1
|
King NR, Martins Freire C, Touhami J, Sitbon M, Toye AM, Satchwell TJ. Basigin mediation of Plasmodium falciparum red blood cell invasion does not require its transmembrane domain or interaction with monocarboxylate transporter 1. PLoS Pathog 2024; 20:e1011989. [PMID: 38315723 PMCID: PMC10868855 DOI: 10.1371/journal.ppat.1011989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 02/15/2024] [Accepted: 01/19/2024] [Indexed: 02/07/2024] Open
Abstract
Plasmodium falciparum invasion of the red blood cell is reliant upon the essential interaction of PfRh5 with the host receptor protein basigin. Basigin exists as part of one or more multiprotein complexes, most notably through interaction with the monocarboxylate transporter MCT1. However, the potential requirement for basigin association with MCT1 and the wider role of basigin host membrane context and lateral protein associations during merozoite invasion has not been established. Using genetically manipulated in vitro derived reticulocytes, we demonstrate the ability to uncouple basigin ectodomain presentation from its transmembrane domain-mediated interactions, including with MCT1. Merozoite invasion of reticulocytes is unaffected by disruption of basigin-MCT1 interaction and by removal or replacement of the basigin transmembrane helix. Therefore, presentation of the basigin ectodomain at the red blood cell surface, independent of its native association with MCT1 or other interactions mediated by the transmembrane domain, is sufficient to facilitate merozoite invasion.
Collapse
Affiliation(s)
- Nadine R. King
- School of Biochemistry, University of Bristol, Bristol, United Kingdom
| | | | - Jawida Touhami
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
| | - Marc Sitbon
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Ashley M. Toye
- School of Biochemistry, University of Bristol, Bristol, United Kingdom
| | | |
Collapse
|
2
|
Lohia R, Allegrini B, Berry L, Guizouarn H, Cerdan R, Abkarian M, Douguet D, Honoré E, Wengelnik K. Pharmacological activation of PIEZO1 in human red blood cells prevents Plasmodium falciparum invasion. Cell Mol Life Sci 2023; 80:124. [PMID: 37071200 PMCID: PMC10113305 DOI: 10.1007/s00018-023-04773-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 03/30/2023] [Indexed: 04/19/2023]
Abstract
An inherited gain-of-function variant (E756del) in the mechanosensitive cationic channel PIEZO1 was shown to confer a significant protection against severe malaria. Here, we demonstrate in vitro that human red blood cell (RBC) infection by Plasmodium falciparum is prevented by the pharmacological activation of PIEZO1. Yoda1 causes an increase in intracellular calcium associated with rapid echinocytosis that inhibits RBC invasion, without affecting parasite intraerythrocytic growth, division or egress. Notably, Yoda1 treatment significantly decreases merozoite attachment and subsequent RBC deformation. Intracellular Na+/K+ imbalance is unrelated to the mechanism of protection, although delayed RBC dehydration observed in the standard parasite culture medium RPMI/albumax further enhances the resistance to malaria conferred by Yoda1. The chemically unrelated Jedi2 PIEZO1 activator similarly causes echinocytosis and RBC dehydration associated with resistance to malaria invasion. Spiky outward membrane projections are anticipated to reduce the effective surface area required for both merozoite attachment and internalization upon pharmacological activation of PIEZO1. Globally, our findings indicate that the loss of the typical biconcave discoid shape of RBCs, together with an altered optimal surface to volume ratio, induced by PIEZO1 pharmacological activation prevent efficient P. falciparum invasion.
Collapse
Affiliation(s)
- Rakhee Lohia
- LPHI, University of Montpellier, CNRS UMR5294, Montpellier, France
| | | | - Laurence Berry
- LPHI, University of Montpellier, CNRS UMR5294, Montpellier, France
| | | | - Rachel Cerdan
- LPHI, University of Montpellier, CNRS UMR5294, Montpellier, France
| | - Manouk Abkarian
- Centre de Biologie Structurale, CNRS UMR5048, INSERM U1054, University of Montpellier, Montpellier, France
| | - Dominique Douguet
- IPMC, University Côte d'Azur, CNRS, INSERM, UMR7275, Labex ICST, Valbonne, France
| | - Eric Honoré
- IPMC, University Côte d'Azur, CNRS, INSERM, UMR7275, Labex ICST, Valbonne, France.
| | - Kai Wengelnik
- LPHI, University of Montpellier, CNRS UMR5294, INSERM, Montpellier, France.
| |
Collapse
|
3
|
Msosa C, Abdalrahman T, Franz T. An analytical model describing the mechanics of erythrocyte membrane wrapping during active invasion of a plasmodium falciparum merozoite. J Mech Behav Biomed Mater 2023; 140:105685. [PMID: 36746046 DOI: 10.1016/j.jmbbm.2023.105685] [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: 06/14/2022] [Revised: 12/22/2022] [Accepted: 01/23/2023] [Indexed: 01/27/2023]
Abstract
The invasion of a merozoite into an erythrocyte by membrane wrapping is a hallmark of malaria pathogenesis. The invasion involves biomechanical interactions whereby the merozoite exerts actomyosin-based forces to push itself into and through the erythrocyte membrane while concurrently inducing biochemical damage to the erythrocyte membrane. Whereas the biochemical damage process has been investigated, the detailed mechanistic understanding of the invasion mechanics remains limited. Thus, the current study aimed to develop a mathematical model describing the mechanical factors involved in the merozoite invasion into an erythrocyte and explore the invasion mechanics. A shell theory model was developed comprising constitutive, equilibrium and governing equations of the deformable erythrocyte membrane to predict membrane mechanics during the wrapping of an entire non-deformable ellipsoidal merozoite. Predicted parameters include principal erythrocyte membrane deformations and stresses, wrapping and indentation forces, and indentation work. The numerical investigations considered two limits for the erythrocyte membrane deformation during wrapping (4% and 51% areal strain) and erythrocyte membrane phosphorylation (decrease of membrane elastic modulus from 1 to 0.5 kPa). For an intact erythrocyte, the maximum indentation force was 1 and 8.5 pN, and the indentation work was 1.92 × 10-18 and 1.40 × 10-17 J for 4% and 51% areal membrane strain. Phosphorylation damage in the erythrocyte membrane reduced the required indentation work by 50% to 0.97 × 10-18 and 0.70 × 10-17 J for 4% and 51% areal strain. The current study demonstrated the developed model's feasibility to provide new knowledge on the physical mechanisms of the merozoite invasion process that contribute to the invasion efficiency towards the discovery of new invasion-blocking anti-malaria drugs.
Collapse
Affiliation(s)
- Chimwemwe Msosa
- Biomedical Engineering Research Centre, Division of Biomedical Engineering, Department of Human Biology, University of Cape Town, Observatory, 7925, South Africa; Faculty of Engineering, Department of Electrical Engineering, Malawi University of Business and Applied Sciences, Blantyre, Malawi.
| | - Tamer Abdalrahman
- Biomedical Engineering Research Centre, Division of Biomedical Engineering, Department of Human Biology, University of Cape Town, Observatory, 7925, South Africa; Computational Mechanobiology, Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Charité Universitätsmedizin, Berlin, 13353, Germany
| | - Thomas Franz
- Biomedical Engineering Research Centre, Division of Biomedical Engineering, Department of Human Biology, University of Cape Town, Observatory, 7925, South Africa; Bioengineering Science Research Group, Engineering Sciences, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, SO171BJ, UK
| |
Collapse
|
4
|
Satchwell TJ. Generation of red blood cells from stem cells: Achievements, opportunities and perspectives for malaria research. Front Cell Infect Microbiol 2022; 12:1039520. [PMID: 36452302 PMCID: PMC9702814 DOI: 10.3389/fcimb.2022.1039520] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 10/21/2022] [Indexed: 06/22/2024] Open
Abstract
Parasites of the genus Plasmodium that cause malaria survive within humans by invasion of, and proliferation within, the most abundant cell type in the body, the red blood cell. As obligate, intracellular parasites, interactions between parasite and host red blood cell components are crucial to multiple aspects of the blood stage malaria parasite lifecycle. The requirement for, and involvement of, an array of red blood cell proteins in parasite invasion and intracellular development is well established. Nevertheless, detailed mechanistic understanding of host cell protein contributions to these processes are hampered by the genetic intractability of the anucleate red blood cell. The advent of stem cell technology and more specifically development of methods that recapitulate in vitro the process of red blood cell development known as erythropoiesis has enabled the generation of erythroid cell stages previously inaccessible in large numbers for malaria studies. What is more, the capacity for genetic manipulation of nucleated erythroid precursors that can be differentiated to generate modified red blood cells has opened new horizons for malaria research. This review summarises current methodologies that harness in vitro erythroid differentiation of stem cells for generation of cells that are susceptible to malaria parasite invasion; discusses existing and emerging approaches to generate novel red blood cell phenotypes and explores the exciting potential of in vitro derived red blood cells for improved understanding the broad role of host red blood cell proteins in malaria pathogenesis.
Collapse
|
5
|
Proteomic Analysis of the Role of the Adenylyl Cyclase-cAMP Pathway in Red Blood Cell Mechanical Responses. Cells 2022; 11:cells11071250. [PMID: 35406814 PMCID: PMC8997765 DOI: 10.3390/cells11071250] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/14/2022] [Accepted: 02/28/2022] [Indexed: 02/01/2023] Open
Abstract
Red blood cell (RBC) deformability is modulated by the phosphorylation status of the cytoskeletal proteins that regulate the interactions of integral transmembrane complexes. Proteomic studies have revealed that receptor-related signaling molecules and regulatory proteins involved in signaling cascades are present in RBCs. In this study, we investigated the roles of the cAMP signaling mechanism in modulating shear-induced RBC deformability and examined changes in the phosphorylation of the RBC proteome. We implemented the inhibitors of adenylyl cyclase (SQ22536), protein kinase A (H89), and phosphodiesterase (PDE) (pentoxifylline) to whole blood samples, applied 5 Pa shear stress (SS) for 300 s with a capillary tubing system, and evaluated RBC deformability using a LORRCA MaxSis. The inhibition of signaling molecules significantly deteriorated shear-induced RBC deformability (p < 0.05). Capillary SS slightly increased the phosphorylation of RBC cytoskeletal proteins. Tyrosine phosphorylation was significantly elevated by the modulation of the cAMP/PKA pathway (p < 0.05), while serine phosphorylation significantly decreased as a result of the inhibition of PDE (p < 0.05). AC is the core element of this signaling pathway, and PDE works as a negative feedback mechanism that could have potential roles in SS-induced RBC deformability. The cAMP/PKA pathway could regulate RBC deformability during capillary transit by triggering significant alterations in the phosphorylation state of RBCs.
Collapse
|
6
|
Groomes PV, Kanjee U, Duraisingh MT. RBC membrane biomechanics and Plasmodium falciparum invasion: probing beyond ligand-receptor interactions. Trends Parasitol 2022; 38:302-315. [PMID: 34991983 PMCID: PMC8917059 DOI: 10.1016/j.pt.2021.12.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/06/2021] [Accepted: 12/07/2021] [Indexed: 10/19/2022]
Abstract
A critical step in malaria blood-stage infections is the invasion of red blood cells (RBCs) by merozoite forms of the Plasmodium parasite. Much progress has been made in defining the parasite ligands and host receptors that mediate this critical step. However, less well understood are the RBC biophysical determinants that influence parasite invasion. In this review we explore how Plasmodium falciparum merozoites interact with the RBC membrane during invasion to modulate RBC deformability and facilitate invasion. We further highlight RBC biomechanics-related polymorphisms that might have been selected for in human populations due to their ability to reduce parasite invasion. Such an understanding will reveal the translational potential of targeting host pathways affecting RBC biomechanical properties for the treatment of malaria.
Collapse
Affiliation(s)
- Patrice V Groomes
- Department of Immunology & Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Usheer Kanjee
- Department of Immunology & Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Manoj T Duraisingh
- Department of Immunology & Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA.
| |
Collapse
|
7
|
Proteome of Stored RBC Membrane and Vesicles from Heterozygous Beta Thalassemia Donors. Int J Mol Sci 2021; 22:ijms22073369. [PMID: 33806028 PMCID: PMC8037027 DOI: 10.3390/ijms22073369] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 01/19/2023] Open
Abstract
Genetic characteristics of blood donors may impact the storability of blood products. Despite higher basal stress, red blood cells (RBCs) from eligible donors that are heterozygous for beta-thalassemia traits (βThal+) possess a differential nitrogen-related metabolism, and cope better with storage stress compared to the control. Nevertheless, not much is known about how storage impacts the proteome of membrane and extracellular vesicles (EVs) in βThal+. For this purpose, RBC units from twelve βThal+ donors were studied through proteomics, immunoblotting, electron microscopy, and functional ELISA assays, versus units from sex- and aged-matched controls. βThal+ RBCs exhibited less irreversible shape modifications. Their membrane proteome was characterized by different levels of structural, lipid raft, transport, chaperoning, redox, and enzyme components. The most prominent findings include the upregulation of myosin proteoforms, arginase-1, heat shock proteins, and protein kinases, but the downregulation of nitrogen-related transporters. The unique membrane proteome was also mirrored, in part, to that of βThal+ EVs. Network analysis revealed interesting connections of membrane vesiculation with storage and stress hemolysis, along with proteome control modulators of the RBC membrane. Our findings, which are in line with the mild but consistent oxidative stress these cells experience in vivo, provide insight into the physiology and aging of stored βThal+ RBCs.
Collapse
|
8
|
20S proteasomes secreted by the malaria parasite promote its growth. Nat Commun 2021; 12:1172. [PMID: 33608523 PMCID: PMC7895969 DOI: 10.1038/s41467-021-21344-8] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 01/20/2021] [Indexed: 12/16/2022] Open
Abstract
Mature red blood cells (RBCs) lack internal organelles and canonical defense mechanisms, making them both a fascinating host cell, in general, and an intriguing choice for the deadly malaria parasite Plasmodium falciparum (Pf), in particular. Pf, while growing inside its natural host, the human RBC, secretes multipurpose extracellular vesicles (EVs), yet their influence on this essential host cell remains unknown. Here we demonstrate that Pf parasites, cultured in fresh human donor blood, secrete within such EVs assembled and functional 20S proteasome complexes (EV-20S). The EV-20S proteasomes modulate the mechanical properties of naïve human RBCs by remodeling their cytoskeletal network. Furthermore, we identify four degradation targets of the secreted 20S proteasome, the phosphorylated cytoskeletal proteins β-adducin, ankyrin-1, dematin and Epb4.1. Overall, our findings reveal a previously unknown 20S proteasome secretion mechanism employed by the human malaria parasite, which primes RBCs for parasite invasion by altering membrane stiffness, to facilitate malaria parasite growth. Plasmodium falciparum secretes extracellular vesicles (EVs) while growing inside red blood cells (RBCs). Here the authors show that these EVs contain assembled and functional 20S proteasome complexes that remodel the cytoskeleton of naïve human RBCs, priming the RBCs for parasite invasion.
Collapse
|
9
|
Chaurasiya A, Garg S, Khanna A, Narayana C, Dwivedi VP, Joshi N, E Anam Z, Singh N, Singhal J, Kaushik S, Kaur Kahlon A, Srivastava P, Marothia M, Kumar M, Kumar S, Kumari G, Munjal A, Gupta S, Singh P, Pati S, Das G, Sagar R, Ranganathan A, Singh S. Pathogen induced subversion of NAD + metabolism mediating host cell death: a target for development of chemotherapeutics. Cell Death Discov 2021; 7:10. [PMID: 33441562 PMCID: PMC7806871 DOI: 10.1038/s41420-020-00366-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/10/2020] [Accepted: 10/19/2020] [Indexed: 02/07/2023] Open
Abstract
Hijacking of host metabolic status by a pathogen for its regulated dissemination from the host is prerequisite for the propagation of infection. M. tuberculosis secretes an NAD+-glycohydrolase, TNT, to induce host necroptosis by hydrolyzing Nicotinamide adenine dinucleotide (NAD+). Herein, we expressed TNT in macrophages and erythrocytes; the host cells for M. tuberculosis and the malaria parasite respectively, and found that it reduced the NAD+ levels and thereby induced necroptosis and eryptosis resulting in premature dissemination of pathogen. Targeting TNT in M. tuberculosis or induced eryptosis in malaria parasite interferes with pathogen dissemination and reduction in the propagation of infection. Building upon our discovery that inhibition of pathogen-mediated host NAD+ modulation is a way forward for regulation of infection, we synthesized and screened some novel compounds that showed inhibition of NAD+-glycohydrolase activity and pathogen infection in the nanomolar range. Overall this study highlights the fundamental importance of pathogen-mediated modulation of host NAD+ homeostasis for its infection propagation and novel inhibitors as leads for host-targeted therapeutics.
Collapse
Affiliation(s)
- Ayushi Chaurasiya
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Swati Garg
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Ashish Khanna
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India
| | - Chintam Narayana
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India
| | - Ved Prakash Dwivedi
- International Centre for Genetic Engineering and Biotechnology, New Delhi, 110067, India
| | - Nishant Joshi
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Greater Noida, 201314, India
| | - Zill E Anam
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Niharika Singh
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Jhalak Singhal
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Shikha Kaushik
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Amandeep Kaur Kahlon
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Pallavi Srivastava
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Manisha Marothia
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Mukesh Kumar
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Santosh Kumar
- International Centre for Genetic Engineering and Biotechnology, New Delhi, 110067, India
| | - Geeta Kumari
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Akshay Munjal
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Sonal Gupta
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Preeti Singh
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Soumya Pati
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Greater Noida, 201314, India
| | - Gobardhan Das
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Ram Sagar
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India.
| | - Anand Ranganathan
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India.
| | - Shailja Singh
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India.
| |
Collapse
|
10
|
Tang B, Kang P, Zhu L, Xuan L, Wang H, Zhang H, Wang X, Xu J. Simvastatin protects heart function and myocardial energy metabolism in pulmonary arterial hypertension induced right heart failure. J Bioenerg Biomembr 2021; 53:1-12. [PMID: 33394312 DOI: 10.1007/s10863-020-09867-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 12/13/2020] [Indexed: 01/10/2023]
Abstract
The favorable effect of simvastatin on pulmonary arterial hypertension (PAH) has been well defined despite the unknown etiology of PAH. However, whether simvastatin exerts similar effects on PAH induced right heart failure (RHF) remains to be determined. We aimed to investigate the function of simvastatin in PAH induced RHF. Rats in the RHF and simvastatin groups were injected intraperitoneally with monocrotaline to establish PAH-induced RHF model. The expression of miR-21-5p in rat myocardium was detected and miR-21-5p expression was inhibited using antagomiRNA. The effect of simvastatin on hemodynamic indexes, ventricular remodeling of myocardial tissues, myocardial energy metabolism, and calmodulin was explored. Dual-luciferase reporter system was used to verify the binding relationship between miR-21-5p and Smad7. In addition, the regulatory role of simvastatin in Smad7, TGFBR1 and Smad2/3 was investigated. Simvastatin treatment improved hemodynamic condition, myocardial tissue remodeling, and myocardial energy metabolism, as well as increasing calmodulin expression in rats with PAH-induced RHF. After simvastatin treatment, the expression of miR-21-5p in myocardium of rats was decreased significantly. miR-21-5p targeted Smad7 and inhibited the expression of Smad7. Compared with RHF rats, the expressions of TGFBR1 and Smad2/3 in myocardium of simvastatin-treated rats were decreased significantly. Collectively, we provided evidence that simvastatin can protect ATPase activity and maintain myocardial ATP energy reserve through the miR-21-5p/Smad/TGF-β axis, thus ameliorating PAH induced RHF.
Collapse
Affiliation(s)
- Bi Tang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, Anhui, People's Republic of China
| | - Pinfang Kang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, Anhui, People's Republic of China
| | - Lei Zhu
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, Anhui, People's Republic of China
| | - Ling Xuan
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, Anhui, People's Republic of China
| | - Hongju Wang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, Anhui, People's Republic of China
| | - Heng Zhang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, Anhui, People's Republic of China
| | - Xiaojing Wang
- Clinical and Basic Provincial Laboratory of Respiratory System Diseases of Anhui Province, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, Anhui, People's Republic of China
| | - Jiali Xu
- Department of Paediatrics, The First Affiliated Hospital of Bengbu Medical College, No. 287, Changhuai Road, Bengbu, 233004, Anhui, People's Republic of China.
| |
Collapse
|
11
|
Paone S, D'Alessandro S, Parapini S, Celani F, Tirelli V, Pourshaban M, Olivieri A. Characterization of the erythrocyte GTPase Rac1 in relation to Plasmodium falciparum invasion. Sci Rep 2020; 10:22054. [PMID: 33328606 PMCID: PMC7744522 DOI: 10.1038/s41598-020-79052-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 11/30/2020] [Indexed: 12/01/2022] Open
Abstract
Malaria is still a devastating disease with 228 million cases globally and 405,000 lethal outcomes in 2018, mainly in children under five years of age. The threat of emerging malaria strains resistant to currently available drugs has made the search for novel drug targets compelling. The process by which Plasmodium falciparum parasites invade the host cell has been widely studied, but only a few erythrocyte proteins involved in this process have been identified so far. The erythrocyte protein Rac1 is a GTPase that plays an important role in host cell invasion by many intracellular pathogens. Here we show that Rac1 is recruited in proximity to the site of parasite entry during P. falciparum invasion process and that subsequently localizes to the parasitophorous vacuole membrane. We also suggest that this GTPase may be involved in erythrocyte invasion by P. falciparum, by testing the effect of specific Rac1 inhibitory compounds. Finally, we suggest a secondary role of the erythrocyte GTPase also in parasite intracellular development. We here characterize a new erythrocyte protein potentially involved in P. falciparum invasion of the host cell and propose the human GTPase Rac1 as a novel and promising antimalarial drug target.
Collapse
Affiliation(s)
- Silvio Paone
- Dipartimento di Malattie Infettive, Istituto Superiore di Sanità, Rome, Italy.,Dipartimento di Sanità Pubblica e Malattie Infettive, Sapienza University of Rome, Rome, Italy
| | - Sarah D'Alessandro
- Dipartimento di Scienze Biomediche, Chirurgiche e Odontoiatriche, University of Milan, Milan, Italy
| | - Silvia Parapini
- Dipartimento di Scienze Biomediche Per La Salute, University of Milan, Milan, Italy
| | - Francesco Celani
- Dipartimento di Malattie Infettive, Istituto Superiore di Sanità, Rome, Italy
| | - Valentina Tirelli
- Dipartimento di Malattie Infettive, Istituto Superiore di Sanità, Rome, Italy
| | | | - Anna Olivieri
- Dipartimento di Malattie Infettive, Istituto Superiore di Sanità, Rome, Italy.
| |
Collapse
|
12
|
The Riveting Cellular Structures of Apicomplexan Parasites. Trends Parasitol 2020; 36:979-991. [PMID: 33011071 DOI: 10.1016/j.pt.2020.09.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/31/2020] [Accepted: 09/03/2020] [Indexed: 12/21/2022]
Abstract
Parasitic protozoa of the phylum Apicomplexa cause a range of human and animal diseases. Their complex life cycles - often heteroxenous with sexual and asexual phases in different hosts - rely on elaborate cytoskeletal structures to enable morphogenesis and motility, organize cell division, and withstand diverse environmental forces. This review primarily focuses on studies using Toxoplasma gondii and Plasmodium spp. as the best studied apicomplexans; however, many cytoskeletal adaptations are broadly conserved and predate the emergence of the parasitic phylum. After decades cataloguing the constituents of such structures, a dynamic picture is emerging of the assembly and maintenance of apicomplexan cytoskeletons, illuminating how they template and orient critical processes during infection. These observations impact our view of eukaryotic diversity and offer future challenges for cell biology.
Collapse
|
13
|
Davies H, Belda H, Broncel M, Ye X, Bisson C, Introini V, Dorin-Semblat D, Semblat JP, Tibúrcio M, Gamain B, Kaforou M, Treeck M. An exported kinase family mediates species-specific erythrocyte remodelling and virulence in human malaria. Nat Microbiol 2020; 5:848-863. [PMID: 32284562 PMCID: PMC7116245 DOI: 10.1038/s41564-020-0702-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 03/05/2020] [Indexed: 01/31/2023]
Abstract
The most severe form of human malaria is caused by Plasmodium falciparum. Its virulence is closely linked to the increase in rigidity of infected erythrocytes and their adhesion to endothelial receptors, obstructing blood flow to vital organs. Unlike other human-infecting Plasmodium species, P. falciparum exports a family of 18 FIKK serine/threonine kinases into the host cell, suggesting that phosphorylation may modulate erythrocyte modifications. We reveal substantial species-specific phosphorylation of erythrocyte proteins by P. falciparum but not by Plasmodium knowlesi, which does not export FIKK kinases. By conditionally deleting all FIKK kinases combined with large-scale quantitative phosphoproteomics we identified unique phosphorylation fingerprints for each kinase, including phosphosites on parasite virulence factors and host erythrocyte proteins. Despite their non-overlapping target sites, a network analysis revealed that some FIKKs may act in the same pathways. Only the deletion of the non-exported kinase FIKK8 resulted in reduced parasite growth, suggesting the exported FIKKs may instead support functions important for survival in the host. We show that one kinase, FIKK4.1, mediates both rigidification of the erythrocyte cytoskeleton and trafficking of the adhesin and key virulence factor PfEMP1 to the host cell surface. This establishes the FIKK family as important drivers of parasite evolution and malaria pathology.
Collapse
Affiliation(s)
- Heledd Davies
- Signalling in Apicomplexan Parasites Laboratory, The Francis Crick Institute, London, UK
| | - Hugo Belda
- Signalling in Apicomplexan Parasites Laboratory, The Francis Crick Institute, London, UK
| | - Malgorzata Broncel
- Signalling in Apicomplexan Parasites Laboratory, The Francis Crick Institute, London, UK
| | - Xingda Ye
- Signalling in Apicomplexan Parasites Laboratory, The Francis Crick Institute, London, UK
- Division of Infectious Diseases, Department of Medicine, Imperial College London, London, UK
| | - Claudine Bisson
- Institute of Structural and Molecular Biology, Birkbeck College, University of London, London, UK
| | - Viola Introini
- Cavendish Laboratory, University of Cambridge, Cambridge, UK
| | - Dominique Dorin-Semblat
- Université de Paris, Biologie Intégrée du Globule Rouge, UMR_S1134, BIGR, INSERM, Paris, France
- Institut National de la Transfusion Sanguine, Paris, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Jean-Philippe Semblat
- Université de Paris, Biologie Intégrée du Globule Rouge, UMR_S1134, BIGR, INSERM, Paris, France
- Institut National de la Transfusion Sanguine, Paris, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Marta Tibúrcio
- Signalling in Apicomplexan Parasites Laboratory, The Francis Crick Institute, London, UK
| | - Benoit Gamain
- Université de Paris, Biologie Intégrée du Globule Rouge, UMR_S1134, BIGR, INSERM, Paris, France
- Institut National de la Transfusion Sanguine, Paris, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Myrsini Kaforou
- Division of Infectious Diseases, Department of Medicine, Imperial College London, London, UK
| | - Moritz Treeck
- Signalling in Apicomplexan Parasites Laboratory, The Francis Crick Institute, London, UK.
| |
Collapse
|
14
|
Horta MF, Andrade LO, Martins-Duarte ÉS, Castro-Gomes T. Cell invasion by intracellular parasites - the many roads to infection. J Cell Sci 2020; 133:133/4/jcs232488. [PMID: 32079731 DOI: 10.1242/jcs.232488] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Intracellular parasites from the genera Toxoplasma, Plasmodium, Trypanosoma, Leishmania and from the phylum Microsporidia are, respectively, the causative agents of toxoplasmosis, malaria, Chagas disease, leishmaniasis and microsporidiosis, illnesses that kill millions of people around the globe. Crossing the host cell plasma membrane (PM) is an obstacle these parasites must overcome to establish themselves intracellularly and so cause diseases. The mechanisms of cell invasion are quite diverse and include (1) formation of moving junctions that drive parasites into host cells, as for the protozoans Toxoplasma gondii and Plasmodium spp., (2) subversion of endocytic pathways used by the host cell to repair PM, as for Trypanosoma cruzi and Leishmania, (3) induction of phagocytosis as for Leishmania or (4) endocytosis of parasites induced by specialized structures, such as the polar tubes present in microsporidian species. Understanding the early steps of cell entry is essential for the development of vaccines and drugs for the prevention or treatment of these diseases, and thus enormous research efforts have been made to unveil their underlying biological mechanisms. This Review will focus on these mechanisms and the factors involved, with an emphasis on the recent insights into the cell biology of invasion by these pathogens.
Collapse
Affiliation(s)
- Maria Fátima Horta
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, CEP 31270-901, Brazil
| | - Luciana Oliveira Andrade
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, CEP 31270-901, Brazil
| | - Érica Santos Martins-Duarte
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, CEP 31270-901, Brazil
| | - Thiago Castro-Gomes
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, CEP 31270-901, Brazil
| |
Collapse
|
15
|
The effects of dyslipidaemia and cholesterol modulation on erythrocyte susceptibility to malaria parasite infection. Malar J 2019; 18:381. [PMID: 31783858 PMCID: PMC6884832 DOI: 10.1186/s12936-019-3016-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Accepted: 11/20/2019] [Indexed: 11/12/2022] Open
Abstract
Background Malaria disease commences when blood-stage parasites, called merozoites, invade human erythrocytes. Whilst the process of invasion is traditionally seen as being entirely merozoite-driven, emerging data suggests erythrocyte biophysical properties markedly influence invasion. Cholesterol is a major determinant of cell membrane biophysical properties demanding its interrogation as a potential mediator of resistance to merozoite invasion of the erythrocyte. Methods Biophysical measurements of erythrocyte deformability by flicker spectroscopy were used to assess changes in erythrocyte bending modulus on forced integration of cholesterol and how these artificial changes affect invasion by human Plasmodium falciparum merozoites. To validate these observations in a natural context, either murine Plasmodium berghei or human Plasmodium falciparum merozoites were tested for their ability to invade erythrocytes from a hypercholesterolaemic mouse model or human clinical erythrocyte samples deriving from patients with a range of serum cholesterol concentrations, respectively. Results Erythrocyte bending modulus (a measure of deformability) was shown to be markedly affected by artificial modulation of cholesterol content and negatively correlated with merozoite invasion efficiency. In an in vitro infection context, however, erythrocytes taken from hypercholesterolaemic mice or from human clinical samples with varying serum cholesterol levels showed little difference in their susceptibility to merozoite invasion. Explaining this, membrane cholesterol levels in both mouse and human hypercholesterolaemia erythrocytes were subsequently found to be no different from matched normal serum controls. Conclusions Based on these observations, serum cholesterol does not appear to impact on erythrocyte susceptibility to merozoite entry. Indeed, no relationship between serum cholesterol and cholesterol content of the erythrocyte is apparent. This work, nonetheless, suggests that native polymorphisms which do affect membrane lipid composition would be expected to affect parasite entry. This supports investigation of erythrocyte biophysical properties in endemic settings, which may yet identify naturally protective lipid-related polymorphisms.
Collapse
|
16
|
Haqshenas G, Doerig C. Targeting of host cell receptor tyrosine kinases by intracellular pathogens. Sci Signal 2019; 12:12/599/eaau9894. [PMID: 31530732 DOI: 10.1126/scisignal.aau9894] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Intracellular pathogens use complex and tightly regulated processes to enter host cells. Upon initial interactions with signaling proteins at the surface of target cells, intracellular microbes activate and co-opt specific host signaling pathways that mediate cell surface-cytosol communications to facilitate pathogen internalization. Here, we discuss the roles of host receptor tyrosine kinases (RTKs) in the establishment of productive infections by major intracellular pathogens. We evaluate the gaps in the current understanding of this process and propose a comprehensive approach for assessing the role of host cell signaling in the biology of intracellular microorganisms and viruses. We also discuss RTK-targeting strategies for the treatment of various infections.
Collapse
Affiliation(s)
- Gholamreza Haqshenas
- Infection and Immunity, Monash Biomedicine Discovery Institute, and Department of Microbiology, Monash University, Clayton, VIC 3800, Australia.
| | - Christian Doerig
- Infection and Immunity, Monash Biomedicine Discovery Institute, and Department of Microbiology, Monash University, Clayton, VIC 3800, Australia. .,Centre for Chronic Infectious and Inflammatory Diseases, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia
| |
Collapse
|
17
|
Host Cytoskeleton Remodeling throughout the Blood Stages of Plasmodium falciparum. Microbiol Mol Biol Rev 2019; 83:83/4/e00013-19. [PMID: 31484690 DOI: 10.1128/mmbr.00013-19] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The asexual intraerythrocytic development of Plasmodium falciparum, causing the most severe form of human malaria, is marked by extensive host cell remodeling. Throughout the processes of invasion, intracellular development, and egress, the erythrocyte membrane skeleton is remodeled by the parasite as required for each specific developmental stage. The remodeling is facilitated by a plethora of exported parasite proteins, and the erythrocyte membrane skeleton is the interface of most of the observed interactions between the parasite and host cell proteins. Host cell remodeling has been extensively described and there is a vast body of information on protein export or the description of parasite-induced structures such as Maurer's clefts or knobs on the host cell surface. Here we specifically review the molecular level of each host cell-remodeling step at each stage of the intraerythrocytic development of P. falciparum We describe key events, such as invasion, knob formation, and egress, and identify the interactions between exported parasite proteins and the host cell cytoskeleton. We discuss each remodeling step with respect to time and specific requirement of the developing parasite to explain host cell remodeling in a stage-specific manner. Thus, we highlight the interaction with the host membrane skeleton as a key event in parasite survival.
Collapse
|
18
|
Pretini V, Koenen MH, Kaestner L, Fens MHAM, Schiffelers RM, Bartels M, Van Wijk R. Red Blood Cells: Chasing Interactions. Front Physiol 2019; 10:945. [PMID: 31417415 PMCID: PMC6684843 DOI: 10.3389/fphys.2019.00945] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 07/09/2019] [Indexed: 12/13/2022] Open
Abstract
Human red blood cells (RBC) are highly differentiated cells that have lost all organelles and most intracellular machineries during their maturation process. RBC are fundamental for the nearly all basic physiologic dynamics and they are key cells in the body's respiratory system by being responsible for the oxygen transport to all cells and tissues, and delivery of carbon dioxide to the lungs. With their flexible structure RBC are capable to deform in order to travel through all blood vessels including very small capillaries. Throughout their in average 120 days lifespan, human RBC travel in the bloodstream and come in contact with a broad range of different cell types. In fact, RBC are able to interact and communicate with endothelial cells (ECs), platelets, macrophages, and bacteria. Additionally, they are involved in the maintenance of thrombosis and hemostasis and play an important role in the immune response against pathogens. To clarify the mechanisms of interaction of RBC and these other cells both in health and disease as well as to highlight the role of important key players, we focused our interest on RBC membrane components such as ion channels, proteins, and phospholipids.
Collapse
Affiliation(s)
- Virginia Pretini
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
- Theoretical Medicine and Biosciences, Saarland University, Homburg, Germany
| | - Mischa H. Koenen
- Department of Laboratory of Translational Immunology and Department of Pediatric Immunology, Wilhelmina Children’s Hospital, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Lars Kaestner
- Theoretical Medicine and Biosciences, Saarland University, Homburg, Germany
- Experimental Physics, Saarland University, Saarbrücken, Germany
| | - Marcel H. A. M. Fens
- Department of Pharmaceutics, Utrecht Institute of Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Raymond M. Schiffelers
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Marije Bartels
- Paediatric Haematology Department, Wilhelmina Children’s Hospital, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Richard Van Wijk
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
19
|
Plasmodium myosin A drives parasite invasion by an atypical force generating mechanism. Nat Commun 2019; 10:3286. [PMID: 31337750 PMCID: PMC6650474 DOI: 10.1038/s41467-019-11120-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 06/11/2019] [Indexed: 12/03/2022] Open
Abstract
Plasmodium parasites are obligate intracellular protozoa and causative agents of malaria, responsible for half a million deaths each year. The lifecycle progression of the parasite is reliant on cell motility, a process driven by myosin A, an unconventional single-headed class XIV molecular motor. Here we demonstrate that myosin A from Plasmodium falciparum (PfMyoA) is critical for red blood cell invasion. Further, using a combination of X-ray crystallography, kinetics, and in vitro motility assays, we elucidate the non-canonical interactions that drive this motor’s function. We show that PfMyoA motor properties are tuned by heavy chain phosphorylation (Ser19), with unphosphorylated PfMyoA exhibiting enhanced ensemble force generation at the expense of speed. Regulated phosphorylation may therefore optimize PfMyoA for enhanced force generation during parasite invasion or for fast motility during dissemination. The three PfMyoA crystallographic structures presented here provide a blueprint for discovery of specific inhibitors designed to prevent parasite infection. Here, Robert-Paganin et al. show that myosin A from Plasmodium falciparum is critical for red blood cell invasion and that non-canonical interactions and regulated phosphorylation are important for force generation during parasite invasion.
Collapse
|
20
|
Pei J, Fu B, Jiang L, Sun T. Biosynthesis, characterization, and anticancer effect of plant-mediated silver nanoparticles using Coptis chinensis. Int J Nanomedicine 2019; 14:1969-1978. [PMID: 30936697 PMCID: PMC6421896 DOI: 10.2147/ijn.s188235] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background Tremendous growth in nanotechnology has opened up new frontiers in fundamental and applied aspects, including the synthesis of nanoscale matter and understanding/utilizing its exotic physicochemical and optoelectronic properties. Green-synthesis methods employing either biological microorganisms or plant extracts have emerged as a simple and alternative to chemical synthesis. Methods In our present study, we aimed to synthesize silver nanoparticles (AgNPs) in combination with an aqueous extract of Coptis chinensis (CC) using a suitable ecofriendly green-synthesis way. Results In our results, ultraviolet-visible spectroscopy revealed a near-absorbance peak at 450 nm, which confirmed the AgNP synthesis. The crystalline nature of the AgNPs was revealed with X-ray diffraction. Transmission electron-microscopy analysis showed spherically dispersed nanoparticles of 6–45 nm diameter. We analyzed the elementary mechanism across A549 lung carcinoma cells ahead of treatment with doses of CC-AgNPs (10 µg/mL and 25 µg/mL). The antiproliferative effect of CC-AgNPs revealed a significant decline in cell viability. Antibacterial assays with both Gram-negative (Escherichia coli) and Gram-positive (Staphylococcus aureus) bacteria exhibited a higher zone of inhibition against S. aureus. Conclusion Furthermore, CC-AgNPs regulated apoptosis using the intrinsic pathway to inhibit A549-cell proliferation. Proliferation migration and invasion were notably inhibited by CC-AgNPs, which promoted apoptosis in lung adenocarcinoma cells by regulating the apoptotic pathway.
Collapse
Affiliation(s)
- Junwen Pei
- Department of Integrated Traditional Chinese and Western Medicine, The Henan Cancer Hospital, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450008, China,
| | - Binfan Fu
- Department of Integrated Traditional Chinese and Western Medicine, The Henan Cancer Hospital, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450008, China,
| | - Lifeng Jiang
- Department of Integrated Traditional Chinese and Western Medicine, The Henan Cancer Hospital, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450008, China,
| | - Taizhen Sun
- Department of Integrated Traditional Chinese and Western Medicine, The Henan Cancer Hospital, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450008, China,
| |
Collapse
|
21
|
Kang H, Hong Z, Zhong M, Klomp J, Bayless KJ, Mehta D, Karginov AV, Hu G, Malik AB. Piezo1 mediates angiogenesis through activation of MT1-MMP signaling. Am J Physiol Cell Physiol 2019; 316:C92-C103. [PMID: 30427721 PMCID: PMC6383143 DOI: 10.1152/ajpcell.00346.2018] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 10/16/2018] [Accepted: 11/07/2018] [Indexed: 11/22/2022]
Abstract
Angiogenesis is initiated in response to a variety of external cues, including mechanical and biochemical stimuli; however, the underlying signaling mechanisms remain unclear. Here, we investigated the proangiogenic role of the endothelial mechanosensor Piezo1. Genetic deletion and pharmacological inhibition of Piezo1 reduced endothelial sprouting and lumen formation induced by wall shear stress and proangiogenic mediator sphingosine 1-phosphate, whereas Piezo1 activation by selective Piezo1 activator Yoda1 enhanced sprouting angiogenesis. Similarly to wall shear stress, sphingosine 1-phosphate functioned by activating the Ca2+ gating function of Piezo1, which in turn signaled the activation of the matrix metalloproteinase-2 and membrane type 1 matrix metalloproteinase during sprouting angiogenesis. Studies in mice in which Piezo1 was conditionally deleted in endothelial cells demonstrated the requisite role of sphingosine 1-phosphate-dependent activation of Piezo1 in mediating angiogenesis in vivo. These results taken together suggest that both mechanical and biochemical stimuli trigger Piezo1-mediated Ca2+ influx and thereby activate matrix metalloproteinase-2 and membrane type 1 matrix metalloproteinase and synergistically facilitate sprouting angiogenesis.
Collapse
Affiliation(s)
- Hojin Kang
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine , Chicago, Illinois
- Department of Anesthesiology, The University of Illinois College of Medicine , Chicago, Illinois
| | - Zhigang Hong
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine , Chicago, Illinois
| | - Ming Zhong
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine , Chicago, Illinois
| | - Jennifer Klomp
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine , Chicago, Illinois
| | - Kayla J Bayless
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center , College Station, Texas
| | - Dolly Mehta
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine , Chicago, Illinois
| | - Andrei V Karginov
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine , Chicago, Illinois
| | - Guochang Hu
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine , Chicago, Illinois
- Department of Anesthesiology, The University of Illinois College of Medicine , Chicago, Illinois
| | - Asrar B Malik
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine , Chicago, Illinois
| |
Collapse
|
22
|
Wanaguru M, Barry DJ, Benton DJ, O’Reilly NJ, Bishop KN. Murine leukemia virus p12 tethers the capsid-containing pre-integration complex to chromatin by binding directly to host nucleosomes in mitosis. PLoS Pathog 2018; 14:e1007117. [PMID: 29906285 PMCID: PMC6021111 DOI: 10.1371/journal.ppat.1007117] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 06/27/2018] [Accepted: 05/22/2018] [Indexed: 12/31/2022] Open
Abstract
The murine leukaemia virus (MLV) Gag cleavage product, p12, is essential for both early and late steps in viral replication. The N-terminal domain of p12 binds directly to capsid (CA) and stabilises the mature viral core, whereas defects in the C-terminal domain (CTD) of p12 can be rescued by addition of heterologous chromatin binding sequences (CBSs). We and others hypothesised that p12 tethers the pre-integration complex (PIC) to host chromatin ready for integration. Using confocal microscopy, we have observed for the first time that CA localises to mitotic chromatin in infected cells in a p12-dependent manner. GST-tagged p12 alone, however, did not localise to chromatin and mass-spectrometry analysis of its interactions identified only proteins known to bind the p12 region of Gag. Surprisingly, the ability to interact with chromatin was conferred by a single amino acid change, M63I, in the p12 CTD. Interestingly, GST-p12_M63I showed increased phosphorylation in mitosis relative to interphase, which correlated with an increased interaction with mitotic chromatin. Mass-spectrometry analysis of GST-p12_M63I revealed nucleosomal histones as primary interactants. Direct binding of MLV p12_M63I peptides to histones was confirmed by biolayer-interferometry (BLI) assays using highly-avid recombinant poly-nucleosomal arrays. Excitingly, using this method, we also observed binding between MLV p12_WT and nucleosomes. Nucleosome binding was additionally detected with p12 orthologs from feline and gibbon ape leukemia viruses using both pull-down and BLI assays, indicating that this a common feature of gammaretroviral p12 proteins. Importantly, p12 peptides were able to block the binding of the prototypic foamy virus CBS to nucleosomes and vice versa, implying that their docking sites overlap and suggesting a conserved mode of chromatin tethering for different retroviral genera. We propose that p12 is acting in a similar capacity to CPSF6 in HIV-1 infection by facilitating initial chromatin targeting of CA-containing PICs prior to integration.
Collapse
Affiliation(s)
- Madushi Wanaguru
- Retroviral Replication Laboratory, The Francis Crick Institute, London, United Kingdom
| | - David J. Barry
- Advanced Light Microscopy Facility, The Francis Crick Institute, London, United Kingdom
| | - Donald J. Benton
- Structural Biology of Disease Processes Laboratory, The Francis Crick Institute, London, United Kingdom
| | | | - Kate N. Bishop
- Retroviral Replication Laboratory, The Francis Crick Institute, London, United Kingdom
| |
Collapse
|
23
|
Miliu A, Lebrun M, Braun-Breton C, Lamarque MH. Shelph2, a bacterial-like phosphatase of the malaria parasite Plasmodium falciparum, is dispensable during asexual blood stage. PLoS One 2017; 12:e0187073. [PMID: 29073264 PMCID: PMC5658161 DOI: 10.1371/journal.pone.0187073] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 10/12/2017] [Indexed: 12/03/2022] Open
Abstract
During the erythrocytic cycle of the malaria parasite Plasmodium falciparum, egress and invasion are essential steps finely controlled by reversible phosphorylation. In contrast to the growing number of kinases identified as key regulators, phosphatases have been poorly studied, and calcineurin is the only one identified so far to play a role in invasion. PfShelph2, a bacterial-like phosphatase, is a promising candidate to participate in the invasion process, as it was reported to be expressed late during the asexual blood stage and to reside within an apical compartment, yet distinct from rhoptry bulb, micronemes, or dense granules. It was also proposed to play a role in the control of the red blood cell membrane deformability at the end of the invasion process. However, genetic studies are still lacking to support this hypothesis. Here, we take advantage of the CRISPR-Cas9 technology to tag shelph2 genomic locus while retaining its endogenous regulatory regions. This new strain allows us to follow the endogenous PfShelph2 protein expression and location during asexual blood stages. We show that PfShelph2 apical location is also distinct from the rhoptry neck or exonemes. We further demonstrate PfShelph2 dispensability during the asexual blood stage by generating PfShelph2-KO parasites using CRISPR-Cas9 machinery. Analyses of the mutant during the course of the erythrocytic development indicate that there are no detectable phenotypic consequences of Pfshelph2 genomic deletion. As this lack of phenotype might be due to functional redundancy, we also explore the likelihood of PfShelph1 (PfShelph2 paralog) being a compensatory phosphatase. We conclude that despite its cyclic expression profile, PfShelph2 is a dispensable phosphatase during the Plasmodium falciparum asexual blood stage, whose function is unlikely substituted by PfShelph1.
Collapse
Affiliation(s)
| | - Maryse Lebrun
- DIMNP, CNRS, Université de Montpellier, Montpellier, France
| | | | - Mauld H. Lamarque
- DIMNP, CNRS, Université de Montpellier, Montpellier, France
- * E-mail:
| |
Collapse
|
24
|
Molecular mechanisms that mediate invasion and egress of malaria parasites from red blood cells. Curr Opin Hematol 2017; 24:208-214. [PMID: 28306665 DOI: 10.1097/moh.0000000000000334] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW Malaria parasites invade and multiply in diverse host cells during their complex life cycle. Some blood stage parasites transform into male and female gametocytes that are transmitted by female anopheline mosquitoes. The gametocytes are activated in the mosquito midgut to form male and female gametes, which egress from RBCs to mate and form a zygote. Here, we will review our current understanding of the molecular mechanisms that mediate invasion and egress by malaria parasites at different life cycle stages. RECENT FINDINGS A number of key effector molecules such as parasite protein ligands for receptor-engagement during invasion as well as proteases and perforin-like proteins that mediate egress have been identified. Interestingly, these parasite-encoded effectors are located in internal, vesicular organelles and are secreted in a highly regulated manner during invasion and egress. Here, we will review our current understanding of the functional roles of these effectors as well as the signaling pathways that regulate their timely secretion with accurate spatiotemporal coordinates. SUMMARY Understanding the molecular basis of key processes such as host cell invasion and egress by malaria parasites could provide novel targets for development of inhibitors to block parasite growth and transmission.
Collapse
|
25
|
Abstract
The erythrocyte contains a network of pathways that regulate salt and water content in the face of extracellular and intracellular osmotic perturbations. This allows the erythrocyte to maintain a narrow range of cell hemoglobin concentration, a process critical for normal red blood cell function and survival. Primary disorders that perturb volume homeostasis jeopardize the erythrocyte and may lead to its premature destruction. These disorders are marked by clinical, laboratory, and physiologic heterogeneity. Recent studies have revealed that these disorders are also marked by genetic heterogeneity. They have implicated roles for several proteins, PIEZO1, a mammalian mechanosensory protein; GLUT1, the glucose transporter; SLC4A1, the anion transporter; RhAG, the Rh-associated glycoprotein; KCNN4, the Gardos channel; and ABCB6, an adenosine triphosphate-binding cassette family member, in the maintenance of erythrocyte volume homeostasis. Secondary disorders of erythrocyte hydration include sickle cell disease, thalassemia, hemoglobin CC, and hereditary spherocytosis, where cellular dehydration may be a significant contributor to disease pathology and clinical complications. Understanding the pathways regulating erythrocyte water and solute content may reveal innovative strategies to maintain normal volume in disorders associated with primary or secondary cellular dehydration. These mechanisms will serve as a paradigm for other cells and may reveal new therapeutic targets for disease prevention and treatment beyond the erythrocyte.
Collapse
|
26
|
|
27
|
Abstract
It is only in the last decade that sub-cellular resolution of red cell invasion by the malaria parasite Plasmodium falciparum has been possible. Here we look back on the development of methodologies that led to this possibility and the subsequent advancements made in understanding this key event in malaria disease.
Collapse
Affiliation(s)
- Jake Baum
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK.
| | - Dave Richard
- Department of Microbiology, Infectious Diseases, and Immunology, Laval University, Quebec City, QC G1V 0A6, Canada
| | - David T Riglar
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| |
Collapse
|
28
|
Goheen MM, Campino S, Cerami C. The role of the red blood cell in host defence against falciparum malaria: an expanding repertoire of evolutionary alterations. Br J Haematol 2017; 179:543-556. [PMID: 28832963 DOI: 10.1111/bjh.14886] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The malaria parasite has co-evolved with its human host as each organism struggles for resources and survival. The scars of this war are carried in the human genome in the form of polymorphisms that confer innate resistance to malaria. Clinical, epidemiological and genome-wide association studies have identified multiple polymorphisms in red blood cell (RBC) proteins that attenuate malaria pathogenesis. These include well-known polymorphisms in haemoglobin, intracellular enzymes, RBC channels, RBC surface markers, and proteins impacting the RBC cytoskeleton and RBC morphology. A better understanding of how changes in RBC physiology impact malaria pathogenesis may uncover new strategies to combat the disease.
Collapse
Affiliation(s)
- Morgan M Goheen
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Susana Campino
- Department of Pathogen Molecular Biology, Faculty of Infectious and Tropical Diseases, The London School of Hygiene & Tropical Medicine, London, UK
| | - Carla Cerami
- MRC International Nutrition Group at Keneba, MRC Unit The Gambia, Banjul, The Gambia
| |
Collapse
|
29
|
Das S, Lemgruber L, Tay CL, Baum J, Meissner M. Multiple essential functions of Plasmodium falciparum actin-1 during malaria blood-stage development. BMC Biol 2017; 15:70. [PMID: 28810863 PMCID: PMC5557482 DOI: 10.1186/s12915-017-0406-2] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 07/14/2017] [Indexed: 01/04/2023] Open
Abstract
Background The phylum Apicomplexa includes intracellular parasites causing immense global disease burden, the deadliest of them being the human malaria parasite Plasmodium falciparum, which invades and replicates within erythrocytes. The cytoskeletal protein actin is well conserved within apicomplexans but divergent from mammalian actins, and was primarily reported to function during host cell invasion. However, novel invasion mechanisms have been described for several apicomplexans, and specific functions of the acto-myosin system are being reinvestigated. Of the two actin genes in P. falciparum, actin-1 (pfact1) is ubiquitously expressed in all life-cycle stages and is thought to be required for erythrocyte invasion, although its functions during parasite development are unknown, and definitive in vivo characterisation during invasion is lacking. Results Here we have used a conditional Cre-lox system to investigate the functions of PfACT1 during P. falciparum blood-stage development and host cell invasion. We demonstrate that PfACT1 is crucially required for segregation of the plastid-like organelle, the apicoplast, and for efficient daughter cell separation during the final stages of cytokinesis. Surprisingly, we observe that egress from the host cell is not an actin-dependent process. Finally, we show that parasites lacking PfACT1 are capable of microneme secretion, attachment and formation of a junction with the erythrocyte, but are incapable of host cell invasion. Conclusions This study provides important mechanistic insights into the definitive essential functions of PfACT1 in P. falciparum, which are not only of biological interest, but owing to functional divergence from mammalian actins, could also form the basis for the development of novel therapeutics against apicomplexans. Electronic supplementary material The online version of this article (doi:10.1186/s12915-017-0406-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sujaan Das
- Wellcome Centre for Molecular Parasitology, Institute of Infection, Immunity & Inflammation, Glasgow Biomedical Research Centre, University of Glasgow, 120 University Place, Glasgow, G12 8TA, UK.
| | - Leandro Lemgruber
- Wellcome Centre for Molecular Parasitology, Institute of Infection, Immunity & Inflammation, Glasgow Biomedical Research Centre, University of Glasgow, 120 University Place, Glasgow, G12 8TA, UK
| | - Chwen L Tay
- Department of Life Sciences, Imperial College London, South Kensington, London, SW7 2AZ, UK
| | - Jake Baum
- Department of Life Sciences, Imperial College London, South Kensington, London, SW7 2AZ, UK
| | - Markus Meissner
- Wellcome Centre for Molecular Parasitology, Institute of Infection, Immunity & Inflammation, Glasgow Biomedical Research Centre, University of Glasgow, 120 University Place, Glasgow, G12 8TA, UK. .,Faculty of Veterinary Medicine, Ludwig-Maximilians-University Munich, Munich, Germany.
| |
Collapse
|
30
|
Tardieux I, Baum J. Reassessing the mechanics of parasite motility and host-cell invasion. J Cell Biol 2017; 214:507-15. [PMID: 27573462 PMCID: PMC5004448 DOI: 10.1083/jcb.201605100] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 08/09/2016] [Indexed: 12/20/2022] Open
Abstract
The capacity to migrate is fundamental to multicellular and single-celled life. Apicomplexan parasites, an ancient protozoan clade that includes malaria parasites (Plasmodium) and Toxoplasma, achieve remarkable speeds of directional cell movement. This rapidity is achieved via a divergent actomyosin motor system, housed within a narrow compartment that lies underneath the length of the parasite plasma membrane. How this motor functions at a mechanistic level during motility and host cell invasion is a matter of debate. Here, we integrate old and new insights toward refining the current model for the function of this motor with the aim of revitalizing interest in the mechanics of how these deadly pathogens move.
Collapse
Affiliation(s)
- Isabelle Tardieux
- Institute of Advanced BioSciences, Institut National de la Santé et de la Recherche Médicale U1209, Centre National de la Recherche Scientifique UMR 5309, Université Grenoble Alpes, 38000, Grenoble, France
| | - Jake Baum
- Department of Life Sciences, Imperial College London, London SW7 2AZ, England, UK
| |
Collapse
|
31
|
Plasmodium falciparum erythrocyte-binding antigen 175 triggers a biophysical change in the red blood cell that facilitates invasion. Proc Natl Acad Sci U S A 2017; 114:4225-4230. [PMID: 28373555 DOI: 10.1073/pnas.1620843114] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Invasion of the red blood cell (RBC) by the Plasmodium parasite defines the start of malaria disease pathogenesis. To date, experimental investigations into invasion have focused predominantly on the role of parasite adhesins or signaling pathways and the identity of binding receptors on the red cell surface. A potential role for signaling pathways within the erythrocyte, which might alter red cell biophysical properties to facilitate invasion, has largely been ignored. The parasite erythrocyte-binding antigen 175 (EBA175), a protein required for entry in most parasite strains, plays a key role by binding to glycophorin A (GPA) on the red cell surface, although the function of this binding interaction is unknown. Here, using real-time deformability cytometry and flicker spectroscopy to define biophysical properties of the erythrocyte, we show that EBA175 binding to GPA leads to an increase in the cytoskeletal tension of the red cell and a reduction in the bending modulus of the cell's membrane. We isolate the changes in the cytoskeleton and membrane and show that reduction in the bending modulus is directly correlated with parasite invasion efficiency. These data strongly imply that the malaria parasite primes the erythrocyte surface through its binding antigens, altering the biophysical nature of the target cell and thus reducing a critical energy barrier to invasion. This finding would constitute a major change in our concept of malaria parasite invasion, suggesting it is, in fact, a balance between parasite and host cell physical forces working together to facilitate entry.
Collapse
|
32
|
Abstract
Mutations in the genes encoding the mechanosensitive cation channels PIEZO1 and PIEZO2 are responsible for multiple hereditary human diseases. Loss-of-function mutations in the human PIEZO1 gene cause autosomal recessive congenital lymphatic dysplasia. Gain-of-function mutations in the human PIEZO1 gene cause the autosomal dominant hemolytic anemia, hereditary xerocytosis (also known as dehydrated stomatocytosis). Loss-of-function mutations in the human PIEZO2 gene cause an autosomal recessive syndrome of muscular atrophy with perinatal respiratory distress, arthrogryposis, and scoliosis. Gain-of-function mutations in the human PIEZO2 gene cause three clinical types of autosomal dominant distal arthrogryposis. This chapter will review the hereditary diseases caused by mutations in the PIEZO genes and will discuss additional physiological systems in which PIEZO channel dysfunction may contribute to human disease pathophysiology.
Collapse
Affiliation(s)
- S L Alper
- Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
33
|
Sisquella X, Nebl T, Thompson JK, Whitehead L, Malpede BM, Salinas ND, Rogers K, Tolia NH, Fleig A, O'Neill J, Tham WH, David Horgen F, Cowman AF. Plasmodium falciparum ligand binding to erythrocytes induce alterations in deformability essential for invasion. eLife 2017; 6. [PMID: 28226242 PMCID: PMC5333951 DOI: 10.7554/elife.21083] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 02/09/2017] [Indexed: 12/31/2022] Open
Abstract
The most lethal form of malaria in humans is caused by Plasmodium falciparum. These parasites invade erythrocytes, a complex process involving multiple ligand-receptor interactions. The parasite makes initial contact with the erythrocyte followed by dramatic deformations linked to the function of the Erythrocyte binding antigen family and P. falciparum reticulocyte binding-like families. We show EBA-175 mediates substantial changes in the deformability of erythrocytes by binding to glycophorin A and activating a phosphorylation cascade that includes erythrocyte cytoskeletal proteins resulting in changes in the viscoelastic properties of the host cell. TRPM7 kinase inhibitors FTY720 and waixenicin A block the changes in the deformability of erythrocytes and inhibit merozoite invasion by directly inhibiting the phosphorylation cascade. Therefore, binding of P. falciparum parasites to the erythrocyte directly activate a signaling pathway through a phosphorylation cascade and this alters the viscoelastic properties of the host membrane conditioning it for successful invasion.
Collapse
Affiliation(s)
- Xavier Sisquella
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Thomas Nebl
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Jennifer K Thompson
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Lachlan Whitehead
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Brian M Malpede
- Molecular Microbiology and Microbial Pathogenesis, Washington University School of Medicine, St. Louis, United States.,Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, United States
| | - Nichole D Salinas
- Molecular Microbiology and Microbial Pathogenesis, Washington University School of Medicine, St. Louis, United States.,Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, United States
| | - Kelly Rogers
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Niraj H Tolia
- Molecular Microbiology and Microbial Pathogenesis, Washington University School of Medicine, St. Louis, United States.,Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, United States
| | - Andrea Fleig
- The Queen's Medical Center and John A. Burns School of Medicine, University of Hawaii, Honolulu, United States
| | - Joseph O'Neill
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Wai-Hong Tham
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - F David Horgen
- Department of Natural Sciences, Hawaii Pacific University, Kaneohe, United States
| | - Alan F Cowman
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Australia
| |
Collapse
|
34
|
Kobayashi K, Kato K. Evaluating the use of heparin for synchronization of in vitro culture of Plasmodium falciparum. Parasitol Int 2016; 65:549-551. [PMID: 27600143 DOI: 10.1016/j.parint.2016.09.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 09/01/2016] [Accepted: 09/02/2016] [Indexed: 11/25/2022]
Abstract
The malaria parasite Plasmodium falciparum infects human erythrocytes and reproduces asexually through an intraerythrocytic developmental cycle. In vitro culture of P. falciparum allows investigation of the parasite's blood-stage development, which spans approximately 48h from the time of invasion to the lysis of mature schizonts to release merozoites. To focus on a specific step in the developmental cycle, synchronization techniques are utilized. d-Sorbitol treatment and the Percoll-sorbitol method have been used; however, these techniques have limitations in terms of the degree of synchronization achieved, the amount of synchronized parasite acquired, convenience, reproducibility, and cost. Here, we evaluated an existing synchronization method involving heparin. Heparin reversibly inhibits erythrocyte invasion by P. falciparum merozoites. We confirm that parasite cultures can be inexpensively, reproducibly, and tightly synchronized by combining a sorbitol step to limit cultures to the ring stages and by adding and removing heparin to manipulate the window during which merozoites can invade erythrocytes.
Collapse
Affiliation(s)
- Kyousuke Kobayashi
- Department of Veterinary Microbiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan; Division of Host-Parasite Interaction, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Kentaro Kato
- Department of Veterinary Microbiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan; National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Hokkaido 080-8555, Japan.
| |
Collapse
|
35
|
Carvalho TG, Morahan B, John von Freyend S, Boeuf P, Grau G, Garcia-Bustos J, Doerig C. The ins and outs of phosphosignalling in Plasmodium: Parasite regulation and host cell manipulation. Mol Biochem Parasitol 2016; 208:2-15. [PMID: 27211241 DOI: 10.1016/j.molbiopara.2016.05.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 05/16/2016] [Indexed: 12/15/2022]
Abstract
Signal transduction and kinomics have been rapidly expanding areas of investigation within the malaria research field. Here, we provide an overview of phosphosignalling pathways that operate in all stages of the Plasmodium life cycle. We review signalling pathways in the parasite itself, in the cells it invades, and in other cells of the vertebrate host with which it interacts. We also discuss the potential of these pathways as novel targets for antimalarial intervention.
Collapse
Affiliation(s)
- Teresa Gil Carvalho
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Victoria 3800, Australia
| | - Belinda Morahan
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Victoria 3800, Australia
| | - Simona John von Freyend
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Victoria 3800, Australia
| | - Philippe Boeuf
- Burnet Institute, Melbourne, Victoria 3004, Australia; The University of Melbourne, Department of Medicine, Melbourne, Victoria 3010, Australia; Victorian Infectious Diseases Service, Royal Melbourne Hospital, Melbourne, Victoria 3010, Australia
| | - Georges Grau
- Vascular Immunology Unit, Department of Pathology, Sydney Medical School, University of Sydney, Camperdown, New South Wales 2050, Australia
| | - Jose Garcia-Bustos
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Victoria 3800, Australia
| | - Christian Doerig
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Victoria 3800, Australia.
| |
Collapse
|
36
|
Plasmodiumfalciparum infection induces dynamic changes in the erythrocyte phospho-proteome. Blood Cells Mol Dis 2016; 58:35-44. [PMID: 27067487 DOI: 10.1016/j.bcmd.2016.02.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 02/05/2016] [Indexed: 11/21/2022]
Abstract
The phosphorylation status of red blood cell proteins is strongly altered during the infection by the malaria parasite Plasmodium falciparum. We identify the key phosphorylation events that occur in the erythrocyte membrane and cytoskeleton during infection, by a comparative analysis of global phospho-proteome screens between infected (obtained at schizont stage) and uninfected RBCs. The meta-analysis of reported mass spectrometry studies revealed a novel compendium of 495 phosphorylation sites in 182 human proteins with regulatory roles in red cell morphology and stability, with about 25% of these sites specific to infected cells. A phosphorylation motif analysis detected 7 unique motifs that were largely mapped to kinase consensus sequences of casein kinase II and of protein kinase A/protein kinase C. This analysis highlighted prominent roles for PKA/PKC involving 78 phosphorylation sites. We then compared the phosphorylation status of PKA (PKC) specific sites in adducin, dematin, Band 3 and GLUT-1 in uninfected RBC stimulated or not by cAMP to their phosphorylation status in iRBC. We showed cAMP-induced phosphorylation of adducin S59 by immunoblotting and we were able to demonstrate parasite-induced phosphorylation for adducin S726, Band 3 and GLUT-1, corroborating the protein phosphorylation status in our erythrocyte phosphorylation site compendium.
Collapse
|
37
|
Koch M, Baum J. The mechanics of malaria parasite invasion of the human erythrocyte - towards a reassessment of the host cell contribution. Cell Microbiol 2016; 18:319-29. [PMID: 26663815 PMCID: PMC4819681 DOI: 10.1111/cmi.12557] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Revised: 11/30/2015] [Accepted: 12/07/2015] [Indexed: 01/15/2023]
Abstract
Despite decades of research, we still know little about the mechanics of Plasmodium host cell invasion. Fundamentally, while the essential or non‐essential nature of different parasite proteins is becoming clearer, their actual function and how each comes together to govern invasion are poorly understood. Furthermore, in recent years an emerging world view is shifting focus away from the parasite actin–myosin motor being the sole force responsible for entry to an appreciation of host cell dynamics and forces and their contribution to the process. In this review, we discuss merozoite invasion of the erythrocyte, focusing on the complex set of pre‐invasion events and how these might prime the red cell to facilitate invasion. While traditionally parasite interactions at this stage have been viewed simplistically as mediating adhesion only, recent work makes it apparent that by interacting with a number of host receptors and signalling pathways, combined with secretion of parasite‐derived lipid material, that the merozoite may initiate cytoskeletal re‐arrangements and biophysical changes in the erythrocyte that greatly reduce energy barriers for entry. Seen in this light Plasmodium invasion may well turn out to be a balance between host and parasite forces, much like that of other pathogen infection mechanisms.
Collapse
Affiliation(s)
- Marion Koch
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, Exhibition Road, South Kensington, London, SW7 2AZ, UK
| | - Jake Baum
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, Exhibition Road, South Kensington, London, SW7 2AZ, UK
| |
Collapse
|