1
|
Iacobucci I, Monaco V, Hovasse A, Dupouy B, Keumoe R, Cichocki B, Elhabiri M, Meunier B, Strub JM, Monti M, Cianférani S, Blandin SA, Schaeffer-Reiss C, Davioud-Charvet E. Proteomic Profiling of Antimalarial Plasmodione Using 3-Benz(o)ylmenadione Affinity-Based Probes. Chembiochem 2024; 25:e202400187. [PMID: 38639212 DOI: 10.1002/cbic.202400187] [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: 02/29/2024] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 04/20/2024]
Abstract
Understanding the mechanisms of drug action in malarial parasites is crucial for the development of new drugs to combat infection and to counteract drug resistance. Proteomics is a widely used approach to study host-pathogen systems and to identify drug protein targets. Plasmodione is an antiplasmodial early-lead drug exerting potent activities against young asexual and sexual blood stages in vitro with low toxicity to host cells. To elucidate its molecular mechanisms, an affinity-based protein profiling (AfBPP) approach was applied to yeast and P. falciparum proteomes. New (pro-) AfBPP probes based on the 3-benz(o)yl-6-fluoro-menadione scaffold were synthesized. With optimized conditions of both photoaffinity labeling and click reaction steps, the AfBPP protocol was then applied to a yeast proteome, yielding 11 putative drug-protein targets. Among these, we found four proteins associated with oxidoreductase activities, the hypothesized type of targets for plasmodione and its metabolites, and other proteins associated with the mitochondria. In Plasmodium parasites, the MS analysis revealed 44 potential plasmodione targets that need to be validated in further studies. Finally, the localization of a 3-benzyl-6-fluoromenadione AfBPP probe was studied in the subcellular structures of the parasite at the trophozoite stage.
Collapse
Affiliation(s)
- Ilaria Iacobucci
- Laboratoire d'Innovation Moléculaire et Applications (LIMA), Team Bio(IN)organic & Medicinal Chemistry, UMR7042 CNRS-Université de Strasbourg-Université Haute-Alsace, European School of Chemistry, Polymers and Materials (ECPM), 25, rue Becquerel, 25, rue Becquerel, F-67087, Strasbourg, France
- Laboratoire de Spectrométrie de Masse BioOrganique, IPHC UMR 7178 CNRS, Université de Strasbourg, 67087, Strasbourg, France
- Infrastructure Nationale de Protéomique ProFI - FR2048, F-67087, Strasbourg, France
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario di Monte Sant' Angelo, Via Cintia 26, I-80126, Napoli, Italy
| | - Vittoria Monaco
- Laboratoire d'Innovation Moléculaire et Applications (LIMA), Team Bio(IN)organic & Medicinal Chemistry, UMR7042 CNRS-Université de Strasbourg-Université Haute-Alsace, European School of Chemistry, Polymers and Materials (ECPM), 25, rue Becquerel, 25, rue Becquerel, F-67087, Strasbourg, France
- Laboratoire de Spectrométrie de Masse BioOrganique, IPHC UMR 7178 CNRS, Université de Strasbourg, 67087, Strasbourg, France
- Infrastructure Nationale de Protéomique ProFI - FR2048, F-67087, Strasbourg, France
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario di Monte Sant' Angelo, Via Cintia 26, I-80126, Napoli, Italy
| | - Agnès Hovasse
- Laboratoire de Spectrométrie de Masse BioOrganique, IPHC UMR 7178 CNRS, Université de Strasbourg, 67087, Strasbourg, France
- Infrastructure Nationale de Protéomique ProFI - FR2048, F-67087, Strasbourg, France
| | - Baptiste Dupouy
- Laboratoire d'Innovation Moléculaire et Applications (LIMA), Team Bio(IN)organic & Medicinal Chemistry, UMR7042 CNRS-Université de Strasbourg-Université Haute-Alsace, European School of Chemistry, Polymers and Materials (ECPM), 25, rue Becquerel, 25, rue Becquerel, F-67087, Strasbourg, France
| | - Rodrigue Keumoe
- Institut de Biologie Moléculaire et Cellulaire, INSERM U1257 - CNRS UPR9022 - Université de Strasbourg, 2, Allée Konrad Roentgen, -67084, Strasbourg, France
| | - Bogdan Cichocki
- Laboratoire d'Innovation Moléculaire et Applications (LIMA), Team Bio(IN)organic & Medicinal Chemistry, UMR7042 CNRS-Université de Strasbourg-Université Haute-Alsace, European School of Chemistry, Polymers and Materials (ECPM), 25, rue Becquerel, 25, rue Becquerel, F-67087, Strasbourg, France
| | - Mourad Elhabiri
- Laboratoire d'Innovation Moléculaire et Applications (LIMA), Team Bio(IN)organic & Medicinal Chemistry, UMR7042 CNRS-Université de Strasbourg-Université Haute-Alsace, European School of Chemistry, Polymers and Materials (ECPM), 25, rue Becquerel, 25, rue Becquerel, F-67087, Strasbourg, France
| | - Brigitte Meunier
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 91198, Gif-Sur-Yvette Cedex, France
| | - Jean-Marc Strub
- Laboratoire de Spectrométrie de Masse BioOrganique, IPHC UMR 7178 CNRS, Université de Strasbourg, 67087, Strasbourg, France
- Infrastructure Nationale de Protéomique ProFI - FR2048, F-67087, Strasbourg, France
| | - Maria Monti
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario di Monte Sant' Angelo, Via Cintia 26, I-80126, Napoli, Italy
| | - Sarah Cianférani
- Laboratoire de Spectrométrie de Masse BioOrganique, IPHC UMR 7178 CNRS, Université de Strasbourg, 67087, Strasbourg, France
- Infrastructure Nationale de Protéomique ProFI - FR2048, F-67087, Strasbourg, France
| | - Stéphanie A Blandin
- Institut de Biologie Moléculaire et Cellulaire, INSERM U1257 - CNRS UPR9022 - Université de Strasbourg, 2, Allée Konrad Roentgen, -67084, Strasbourg, France
| | - Christine Schaeffer-Reiss
- Laboratoire de Spectrométrie de Masse BioOrganique, IPHC UMR 7178 CNRS, Université de Strasbourg, 67087, Strasbourg, France
- Infrastructure Nationale de Protéomique ProFI - FR2048, F-67087, Strasbourg, France
| | - Elisabeth Davioud-Charvet
- Laboratoire d'Innovation Moléculaire et Applications (LIMA), Team Bio(IN)organic & Medicinal Chemistry, UMR7042 CNRS-Université de Strasbourg-Université Haute-Alsace, European School of Chemistry, Polymers and Materials (ECPM), 25, rue Becquerel, 25, rue Becquerel, F-67087, Strasbourg, France
| |
Collapse
|
2
|
Springer E, Heimsch KC, Rahlfs S, Becker K, Przyborski JM. Real-time measurements of ATP dynamics via ATeams in Plasmodium falciparum reveal drug-class-specific response patterns. Antimicrob Agents Chemother 2024; 68:e0169023. [PMID: 38501806 PMCID: PMC11064498 DOI: 10.1128/aac.01690-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 02/26/2024] [Indexed: 03/20/2024] Open
Abstract
Malaria tropica, caused by the parasite Plasmodium falciparum (P. falciparum), remains one of the greatest public health burdens for humankind. Due to its pivotal role in parasite survival, the energy metabolism of P. falciparum is an interesting target for drug design. To this end, analysis of the central metabolite adenosine triphosphate (ATP) is of great interest. So far, only cell-disruptive or intensiometric ATP assays have been available in this system, with various drawbacks for mechanistic interpretation and partly inconsistent results. To address this, we have established fluorescent probes, based on Förster resonance energy transfer (FRET) and known as ATeam, for use in blood-stage parasites. ATeams are capable of measuring MgATP2- levels in a ratiometric manner, thereby facilitating in cellulo measurements of ATP dynamics in real-time using fluorescence microscopy and plate reader detection and overcoming many of the obstacles of established ATP analysis methods. Additionally, we established a superfolder variant of the ratiometric pH sensor pHluorin (sfpHluorin) in P. falciparum to monitor pH homeostasis and control for pH fluctuations, which may affect ATeam measurements. We characterized recombinant ATeam and sfpHluorin protein in vitro and stably integrated the sensors into the genome of the P. falciparum NF54attB cell line. Using these new tools, we found distinct sensor response patterns caused by several different drug classes. Arylamino alcohols increased and redox cyclers decreased ATP; doxycycline caused first-cycle cytosol alkalization; and 4-aminoquinolines caused aberrant proteolysis. Our results open up a completely new perspective on drugs' mode of action, with possible implications for target identification and drug development.
Collapse
Affiliation(s)
- Eric Springer
- Biochemistry and Molecular Biology, Interdisciplinary Research Center, Justus Liebig University, Giessen, Germany
| | - Kim C. Heimsch
- Biochemistry and Molecular Biology, Interdisciplinary Research Center, Justus Liebig University, Giessen, Germany
| | - Stefan Rahlfs
- Biochemistry and Molecular Biology, Interdisciplinary Research Center, Justus Liebig University, Giessen, Germany
| | - Katja Becker
- Biochemistry and Molecular Biology, Interdisciplinary Research Center, Justus Liebig University, Giessen, Germany
| | - Jude M. Przyborski
- Biochemistry and Molecular Biology, Interdisciplinary Research Center, Justus Liebig University, Giessen, Germany
| |
Collapse
|
3
|
Collins JE, Lee JW, Rocamora F, Saggu GS, Wendt KL, Pasaje CFA, Smick S, Santos NM, Paes R, Jiang T, Mittal N, Luth MR, Chin T, Chang H, McLellan JL, Morales-Hernandez B, Hanson KK, Niles JC, Desai SA, Winzeler EA, Cichewicz RH, Chakrabarti D. Antiplasmodial peptaibols act through membrane directed mechanisms. Cell Chem Biol 2024; 31:312-325.e9. [PMID: 37995692 PMCID: PMC10923054 DOI: 10.1016/j.chembiol.2023.10.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 08/29/2023] [Accepted: 10/27/2023] [Indexed: 11/25/2023]
Abstract
Our previous study identified 52 antiplasmodial peptaibols isolated from fungi. To understand their antiplasmodial mechanism of action, we conducted phenotypic assays, assessed the in vitro evolution of resistance, and performed a transcriptome analysis of the most potent peptaibol, HZ NPDG-I. HZ NPDG-I and 2 additional peptaibols were compared for their killing action and stage dependency, each showing a loss of digestive vacuole (DV) content via ultrastructural analysis. HZ NPDG-I demonstrated a stepwise increase in DV pH, impaired DV membrane permeability, and the ability to form ion channels upon reconstitution in planar membranes. This compound showed no signs of cross resistance to targets of current clinical candidates, and 3 independent lines evolved to resist HZ NPDG-I acquired nonsynonymous changes in the P. falciparum multidrug resistance transporter, pfmdr1. Conditional knockdown of PfMDR1 showed varying effects to other peptaibol analogs, suggesting differing sensitivity.
Collapse
Affiliation(s)
- Jennifer E Collins
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL 32826, USA
| | - Jin Woo Lee
- Department of Chemistry and Biochemistry, Institute for Natural Products Applications & Research Technologies, University of Oklahoma, Norman OK 73019, USA
| | - Frances Rocamora
- Division of Host-Microbe Systems & Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA
| | - Gagandeep S Saggu
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, NIH, Rockville, MD 20852, USA
| | - Karen L Wendt
- Department of Chemistry and Biochemistry, Institute for Natural Products Applications & Research Technologies, University of Oklahoma, Norman OK 73019, USA
| | - Charisse Flerida A Pasaje
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Sebastian Smick
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Natalia Mojica Santos
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL 32826, USA
| | - Raphaella Paes
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL 32826, USA
| | - Tiantian Jiang
- Division of Host-Microbe Systems & Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA
| | - Nimisha Mittal
- Division of Host-Microbe Systems & Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA
| | - Madeline R Luth
- Division of Host-Microbe Systems & Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA
| | - Taylor Chin
- Division of Host-Microbe Systems & Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA
| | - Howard Chang
- Division of Host-Microbe Systems & Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA
| | - James L McLellan
- Department of Molecular Microbiology and Immunology and South Texas Center for Emerging Infectious Diseases, University of Texas San Antonio, San Antonio, TX 78249, USA
| | - Beatriz Morales-Hernandez
- Department of Molecular Microbiology and Immunology and South Texas Center for Emerging Infectious Diseases, University of Texas San Antonio, San Antonio, TX 78249, USA
| | - Kirsten K Hanson
- Department of Molecular Microbiology and Immunology and South Texas Center for Emerging Infectious Diseases, University of Texas San Antonio, San Antonio, TX 78249, USA
| | - Jacquin C Niles
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Sanjay A Desai
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, NIH, Rockville, MD 20852, USA
| | - Elizabeth A Winzeler
- Division of Host-Microbe Systems & Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA.
| | - Robert H Cichewicz
- Department of Chemistry and Biochemistry, Institute for Natural Products Applications & Research Technologies, University of Oklahoma, Norman OK 73019, USA.
| | - Debopam Chakrabarti
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL 32826, USA.
| |
Collapse
|
4
|
Alder A, Sanchez CP, Russell MRG, Collinson LM, Lanzer M, Blackman MJ, Gilberger TW, Matz JM. The role of Plasmodium V-ATPase in vacuolar physiology and antimalarial drug uptake. Proc Natl Acad Sci U S A 2023; 120:e2306420120. [PMID: 37463201 PMCID: PMC10372686 DOI: 10.1073/pnas.2306420120] [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: 04/24/2023] [Accepted: 06/20/2023] [Indexed: 07/20/2023] Open
Abstract
To ensure their survival in the human bloodstream, malaria parasites degrade up to 80% of the host erythrocyte hemoglobin in an acidified digestive vacuole. Here, we combine conditional reverse genetics and quantitative imaging approaches to demonstrate that the human malaria pathogen Plasmodium falciparum employs a heteromultimeric V-ATPase complex to acidify the digestive vacuole matrix, which is essential for intravacuolar hemoglobin release, heme detoxification, and parasite survival. We reveal an additional function of the membrane-embedded V-ATPase subunits in regulating morphogenesis of the digestive vacuole independent of proton translocation. We further show that intravacuolar accumulation of antimalarial chemotherapeutics is surprisingly resilient to severe deacidification of the vacuole and that modulation of V-ATPase activity does not affect parasite sensitivity toward these drugs.
Collapse
Affiliation(s)
- Arne Alder
- Cell Biology of Human Parasites Group, Centre for Structural Systems Biology, Hamburg22607, Germany
- Cellular Parasitology Department, Bernhard Nocht Institute for Tropical Medicine, Hamburg20359, Germany
- Department of Biology, University of Hamburg, Hamburg20146, Germany
| | - Cecilia P. Sanchez
- Center of Infectious Diseases, Parasitology, Heidelberg University Hospital, Heidelberg69120, Germany
| | - Matthew R. G. Russell
- Electron Microscopy Science Technology Platform, The Francis Crick Institute, LondonNW1 1AT, United Kingdom
- Centre for Ultrastructural Imaging, King’s College London, LondonSE1 1UL, United Kingdom
| | - Lucy M. Collinson
- Electron Microscopy Science Technology Platform, The Francis Crick Institute, LondonNW1 1AT, United Kingdom
| | - Michael Lanzer
- Center of Infectious Diseases, Parasitology, Heidelberg University Hospital, Heidelberg69120, Germany
| | - Michael J. Blackman
- Malaria Biochemistry Laboratory, The Francis Crick Institute, LondonNW1 1AT, United Kingdom
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, LondonWC1E 7HT, United Kingdom
| | - Tim-Wolf Gilberger
- Cell Biology of Human Parasites Group, Centre for Structural Systems Biology, Hamburg22607, Germany
- Cellular Parasitology Department, Bernhard Nocht Institute for Tropical Medicine, Hamburg20359, Germany
- Department of Biology, University of Hamburg, Hamburg20146, Germany
| | - Joachim M. Matz
- Cell Biology of Human Parasites Group, Centre for Structural Systems Biology, Hamburg22607, Germany
- Cellular Parasitology Department, Bernhard Nocht Institute for Tropical Medicine, Hamburg20359, Germany
- Malaria Biochemistry Laboratory, The Francis Crick Institute, LondonNW1 1AT, United Kingdom
| |
Collapse
|
5
|
Markus MB. Putative Contribution of 8-Aminoquinolines to Preventing Recrudescence of Malaria. Trop Med Infect Dis 2023; 8:278. [PMID: 37235326 PMCID: PMC10223033 DOI: 10.3390/tropicalmed8050278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/07/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
Enhanced therapeutic efficacy achieved in treating Plasmodium vivax malaria with an 8-aminoquinoline (8-AQ) drug such as primaquine (PQ) together with a partner drug such as chloroquine (CQ) is usually explained as CQ inhibiting asexual parasites in the bloodstream and PQ acting against liver stages. However, PQ's contribution, if any, to inactivating non-circulating, extra-hepatic asexual forms, which make up the bulk of the parasite biomass in chronic P. vivax infections, remains unclear. In this opinion article, I suggest that, considering its newly described mode of action, PQ might be doing something of which we are currently unaware.
Collapse
Affiliation(s)
- Miles B. Markus
- Wits Research Institute for Malaria, School of Pathology, Faculty of Health Sciences, University of Witwatersrand, Johannesburg 2193, South Africa;
- School of Animal, Plant and Environmental Sciences, Faculty of Science, University of Witwatersrand, Johannesburg 2001, South Africa
| |
Collapse
|
6
|
Coronado LM, Stoute JA, Nadovich CT, Cheng J, Correa R, Chaw K, González G, Zambrano M, Gittens RA, Agrawal DK, Jemison WD, Donado Morcillo CA, Spadafora C. Microwaves can kill malaria parasites non-thermally. Front Cell Infect Microbiol 2023; 13:955134. [PMID: 36816585 PMCID: PMC9932958 DOI: 10.3389/fcimb.2023.955134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 01/17/2023] [Indexed: 02/05/2023] Open
Abstract
Malaria, which infected more than 240 million people and killed around six hundred thousand only in 2021, has reclaimed territory after the SARS-CoV-2 pandemic. Together with parasite resistance and a not-yet-optimal vaccine, the need for new approaches has become critical. While earlier, limited, studies have suggested that malaria parasites are affected by electromagnetic energy, the outcomes of this affectation vary and there has not been a study that looks into the mechanism of action behind these responses. In this study, through development and implementation of custom applicators for in vitro experimentation, conditions were generated in which microwave energy (MW) killed more than 90% of the parasites, not by a thermal effect but via a MW energy-induced programmed cell death that does not seem to affect mammalian cell lines. Transmission electron microscopy points to the involvement of the haemozoin-containing food vacuole, which becomes destroyed; while several other experimental approaches demonstrate the involvement of calcium signaling pathways in the resulting effects of exposure to MW. Furthermore, parasites were protected from the effects of MW by calcium channel blockers calmodulin and phosphoinositol. The findings presented here offer a molecular insight into the elusive interactions of oscillating electromagnetic fields with P. falciparum, prove that they are not related to temperature, and present an alternative technology to combat this devastating disease.
Collapse
Affiliation(s)
- Lorena M. Coronado
- Biomedical Physics and Engineering Unit, Center of Cellular and Molecular Biology of Diseases (CBCMe), Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Panama City, Panama,Department of Biotechnology, Acharya Nagarjuna University, Guntur, India,Biomedical Physics and Engineering (BiomedφEngine) Group, Panama City, Panama
| | - José A. Stoute
- Department of Medicine, Division of Infectious Diseases and Epidemiology, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Christopher T. Nadovich
- Electrical and Computer Engineering, Lafayette College, Easton, PA, United States,Wallace H. Coulter School of Engineering, Clarkson University, Potsdam, NY, United States
| | - Jiping Cheng
- Department of Material Science and Engineering, Pennsylvania State University, University Park, PA, United States
| | - Ricardo Correa
- Biomedical Physics and Engineering Unit, Center of Cellular and Molecular Biology of Diseases (CBCMe), Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Panama City, Panama,Department of Biotechnology, Acharya Nagarjuna University, Guntur, India,Biomedical Physics and Engineering (BiomedφEngine) Group, Panama City, Panama
| | - Kevin Chaw
- Biomedical Physics and Engineering Unit, Center of Cellular and Molecular Biology of Diseases (CBCMe), Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Panama City, Panama,Biomedical Physics and Engineering (BiomedφEngine) Group, Panama City, Panama,School of Technology and Engineering, Universidad Católica Santa María La Antigua, Panama City, Panama
| | - Guadalupe González
- Biomedical Physics and Engineering (BiomedφEngine) Group, Panama City, Panama,School of Electrical Engineering, Universidad Tecnológica de Panamá, Panama City, Panama
| | - Maytee Zambrano
- Biomedical Physics and Engineering (BiomedφEngine) Group, Panama City, Panama,School of Electrical Engineering, Universidad Tecnológica de Panamá, Panama City, Panama
| | - Rolando A. Gittens
- Biomedical Physics and Engineering Unit, Center of Cellular and Molecular Biology of Diseases (CBCMe), Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Panama City, Panama,Biomedical Physics and Engineering (BiomedφEngine) Group, Panama City, Panama
| | - Dinesh K. Agrawal
- Department of Material Science and Engineering, Pennsylvania State University, University Park, PA, United States
| | - William D. Jemison
- Wallace H. Coulter School of Engineering, Clarkson University, Potsdam, NY, United States
| | - Carlos A. Donado Morcillo
- Biomedical Physics and Engineering Unit, Center of Cellular and Molecular Biology of Diseases (CBCMe), Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Panama City, Panama,Biomedical Physics and Engineering (BiomedφEngine) Group, Panama City, Panama,School of Technology and Engineering, Universidad Católica Santa María La Antigua, Panama City, Panama
| | - Carmenza Spadafora
- Biomedical Physics and Engineering Unit, Center of Cellular and Molecular Biology of Diseases (CBCMe), Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Panama City, Panama,Biomedical Physics and Engineering (BiomedφEngine) Group, Panama City, Panama,*Correspondence: Carmenza Spadafora,
| |
Collapse
|
7
|
Jin X, Zhou M, Chen S, Li D, Cao X, Liu B. Effects of pH alterations on stress- and aging-induced protein phase separation. Cell Mol Life Sci 2022; 79:380. [PMID: 35750966 PMCID: PMC9232405 DOI: 10.1007/s00018-022-04393-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/26/2022] [Accepted: 05/21/2022] [Indexed: 01/18/2023]
Abstract
Upon stress challenges, proteins/RNAs undergo liquid–liquid phase separation (LLPS) to fine-tune cell physiology and metabolism to help cells adapt to adverse environments. The formation of LLPS has been recently linked with intracellular pH, and maintaining proper intracellular pH homeostasis is known to be essential for the survival of organisms. However, organisms are constantly exposed to diverse stresses, which are accompanied by alterations in the intracellular pH. Aging processes and human diseases are also intimately linked with intracellular pH alterations. In this review, we summarize stress-, aging-, and cancer-associated pH changes together with the mechanisms by which cells regulate cytosolic pH homeostasis. How critical cell components undergo LLPS in response to pH alterations is also discussed, along with the functional roles of intracellular pH fluctuation in the regulation of LLPS. Further studies investigating the interplay of pH with other stressors in LLPS regulation and identifying protein responses to different pH levels will provide an in-depth understanding of the mechanisms underlying pH-driven LLPS in cell adaptation. Moreover, deciphering aging and disease-associated pH changes that influence LLPS condensate formation could lead to a deeper understanding of the functional roles of biomolecular condensates in aging and aging-related diseases.
Collapse
Affiliation(s)
- Xuejiao Jin
- State Key Laboratory of Subtropical Silviculture, School of Forestry and Biotechnology, Zhejiang A&F University, Lin'an, Hangzhou, 311300, China
| | - Min Zhou
- State Key Laboratory of Subtropical Silviculture, School of Forestry and Biotechnology, Zhejiang A&F University, Lin'an, Hangzhou, 311300, China
| | - Shuxin Chen
- State Key Laboratory of Subtropical Silviculture, School of Forestry and Biotechnology, Zhejiang A&F University, Lin'an, Hangzhou, 311300, China
| | - Danqi Li
- State Key Laboratory of Subtropical Silviculture, School of Forestry and Biotechnology, Zhejiang A&F University, Lin'an, Hangzhou, 311300, China
| | - Xiuling Cao
- State Key Laboratory of Subtropical Silviculture, School of Forestry and Biotechnology, Zhejiang A&F University, Lin'an, Hangzhou, 311300, China.
| | - Beidong Liu
- State Key Laboratory of Subtropical Silviculture, School of Forestry and Biotechnology, Zhejiang A&F University, Lin'an, Hangzhou, 311300, China. .,Department of Chemistry and Molecular Biology, University of Gothenburg, Medicinaregatan 9C, 413 90, Goteborg, Sweden. .,Center for Large-Scale Cell-Based Screening, Faculty of Science, University of Gothenburg, Medicinaregatan 9C, 413 90, Goteborg, Sweden.
| |
Collapse
|
8
|
Gutierrez JI, Brittingham GP, Karadeniz YB, Tran KD, Dutta A, Holehouse AS, Peterson CL, Holt LJ. SWI/SNF senses carbon starvation with a pH-sensitive low complexity sequence. eLife 2022; 11:70344. [PMID: 35129437 PMCID: PMC8890752 DOI: 10.7554/elife.70344] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 02/06/2022] [Indexed: 11/16/2022] Open
Abstract
It is increasingly appreciated that intracellular pH changes are important biological signals. This motivates the elucidation of molecular mechanisms of pH sensing. We determined that a nucleocytoplasmic pH oscillation was required for the transcriptional response to carbon starvation in Saccharomyces cerevisiae. The SWI/SNF chromatin remodeling complex is a key mediator of this transcriptional response. A glutamine-rich low-complexity domain (QLC) in the SNF5 subunit of this complex, and histidines within this sequence, was required for efficient transcriptional reprogramming. Furthermore, the SNF5 QLC mediated pH-dependent recruitment of SWI/SNF to an acidic transcription factor in a reconstituted nucleosome remodeling assay. Simulations showed that protonation of histidines within the SNF5 QLC leads to conformational expansion, providing a potential biophysical mechanism for regulation of these interactions. Together, our results indicate that pH changes are a second messenger for transcriptional reprogramming during carbon starvation and that the SNF5 QLC acts as a pH sensor.
Collapse
Affiliation(s)
| | - Gregory P Brittingham
- Institute for Systems Genetics, New York University Langone Health, New York, United States
| | - Yonca B Karadeniz
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, United States
| | - Kathleen D Tran
- Department of Cell and Molecular Biology, University of Rhode Island, South Kingstown, United States
| | - Arnob Dutta
- Department of Cell and Molecular Biology, University of Rhode Island, South Kingstown, United States
| | - Alex S Holehouse
- Department of Biochemistry and Molecular Biophysics, Washington University in St. Louis, St Louis, United States
| | - Craig L Peterson
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, United States
| | - Liam J Holt
- Institute for Systems Genetics, New York University Langone Health, New York, United States
| |
Collapse
|
9
|
Birrell GW, Challis MP, De Paoli A, Anderson D, Devine SM, Heffernan GD, Jacobus DP, Edstein MD, Siddiqui G, Creek DJ. Multi-omic Characterization of the Mode of Action of a Potent New Antimalarial Compound, JPC-3210, Against Plasmodium falciparum. Mol Cell Proteomics 2020; 19:308-325. [PMID: 31836637 PMCID: PMC7000111 DOI: 10.1074/mcp.ra119.001797] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 11/17/2019] [Indexed: 01/22/2023] Open
Abstract
The increasing incidence of antimalarial drug resistance to the first-line artemisinin combination therapies underpins an urgent need for new antimalarial drugs, ideally with a novel mode of action. The recently developed 2-aminomethylphenol, JPC-3210, (MMV 892646) is an erythrocytic schizonticide with potent in vitro antimalarial activity against multidrug-resistant Plasmodium falciparum lines, low cytotoxicity, potent in vivo efficacy against murine malaria, and favorable preclinical pharmacokinetics including a lengthy plasma elimination half-life. To investigate the impact of JPC-3210 on biochemical pathways within P. falciparum-infected red blood cells, we have applied a "multi-omics" workflow based on high resolution orbitrap mass spectrometry combined with biochemical approaches. Metabolomics, peptidomics and hemoglobin fractionation analyses revealed a perturbation in hemoglobin metabolism following JPC-3210 exposure. The metabolomics data demonstrated a specific depletion of short hemoglobin-derived peptides, peptidomics analysis revealed a depletion of longer hemoglobin-derived peptides, and the hemoglobin fractionation assay demonstrated decreases in hemoglobin, heme and hemozoin levels. To further elucidate the mechanism responsible for inhibition of hemoglobin metabolism, we used in vitro β-hematin polymerization assays and showed JPC-3210 to be an intermediate inhibitor of β-hematin polymerization, about 10-fold less potent then the quinoline antimalarials, such as chloroquine and mefloquine. Further, quantitative proteomics analysis showed that JPC-3210 treatment results in a distinct proteomic signature compared with other known antimalarials. While JPC-3210 clustered closely with mefloquine in the metabolomics and proteomics analyses, a key differentiating signature for JPC-3210 was the significant enrichment of parasite proteins involved in regulation of translation. These studies revealed that the mode of action for JPC-3210 involves inhibition of the hemoglobin digestion pathway and elevation of regulators of protein translation. Importantly, JPC-3210 demonstrated rapid parasite killing kinetics compared with other quinolones, suggesting that JPC-3210 warrants further investigation as a potentially long acting partner drug for malaria treatment.
Collapse
Affiliation(s)
- Geoffrey W Birrell
- Australian Defense Force Malaria and Infectious Disease Institute, Brisbane, Australia
| | - Matthew P Challis
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Amanda De Paoli
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Dovile Anderson
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Shane M Devine
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | | | | | - Michael D Edstein
- Australian Defense Force Malaria and Infectious Disease Institute, Brisbane, Australia
| | - Ghizal Siddiqui
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia.
| | - Darren J Creek
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| |
Collapse
|
10
|
Chan LY, Teo JDW, Tan KSW, Sou K, Kwan WL, Lee CLK. Near Infrared Fluorophore-Tagged Chloroquine in Plasmodium falciparum Diagnostic Imaging. Molecules 2018; 23:molecules23102635. [PMID: 30322183 PMCID: PMC6222297 DOI: 10.3390/molecules23102635] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 10/11/2018] [Accepted: 10/12/2018] [Indexed: 12/16/2022] Open
Abstract
Chloroquine was among the first of several effective drug treatments against malaria until the onset of chloroquine resistance. In light of diminished clinical efficacy of chloroquine as an antimalarial therapeutic, there is potential in efforts to adapt chloroquine for other clinical applications, such as in combination therapies and in diagnostics. In this context, we designed and synthesized a novel asymmetrical squaraine dye coupled with chloroquine (SQR1-CQ). In this study, SQR1-CQ was used to label live Plasmodium falciparum (P. falciparum) parasite cultures of varying sensitivities towards chloroquine. SQR1-CQ positively stained ring, mature trophozoite and schizont stages of both chloroquine⁻sensitive and chloroquine⁻resistant P. falciparum strains. In addition, SQR1-CQ exhibited significantly higher fluorescence, when compared to the commercial chloroquine-BODIPY (borondipyrromethene) conjugate CQ-BODIPY. We also achieved successful SQR1-CQ labelling of P. falciparum directly on thin blood smear preparations. Drug efficacy experiments measuring half-maximal inhibitory concentration (IC50) showed lower concentration of effective inhibition against resistant strain K1 by SQR1-CQ compared to conventional chloroquine. Taken together, the versatile and highly fluorescent labelling capability of SQR1-CQ and promising preliminary IC50 findings makes it a great candidate for further development as diagnostic tool with drug efficacy against chloroquine-resistant P. falciparum.
Collapse
Affiliation(s)
- Li Yan Chan
- Department of Technology, Innovation and Enterprise (TIE), Singapore Polytechnic, 500 Dover Road, Singapore 139651, Singapore.
| | - Joshua Ding Wei Teo
- Department of Technology, Innovation and Enterprise (TIE), Singapore Polytechnic, 500 Dover Road, Singapore 139651, Singapore.
| | - Kevin Shyong-Wei Tan
- Laboratory of Molecular and Cellular Parasitology, Department of Microbiology and Immunology, National University of Singapore, 5 Science Drive 2 Block MD4, Level 3, Singapore 117545, Singapore.
| | - Keitaro Sou
- Research Institute for Science and Engineering, Waseda University, 3-4-1 Ohkubo, Shinjuku-ku, Tokyo 169-8555, Japan.
| | - Wei Lek Kwan
- Engineering Product Development, Singapore University of Technology and Design, 8 Somapah Road, Singapore 487372, Singapore.
| | - Chi-Lik Ken Lee
- Department of Technology, Innovation and Enterprise (TIE), Singapore Polytechnic, 500 Dover Road, Singapore 139651, Singapore.
| |
Collapse
|
11
|
Araujo JSC, de Souza BC, Costa Junior DB, Oliveira LDM, Santana IB, Duarte AA, Lacerda PS, dos Santos Junior MC, Leite FHA. Identification of new promising Plasmodium falciparum superoxide dismutase allosteric inhibitors through hierarchical pharmacophore-based virtual screening and molecular dynamics. J Mol Model 2018; 24:220. [DOI: 10.1007/s00894-018-3746-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 06/27/2018] [Indexed: 12/13/2022]
|
12
|
Anti-apoptotic effects of Sonic hedgehog signalling through oxidative stress reduction in astrocytes co-cultured with excretory-secretory products of larval Angiostrongylus cantonensis. Sci Rep 2017; 7:41574. [PMID: 28169282 PMCID: PMC5294578 DOI: 10.1038/srep41574] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 12/20/2016] [Indexed: 12/21/2022] Open
Abstract
Angiostrongylus cantonensis, the rat lungworm, is an important aetiologic agent of eosinophilic meningitis and meningoencephalitis in humans. Co-culturing astrocytes with soluble antigens of A. cantonensis activated the Sonic hedgehog (Shh) signalling pathway and inhibited the apoptosis of astrocytes via the activation of Bcl-2. This study was conducted to determine the roles of the Shh signalling pathway, apoptosis, and oxidative stress in astrocytes after treatment with excretory-secretory products (ESP) from A. cantonensis fifth-stage larvae. Although astrocyte viability was significantly decreased after ESP treatment, the expression of Shh signalling pathway related proteins (Shh, Ptch-1 and Gli-1) was significantly increased. However, apoptosis in astrocytes was significantly decreased after activation of the Shh signalling pathway. Moreover, superoxide and hydrogen superoxide levels in astrocytes were significantly reduced after the activation of Shh pathway signalling due to increasing levels of the antioxidants catalase and superoxide dismutase. These findings indicate that the anti-apoptotic effects of the Shh signalling pathway in the astrocytes of mice infected with A. cantonensis are due to reduced levels of oxidative stress caused by the activation of antioxidants.
Collapse
|
13
|
Colacurcio DJ, Nixon RA. Disorders of lysosomal acidification-The emerging role of v-ATPase in aging and neurodegenerative disease. Ageing Res Rev 2016; 32:75-88. [PMID: 27197071 DOI: 10.1016/j.arr.2016.05.004] [Citation(s) in RCA: 322] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 05/02/2016] [Accepted: 05/13/2016] [Indexed: 12/21/2022]
Abstract
Autophagy and endocytosis deliver unneeded cellular materials to lysosomes for degradation. Beyond processing cellular waste, lysosomes release metabolites and ions that serve signaling and nutrient sensing roles, linking the functions of the lysosome to various pathways for intracellular metabolism and nutrient homeostasis. Each of these lysosomal behaviors is influenced by the intraluminal pH of the lysosome, which is maintained in the low acidic range by a proton pump, the vacuolar ATPase (v-ATPase). New reports implicate altered v-ATPase activity and lysosomal pH dysregulation in cellular aging, longevity, and adult-onset neurodegenerative diseases, including forms of Parkinson disease and Alzheimer disease. Genetic defects of subunits composing the v-ATPase or v-ATPase-related proteins occur in an increasingly recognized group of familial neurodegenerative diseases. Here, we review the expanding roles of the v-ATPase complex as a platform regulating lysosomal hydrolysis and cellular homeostasis. We discuss the unique vulnerability of neurons to persistent low level lysosomal dysfunction and review recent clinical and experimental studies that link dysfunction of the v-ATPase complex to neurodegenerative diseases across the age spectrum.
Collapse
|
14
|
Moles E, Moll K, Ch'ng JH, Parini P, Wahlgren M, Fernàndez-Busquets X. Development of drug-loaded immunoliposomes for the selective targeting and elimination of rosetting Plasmodium falciparum-infected red blood cells. J Control Release 2016; 241:57-67. [PMID: 27620073 DOI: 10.1016/j.jconrel.2016.09.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 09/06/2016] [Accepted: 09/07/2016] [Indexed: 11/20/2022]
Abstract
Parasite proteins exported to the surface of Plasmodium falciparum-parasitized red blood cells (pRBCs) have a major role in severe malaria clinical manifestation, where pRBC cytoadhesion and rosetting processes have been strongly linked with microvascular sequestration while avoiding both spleen filtration and immune surveillance. The parasite-derived and pRBC surface-exposed PfEMP1 protein has been identified as one of the responsible elements for rosetting and, therefore, considered as a promising vaccine candidate for the generation of rosette-disrupting antibodies against severe malaria. However, the potential role of anti-rosetting antibodies as targeting molecules for the functionalization of antimalarial drug-loaded nanovectors has never been studied. Our manuscript presents a proof-of-concept study where the activity of an immunoliposomal vehicle with a dual performance capable of specifically recognizing and disrupting rosettes while simultaneously eliminating those pRBCs forming them has been assayed in vitro. A polyclonal antibody against the NTS-DBL1α N-terminal domain of a rosetting PfEMP1 variant has been selected as targeting molecule and lumefantrine as the antimalarial payload. After 30min incubation with 2μM encapsulated drug, a 70% growth inhibition for all parasitic forms in culture (IC50: 414nM) and a reduction in ca. 60% of those pRBCs with a rosetting phenotype (IC50: 747nM) were achieved. This immunoliposomal approach represents an innovative combination therapy for the improvement of severe malaria therapeutics having a broader spectrum of activity than either anti-rosetting antibodies or free drugs on their own.
Collapse
Affiliation(s)
- Ernest Moles
- Nanomalaria Group, Institute for Bioengineering of Catalonia (IBEC), Baldiri Reixac 10-12, ES-08028 Barcelona, Spain; Barcelona Institute for Global Health (ISGlobal), Barcelona Center for International Health Research (CRESIB, Hospital Clínic-Universitat de Barcelona), Rosselló 149-153, ES-08036 Barcelona, Spain; Nanoscience and Nanotechnology Institute (IN2UB), University of Barcelona, Martí i Franquès 1, ES-08028 Barcelona, Spain.
| | - Kirsten Moll
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Jun-Hong Ch'ng
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden; Department of Microbiology, National University of Singapore, Singapore
| | - Paolo Parini
- Department of Laboratory Medicine (LABMED), H5, Division of Clinical Chemistry, Karolinska Institutet, Huddinge, Sweden
| | - Mats Wahlgren
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Xavier Fernàndez-Busquets
- Nanomalaria Group, Institute for Bioengineering of Catalonia (IBEC), Baldiri Reixac 10-12, ES-08028 Barcelona, Spain; Barcelona Institute for Global Health (ISGlobal), Barcelona Center for International Health Research (CRESIB, Hospital Clínic-Universitat de Barcelona), Rosselló 149-153, ES-08036 Barcelona, Spain; Nanoscience and Nanotechnology Institute (IN2UB), University of Barcelona, Martí i Franquès 1, ES-08028 Barcelona, Spain.
| |
Collapse
|
15
|
Abstract
Some hours after invading the erythrocytes of its human host, the malaria parasite Plasmodium falciparum induces an increase in the permeability of the erythrocyte membrane to monovalent ions. The resulting net influx of Na(+) and net efflux of K(+), down their respective concentration gradients, converts the erythrocyte cytosol from an initially high-K(+), low-Na(+) solution to a high-Na(+), low-K(+) solution. The intraerythrocytic parasite itself exerts tight control over its internal Na(+), K(+), Cl(-), and Ca(2+) concentrations and its intracellular pH through the combined actions of a range of membrane transport proteins. The molecular mechanisms underpinning ion regulation in the parasite are receiving increasing attention, not least because PfATP4, a P-type ATPase postulated to be involved in Na(+) regulation, has emerged as a potential antimalarial drug target, susceptible to inhibition by a wide range of chemically unrelated compounds.
Collapse
Affiliation(s)
- Kiaran Kirk
- Research School of Biology, The Australian National University, Canberra, ACT 2601, Australia;
| |
Collapse
|
16
|
Sun X, Qi W, Yue Y, Ling H, Wang G, Song R. Maize ZmVPP5 is a truncated Vacuole H(+) -PPase that confers hypersensitivity to salt stress. JOURNAL OF INTEGRATIVE PLANT BIOLOGY 2016; 58:518-528. [PMID: 26728417 PMCID: PMC5071666 DOI: 10.1111/jipb.12462] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 12/31/2015] [Indexed: 05/30/2023]
Abstract
In plants, Vacuole H(+) -PPases (VPPs) are important proton pumps and encoded by multiple genes. In addition to full-length VPPs, several truncated forms are expressed, but their biological functions are unknown. In this study, we functionally characterized maize vacuole H(+) -PPase 5 (ZmVPP5), a truncated VPP in the maize genome. Although ZmVPP5 shares high sequence similarity with ZmVPP1, ZmVPP5 lacks the complete structure of the conserved proton transport and the inorganic pyrophosphatase-related domain. Phylogenetic analysis suggests that ZmVPP5 might be derived from an incomplete gene duplication event. ZmVPP5 is expressed in multiple tissues, and ZmVPP5 was detected in the plasma membrane, vacuole membrane and nuclei of maize cells. The overexpression of ZmVPP5 in yeast cells caused a hypersensitivity to salt stress. Transgenic maize lines with overexpressed ZmVPP5 also exhibited the salt hypersensitivity phenotype. A yeast two-hybrid analysis identified the ZmBag6-like protein as a putative ZmVPP5-interacting protein. The results of bimolecular luminescence complementation (BiLC) assay suggest an interaction between ZmBag6-like protein and ZmVPP5 in vivo. Overall, this study suggests that ZmVPP5 might act as a VPP antagonist and participate in the cellular response to salt stress. Our study of ZmVPP5 has expanded the understanding of the origin and functions of truncated forms of plant VPPs.
Collapse
Affiliation(s)
- Xiaoliang Sun
- Shanghai key Laboratory of Bio-Energy Crops, School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Weiwei Qi
- Shanghai key Laboratory of Bio-Energy Crops, School of Life Sciences, Shanghai University, Shanghai 200444, China
- Coordinated Crop Biology Research Center(CBRC), Beijing 100193, China
| | - Yihong Yue
- Shanghai key Laboratory of Bio-Energy Crops, School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Huiling Ling
- Shanghai key Laboratory of Bio-Energy Crops, School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Gang Wang
- Shanghai key Laboratory of Bio-Energy Crops, School of Life Sciences, Shanghai University, Shanghai 200444, China
- Coordinated Crop Biology Research Center(CBRC), Beijing 100193, China
| | - Rentao Song
- Shanghai key Laboratory of Bio-Energy Crops, School of Life Sciences, Shanghai University, Shanghai 200444, China
- Coordinated Crop Biology Research Center(CBRC), Beijing 100193, China
| |
Collapse
|
17
|
Mallo N, Lamas J, DeFelipe AP, Sueiro RA, Fontenla F, Leiro JM. Enzymes Involved in Pyrophosphate and Calcium Metabolism as Targets for Anti-scuticociliate Chemotherapy. J Eukaryot Microbiol 2016; 63:505-15. [DOI: 10.1111/jeu.12294] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Revised: 12/18/2015] [Accepted: 01/05/2016] [Indexed: 12/20/2022]
Affiliation(s)
- Natalia Mallo
- Departamento de Microbiología y Parasitología; Instituto de Investigación y Análisis Alimentarios; Universidad de Santiago de Compostela; Santiago de Compostela Spain
| | - Jesús Lamas
- Departamento de Biología Celular y Ecología; Facultad de Biología; Instituto de Acuicultura; Universidad de Santiago de Compostela; Santiago de Compostela Spain
| | - Ana-Paula DeFelipe
- Departamento de Microbiología y Parasitología; Instituto de Investigación y Análisis Alimentarios; Universidad de Santiago de Compostela; Santiago de Compostela Spain
| | - Rosa-Ana Sueiro
- Departamento de Microbiología y Parasitología; Instituto de Investigación y Análisis Alimentarios; Universidad de Santiago de Compostela; Santiago de Compostela Spain
- Departamento de Biología Celular y Ecología; Facultad de Biología; Instituto de Acuicultura; Universidad de Santiago de Compostela; Santiago de Compostela Spain
| | - Francisco Fontenla
- Departamento de Biología Celular y Ecología; Facultad de Biología; Instituto de Acuicultura; Universidad de Santiago de Compostela; Santiago de Compostela Spain
| | - José-Manuel Leiro
- Departamento de Microbiología y Parasitología; Instituto de Investigación y Análisis Alimentarios; Universidad de Santiago de Compostela; Santiago de Compostela Spain
| |
Collapse
|
18
|
Skorokhod OA, Davalos-Schafler D, Gallo V, Valente E, Ulliers D, Notarpietro A, Mandili G, Novelli F, Persico M, Taglialatela-Scafati O, Arese P, Schwarzer E. Oxidative stress-mediated antimalarial activity of plakortin, a natural endoperoxide from the tropical sponge Plakortis simplex. Free Radic Biol Med 2015; 89:624-37. [PMID: 26459031 DOI: 10.1016/j.freeradbiomed.2015.10.399] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 09/27/2015] [Accepted: 10/07/2015] [Indexed: 10/22/2022]
Abstract
Plakortin, a polyketide endoperoxide from the sponge Plakortis simplex has antiparasitic activity against P. falciparum. Similar to artemisinin, its activity depends on the peroxide functionality. Plakortin induced stage-, dose- and time-dependent morphologic anomalies, early maturation delay, ROS generation and lipid peroxidation in the parasite. Ring damage by 1 and 10 µM plakortin led to parasite death before schizogony at 20 and 95%, respectively. Treatment of late schizonts with 1, 2, 5 and 10 µM plakortin resulted in decreased reinfection rates by 30, 50, 61 and 65%, respectively. In both rings and trophozoites, plakortin induced a dose- and time-dependent ROS production as well as a significant lipid peroxidation and up to 4-fold increase of the lipoperoxide breakdown product 4-hydroxynonenal (4-HNE). Antioxidants and the free radical scavengers trolox and N-acetylcysteine significantly attenuated the parasite damage. Plakortin generated 4-HNE conjugates with the P. falciparum proteins: heat shock protein Hsp70-1, endoplasmatic reticulum-standing Hsp70-2 (BiP analogue), V-type proton ATPase catalytic subunit A, enolase, the putative vacuolar protein sorting-associated protein 11, and the dynein heavy chain-like protein, whose specific binding sites were identified by mass spectrometry. These proteins are crucially involved in protein trafficking, transmembrane and vesicular transport and parasite survival. We hypothesize that binding of 4-HNE to functionally relevant parasite proteins may explain the observed plakortin-induced morphologic aberrations and parasite death. The identification of 4-HNE-protein conjugates may generate a novel paradigm to explain the mechanism of action of pro-oxidant, peroxide-based antimalarials such as plakortin, artemisinins and synthetic endoperoxides.
Collapse
Affiliation(s)
- Oleksii A Skorokhod
- Department of Oncology, University of Torino, Via Santena 5bis, 10126 Torino, Italy.
| | | | - Valentina Gallo
- Department of Oncology, University of Torino, Via Santena 5bis, 10126 Torino, Italy.
| | - Elena Valente
- Department of Oncology, University of Torino, Via Santena 5bis, 10126 Torino, Italy.
| | - Daniela Ulliers
- Department of Oncology, University of Torino, Via Santena 5bis, 10126 Torino, Italy.
| | - Agata Notarpietro
- Department of Oncology, University of Torino, Via Santena 5bis, 10126 Torino, Italy.
| | - Giorgia Mandili
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino Medical School, Via Nizza 52, 10126 Torino, Italy; Center for Experimental Research and Medical Studies (CeRMS), Città della Salute e della Scienza, Ospedale San Giovanni Battista, Via Cherasco 15, 10126 Torino, Italy.
| | - Francesco Novelli
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino Medical School, Via Nizza 52, 10126 Torino, Italy; Center for Experimental Research and Medical Studies (CeRMS), Città della Salute e della Scienza, Ospedale San Giovanni Battista, Via Cherasco 15, 10126 Torino, Italy.
| | - Marco Persico
- Department of Pharmacy, University of Napoli 'Federico II', Via D. Montesano 49, 80131 Napoli, Italy.
| | | | - Paolo Arese
- Department of Oncology, University of Torino, Via Santena 5bis, 10126 Torino, Italy.
| | - Evelin Schwarzer
- Department of Oncology, University of Torino, Via Santena 5bis, 10126 Torino, Italy.
| |
Collapse
|
19
|
Okombo J, Chibale K. Antiplasmodial drug targets: a patent review (2000 – 2013). Expert Opin Ther Pat 2015; 26:107-30. [DOI: 10.1517/13543776.2016.1113258] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
20
|
Abstract
As it grows and replicates within the erythrocytes of its host the malaria parasite takes up nutrients from the extracellular medium, exports metabolites and maintains a tight control over its internal ionic composition. These functions are achieved via membrane transport proteins, integral membrane proteins that mediate the passage of solutes across the various membranes that separate the biochemical machinery of the parasite from the extracellular environment. Proteins of this type play a key role in antimalarial drug resistance, as well as being candidate drug targets in their own right. This review provides an overview of recent work on the membrane transport biology of the malaria parasite-infected erythrocyte, encompassing both the parasite-induced changes in the membrane transport properties of the host erythrocyte and the cell physiology of the intracellular parasite itself.
Collapse
|
21
|
Antoine T, Fisher N, Amewu R, O'Neill PM, Ward SA, Biagini GA. Rapid kill of malaria parasites by artemisinin and semi-synthetic endoperoxides involves ROS-dependent depolarization of the membrane potential. J Antimicrob Chemother 2013; 69:1005-16. [PMID: 24335485 PMCID: PMC3956377 DOI: 10.1093/jac/dkt486] [Citation(s) in RCA: 88] [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: 11/27/2022] Open
Abstract
Objectives Artemisinin and artemisinin semi-synthetic derivatives (collectively known as endoperoxides) are first-line antimalarials for the treatment of uncomplicated and severe malaria. Endoperoxides display very fast killing rates and are generally recalcitrant to parasite resistance development. These key pharmacodynamic features are a result of a complex mechanism of action, the details of which lack consensus. Here, we report on the primary physiological events leading to parasite death. Methods Parasite mitochondrial (ΔΨm) and plasma membrane (ΔΨp) electrochemical potentials were measured using real-time single-cell imaging following exposure to pharmacologically relevant concentrations of endoperoxides (artemisinin, dihydroartemisinin, artesunate and the synthetic tetraoxane RKA182). In addition, mitochondrial electron transport chain components NADH:quinone oxidoreductase (alternative complex I), bc1 (complex III) and cytochrome oxidase (complex IV) were investigated to determine their functional sensitivity to the various endoperoxides. Results Parasite exposure to endoperoxides resulted in rapid depolarization of parasite ΔΨm and ΔΨp. The rate of depolarization was decreased in the presence of a reactive oxygen species (ROS) scavenger and Fe3+ chelators. Depolarization of ΔΨm by endoperoxides is not believed to be through the inhibition of mitochondrial electron transport chain components, owing to the lack of significant inhibition when assayed directly. Conclusions The depolarization of ΔΨm and ΔΨp is shown to be mediated via the generation of ROS that are initiated by iron bioactivation of endoperoxides and/or catalysed by iron-dependent oxidative stress. These data are discussed in the context of current hypotheses concerning the mode of action of endoperoxides.
Collapse
Affiliation(s)
- Thomas Antoine
- Liverpool School of Tropical Medicine, Pembroke Place, Liverpool L3 5QA, UK
| | - Nicholas Fisher
- Liverpool School of Tropical Medicine, Pembroke Place, Liverpool L3 5QA, UK
| | - Richard Amewu
- Department of Chemistry, University of Liverpool, Liverpool L69 7ZD, UK
| | - Paul M. O'Neill
- Department of Chemistry, University of Liverpool, Liverpool L69 7ZD, UK
| | - Stephen A. Ward
- Liverpool School of Tropical Medicine, Pembroke Place, Liverpool L3 5QA, UK
| | - Giancarlo A. Biagini
- Liverpool School of Tropical Medicine, Pembroke Place, Liverpool L3 5QA, UK
- Corresponding author. Tel: +44-151-7053151; Fax: +44-151-7053371; E-mail:
| |
Collapse
|