš
- Institute of Parasitology, Biology Centre, České Budějovice (Budweis), Czech Republic; University of South Bohemia, Faculty of Science, České Budějovice (Budweis), Czech Republic; Canadian Institute for Advanced Research, Toronto, Canada.
| |
Collapse
|
30
|
Schnermann MJ, Shenvi RA. Syntheses and biological studies of marine terpenoids derived from inorganic cyanide. Nat Prod Rep 2015; 32:543-77. [PMID: 25514696 DOI: 10.1039/c4np00109e] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Isocyanoterpenes (ICTs) are marine natural products biosynthesized through an unusual pathway that adorns terpene scaffolds with nitrogenous functionality derived from cyanide. The appendage of nitrogen functional groups - isonitriles in particular - onto stereochemically-rich carbocyclic ring systems provides enigmatic, bioactive molecules that have required innovative chemical syntheses. This review discusses the challenges inherent to the synthesis of this diverse family and details the development of the field. We also present recent progress in isolation and discuss key aspects of the remarkable biological activity of these compounds.
Collapse
Affiliation(s)
- Martin J Schnermann
- Chemical Biology Laboratory, National Cancer Institute, Frederick, MD 21701, USA.
| | | |
Collapse
|
31
|
Sigala PA, Crowley JR, Henderson JP, Goldberg DE. Deconvoluting heme biosynthesis to target blood-stage malaria parasites. eLife 2015; 4. [PMID: 26173178 PMCID: PMC4532139 DOI: 10.7554/elife.09143] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 07/13/2015] [Indexed: 12/03/2022] Open
Abstract
Heme metabolism is central to blood-stage infection by the malaria parasite Plasmodium falciparum. Parasites retain a heme biosynthesis pathway but do not require its activity during infection of heme-rich erythrocytes, where they can scavenge host heme to meet metabolic needs. Nevertheless, heme biosynthesis in parasite-infected erythrocytes can be potently stimulated by exogenous 5-aminolevulinic acid (ALA), resulting in accumulation of the phototoxic intermediate protoporphyrin IX (PPIX). Here we use photodynamic imaging, mass spectrometry, parasite gene disruption, and chemical probes to reveal that vestigial host enzymes in the cytoplasm of Plasmodium-infected erythrocytes contribute to ALA-stimulated heme biosynthesis and that ALA uptake depends on parasite-established permeability pathways. We show that PPIX accumulation in infected erythrocytes can be harnessed for antimalarial chemotherapy using luminol-based chemiluminescence and combinatorial stimulation by low-dose artemisinin to photoactivate PPIX to produce cytotoxic reactive oxygen. This photodynamic strategy has the advantage of exploiting host enzymes refractory to resistance-conferring mutations. DOI:http://dx.doi.org/10.7554/eLife.09143.001 Malaria is a devastating infectious disease that is caused by single-celled parasites called Plasmodium that can live inside red blood cells. Several important proteins from these parasites require a small molecule called heme in order to work. The parasites have enzymes that make heme via a series of intermediate steps. However, it remains unclear exactly how important this ‘pathway’ of enzymes is for the parasite, and whether this pathway could be targeted by drugs to treat malaria. Now Sigala et al. have used a range of genetic and biochemical approaches to better understand the production of heme molecules in Plasmodium-infected red blood cells. First, several parasite genes that encode the enzymes used to make heme molecules were deleted. Unexpectedly, these gene deletions did not affect the ability of the infected blood cells to make heme. This result suggested that the parasites do not use their own pathway to produce heme while they are growing in the bloodstream. Sigala et al. then showed that human enzymes involved in making heme, most of which are also found within the infected red blood cells, are still active. These human enzymes provide a parallel pathway that can link up with the final parasite enzyme to generate heme. Further experiments revealed that the activity of the human enzymes could be strongly stimulated by providing the pathway with one of the building blocks used to make heme. This stimulation led to the build-up of an intermediate molecule called PPIX. This intermediate molecule can kill cells when it is exposed to light—a property that is called ‘phototoxicity’. Sigala et al. showed that treating infected red blood cells with a new combination of non-toxic chemicals that emit light can activate PPIX in the bloodstream and can selectively kill the malaria parasites while leaving uninfected cells intact. These findings suggest a new treatment that could be effective against blood-stage malaria. Furthermore, the parasite will be unable to easily mutate to avoid the effects of this treatment because it relies on human proteins that are already made. Future work is now needed to optimize the dosage and the combination of drugs that could provide such a treatment. DOI:http://dx.doi.org/10.7554/eLife.09143.002
Collapse
Affiliation(s)
- Paul A Sigala
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, United States
| | - Jan R Crowley
- Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, United States
| | - Jeffrey P Henderson
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, United States
| | - Daniel E Goldberg
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, United States
| |
Collapse
|
32
|
Rücker H, Amslinger S. Identification of heme oxygenase-1 stimulators by a convenient ELISA-based bilirubin quantification assay. Free Radic Biol Med 2015; 78:135-46. [PMID: 25462643 DOI: 10.1016/j.freeradbiomed.2014.10.506] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 10/10/2014] [Accepted: 10/13/2014] [Indexed: 02/09/2023]
Abstract
The upregulation of heme oxygenase-1 (HO-1) has proven to be a useful tool for fighting inflammation. In order to identify new HO-1 inducers, an efficient screening method was developed which can provide new lead structures for drug research. We designed a simple ELISA-based HO-1 enzyme activity assay, which allows for the screening of 12 compounds in parallel in the setting of a 96-well plate. The well-established murine macrophage cell line RAW264.7 is used and only about 26µg of protein from whole cell lysates is needed for the analysis of HO-1 activity. The quantification of HO-1 activity is based on an indirect ELISA using the specific anti-bilirubin antibody 24G7 to quantify directly bilirubin in the whole cell lysate, applying a horseradish peroxidase-tagged antibody together with ortho-phenylenediamine and H2O2 for detection. The bilirubin is produced on the action of HO enzymes by converting their substrate heme to biliverdin and additional recombinant biliverdin reductase together with NADPH at pH 7.4 in buffer. This sensitive assay allows for the detection of 0.57-82pmol bilirubin per sample in whole cell lysates. Twenty-three small molecules, mainly natural products with an α,β-unsaturated carbonyl unit such as polyphenols, including flavonoids and chalcones, terpenes, an isothiocyanate, and the drug oltipraz were tested at typically 6 or 24h incubation with RAW264.7 cells. The activity of known HO-1 inducers was confirmed, while the chalcones cardamonin, flavokawain A, calythropsin, 2',3,4'-trihydroxy-4-methoxychalcone (THMC), and 2',4'-dihydroxy-3,4-dimethoxychalcone (DHDMC) were identified as new potent HO-1 inducers. The highest inductive power after 6h incubation was found at 10µM for DHDMC (6.1-fold), carnosol (3.9-fold), butein (3.1-fold), THMC (2.9-fold), and zerumbone (2.5-fold). Moreover, the time dependence of HO-1 protein production for DHDMC was compared to its enzyme activity, which was further evaluated in the presence of lipopolysaccharide and the specific HO-1 inhibitor tin protoporphyrin IX. Taken together, we developed a convenient and highly sensitive ELISA-based HO-1 enzyme activity assay, allowing the identification and characterization of molecules potentially useful for the treatment of inflammatory and autoimmune diseases.
Collapse
Affiliation(s)
- Hannelore Rücker
- Institute of Organic Chemistry, University of Regensburg, Universitätsstraße 31, 93053 Regensburg, Germany
| | - Sabine Amslinger
- Institute of Organic Chemistry, University of Regensburg, Universitätsstraße 31, 93053 Regensburg, Germany.
| |
Collapse
|
33
|
Ke H, Sigala PA, Miura K, Morrisey JM, Mather MW, Crowley JR, Henderson JP, Goldberg DE, Long CA, Vaidya AB. The heme biosynthesis pathway is essential for Plasmodium falciparum development in mosquito stage but not in blood stages. J Biol Chem 2014; 289:34827-37. [PMID: 25352601 DOI: 10.1074/jbc.m114.615831] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Heme is an essential cofactor for aerobic organisms. Its redox chemistry is central to a variety of biological functions mediated by hemoproteins. In blood stages, malaria parasites consume most of the hemoglobin inside the infected erythrocytes, forming nontoxic hemozoin crystals from large quantities of heme released during digestion. At the same time, the parasites possess a heme de novo biosynthetic pathway. This pathway in the human malaria parasite Plasmodium falciparum has been considered essential and is proposed as a potential drug target. However, we successfully disrupted the first and last genes of the pathway, individually and in combination. These knock-out parasite lines, lacking 5-aminolevulinic acid synthase and/or ferrochelatase (FC), grew normally in blood-stage culture and exhibited no changes in sensitivity to heme-related antimalarial drugs. We developed a sensitive LC-MS/MS assay to monitor stable isotope incorporation into heme from its precursor 5-[(13)C4]aminolevulinic acid, and this assay confirmed that de novo heme synthesis was ablated in FC knock-out parasites. Disrupting the FC gene also caused no defects in gametocyte generation or maturation but resulted in a greater than 70% reduction in male gamete formation and completely prevented oocyst formation in female Anopheles stephensi mosquitoes. Our data demonstrate that the heme biosynthesis pathway is not essential for asexual blood-stage growth of P. falciparum parasites but is required for mosquito transmission. Drug inhibition of pathway activity is therefore unlikely to provide successful antimalarial therapy. These data also suggest the existence of a parasite mechanism for scavenging host heme to meet metabolic needs.
Collapse
Affiliation(s)
- Hangjun Ke
- From the Center for Molecular Parasitology, Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania 19129
| | - Paul A Sigala
- the Department of Molecular Microbiology and the Howard Hughes Medical Institute, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Kazutoyo Miura
- the Laboratory of Malaria and Vector Research, NIAID, National Institutes of Health, Rockville, Maryland 20852, and
| | - Joanne M Morrisey
- From the Center for Molecular Parasitology, Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania 19129
| | - Michael W Mather
- From the Center for Molecular Parasitology, Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania 19129
| | - Jan R Crowley
- the Center for Women's Infectious Disease Research and
| | - Jeffrey P Henderson
- the Center for Women's Infectious Disease Research and Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Daniel E Goldberg
- the Department of Molecular Microbiology and the Howard Hughes Medical Institute, Washington University School of Medicine, St. Louis, Missouri 63110, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Carole A Long
- the Laboratory of Malaria and Vector Research, NIAID, National Institutes of Health, Rockville, Maryland 20852, and
| | - Akhil B Vaidya
- From the Center for Molecular Parasitology, Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania 19129,
| |
Collapse
|
34
|
Affiliation(s)
- Paul A. Sigala
- Departments of Medicine and Molecular Microbiology and the Howard Hughes Medical Institute, Washington University School of Medicine, St. Louis, Missouri 63110; ,
| | - Daniel E. Goldberg
- Departments of Medicine and Molecular Microbiology and the Howard Hughes Medical Institute, Washington University School of Medicine, St. Louis, Missouri 63110; ,
| |
Collapse
|
35
|
Identification of essential histidine residues involved in heme binding and Hemozoin formation in heme detoxification protein from Plasmodium falciparum. Sci Rep 2014; 4:6137. [PMID: 25138161 PMCID: PMC4138515 DOI: 10.1038/srep06137] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 07/31/2014] [Indexed: 12/22/2022] Open
Abstract
Malaria parasites digest hemoglobin within a food vacuole to supply amino acids, releasing the toxic product heme. During the detoxification, toxic free heme is converted into an insoluble crystalline form called hemozoin (Hz). Heme detoxification protein (HDP) in Plasmodium falciparum is one of the most potent of the hemozoin-producing enzymes. However, the reaction mechanisms of HDP are poorly understood. We identified the active site residues in HDP using a combination of Hz formation assay and spectroscopic characterization of mutant proteins. Replacement of the critical histidine residues His122, His172, His175, and His197 resulted in a reduction in the Hz formation activity to approximately 50% of the wild-type protein. Spectroscopic characterization of histidine-substituted mutants revealed that His122 binds heme and that His172 and His175 form a part of another heme-binding site. Our results show that the histidine residues could be present in the individual active sites and could be ligated to each heme. The interaction between heme and the histidine residues would serve as a molecular tether, allowing the proper positioning of two hemes to enable heme dimer formation. The heme dimer would act as a seed for the crystal growth of Hz in P. falciparum.
Collapse
|
36
|
Cupello MP, Souza CF, Menna-Barreto RF, Nogueira NPA, Laranja GAT, Sabino KCC, Coelho MGP, Oliveira MM, Paes MC. Trypanosomatid essential metabolic pathway: new approaches about heme fate in Trypanosoma cruzi. Biochem Biophys Res Commun 2014; 449:216-21. [PMID: 24824181 DOI: 10.1016/j.bbrc.2014.05.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Accepted: 05/02/2014] [Indexed: 11/27/2022]
Abstract
Trypanosoma cruzi, the causal agent of Chagas disease, has a complex life cycle and depends on hosts for its nutritional needs. Our group has investigated heme (Fe-protoporphyrin IX) internalization and the effects on parasite growth, following the fate of this porphyrin in the parasite. Here, we show that epimastigotes cultivated with heme yielded the compounds α-meso-hydroxyheme, verdoheme and biliverdin (as determined by HPLC), suggesting an active heme degradation pathway in this parasite. Furthermore, through immunoprecipitation and immunoblotting assays of epimastigote extracts, we observed recognition by an antibody against mammalian HO-1. We also detected the localization of the HO-1-like protein in the parasite using immunocytochemistry, with antibody staining primarily in the cytoplasm. Although HO has not been described in the parasite's genome, our results offer new insights into heme metabolism in T. cruzi, revealing potential future therapeutic targets.
Collapse
Affiliation(s)
- M P Cupello
- Laboratório de Interação Tripanossomatídeos e Vetores, Departamento de Bioquímica, Instituto de Biologia Roberto Alcântara Gomes (IBRAG), UERJ, Rio de Janeiro, Brazil
| | - C F Souza
- Laboratório de Interação Tripanossomatídeos e Vetores, Departamento de Bioquímica, Instituto de Biologia Roberto Alcântara Gomes (IBRAG), UERJ, Rio de Janeiro, Brazil
| | - R F Menna-Barreto
- Laboratório de Biologia Celular, IOC, FIOCRUZ, Rio de Janeiro, Brazil
| | - N P A Nogueira
- Laboratório de Interação Tripanossomatídeos e Vetores, Departamento de Bioquímica, Instituto de Biologia Roberto Alcântara Gomes (IBRAG), UERJ, Rio de Janeiro, Brazil
| | - G A T Laranja
- Laboratório de Interação Tripanossomatídeos e Vetores, Departamento de Bioquímica, Instituto de Biologia Roberto Alcântara Gomes (IBRAG), UERJ, Rio de Janeiro, Brazil
| | - K C C Sabino
- Laboratório de Imunologia Aplicada à Bioquímica e Produtos Naturais, Departamento de Bioquímica, IBRAG, UERJ, Rio de Janeiro, Brazil
| | - M G P Coelho
- Laboratório de Imunologia Aplicada à Bioquímica e Produtos Naturais, Departamento de Bioquímica, IBRAG, UERJ, Rio de Janeiro, Brazil
| | - M M Oliveira
- Instituto de Biofísica Carlos Chagas Filho (IBCCF), UFRJ, Rio de Janeiro, Brazil
| | - M C Paes
- Laboratório de Interação Tripanossomatídeos e Vetores, Departamento de Bioquímica, Instituto de Biologia Roberto Alcântara Gomes (IBRAG), UERJ, Rio de Janeiro, Brazil; Instituto Nacional de Ciência e Tecnologia - Entomologia Molecular (INCT-EM), Brazil.
| |
Collapse
|
37
|
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
|
38
|
Abstract
Recent initiatives to develop more effective and affordable drugs, controlling mosquitoes and development of a preventative vaccine have been launched with the goal of completely eradicating malaria. To this end, Novartis (Surrey, UK) and GlaxoSmithKline (Middlesex, UK) screened their chemical libraries of approximately two million small molecules for antimalarial properties, which resulted in a set of over 20,000 'highly druggable' initial hits. Efforts in academia are centered on specific pathway targets. One such high-throughput screening effort has been focused on hemozoin formation, a unique heme detoxification pathway found in the malaria parasite. This review discusses the current approaches and limitations of high-throughput screening discovery of hemozoin inhibitors. In the future, new methods must be developed to validate the mechanism of action of these hit compounds within the parasite.
Collapse
|
39
|
Walcourt A, Kurantsin-Mills J, Kwagyan J, Adenuga BB, Kalinowski DS, Lovejoy DB, Lane DJR, Richardson DR. Anti-plasmodial activity of aroylhydrazone and thiosemicarbazone iron chelators: effect on erythrocyte membrane integrity, parasite development and the intracellular labile iron pool. J Inorg Biochem 2013; 129:43-51. [PMID: 24028863 PMCID: PMC3838870 DOI: 10.1016/j.jinorgbio.2013.08.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Revised: 08/19/2013] [Accepted: 08/19/2013] [Indexed: 01/10/2023]
Abstract
Iron chelators inhibit the growth of the malaria parasite, Plasmodium falciparum, in culture and in animal and human studies. We previously reported the anti-plasmodial activity of the chelators, 2-hydroxy-1-naphthylaldehyde isonicotinoyl hydrazone (311), 2-hydroxy-1-naphthylaldehyde 4-methyl-3-thiosemicarbazone (N4mT), and 2-hydroxy-1-naphthylaldehyde 4-phenyl-3-thiosemicarbazone (N4pT). In fact, these ligands showed greater growth inhibition of chloroquine-sensitive (3D7) and chloroquine-resistant (7G8) strains of P. falciparum in culture compared to desferrioxamine (DFO). The present study examined the effects of 311, N4mT and N4pT on erythrocyte membrane integrity and asexual parasite development. While the characteristic biconcave disk shape of the erythrocytes was unaffected, the chelators caused very slight hemolysis at IC50 values that inhibited parasite growth. The chelators 311, N4mT and N4pT affected all stages of the intra-erythrocytic development cycle (IDC) of P. falciparum in culture. However, while these ligands primarily affected the ring-stage, DFO inhibited primarily trophozoite and schizont-stages. Ring, trophozoite and schizont-stages of the IDC were inhibited by significantly lower concentrations of 311, N4mT, and N4pT (IC50=4.45±1.70, 10.30±4.40, and 3.64±2.00μM, respectively) than DFO (IC50=23.43±3.40μM). Complexation of 311, N4mT and N4pT with iron reduced their anti-plasmodial activity. Estimation of the intracellular labile iron pool (LIP) in erythrocytes showed that the chelation efficacy of 311, N4mT and N4pT corresponded to their anti-plasmodial activities, suggesting that the LIP may be a potential source of non-heme iron for parasite metabolism within the erythrocyte. This study has implications for malaria chemotherapy that specifically disrupts parasite iron utilization.
Collapse
Affiliation(s)
- Asikiya Walcourt
- Department of Physiology and Biophysics, Howard University College of Medicine, Washington, DC, 20059
| | - Joseph Kurantsin-Mills
- Department of Physiology and Biophysics, Howard University College of Medicine, Washington, DC, 20059
- Departments of Medicine, Pharmacology and Physiology, The George Washington University Medical Center, Washington, DC, 20037
| | - John Kwagyan
- Design, Biostatistics & Population Studies, Center for Clinical & Translation Science and Department of Community and Family Medicine, Howard University College of Medicine, Washington, DC,20059
| | | | - Danuta S. Kalinowski
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, Blackburn Building (D06), University of Sydney, Sydney, New South Wales, 2006 Australia
| | - David B. Lovejoy
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, Blackburn Building (D06), University of Sydney, Sydney, New South Wales, 2006 Australia
| | - Darius J. R. Lane
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, Blackburn Building (D06), University of Sydney, Sydney, New South Wales, 2006 Australia
| | - Des R. Richardson
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, Blackburn Building (D06), University of Sydney, Sydney, New South Wales, 2006 Australia
| |
Collapse
|
40
|
Komatsuya K, Hata M, Balogun EO, Hikosaka K, Suzuki S, Takahashi K, Tanaka T, Nakajima M, Ogura SI, Sato S, Kita K. Synergy of ferrous ion on 5-aminolevulinic acid-mediated growth inhibition of Plasmodium falciparum. J Biochem 2013; 154:501-4. [PMID: 24158489 DOI: 10.1093/jb/mvt096] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Haem biosynthesis appeared to be a target of malaria therapy because 5-aminolevulinic acid (ALA), a haem biosynthesis starting material, with light exposure or a high amount of ALA alone reduced Plasmodium falciparum growth to undetectable level. However, the administration of a high dose of ALA is unrealistic for clinical therapy. We found that Fe(2+) enhanced P. falciparum-killing potency of ALA and significantly inhibited the parasite growth. The intermediates of haem biosynthesis localized to the parasite organelles, and coproporphyrin III was the most accumulated intermediate. These novel findings may lead to development of a new anti-malarial drug using ALA and Fe(2+).
Collapse
Affiliation(s)
- Keisuke Komatsuya
- Department of Biomedical Chemistry, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033; Department of Applied Biology, Graduate School of Science and Technology, Kyoto Institute of Technology, Sakyo-ku, Kyoto 606-8585, Japan; Department of Biochemistry, Ahmadu Bello University, Zaria 2222, Nigeria; SBI Pharmaceuticals Co, LTD, Izumi Garden Tower 20F, 1-6-1, Roppongi, Minato-ku, Tokyo; Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, 4259-B102 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan; and Division of Parasitology, MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Heme-binding properties of heme detoxification protein from Plasmodium falciparum. Biochem Biophys Res Commun 2013; 439:477-80. [PMID: 24025682 DOI: 10.1016/j.bbrc.2013.08.100] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 08/30/2013] [Indexed: 12/15/2022]
Abstract
The heme detoxification protein of the malaria parasite Plasmodium falciparum is involved in the formation of hemozoin, an insoluble crystalline form of heme. Although the disruption of hemozoin formation is the most widely used strategy for controlling the malaria parasite, the heme-binding properties of heme detoxification protein are poorly characterized. In this study, we established a method for the expression and purification of the non-tagged protein and characterized heme-binding properties. The spectroscopic features of non-tagged protein differ from those of the His-tagged protein, suggesting that the artificial tag interferes with the properties of the recombinant protein. The purified recombinant non-tagged heme detoxification protein had two heme-binding sites and exhibited a spectrum typical of heme proteins. A mechanism for hemozoin formation is proposed.
Collapse
|
42
|
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
|
43
|
Nagaraj VA, Sundaram B, Varadarajan NM, Subramani PA, Kalappa DM, Ghosh SK, Padmanaban G. Malaria parasite-synthesized heme is essential in the mosquito and liver stages and complements host heme in the blood stages of infection. PLoS Pathog 2013; 9:e1003522. [PMID: 23935500 PMCID: PMC3731253 DOI: 10.1371/journal.ppat.1003522] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2013] [Accepted: 06/10/2013] [Indexed: 01/21/2023] Open
Abstract
Heme metabolism is central to malaria parasite biology. The parasite acquires heme from host hemoglobin in the intraerythrocytic stages and stores it as hemozoin to prevent free heme toxicity. The parasite can also synthesize heme de novo, and all the enzymes in the pathway are characterized. To study the role of the dual heme sources in malaria parasite growth and development, we knocked out the first enzyme, δ-aminolevulinate synthase (ALAS), and the last enzyme, ferrochelatase (FC), in the heme-biosynthetic pathway of Plasmodium berghei (Pb). The wild-type and knockout (KO) parasites had similar intraerythrocytic growth patterns in mice. We carried out in vitro radiolabeling of heme in Pb-infected mouse reticulocytes and Plasmodium falciparum-infected human RBCs using [4-14C] aminolevulinic acid (ALA). We found that the parasites incorporated both host hemoglobin-heme and parasite-synthesized heme into hemozoin and mitochondrial cytochromes. The similar fates of the two heme sources suggest that they may serve as backup mechanisms to provide heme in the intraerythrocytic stages. Nevertheless, the de novo pathway is absolutely essential for parasite development in the mosquito and liver stages. PbKO parasites formed drastically reduced oocysts and did not form sporozoites in the salivary glands. Oocyst production in PbALASKO parasites recovered when mosquitoes received an ALA supplement. PbALASKO sporozoites could infect mice only when the mice received an ALA supplement. Our results indicate the potential for new therapeutic interventions targeting the heme-biosynthetic pathway in the parasite during the mosquito and liver stages. We demonstrated about two decades ago that the malaria parasite could make heme on its own, although it imports heme from red blood cell hemoglobin during the blood stages of infection. We investigated the role of parasite-synthesized heme in all stages of parasite growth by knocking out two genes in the heme-biosynthetic pathway of Plasmodium berghei that infects mice. We found that the parasite-synthesized heme complements the function of hemoglobin-heme during the blood stages. The parasite-synthesized heme appears to be a backup mechanism. The parasite incorporates both sources of heme into hemozoin, a detoxification product, and into mitochondrial cytochromes. The parasite-synthesized heme is, however, absolutely essential for parasite growth during the mosquito and liver stages. We restored the sporozoite formation and liver-stage development of the knockout parasites by providing the missing metabolite. Thus, the heme-biosynthetic pathway could be a target for antimalarial therapies in the mosquito and liver stages of infection. The knockout parasite could also be tested for its potential as a genetically attenuated sporozoite vaccine.
Collapse
|
44
|
Gorka AP, de Dios A, Roepe PD. Quinoline drug-heme interactions and implications for antimalarial cytostatic versus cytocidal activities. J Med Chem 2013; 56:5231-46. [PMID: 23586757 DOI: 10.1021/jm400282d] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Historically, the most successful molecular target for antimalarial drugs has been heme biomineralization within the malarial parasite digestive vacuole. Heme released from catabolized host red blood cell hemoglobin is toxic, so malarial parasites crystallize heme to nontoxic hemozoin. For years it has been accepted that a number of effective quinoline antimalarial drugs (e.g., chloroquine, quinine, amodiaquine) function by preventing hemozoin crystallization. However, recent studies over the past decade have revealed a surprising molecular diversity in quinoline-heme molecular interactions. This diversity shows that even closely related quinoline drugs may have quite different molecular pharmacology. This paper reviews the molecular diversity and highlights important implications for understanding quinoline antimalarial drug resistance and for future drug design.
Collapse
Affiliation(s)
- Alexander P Gorka
- Department of Chemistry, Department of Biochemistry, Cellular, and Molecular Biology, and Center for Infectious Diseases, Georgetown University , 37th and O Streets, NW, Washington, D.C. 20057, United States
| | | | | |
Collapse
|
45
|
van Santen S, de Mast Q, Swinkels DW, van der Ven AJAM. The iron link between malaria and invasive non-typhoid Salmonella infections. Trends Parasitol 2013; 29:220-7. [PMID: 23601932 DOI: 10.1016/j.pt.2013.03.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Revised: 03/11/2013] [Accepted: 03/12/2013] [Indexed: 02/06/2023]
Abstract
Epidemiological studies have demonstrated an association between malaria and invasive non-typhoid Salmonella (NTS) infections, especially in children. We explore the role of iron as a possible cofactor in this association. Malarial disease, among others, is associated with enhanced erythrophagocytosis and inflammation, which increases the iron content of macrophages and thereby also the survival of Salmonella spp. within macrophages. Whether iron supplementation programs augment the risk of invasive NTS infections in malaria-endemic regions is an important global health issue that still needs to be determined.
Collapse
Affiliation(s)
- Susanne van Santen
- Department of General Internal Medicine, Nijmegen Institute for International Health (456), Radboud University Medical Center, Geert Grooteplein Zuid 8, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| | | | | | | |
Collapse
|
46
|
Torda AE, Groves MR, Wedderhoff I, Ortiz de Orué Lucana D. Elucidation of haem-binding sites in the actinobacterial protein HbpS. FEMS Microbiol Lett 2013; 342:106-12. [PMID: 23373615 DOI: 10.1111/1574-6968.12093] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Revised: 01/28/2013] [Accepted: 01/28/2013] [Indexed: 11/28/2022] Open
Abstract
The extracellular haem-binding protein from Streptomyces reticuli (HbpS) has been shown to be involved in redox sensing and to bind haem. However, the residues involved in haem coordination are unknown. Structural alignments to distantly related haem-binding proteins from Mycobacterium tuberculosis were used to identify a candidate haem-coordinating residue, and site-directed mutagenesis with UV/Vis spectroscopy was used to assess haem binding in vivo and in vitro. We present strong evidence that HbpS belongs to the small set of proteins, which do not use histidine to coordinate the metal in the haem group. Further spectroscopic evidence strongly indicates that threonine 113 is actively involved in coordination of haem. Subsequent protein/haem titration experiments show a 1 : 2, protein/haem stoichiometry. We also present data showing the degradation of haem by HbpS in vivo. Because HbpS is conserved in many Actinobacteria, the presented results are applicable to related species.
Collapse
Affiliation(s)
- Andrew E Torda
- Centre for Bioinformatics, Hamburg University, Hamburg, Germany
| | | | | | | |
Collapse
|
47
|
Clark M, Fisher NC, Kasthuri R, Cerami Hand C. Parasite maturation and host serum iron influence the labile iron pool of erythrocyte stage Plasmodium falciparum. Br J Haematol 2013; 161:262-9. [PMID: 23398516 DOI: 10.1111/bjh.12234] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Accepted: 12/20/2012] [Indexed: 12/30/2022]
Abstract
Iron is a critical and tightly regulated nutrient for both the malaria parasite and its human host. The importance of the relationship between host iron and the parasite has been underscored recently by studies showing that host iron supplementation may increase the risk of falciparum malaria. It is unclear what host iron sources the parasite is able to access. We developed a flow cytometry-based method for measuring the labile iron pool (LIP) of parasitized erythrocytes using the nucleic acid dye STYO 61 and the iron sensitive dye, calcein acetoxymethyl ester (CA-AM). This new approach enabled us to measure the LIP of P. falciparum through the course of its erythrocytic life cycle and in response to the addition of host serum iron sources. We found that the LIP increases as the malaria parasite develops from early ring to late schizont stage, and that the addition of either transferrin or ferric citrate to culture media increases the LIP of trophozoites. Our method for detecting the LIP within malaria parasitized RBCs provides evidence that the parasite is able to access serum iron sources as part of the host vs. parasite arms race for iron.
Collapse
Affiliation(s)
- Martha Clark
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, USA, United States
| | | | | | | |
Collapse
|