1
|
Kelly B, Boudreau JE, Beyea S, Brewer K. Molecular imaging of viral pathogenesis and opportunities for the future. NPJ IMAGING 2025; 3:3. [PMID: 39872292 PMCID: PMC11761071 DOI: 10.1038/s44303-024-00056-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 10/24/2024] [Indexed: 01/30/2025]
Abstract
Molecular imaging is used in clinical and research settings. Since tools to study viral pathogenesis longitudinally and systemically are limited, molecular imaging is an attractive and largely unexplored tool. This review discusses molecular imaging probes and techniques for studying viruses, particularly those currently used in oncology that are applicable to virology. Expanding the repertoire of probes to better detect viral disease may make imaging even more valuable in (pre-)clinical settings.
Collapse
Affiliation(s)
- Brianna Kelly
- Biomedical MRI Research Laboratory (BMRL), IWK Health Centre, Halifax, NS Canada
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS Canada
| | - Jeanette E. Boudreau
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS Canada
- Department of Pathology, Dalhousie University, Halifax, NS Canada
- Beatrice Hunter Cancer Research Institute (BHCRI), Halifax, NS Canada
| | - Steven Beyea
- IWK Health Centre, Halifax, NS Canada
- Department of Diagnostic Radiology, Dalhousie University, Halifax, NS Canada
- School of Biomedical Engineering, Dalhousie University, Halifax, NS Canada
- Department of Physics & Atmospheric Science, Dalhousie University, Halifax, NS Canada
| | - Kimberly Brewer
- Biomedical MRI Research Laboratory (BMRL), IWK Health Centre, Halifax, NS Canada
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS Canada
- IWK Health Centre, Halifax, NS Canada
- Department of Diagnostic Radiology, Dalhousie University, Halifax, NS Canada
- School of Biomedical Engineering, Dalhousie University, Halifax, NS Canada
- Department of Physics & Atmospheric Science, Dalhousie University, Halifax, NS Canada
| |
Collapse
|
2
|
de Souza Cardoso R, Ono A. The Effects of Viral Structural Proteins on Acidic Phospholipids in Host Membranes. Viruses 2024; 16:1714. [PMID: 39599829 PMCID: PMC11599007 DOI: 10.3390/v16111714] [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: 09/13/2024] [Revised: 10/23/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024] Open
Abstract
Enveloped viruses rely on host membranes for trafficking and assembly. A substantial body of literature published over the years supports the involvement of cellular membrane lipids in the enveloped virus assembly processes. In particular, the knowledge regarding the relationship between viral structural proteins and acidic phospholipids has been steadily increasing in recent years. In this review, we will briefly review the cellular functions of plasma membrane-associated acidic phospholipids and the mechanisms that regulate their local distribution within this membrane. We will then explore the interplay between viruses and the plasma membrane acidic phospholipids in the context of the assembly process for two enveloped viruses, the influenza A virus (IAV) and the human immunodeficiency virus type 1 (HIV-1). Among the proteins encoded by these viruses, three viral structural proteins, IAV hemagglutinin (HA), IAV matrix protein-1 (M1), and HIV-1 Gag protein, are known to interact with acidic phospholipids, phosphatidylserine and/or phosphatidylinositol (4,5)-bisphosphate. These interactions regulate the localization of the viral proteins to and/or within the plasma membrane and likely facilitate the clustering of the proteins. On the other hand, these viral proteins, via their ability to multimerize, can also alter the distribution of the lipids and may induce acidic-lipid-enriched membrane domains. We will discuss the potential significance of these interactions in the virus assembly process and the property of the progeny virions. Finally, we will outline key outstanding questions that need to be answered for a better understanding of the relationships between enveloped virus assembly and acidic phospholipids.
Collapse
Affiliation(s)
| | - Akira Ono
- Department of Microbiology and Immunology, The University of Michigan, Ann Arbor, MI 48109, USA;
| |
Collapse
|
3
|
Banerjee P, Qu K, Briggs JAG, Voth GA. Molecular dynamics simulations of HIV-1 matrix-membrane interactions at different stages of viral maturation. Biophys J 2024; 123:389-406. [PMID: 38196190 PMCID: PMC10870173 DOI: 10.1016/j.bpj.2024.01.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/05/2023] [Accepted: 01/04/2024] [Indexed: 01/11/2024] Open
Abstract
Although the structural rearrangement of the membrane-bound matrix (MA) protein trimers upon HIV-1 maturation has been reported, the consequences of MA maturation on the MA-lipid interactions are not well understood. Long-timescale molecular dynamics simulations of the MA multimeric assemblies of immature and mature virus particles with our realistic asymmetric membrane model have explored MA-lipid interactions and lateral organization of lipids around MA complexes. The number of stable MA-phosphatidylserine and MA-phosphatidylinositol 4,5-bisphosphate (PIP2) interactions at the trimeric interface of the mature MA complex is observed to be greater compared to that of the immature MA complex. Our simulations identified an alternative PIP2-binding site in the immature MA complex where the multivalent headgroup of a PIP2 lipid with a greater negative charge binds to multiple basic amino acid residues such as ARG3 residues of both the MA monomers at the trimeric interface and highly basic region (HBR) residues (LYS29, LYS31) of one of the MA monomers. Our enhanced sampling simulations have explored the conformational space of phospholipids at different binding sites of the trimer-trimer interface of MA complexes that are not accessible by conventional unbiased molecular dynamics. Unlike the immature MA complex, the 2' acyl tail of two PIP2 lipids at the trimeric interface of the mature MA complex is observed to sample stable binding pockets of MA consisting of helix-4 residues. Together, our results provide molecular-level insights into the interactions of MA trimeric complexes with membrane and different lipid conformations at the specific binding sites of MA protein before and after viral maturation.
Collapse
Affiliation(s)
- Puja Banerjee
- Department of Chemistry, Chicago Center for Theoretical Chemistry, Institute for Biophysical Dynamics, and James Franck Institute, The University of Chicago, Chicago, Illinois
| | - Kun Qu
- Infectious Diseases Translational Research Programme, Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - John A G Briggs
- Department of Cell and Virus Structure, Max Planck Institute of Biochemistry, Planegg, Germany
| | - Gregory A Voth
- Department of Chemistry, Chicago Center for Theoretical Chemistry, Institute for Biophysical Dynamics, and James Franck Institute, The University of Chicago, Chicago, Illinois.
| |
Collapse
|
4
|
Kawano K, Hashikura Y, Umekita K. Purification Method of Extracellular Vesicles Derived from Human T-Cell Leukemia Virus Type 1-Infected Cells without Virions. Viruses 2024; 16:249. [PMID: 38400025 PMCID: PMC10892183 DOI: 10.3390/v16020249] [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: 11/17/2023] [Revised: 01/19/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
To mediate intercellular communication, cells produce extracellular vesicles (EVs). These EVs transport many biomolecules such as proteins, nucleic acids, and lipids between cells and regulate pathophysiological actions in the recipient cell. However, EVs and virus particles produced from virus-infected cells are of similar size and specific gravity; therefore, the separation and purification of these two particles is often controversial. When analyzing the physiological functions of EVs from virus-infected cells, the presence or absence of virus particle contamination must always be verified. The human T-cell leukemia virus type 1 (HTLV-1)-infected cell line, MT-2, produces EVs and virus particles. Here, we validated a method for purifying EVs from MT-2 cell culture supernatants while avoiding HTLV-1 viral particle contamination. EV fractions were collected using a combination of immunoprecipitation with Tim-4, which binds to phosphatidylserine, and polymer precipitation. The HTLV-1 viral envelope protein, gp46, was not detected in the EV fraction. Proteomic analysis revealed that EV-constituted proteins were predominant in this EV fraction. Furthermore, the EVs were found to contain the HTLV-1 viral genome. The proposed method can purify EVs while avoiding virus particle contamination and is expected to contribute to future research on EVs derived from HTLV-1-infected cells.
Collapse
Affiliation(s)
- Katsumi Kawano
- Division of Respirology, Rheumatology, Infectious Diseases and Neurology, Department of Internal Medicine, University of Miyazaki, Kihara 5200, Kiyotake, Miyazaki 889-1692, Japan;
- Clinical Laboratory, University of Miyazaki of Hospital, Kihara 5200, Kiyotake, Miyazaki 889-1692, Japan;
| | - Yuki Hashikura
- Clinical Laboratory, University of Miyazaki of Hospital, Kihara 5200, Kiyotake, Miyazaki 889-1692, Japan;
| | - Kunihiko Umekita
- Division of Respirology, Rheumatology, Infectious Diseases and Neurology, Department of Internal Medicine, University of Miyazaki, Kihara 5200, Kiyotake, Miyazaki 889-1692, Japan;
- Clinical Laboratory, University of Miyazaki of Hospital, Kihara 5200, Kiyotake, Miyazaki 889-1692, Japan;
| |
Collapse
|
5
|
Orosco FL. Host immune responses against African swine fever virus: Insights and challenges for vaccine development. Open Vet J 2023; 13:1517-1535. [PMID: 38292721 PMCID: PMC10824091 DOI: 10.5455/ovj.2023.v13.i12.2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 11/22/2023] [Indexed: 02/01/2024] Open
Abstract
The African swine fever virus (ASFV) poses a serious threat to global swine populations, underscoring the urgent need for effective preventive strategies. This comprehensive review investigates the intricate interplay between innate, cellular, and humoral immunity against ASFV, with a focus on their relevance to vaccine development. By delving into immunopathogenesis and immunological challenges, this review article aims to provide a holistic perspective on the complexities of ASFV infections and immune evasion. Key findings underscore the critical role of innate immune recognition in shaping subsequent adaptive immune defenses, potential protective antigens, and the multifaceted nature of ASFV-specific antibodies and cytotoxic T-cell responses. Despite advancements, the unique attributes of ASFV present hurdles in the development of a successful vaccine. In conclusion, this review examines the current state of ASFV immune responses and offers insights into future research directions, fostering the development of effective interventions against this devastating pathogen.
Collapse
Affiliation(s)
- Fredmoore L. Orosco
- Virology and Vaccine Institute of the Philippines Program, Department of Science and Technology, Industrial Technology Development Institute, Taguig, Philippines
- S&T Fellows Program, Department of Science and Technology, Taguig, Philippines
- Department of Biology, College of Arts and Sciences, University of the Philippines Manila, Manila, Philippines
| |
Collapse
|
6
|
Mohammad-Rafiei F, Moadab F, Mahmoudi A, Navashenaq JG, Gheibihayat SM. Efferocytosis: a double-edged sword in microbial immunity. Arch Microbiol 2023; 205:370. [PMID: 37925389 DOI: 10.1007/s00203-023-03704-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 09/30/2023] [Accepted: 10/10/2023] [Indexed: 11/06/2023]
Abstract
Efferocytosis is characterized as the rapid and efficient process by which dying or dead cells are removed. This type of clearance is initiated via "find-me" signals, and then, carries on by "eat-me" and "don't-eat-me" ones. Efferocytosis has a critical role to play in tissue homeostasis and innate immunity. However, some evidence suggests it as a double-edged sword in microbial immunity. In other words, some pathogens have degraded efferocytosis by employing efferocytic mechanisms to bypass innate immune detection and promote infection, despite the function of this process for the control and clearance of pathogens. In this review, the efferocytosis mechanisms from the recognition of dying cells to phagocytic engulfment are initially presented, and then, its diverse roles in inflammation and immunity are highlighted. In this case, much focus is also laid on some bacterial, viral, and parasitic infections caused by Mycobacterium tuberculosis (M. tb), Mycobacterium marinum (M. marinum), Listeria monocytogenes (L. monocytogenes), Chlamydia pneumoniae (CP), Klebsiella pneumoniae (KP), Influenza A virus (IAV), human immunodeficiency virus (HIV), and Leishmania, respectively.
Collapse
Affiliation(s)
- Fatemeh Mohammad-Rafiei
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fatemeh Moadab
- Division of Rheumatology, Department of Medicine, University of Washington, Seattle, USA
| | - Ali Mahmoudi
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Science, Mashhad, Iran
| | | | - Seyed Mohammad Gheibihayat
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
7
|
Reis AL, Rathakrishnan A, Goulding LV, Barber C, Goatley LC, Dixon LK. Deletion of the gene for the African swine fever virus BCL-2 family member A179L increases virus uptake and apoptosis but decreases virus spread in macrophages and reduces virulence in pigs. J Virol 2023; 97:e0110623. [PMID: 37796125 PMCID: PMC10617521 DOI: 10.1128/jvi.01106-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 08/23/2023] [Indexed: 10/06/2023] Open
Abstract
IMPORTANCE African swine fever virus (ASFV) causes a lethal disease of pigs with high economic impact in affected countries in Africa, Europe, and Asia. The virus encodes proteins that inhibit host antiviral defenses, including the type I interferon response. Host cells also activate cell death through a process called apoptosis to limit virus replication. We showed that the ASFV A179L protein, a BCL-2 family apoptosis inhibitor, is important in reducing apoptosis in infected cells since deletion of this gene increased cell death and reduced virus replication in cells infected with the A179L gene-deleted virus. Pigs immunized with the BeninΔA179L virus showed no clinical signs and a weak immune response but were not protected from infection with the deadly parental virus. The results show an important role for the A179L protein in virus replication in macrophages and virulence in pigs and suggest manipulation of apoptosis as a possible route to control infection.
Collapse
Affiliation(s)
| | | | | | - Claire Barber
- The Pirbright Institute, Woking, Surrey, United Kingdom
| | | | | |
Collapse
|
8
|
Herrera SA, Günther Pomorski T. Reconstitution of ATP-dependent lipid transporters: gaining insight into molecular characteristics, regulation, and mechanisms. Biosci Rep 2023; 43:BSR20221268. [PMID: 37417269 PMCID: PMC10412526 DOI: 10.1042/bsr20221268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/30/2023] [Accepted: 07/06/2023] [Indexed: 07/08/2023] Open
Abstract
Lipid transporters play a crucial role in supporting essential cellular processes such as organelle assembly, vesicular trafficking, and lipid homeostasis by driving lipid transport across membranes. Cryo-electron microscopy has recently resolved the structures of several ATP-dependent lipid transporters, but functional characterization remains a major challenge. Although studies of detergent-purified proteins have advanced our understanding of these transporters, in vitro evidence for lipid transport is still limited to a few ATP-dependent lipid transporters. Reconstitution into model membranes, such as liposomes, is a suitable approach to study lipid transporters in vitro and to investigate their key molecular features. In this review, we discuss the current approaches for reconstituting ATP-driven lipid transporters into large liposomes and common techniques used to study lipid transport in proteoliposomes. We also highlight the existing knowledge on the regulatory mechanisms that modulate the activity of lipid transporters, and finally, we address the limitations of the current approaches and future perspectives in this field.
Collapse
Affiliation(s)
- Sara Abad Herrera
- Department of Molecular Biochemistry, Faculty of Chemistry and Biochemistry, Ruhr University Bochum, Bochum, Germany
| | - Thomas Günther Pomorski
- Department of Molecular Biochemistry, Faculty of Chemistry and Biochemistry, Ruhr University Bochum, Bochum, Germany
- Department of Plant and Environmental Sciences, University of Copenhagen, Frederiksberg, Denmark
| |
Collapse
|
9
|
Ward AE, Sokovikova D, Waxham MN, Heberle FA, Levental I, Levental KR, Kiessling V, White JM, Tamm LK. Serinc5 Restricts HIV Membrane Fusion by Altering Lipid Order and Heterogeneity in the Viral Membrane. ACS Infect Dis 2023; 9:773-784. [PMID: 36946615 PMCID: PMC10366416 DOI: 10.1021/acsinfecdis.2c00478] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Abstract
The host restriction factor, Serinc5, incorporates into budding HIV particles and inhibits their infection by an incompletely understood mechanism. We have previously reported that Serinc5 but not its paralogue, Serinc2, blocks HIV cell entry by membrane fusion, specifically by inhibiting fusion pore formation and dilation. A body of work suggests that Serinc5 may alter the conformation and clustering of the HIV fusion protein, Env. To contribute an additional perspective to the developing model of Serinc5 restriction, we assessed Serinc2 and Serinc5's effects on HIV pseudoviral membranes. By measuring pseudoviral membrane thickness via cryo-electron microscopy and order via the fluorescent dye, FLIPPER-TR, Serinc5 was found to increase membrane heterogeneity, skewing the distribution toward a larger fraction of the viral membrane in an ordered phase. We also directly observed for the first time the coexistence of membrane domains within individual viral membrane envelopes. Using a total internal reflection fluorescence-based single particle fusion assay, we found that treatment of HIV pseudoviral particles with phosphatidylethanolamine (PE) rescued HIV pseudovirus fusion from restriction by Serinc5, which was accompanied by decreased membrane heterogeneity and order. This effect was specific for PE and did not depend on acyl chain length or saturation. Together, these data suggest that Serinc5 alters multiple interrelated properties of the viral membrane─lipid chain order, rigidity, line tension, and lateral pressure─which decrease the accessibility of fusion intermediates and disfavor completion of fusion. These biophysical insights into Serinc5 restriction of HIV infectivity could contribute to the development of novel antivirals that exploit the same weaknesses.
Collapse
Affiliation(s)
- Amanda E. Ward
- Department of Molecular Physiology and Biological Physics, University of Virginia Health System, Charlottesville, VA 22908
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA 22908
| | - Daria Sokovikova
- Department of Molecular Physiology and Biological Physics, University of Virginia Health System, Charlottesville, VA 22908
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA 22908
| | - Melvin Neal Waxham
- Department of Neurobiology and Anatomy, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX 77030
| | | | - Ilya Levental
- Department of Molecular Physiology and Biological Physics, University of Virginia Health System, Charlottesville, VA 22908
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA 22908
| | - Kandice R. Levental
- Department of Molecular Physiology and Biological Physics, University of Virginia Health System, Charlottesville, VA 22908
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA 22908
| | - Volker Kiessling
- Department of Molecular Physiology and Biological Physics, University of Virginia Health System, Charlottesville, VA 22908
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA 22908
| | - Judith M. White
- Department of Cell Biology, University of Virginia Health System, Charlottesville, VA 22908
| | - Lukas K. Tamm
- Department of Molecular Physiology and Biological Physics, University of Virginia Health System, Charlottesville, VA 22908
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA 22908
| |
Collapse
|
10
|
Corey D, Haeseleer F, Hou J, Corey L. Novel engineered chimeric engulfment receptors trigger T cell effector functions against SIV-infected CD4+ T cells. Mol Ther Methods Clin Dev 2023; 28:1-10. [PMID: 36514789 PMCID: PMC9720250 DOI: 10.1016/j.omtm.2022.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 11/14/2022] [Indexed: 11/17/2022]
Abstract
Adoptive therapy with genetically engineered T cells offers potential for infectious disease treatment in immunocompromised persons. HIV/simian immunodeficiency virus (SIV)-infected cells express phosphatidylserine (PS) early post infection. We tested whether chimeric engulfment receptor (CER) T cells designed to recognize PS-expressing cells could eliminate SIV-infected cells. Lentiviral CER constructs composed of the extracellular domain of T cell immunoglobulin and mucin domain containing 4 (TIM-4), the PS receptor, and engulfment signaling domains were transduced into primary rhesus macaque (RM) T cells. We measured PS binding and T cell engulfment of RM CD4+ T cells infected with SIV expressing GFP and in vitro, TIM-4 CER CD4+ T cells effectively killed SIV-infected cells, which was dependent on TIM-4 binding to PS. Enhanced killing of SIV-infected CD4+ T cells by CER and chimeric antigen receptor T cell combinations was also observed. This installation of innate immune functions into T cells presents an opportunity to enhance elimination of SIV-infected cells, and studies to evaluate their effect in vivo are warranted.
Collapse
Affiliation(s)
- Daniel Corey
- CERo Therapeutics, 201 Haskins Way, Suite 230, San Francisco, CA 94080, USA
| | - Francoise Haeseleer
- Department of Laboratory Medicine, University of Washington, Seattle, WA 98195, USA
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Joe Hou
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Lawrence Corey
- Department of Laboratory Medicine, University of Washington, Seattle, WA 98195, USA
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| |
Collapse
|
11
|
Targeting Human Proteins for Antiviral Drug Discovery and Repurposing Efforts: A Focus on Protein Kinases. Viruses 2023; 15:v15020568. [PMID: 36851782 PMCID: PMC9966946 DOI: 10.3390/v15020568] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/07/2023] [Accepted: 02/09/2023] [Indexed: 02/22/2023] Open
Abstract
Despite the great technological and medical advances in fighting viral diseases, new therapies for most of them are still lacking, and existing antivirals suffer from major limitations regarding drug resistance and a limited spectrum of activity. In fact, most approved antivirals are directly acting antiviral (DAA) drugs, which interfere with viral proteins and confer great selectivity towards their viral targets but suffer from resistance and limited spectrum. Nowadays, host-targeted antivirals (HTAs) are on the rise, in the drug discovery and development pipelines, in academia and in the pharmaceutical industry. These drugs target host proteins involved in the virus life cycle and are considered promising alternatives to DAAs due to their broader spectrum and lower potential for resistance. Herein, we discuss an important class of HTAs that modulate signal transduction pathways by targeting host kinases. Kinases are considered key enzymes that control virus-host interactions. We also provide a synopsis of the antiviral drug discovery and development pipeline detailing antiviral kinase targets, drug types, therapeutic classes for repurposed drugs, and top developing organizations. Furthermore, we detail the drug design and repurposing considerations, as well as the limitations and challenges, for kinase-targeted antivirals, including the choice of the binding sites, physicochemical properties, and drug combinations.
Collapse
|
12
|
Brinkhuizen C, Shapman D, Lebon A, Bénard M, Tardivel M, Dubuquoy L, Galas L, Carpentier R. Dipalmitoyl-phosphatidylserine-filled cationic maltodextrin nanoparticles exhibit enhanced efficacy for cell entry and intracellular protein delivery in phagocytic THP-1 cells. Biomol Concepts 2023; 14:bmc-2022-0029. [PMID: 37377352 DOI: 10.1515/bmc-2022-0029] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 05/08/2023] [Indexed: 06/29/2023] Open
Abstract
Vaccination through the upper respiratory tract is a promising strategy, and particulate antigens, such as antigens associated with nanoparticles, triggered a stronger immune response than the sole antigens. Cationic maltodextrin-based nanoparticles loaded with phosphatidylglycerol (NPPG) are efficient for intranasal vaccination but non-specific to trigger immune cells. Here we focused on phosphatidylserine (PS) receptors, specifically expressed by immune cells including macrophages, to improve nanoparticle targeting through an efferocytosis-like mechanism. Consequently, the lipids associated with NPPG have been substituted by PS to generate cationic maltodextrin-based nanoparticles with dipalmitoyl-phosphatidylserine (NPPS). Both NPPS and NPPG exhibited similar physical characteristics and intracellular distribution in THP-1 macrophages. NPPS cell entry was faster and higher (two times more) than NPPG. Surprisingly, competition of PS receptors with phospho-L-serine did not alter NPPS cell entry and annexin V did not preferentially interact with NPPS. Although the protein association is similar, NPPS delivered more proteins than NPPG in cells. On the contrary, the proportion of mobile nanoparticles (50%), the movement speed of nanoparticles (3 µm/5 min), and protein degradation kinetics in THP-1 were not affected by lipid substitution. Together, the results indicate that NPPS enter cells and deliver protein better than NPPG, suggesting that modification of the lipids of cationic maltodextrin-based nanoparticles may be a useful strategy to enhance nanoparticle efficacy for mucosal vaccination.
Collapse
Affiliation(s)
- Clément Brinkhuizen
- University Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Damien Shapman
- University of Rouen Normandy, INSERM US 51, CNRS UAR 2026, HeRacLeS-PRIMACEN, Normandie Université, 76000 Rouen, France
| | - Alexis Lebon
- University of Rouen Normandy, INSERM US 51, CNRS UAR 2026, HeRacLeS-PRIMACEN, Normandie Université, 76000 Rouen, France
| | - Magalie Bénard
- University of Rouen Normandy, INSERM US 51, CNRS UAR 2026, HeRacLeS-PRIMACEN, Normandie Université, 76000 Rouen, France
| | - Meryem Tardivel
- University Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US 41 - UAR 2014 - PLBS, F-59000 Lille, France
| | - Laurent Dubuquoy
- University Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Ludovic Galas
- University of Rouen Normandy, INSERM US 51, CNRS UAR 2026, HeRacLeS-PRIMACEN, Normandie Université, 76000 Rouen, France
| | - Rodolphe Carpentier
- University Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000 Lille, France
| |
Collapse
|
13
|
African Swine Fever Vaccinology: The Biological Challenges from Immunological Perspectives. Viruses 2022; 14:v14092021. [PMID: 36146827 PMCID: PMC9505361 DOI: 10.3390/v14092021] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 08/22/2022] [Accepted: 09/08/2022] [Indexed: 11/17/2022] Open
Abstract
African swine fever virus (ASFV), a nucleocytoplasmic large DNA virus (NCLDV), causes African swine fever (ASF), an acute hemorrhagic disease with mortality rates up to 100% in domestic pigs. ASF is currently epidemic or endemic in many countries and threatening the global swine industry. Extensive ASF vaccine research has been conducted since the 1920s. Like inactivated viruses of other NCLDVs, such as vaccinia virus, inactivated ASFV vaccine candidates did not induce protective immunity. However, inactivated lumpy skin disease virus (poxvirus) vaccines are protective in cattle. Unlike some experimental poxvirus subunit vaccines that induced protection, ASF subunit vaccine candidates implemented with various platforms containing several ASFV structural genes or proteins failed to protect pigs effectively. Only some live attenuated viruses (LAVs) are able to protect pigs with high degrees of efficacy. There are currently several LAV ASF vaccine candidates. Only one commercial LAV vaccine is approved for use in Vietnam. LAVs, as ASF vaccines, have not yet been widely tested. Reports thus far show that the onset and duration of protection induced by the LAVs are late and short, respectively, compared to LAV vaccines for other diseases. In this review, the biological challenges in the development of ASF vaccines, especially subunit platforms, are discussed from immunological perspectives based on several unusual ASFV characteristics shared with HIV and poxviruses. These characteristics, including multiple distinct infectious virions, extremely high glycosylation and low antigen surface density of envelope proteins, immune evasion, and possible apoptotic mimicry, could pose enormous challenges to the development of ASF vaccines, especially subunit platforms designed to induce humoral immunity.
Collapse
|
14
|
Lao D, Liu R, Liang J. Study on plasma metabolomics for HIV/AIDS patients treated by HAART based on LC/MS-MS. Front Pharmacol 2022; 13:885386. [PMID: 36105186 PMCID: PMC9465010 DOI: 10.3389/fphar.2022.885386] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 07/21/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Metabolomics can be applied to the clinical diagnosis and treatment evaluation of acquired immune deficiency syndrome (AIDS). AIDS biomarkers have become a new direction of AIDS research providing clinical guidance for diagnosis. Objective: We sought to apply both untargeted and targeted metabolomic profiling to identify potential biomarkers for AIDS patients. Methods: A liquid chromatography-tandem mass spectrometry (LC-MS/MS) based untargeted metabolomic profiling was performed on plasma samples of patients before and after highly active antiretroviral therapy (HAART) treatment as well as healthy volunteers to identify potential AIDS biomarkers. Targeted quantitative analysis was performed on the potential biomarkers screened from untargeted metabolic profiling for verification. Results: Using the Mass Profiler Professional and the MassHunter, several potential biomarkers have been found by LC-MS/MS in the untargeted metabolomic study. High-resolution MS and MS/MS were used to analyze fragmentation rules of the metabolites, with comparisons of related standards. Several potential biomarkers have been identified, including PS(O-18:0/0:0), sphingosine, PE (21:0/0:0), and 1-Linoleoyl Glycerol. Targeted quantitative analysis showed that sphingosine and 1-Linoleoyl Glycerol might be closely related to HIV/AIDS, which may be the potential biomarkers to the diagnosis. Conclusion: We conducted untargeted metabolomic profiling, which indicates that several metabolites should be considered potential biomarkers for AIDS patients. Further targeted metabolomic research verified that d-Sphingosine and 1-Linoleoyl glycerol as the diagnostic biomarker of AIDS.
Collapse
Affiliation(s)
- Donghui Lao
- Department of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Rong Liu
- Department of Pharmacy, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | | |
Collapse
|
15
|
Ma X, Li X, Wang W, Zhang M, Yang B, Miao Z. Phosphatidylserine, inflammation, and central nervous system diseases. Front Aging Neurosci 2022; 14:975176. [PMID: 35992593 PMCID: PMC9382310 DOI: 10.3389/fnagi.2022.975176] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 07/15/2022] [Indexed: 11/13/2022] Open
Abstract
Phosphatidylserine (PS) is an anionic phospholipid in the eukaryotic membrane and is abundant in the brain. Accumulated studies have revealed that PS is involved in the multiple functions of the brain, such as activation of membrane signaling pathways, neuroinflammation, neurotransmission, and synaptic refinement. Those functions of PS are related to central nervous system (CNS) diseases. In this review, we discuss the metabolism of PS, the anti-inflammation function of PS in the brain; the alterations of PS in different CNS diseases, and the possibility of PS to serve as a therapeutic agent for diseases. Clinical studies have showed that PS has no side effects and is well tolerated. Therefore, PS and PS liposome could be a promising supplementation for these neurodegenerative and neurodevelopmental diseases.
Collapse
Affiliation(s)
- Xiaohua Ma
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Neuroscience, Soochow University, Suzhou, China
| | - Xiaojing Li
- Suzhou Science and Technology Town Hospital, Suzhou, China
| | - Wenjuan Wang
- Institute of Neuroscience, Soochow University, Suzhou, China
| | - Meng Zhang
- Institute of Neuroscience, Soochow University, Suzhou, China
| | - Bo Yang
- Department of Anesthesiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- *Correspondence: Bo Yang,
| | - Zhigang Miao
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Neuroscience, Soochow University, Suzhou, China
- Zhigang Miao,
| |
Collapse
|
16
|
Canter JA, Aponte T, Ramirez-Medina E, Pruitt S, Gladue DP, Borca MV, Zhu JJ. Serum Neutralizing and Enhancing Effects on African Swine Fever Virus Infectivity in Adherent Pig PBMC. Viruses 2022; 14:v14061249. [PMID: 35746720 PMCID: PMC9229155 DOI: 10.3390/v14061249] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/03/2022] [Accepted: 06/07/2022] [Indexed: 12/28/2022] Open
Abstract
African swine fever virus (ASFV) causes hemorrhagic fever with mortality rates of up to 100% in domestic pigs. Currently, there are no commercial vaccines for the disease. Only some live-attenuated viruses have been able to protect pigs from ASFV infection. The immune mechanisms involved in the protection are unclear. Immune sera can neutralize ASFV but incompletely. The mechanisms involved are not fully understood. Currently, there is no standardized protocol for ASFV neutralization assays. In this study, a flow cytometry-based ASFV neutralization assay was developed and tested in pig adherent PBMC using a virulent ASFV containing a fluorescent protein gene as a substrate for neutralization. As with previous studies, the percentage of infected macrophages was approximately five time higher than that of infected monocytes, and nearly all infected cells displayed no staining with anti-CD16 antibodies. Sera from naïve pigs and pigs immunized with a live-attenuated ASFV and fully protected against parental virus were used in the assay. The sera displayed incomplete neutralization with MOI-dependent neutralizing efficacies. Extracellular, but not intracellular, virions suspended in naïve serum were more infectious than those in the culture medium, as reported for some enveloped viruses, suggesting a novel mechanism of ASFV infection in macrophages. Both the intracellular and extracellular virions could not be completely neutralized.
Collapse
Affiliation(s)
- Jessica A. Canter
- Foreign Animal Disease Research Unit, Plum Island Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Orient, NY 11957, USA; (J.A.C.); (T.A.); (E.R.-M.); (S.P.)
- Plum Island Animal Disease Center, Oak Ridge Institute for Science and Education, Oak Ridge, TN 37830, USA
| | - Theresa Aponte
- Foreign Animal Disease Research Unit, Plum Island Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Orient, NY 11957, USA; (J.A.C.); (T.A.); (E.R.-M.); (S.P.)
- Plum Island Animal Disease Center, Oak Ridge Institute for Science and Education, Oak Ridge, TN 37830, USA
| | - Elizabeth Ramirez-Medina
- Foreign Animal Disease Research Unit, Plum Island Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Orient, NY 11957, USA; (J.A.C.); (T.A.); (E.R.-M.); (S.P.)
| | - Sarah Pruitt
- Foreign Animal Disease Research Unit, Plum Island Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Orient, NY 11957, USA; (J.A.C.); (T.A.); (E.R.-M.); (S.P.)
| | - Douglas P. Gladue
- Foreign Animal Disease Research Unit, Plum Island Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Orient, NY 11957, USA; (J.A.C.); (T.A.); (E.R.-M.); (S.P.)
- Correspondence: (D.P.G.); (M.V.B.); (J.J.Z.); Tel.: +1-631-323-3131 (D.P.G.); +1-631-323-3035 (M.V.B.); +1-631-323-3186 (J.J.Z.)
| | - Manuel V. Borca
- Foreign Animal Disease Research Unit, Plum Island Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Orient, NY 11957, USA; (J.A.C.); (T.A.); (E.R.-M.); (S.P.)
- Correspondence: (D.P.G.); (M.V.B.); (J.J.Z.); Tel.: +1-631-323-3131 (D.P.G.); +1-631-323-3035 (M.V.B.); +1-631-323-3186 (J.J.Z.)
| | - James J. Zhu
- Foreign Animal Disease Research Unit, Plum Island Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Orient, NY 11957, USA; (J.A.C.); (T.A.); (E.R.-M.); (S.P.)
- Correspondence: (D.P.G.); (M.V.B.); (J.J.Z.); Tel.: +1-631-323-3131 (D.P.G.); +1-631-323-3035 (M.V.B.); +1-631-323-3186 (J.J.Z.)
| |
Collapse
|
17
|
Rajah MM, Bernier A, Buchrieser J, Schwartz O. The Mechanism and Consequences of SARS-CoV-2 Spike-Mediated Fusion and Syncytia Formation. J Mol Biol 2022; 434:167280. [PMID: 34606831 PMCID: PMC8485708 DOI: 10.1016/j.jmb.2021.167280] [Citation(s) in RCA: 94] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/22/2021] [Accepted: 09/27/2021] [Indexed: 12/12/2022]
Abstract
Syncytia are formed when individual cells fuse. SARS-CoV-2 induces syncytia when the viral spike (S) protein on the surface of an infected cell interacts with receptors on neighboring cells. Syncytia may potentially contribute to pathology by facilitating viral dissemination, cytopathicity, immune evasion, and inflammatory response. SARS-CoV-2 variants of concern possess several mutations within the S protein that enhance receptor interaction, fusogenicity and antibody binding. In this review, we discuss the molecular determinants of S mediated fusion and the antiviral innate immunity components that counteract syncytia formation. Several interferon-stimulated genes, including IFITMs and LY6E act as barriers to S protein-mediated fusion by altering the composition or biophysical properties of the target membrane. We also summarize the effect that the mutations associated with the variants of concern have on S protein fusogenicity. Altogether, this review contextualizes the current understanding of Spike fusogenicity and the role of syncytia during SARS-CoV-2 infection and pathology.
Collapse
Affiliation(s)
- Maaran Michael Rajah
- Virus & Immunity Unit, Department of Virology, Institut Pasteur, CNRS UMR 3569, Paris, France; Université de Paris, Sorbonne Paris Cité, Paris, France. https://twitter.com/MaaranRajah
| | - Annie Bernier
- Institut Curie, INSERM U932, Paris, France. https://twitter.com/nini_bernier
| | - Julian Buchrieser
- Virus & Immunity Unit, Department of Virology, Institut Pasteur, CNRS UMR 3569, Paris, France. https://twitter.com/JBuchrieser
| | - Olivier Schwartz
- Virus & Immunity Unit, Department of Virology, Institut Pasteur, CNRS UMR 3569, Paris, France; Université de Paris, Sorbonne Paris Cité, Paris, France; Vaccine Research Institute, Creteil, France.
| |
Collapse
|
18
|
Acciani MD, Brindley MA. Scrambled or flipped: 5 facts about how cellular phosphatidylserine localization can mediate viral replication. PLoS Pathog 2022; 18:e1010352. [PMID: 35245334 PMCID: PMC8896693 DOI: 10.1371/journal.ppat.1010352] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Affiliation(s)
- Marissa Danielle Acciani
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia, United States of America
| | - Melinda Ann Brindley
- Department of Infectious Diseases, Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, Georgia, United States of America
- * E-mail:
| |
Collapse
|
19
|
Zhang Z, Trypsteen W, Blaauw M, Chu X, Rutsaert S, Vandekerckhove L, van der Heijden W, Dos Santos JC, Xu CJ, Swertz MA, van der Ven A, Li Y. IRF7 and RNH1 are modifying factors of HIV-1 reservoirs: a genome-wide association analysis. BMC Med 2021; 19:282. [PMID: 34781942 PMCID: PMC8594146 DOI: 10.1186/s12916-021-02156-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 10/07/2021] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Combination antiretroviral treatment (cART) cannot eradicate HIV-1 from the body due to the establishment of persisting viral reservoirs which are not affected by therapy and reinitiate new rounds of HIV-1 replication after treatment interruption. These HIV-1 reservoirs mainly comprise long-lived resting memory CD4+ T cells and are established early after infection. There is a high variation in the size of these viral reservoirs among virally suppressed individuals. Identification of host factors that contribute to or can explain this observed variation could open avenues for new HIV-1 treatment strategies. METHODS In this study, we conducted a genome-wide quantitative trait locus (QTL) analysis to probe functionally relevant genetic variants linked to levels of cell-associated (CA) HIV-1 DNA, CA HIV-1 RNA, and RNA:DNA ratio in CD4+ T cells isolated from blood from a cohort of 207 (Caucasian) people living with HIV-1 (PLHIV) on long-term suppressive antiretroviral treatment (median = 6.6 years). CA HIV-1 DNA and CA HIV-1 RNA levels were measured with corresponding droplet digital PCR (ddPCR) assays, and genotype information of 522,455 single-nucleotide variants was retrieved via the Infinium Global Screening array platform. RESULTS The analysis resulted in one significant association with CA HIV-1 DNA (rs2613996, P < 5 × 10-8) and two suggestive associations with RNA:DNA ratio (rs7113204 and rs7817589, P < 5 × 10-7). Then, we prioritized PTDSS2, IRF7, RNH1, and DEAF1 as potential HIV-1 reservoir modifiers and validated that higher expressions of IRF7 and RNH1 were accompanied by rs7113204-G. Moreover, RNA:DNA ratio, indicating relative HIV-1 transcription activity, was lower in PLHIV carrying this variant. CONCLUSIONS The presented data suggests that the amount of CA HIV-1 DNA and RNA:DNA ratio can be influenced through PTDSS2, RNH1, and IRF7 that were anchored by our genome-wide association analysis. Further, these observations reveal potential host genetic factors affecting the size and transcriptional activity of HIV-1 reservoirs and could indicate new targets for HIV-1 therapeutic strategies.
Collapse
Affiliation(s)
- Zhenhua Zhang
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525HP, Nijmegen, the Netherlands.,Department of Genetics, University Medical Center Groningen, 9700RB, Groningen, the Netherlands.,Genomics Coordination Center, University Medical Center Groningen, 9700RB, Groningen, the Netherlands.,Department of Computational Biology for Individualised Medicine, Centre for Individualised Infection Medicine, CiiM, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
| | - Wim Trypsteen
- HIV Cure Research Center, Department of Internal Medicine, and Pediatrics, Ghent University and Ghent University Hospital, Ghent, Belgium
| | - Marc Blaauw
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525HP, Nijmegen, the Netherlands
| | - Xiaojing Chu
- Department of Genetics, University Medical Center Groningen, 9700RB, Groningen, the Netherlands.,Department of Computational Biology for Individualised Medicine, Centre for Individualised Infection Medicine, CiiM, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany.,TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
| | - Sofie Rutsaert
- HIV Cure Research Center, Department of Internal Medicine, and Pediatrics, Ghent University and Ghent University Hospital, Ghent, Belgium
| | - Linos Vandekerckhove
- HIV Cure Research Center, Department of Internal Medicine, and Pediatrics, Ghent University and Ghent University Hospital, Ghent, Belgium
| | - Wouter van der Heijden
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525HP, Nijmegen, the Netherlands
| | - Jéssica Cristina Dos Santos
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525HP, Nijmegen, the Netherlands
| | - Cheng-Jian Xu
- Department of Computational Biology for Individualised Medicine, Centre for Individualised Infection Medicine, CiiM, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany.,TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
| | - Morris A Swertz
- Department of Genetics, University Medical Center Groningen, 9700RB, Groningen, the Netherlands.,Genomics Coordination Center, University Medical Center Groningen, 9700RB, Groningen, the Netherlands
| | - Andre van der Ven
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525HP, Nijmegen, the Netherlands.
| | - Yang Li
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525HP, Nijmegen, the Netherlands. .,Department of Genetics, University Medical Center Groningen, 9700RB, Groningen, the Netherlands. .,Department of Computational Biology for Individualised Medicine, Centre for Individualised Infection Medicine, CiiM, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany. .,TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany.
| |
Collapse
|
20
|
Song DH, Garcia G, Situ K, Chua BA, Hong MLO, Do EA, Ramirez CM, Harui A, Arumugaswami V, Morizono K. Development of a blocker of the universal phosphatidylserine- and phosphatidylethanolamine-dependent viral entry pathways. Virology 2021; 560:17-33. [PMID: 34020328 DOI: 10.1016/j.virol.2021.04.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/16/2021] [Accepted: 04/22/2021] [Indexed: 12/28/2022]
Abstract
Envelope phosphatidylserine (PtdSer) and phosphatidylethanolamine (PtdEtr) have been shown to mediate binding of enveloped viruses. However, commonly used PtdSer binding molecules such as Annexin V cannot block PtdSer-mediated viral infection. Lack of reagents that can conceal envelope PtdSer and PtdEtr and subsequently inhibit infection hinders elucidation of the roles of the envelope phospholipids in viral infection. Here, we developed sTIM1dMLDR801, a reagent capable of blocking PtdSer- and PtdEtr-dependent infection of enveloped viruses. Using sTIM1dMLDR801, we found that envelope PtdSer and/or PtdEtr can support ZIKV infection of not only human but also mosquito cells. In a mouse model for ZIKV infection, sTIM1dMLDR801 reduced ZIKV load in serum and the spleen, indicating envelope PtdSer and/or PtdEtr support in viral infection in vivo. sTIM1dMLDR801 will enable elucidation of the roles of envelope PtdSer and PtdEtr in infection of various virus species, thereby facilitating identification of their receptors and transmission mechanisms.
Collapse
Affiliation(s)
- Da-Hoon Song
- Division of Hematology and Oncology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA; UCLA AIDS Institute, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Gustavo Garcia
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, 90095, USA
| | - Kathy Situ
- Division of Hematology and Oncology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA; UCLA AIDS Institute, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Bernadette A Chua
- Division of Hematology and Oncology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA; UCLA AIDS Institute, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Madeline Lauren O Hong
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, 90095, USA
| | - Elyza A Do
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, 90095, USA
| | - Christina M Ramirez
- Department of Biostatistics, UCLA Fielding School of Public Health, University of California, Los Angeles, CA, 90095, USA
| | - Airi Harui
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Vaithilingaraja Arumugaswami
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, 90095, USA
| | - Kouki Morizono
- Division of Hematology and Oncology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA; UCLA AIDS Institute, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA.
| |
Collapse
|
21
|
Haase S, Condron M, Miller D, Cherkaoui D, Jordan S, Gulbis JM, Baum J. Identification and characterisation of a phospholipid scramblase in the malaria parasite Plasmodium falciparum. Mol Biochem Parasitol 2021; 243:111374. [PMID: 33974939 PMCID: PMC8202325 DOI: 10.1016/j.molbiopara.2021.111374] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/27/2021] [Accepted: 05/06/2021] [Indexed: 02/07/2023]
Abstract
Recent studies highlight the emerging role of lipids as important messengers in malaria parasite biology. In an attempt to identify interacting proteins and regulators of these dynamic and versatile molecules, we hypothesised the involvement of phospholipid translocases and their substrates in the infection of the host erythrocyte by the malaria parasite Plasmodium spp. Here, using a data base searching approach of the Plasmodium Genomics Resources (www.plasmodb.org), we have identified a putative phospholipid (PL) scramblase in P. falciparum (PfPLSCR) that is conserved across the genus and in closely related unicellular algae. By reconstituting recombinant PfPLSCR into liposomes, we demonstrate metal ion dependent PL translocase activity and substrate preference, confirming PfPLSCR as a bona fide scramblase. We show that PfPLSCR is expressed during asexual and sexual parasite development, localising to different membranous compartments of the parasite throughout the intra-erythrocytic life cycle. Two different gene knockout approaches, however, suggest that PfPLSCR is not essential for erythrocyte invasion and asexual parasite development, pointing towards a possible role in other stages of the parasite life cycle.
Collapse
Affiliation(s)
- Silvia Haase
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, Exhibition Road, South Kensington, London, UK.
| | - Melanie Condron
- Division of Infection and Immunity, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - David Miller
- Division of Structural Biology, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Dounia Cherkaoui
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, Exhibition Road, South Kensington, London, UK
| | - Sarah Jordan
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, Exhibition Road, South Kensington, London, UK
| | - Jacqueline M Gulbis
- Division of Structural Biology, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia; Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Jake Baum
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, Exhibition Road, South Kensington, London, UK.
| |
Collapse
|
22
|
Xie S, Zhang H, Liang Z, Yang X, Cao R. AXL, an Important Host Factor for DENV and ZIKV Replication. Front Cell Infect Microbiol 2021; 11:575346. [PMID: 33954117 PMCID: PMC8092360 DOI: 10.3389/fcimb.2021.575346] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 02/18/2021] [Indexed: 12/13/2022] Open
Abstract
Flaviviruses, as critically important pathogens, are still major public health problems all over the world. For instance, the evolution of ZIKV led to large-scale outbreaks in the Yap island in 2007. DENV was considered by the World Health Organization (WHO) as one of the 10 threats to global health in 2019. Enveloped viruses hijack a variety of host factors to complete its replication cycle. Phosphatidylserine (PS) receptor, AXL, is considered to be a candidate receptor for flavivirus invasion. In this review, we discuss the molecular structure of ZIKV and DENV, and how they interact with AXL to successfully invade host cells. A more comprehensive understanding of the molecular mechanisms of flavivirus-AXL interaction will provide crucial insights into the virus infection process and the development of anti-flavivirus therapeutics.
Collapse
Affiliation(s)
- Shengda Xie
- Ministry of Education (MOE) Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Huiru Zhang
- Ministry of Education (MOE) Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Zhenjie Liang
- Ministry of Education (MOE) Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xingmiao Yang
- Ministry of Education (MOE) Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Ruibing Cao
- Ministry of Education (MOE) Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
23
|
Flagging fusion: Phosphatidylserine signaling in cell-cell fusion. J Biol Chem 2021; 296:100411. [PMID: 33581114 PMCID: PMC8005811 DOI: 10.1016/j.jbc.2021.100411] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 02/06/2023] Open
Abstract
Formations of myofibers, osteoclasts, syncytiotrophoblasts, and fertilized zygotes share a common step, cell–cell fusion. Recent years have brought about considerable progress in identifying some of the proteins involved in these and other cell-fusion processes. However, even for the best-characterized cell fusions, we still do not know the mechanisms that regulate the timing of cell-fusion events. Are they fully controlled by the expression of fusogenic proteins or do they also depend on some triggering signal that activates these proteins? The latter scenario would be analogous to the mechanisms that control the timing of exocytosis initiated by Ca2+ influx and virus-cell fusion initiated by low pH- or receptor interaction. Diverse cell fusions are accompanied by the nonapoptotic exposure of phosphatidylserine at the surface of fusing cells. Here we review data on the dependence of membrane remodeling in cell fusion on phosphatidylserine and phosphatidylserine-recognizing proteins and discuss the hypothesis that cell surface phosphatidylserine serves as a conserved “fuse me” signal regulating the time and place of cell-fusion processes.
Collapse
|
24
|
Bruxelle JF, Trattnig N, Mureithi MW, Landais E, Pantophlet R. HIV-1 Entry and Prospects for Protecting against Infection. Microorganisms 2021; 9:microorganisms9020228. [PMID: 33499233 PMCID: PMC7911371 DOI: 10.3390/microorganisms9020228] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 12/19/2022] Open
Abstract
Human Immunodeficiency Virus type-1 (HIV-1) establishes a latent viral reservoir soon after infection, which poses a major challenge for drug treatment and curative strategies. Many efforts are therefore focused on blocking infection. To this end, both viral and host factors relevant to the onset of infection need to be considered. Given that HIV-1 is most often transmitted mucosally, strategies designed to protect against infection need to be effective at mucosal portals of entry. These strategies need to contend also with cell-free and cell-associated transmitted/founder (T/F) virus forms; both can initiate and establish infection. This review will discuss how insight from the current model of HIV-1 mucosal transmission and cell entry has highlighted challenges in developing effective strategies to prevent infection. First, we examine key viral and host factors that play a role in transmission and infection. We then discuss preventive strategies based on antibody-mediated protection, with emphasis on targeting T/F viruses and mucosal immunity. Lastly, we review treatment strategies targeting viral entry, with focus on the most clinically advanced entry inhibitors.
Collapse
Affiliation(s)
- Jean-François Bruxelle
- Faculty of Health Sciences, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
- Correspondence: (J.-F.B.); (R.P.)
| | - Nino Trattnig
- Chemical Biology and Drug Discovery, Utrecht University, 3584 CG Utrecht, The Netherlands;
| | - Marianne W. Mureithi
- KAVI—Institute of Clinical Research, College of Health Sciences, University of Nairobi, P.O. Box, Nairobi 19676–00202, Kenya;
| | - Elise Landais
- IAVI Neutralizing Antibody Center, La Jolla, CA 92037, USA;
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA
| | - Ralph Pantophlet
- Faculty of Health Sciences, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
- Correspondence: (J.-F.B.); (R.P.)
| |
Collapse
|
25
|
Lamkin DM, Bradshaw KP, Chang J, Epstein M, Gomberg J, Prajapati KP, Soliman VH, Sylviana T, Wong Y, Morizono K, Sloan EK, Cole SW. Physical activity modulates mononuclear phagocytes in mammary tissue and inhibits tumor growth in mice. PeerJ 2021; 9:e10725. [PMID: 33552733 PMCID: PMC7821756 DOI: 10.7717/peerj.10725] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 12/16/2020] [Indexed: 12/29/2022] Open
Abstract
The risk for breast cancer is significantly reduced in persons who engage in greater amounts of physical activity, and greater physical activity before or after diagnosis associates with reduced disease-specific mortality. Previous mechanistic studies indicate that components of innate immunity can mediate an inhibitory effect of physical activity on several types of tumor. However, in breast cancer specifically, the myeloid compartment of innate immunity is thought to exhibit high propensity for an immunosuppressive role that obstructs anti-tumor immunity. Thus, we tested the notion that greater physical activity alters mononuclear phagocytes in mammary tissue when inhibiting nascent tumor in a murine model of breast cancer. To model greater physical activity, we placed an angled running wheel in each mouse's home cage for two weeks before tumor engraftment with EO771 mammary cancer cells that express luciferase for bioluminescent detection. Fully immunocompetent mice and mice with compromised adaptive immunity showed significantly less mammary tumor signal when given access to running wheels, although the effect size was smaller in this latter group. To investigate the role of the myeloid compartment, mononuclear phagocytes were ablated by systemic injection of clodronate liposomes at 24 h before tumor engraftment and again at the time of tumor engraftment, and this treatment reversed the inhibition in wheel running mice. However, clodronate also inhibited mammary tumor signal in sedentary mice, in conjunction with an expected decrease in gene and protein expression of the myeloid antigen, F4/80 (Adgre1), in mammary tissue. Whole transcriptome digital cytometry with CIBERSORTx was used to analyze myeloid cell populations in mammary tissue following voluntary wheel running and clodronate treatment, and this approach found significant changes in macrophage and monocyte populations. In exploratory analyses, whole transcriptome composite scores for monocytic myeloid-derived suppressor cell (M-MDSC), macrophage lactate timer, and inflammation resolution gene expression programs were significantly altered. Altogether, the results support the hypothesis that physical activity inhibits nascent mammary tumor growth by enhancing the anti-tumor potential of mononuclear phagocytes in mammary tissue.
Collapse
Affiliation(s)
- Donald M. Lamkin
- Norman Cousins Center for PNI, Semel Institute for Neuroscience, University of California, Los Angeles, CA, United States of America
- Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, CA, United States of America
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, United States of America
| | - Karen P. Bradshaw
- Norman Cousins Center for PNI, Semel Institute for Neuroscience, University of California, Los Angeles, CA, United States of America
- Department of Neuroscience, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Janice Chang
- Norman Cousins Center for PNI, Semel Institute for Neuroscience, University of California, Los Angeles, CA, United States of America
| | - Ma’ayan Epstein
- Norman Cousins Center for PNI, Semel Institute for Neuroscience, University of California, Los Angeles, CA, United States of America
| | - Jack Gomberg
- Norman Cousins Center for PNI, Semel Institute for Neuroscience, University of California, Los Angeles, CA, United States of America
| | - Krupa P. Prajapati
- Norman Cousins Center for PNI, Semel Institute for Neuroscience, University of California, Los Angeles, CA, United States of America
| | - Veronica H. Soliman
- Norman Cousins Center for PNI, Semel Institute for Neuroscience, University of California, Los Angeles, CA, United States of America
| | - Thezia Sylviana
- Norman Cousins Center for PNI, Semel Institute for Neuroscience, University of California, Los Angeles, CA, United States of America
| | - Yinnie Wong
- Norman Cousins Center for PNI, Semel Institute for Neuroscience, University of California, Los Angeles, CA, United States of America
| | - Kouki Morizono
- Divison of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, United States of America
- UCLA AIDS Institute, David Geffen School of Medicine, University of California, Los Angeles, CA, United States of America
| | - Erica K. Sloan
- Norman Cousins Center for PNI, Semel Institute for Neuroscience, University of California, Los Angeles, CA, United States of America
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, United States of America
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Division of Cancer Surgery, Peter MacCallum Cancer Centre-Victorian Comprehensive Cancer Centre, Melbourne, Victoria, Austalia
| | - Steve W. Cole
- Norman Cousins Center for PNI, Semel Institute for Neuroscience, University of California, Los Angeles, CA, United States of America
- Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, CA, United States of America
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, United States of America
- Divison of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, United States of America
| |
Collapse
|
26
|
Zhang L, Richard AS, Jackson CB, Ojha A, Choe H. Phosphatidylethanolamine and Phosphatidylserine Synergize To Enhance GAS6/AXL-Mediated Virus Infection and Efferocytosis. J Virol 2020; 95:e02079-20. [PMID: 33115868 PMCID: PMC7944455 DOI: 10.1128/jvi.02079-20] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 10/23/2020] [Indexed: 12/13/2022] Open
Abstract
Phosphatidylserine (PS) receptors mediate clearance of apoptotic cells-efferocytosis-by recognizing the PS exposed on those cells. They also mediate the entry of enveloped viruses by binding PS in the virion membrane. Here, we show that phosphatidylethanolamine (PE) synergizes with PS to enhance PS receptor-mediated efferocytosis and virus entry. The presence of PE on the same surface as PS dramatically enhances recognition of PS by PS-binding proteins such as GAS6, PROS, and TIM1. Liposomes containing both PE and PS bound to GAS6 and were engulfed by AXL-expressing cells much more efficiently than those containing PS alone. Further, infection of AXL-expressing cells by infectious Zika virus or Ebola, Chikungunya, or eastern equine encephalitis pseudoviruses was inhibited with greater efficiency by the liposomes containing both PS and PE compared to a mixture of liposomes separately composed of PS and PE. These data demonstrate that simultaneous recognition of PE and PS maximizes PS receptor-mediated virus entry and efferocytosis and underscore the important contribution of PE in these major biological processes.IMPORTANCE Phosphatidylserine (PS) and phosphatidylethanolamine (PE) are usually sequestered to the inner leaflet of the plasma membrane of the healthy eukaryotic cells. During apoptosis, these phospholipids move to the cell's outer leaflet where they are recognized by so-called PS receptors on surveilling phagocytes. Several pathogenic families of enveloped viruses hijack these PS receptors to gain entry into their target cells. Here, we show that efficiency of these processes is enhanced, namely, PE synergizes with PS to promote PS receptor-mediated virus infection and clearance of apoptotic cells. These findings deepen our understanding of how these fundamental biological processes are executed.
Collapse
Affiliation(s)
- Lizhou Zhang
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, Florida, USA
| | - Audrey S Richard
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, Florida, USA
| | - Cody B Jackson
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, Florida, USA
| | - Amrita Ojha
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, Florida, USA
| | - Hyeryun Choe
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, Florida, USA
| |
Collapse
|
27
|
Chang W, Fa H, Xiao D, Wang J. Targeting phosphatidylserine for Cancer therapy: prospects and challenges. Theranostics 2020; 10:9214-9229. [PMID: 32802188 PMCID: PMC7415799 DOI: 10.7150/thno.45125] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 07/13/2020] [Indexed: 02/06/2023] Open
Abstract
Cancer is a leading cause of mortality and morbidity worldwide. Despite major improvements in current therapeutic methods, ideal therapeutic strategies for improved tumor elimination are still lacking. Recently, immunotherapy has attracted much attention, and many immune-active agents have been approved for clinical use alone or in combination with other cancer drugs. However, some patients have a poor response to these agents. New agents and strategies are needed to overcome such deficiencies. Phosphatidylserine (PS) is an essential component of bilayer cell membranes and is normally present in the inner leaflet. In the physiological state, PS exposure on the external leaflet not only acts as an engulfment signal for phagocytosis in apoptotic cells but also participates in blood coagulation, myoblast fusion and immune regulation in nonapoptotic cells. In the tumor microenvironment, PS exposure is significantly increased on the surface of tumor cells or tumor cell-derived microvesicles, which have innate immunosuppressive properties and facilitate tumor growth and metastasis. To date, agents targeting PS have been developed, some of which are under investigation in clinical trials as combination drugs for various cancers. However, controversial results are emerging in laboratory research as well as in clinical trials, and the efficiency of PS-targeting agents remains uncertain. In this review, we summarize recent progress in our understanding of the physiological and pathological roles of PS, with a focus on immune suppressive features. In addition, we discuss current drug developments that are based on PS-targeting strategies in both experimental and clinical studies. We hope to provide a future research direction for the development of new agents for cancer therapy.
Collapse
Affiliation(s)
- Wenguang Chang
- Institute for Translational Medicine, The Affiliated Hospital, College of medicine, Qingdao University, Qingdao, China
| | - Hongge Fa
- Institute for Translational Medicine, The Affiliated Hospital, College of medicine, Qingdao University, Qingdao, China
- School of Basic Medical Sciences, College of medicine, Qingdao University, Qingdao, China
| | - Dandan Xiao
- Institute for Translational Medicine, The Affiliated Hospital, College of medicine, Qingdao University, Qingdao, China
- School of Basic Medical Sciences, College of medicine, Qingdao University, Qingdao, China
| | - Jianxun Wang
- School of Basic Medical Sciences, College of medicine, Qingdao University, Qingdao, China
| |
Collapse
|
28
|
Schwartz D, Iyengar S. Recognition of Apoptotic Cells by Viruses and Cytolytic Lymphocytes: Target Selection in the Fog of War. Viral Immunol 2020; 33:188-196. [PMID: 32286181 PMCID: PMC7185367 DOI: 10.1089/vim.2019.0173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Viruses and cytolytic lymphocytes operate in an environment filled with dying and dead cells, and cell fragments. For viruses, irreversible fusion with doomed cells is suicide. For cytotoxic T lymphocyte and natural killer cells, time and limited lytic resources spent on apoptotic targets is wasteful and may result in death of the host. We make the case that the target membrane cytoskeleton is the best source of information regarding the suitability of potential targets for engagement for both viruses and lytic effector cells, and we present experimental evidence for detection of apoptotic cells by HIV, without loss of infectivity.
Collapse
Affiliation(s)
- David Schwartz
- Jurist Research Department, Hackensack University Medical Center, Hackensack, New Jersey
| | - Sujatha Iyengar
- Jurist Research Department, Hackensack University Medical Center, Hackensack, New Jersey
| |
Collapse
|
29
|
Calianese DC, Birge RB. Biology of phosphatidylserine (PS): basic physiology and implications in immunology, infectious disease, and cancer. Cell Commun Signal 2020; 18:41. [PMID: 32160904 PMCID: PMC7065380 DOI: 10.1186/s12964-020-00543-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 02/26/2020] [Indexed: 12/14/2022] Open
Abstract
Phosphatidylserine (PS) is an anionic phospholipid found on the membranes of a variety of organelles throughout the cell, most notably the plasma membrane. Under homeostatic conditions, PS is typically restricted to the inner leaflet of the plasma membrane. However, during cellular activation and/or induction of cell death, PS is externalized on the outer surface via the activation of phospholipid scramblases. Externalized PS not only changes the biochemical and biophysical properties of the plasma membrane but also initiates a series of interactions between endogenous extracellular proteins as well as receptors on neighboring cells to stimulate engulfment (efferocytosis) that influence the surrounding immune milieu. In this thematic series published in Cell Communication and Signaling, we feature review articles that highlight recent work in the field of PS biology, including the biochemistry and physiological significance of PS externalization, therapeutic applications and efforts to target PS, as well as posit open questions that remain in the field.
Collapse
Affiliation(s)
- David C Calianese
- Rutgers University, The State University of New Jersey, Newark, New Jersey, USA
| | - Raymond B Birge
- Rutgers University, The State University of New Jersey, Newark, New Jersey, USA.
| |
Collapse
|
30
|
Ghosh Roy S. TAM receptors: A phosphatidylserine receptor family and its implications in viral infections. TAM RECEPTORS IN HEALTH AND DISEASE 2020; 357:81-122. [DOI: 10.1016/bs.ircmb.2020.09.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|