1
|
Saribas AS, Jensen LE, Safak M. Recent advances in discovery and functional analysis of the small proteins and microRNA expressed by polyomaviruses. Virology 2025; 602:110310. [PMID: 39612622 DOI: 10.1016/j.virol.2024.110310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 11/13/2024] [Accepted: 11/18/2024] [Indexed: 12/01/2024]
Abstract
The polyomavirus family consists of a highly diverse group of small DNA viruses isolated from various species, including humans. Some family members have been used as model systems to understand the fundamentals of modern biology. After the discovery of the first two human polyomaviruses (JC virus and BK virus) during the early 1970s, their current number reached 14 today. Some family members cause considerably severe human diseases, including polyomavirus-associated nephropathy (PVAN), progressive multifocal leukoencephalopathy (PML), trichodysplasia spinulosa (TS) and Merkel cell carcinoma (MCC). Polyomaviruses encode universal regulatory and structural proteins, but some members express additional virus-specific proteins and microRNA, which significantly contribute to the viral biology, cell transformation, and perhaps progression of the disease that they are associated with. In the current review, we summarized the recent advances in discovery, and functional and structural analysis of those viral proteins and microRNA.
Collapse
Affiliation(s)
- A Sami Saribas
- Lewis Katz School of Medicine at Temple University, Department of Microbiology, Immunology and Inflammation Center for Neurovirology and Gene Editing, 3500 N. Broad Street, Philadelphia, PA, 19140, USA.
| | - Liselotte E Jensen
- Lewis Katz School of Medicine at Temple University, Department of Microbiology, Immunology and Inflammation, Center for Inflammation and Lung Research, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Mahmut Safak
- Lewis Katz School of Medicine at Temple University, Department of Microbiology, Immunology and Inflammation Center for Neurovirology and Gene Editing, 3500 N. Broad Street, Philadelphia, PA, 19140, USA.
| |
Collapse
|
2
|
Mocarski ES. Cytomegalovirus Biology Viewed Through a Cell Death Suppression Lens. Viruses 2024; 16:1820. [PMID: 39772130 PMCID: PMC11680106 DOI: 10.3390/v16121820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/22/2024] [Accepted: 11/22/2024] [Indexed: 01/11/2025] Open
Abstract
Cytomegaloviruses, species-specific members of the betaherpesviruses, encode an impressive array of immune evasion strategies committed to the manipulation of the host immune system enabling these viruses to remain for life in a stand-off with host innate and adaptive immune mechanisms. Even though they are species-restricted, cytomegaloviruses are distributed across a wide range of different mammalian species in which they cause systemic infection involving many different cell types. Regulated, or programmed cell death has a recognized potential to eliminate infected cells prior to completion of viral replication and release of progeny. Cell death also naturally terminates replication during the final stages of replication. Over the past two decades, the host defense potential of known programmed cell death pathways (apoptosis, necroptosis, and pyroptosis), as well as a novel mitochondrial serine protease pathway have been defined through studies of cytomegalovirus-encoded cell death suppressors. Such virus-encoded inhibitors prevent virus-induced, cytokine-induced, and stress-induced death of infected cells while also moderating inflammation. By evading cell death and consequent inflammation as well as innate and adaptive immune clearance, cytomegaloviruses represent successful pathogens that become a critical disease threat when the host immune system is compromised. This review will discuss cell death programs acquired for mammalian host defense against cytomegaloviruses and enumerate the range of modulatory strategies this type of virus employs to balance host defense in favor of lifelong persistence.
Collapse
Affiliation(s)
- Edward S. Mocarski
- Department of Microbiology & Immunology, Stanford Medical School, Stanford University, Stanford, CA 94305, USA;
- Department of Microbiology & Immunology, Emory Medical School, Emory Vaccine Center, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
3
|
Morovati S, Mohammadi A, Masoudi R, Heidari AA, Asad Sangabi M. The power of mumps virus: Matrix protein activates apoptotic pathways in human colorectal cell lines. PLoS One 2023; 18:e0295819. [PMID: 38091318 PMCID: PMC10718445 DOI: 10.1371/journal.pone.0295819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 11/30/2023] [Indexed: 12/18/2023] Open
Abstract
New therapeutic approaches can significantly impact the control of colorectal cancer (CRC), which is increasing worldwide. In this study, we investigated the potential of targeting viral proteins to combat cancer cells. Specifically, we examined the anticancer potential of the matrix (M) protein of the mumps virus Hoshino strain in SW480 CRC cell lines. To begin, we individually transfected SW480 cells with pcDNA3 plasmids containing the mumps virus M gene. We then investigated the percentage of cell death, caspase activity, and the expression levels of genes involved in apoptosis pathways. Following this, we performed bioinformatics analysis on the M protein to identify any similarities with Bcl-2 family members and their viral homologs. Our diagnostic methods showed that treatment with the mumps M protein induced apoptosis and upregulated the expression and activity of pro-apoptotic proteins in SW480 CRC cells compared to the control and vector groups. Based on our bioinformatics studies, we proposed that the BH3 motif in the M protein may trigger apoptosis in CRC cells by interacting with cellular Bax. Overall, our study showed for the first time that the mumps virus M protein could be considered as a targeted treatment for CRC by inducing apoptotic pathways.
Collapse
Affiliation(s)
- Solmaz Morovati
- Department of Pathobiology, Division of Biotechnology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Ali Mohammadi
- Department of Pathobiology, Division of Virology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Ramin Masoudi
- Department of Pathobiology, Division of Biotechnology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Amir Ali Heidari
- Department of Clinical Sciences, Division of Aquatic Animal Health and Diseases, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Mehdi Asad Sangabi
- Department of Pathobiology, Division of Virology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| |
Collapse
|
4
|
Ferrara G, Longobardi C, Sgadari MF, Restucci B, Iovane G, Ciarcia R, Pagnini U, Montagnaro S. Apoptosis is mediated by FeHV-1 through the intrinsic pathway and interacts with the autophagic process. Virol J 2023; 20:295. [PMID: 38087282 PMCID: PMC10716993 DOI: 10.1186/s12985-023-02267-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Accepted: 12/09/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Although FeHV-1 is a primary feline pathogen, little is known about its interactions with host cells. Its relationship with several cellular pathways has recently been described, whereas its interplay with the apoptotic process, unlike other herpesviruses, has not yet been clarified. The aim of this work was to evaluate whether FeHV-1 induces apoptosis in its permissive cells, as well as the pathway involved and the effects of induction and inhibition of apoptosis on viral replication. METHODS Monolayers of CRFK cells were infected at different times with different viral doses. A cytofluorimetric approach allowed the quantification of cells in early and late apoptosis. All infections and related controls were also subjected to Western blot analysis to assess the expression of apoptotic markers (caspase 3-8-9, Bcl-2, Bcl-xL, NF-κB). An inhibitor (Z-VAD-FMK) and an inducer (ionomycin) were used to evaluate the role of apoptosis in viral replication. Finally, the expression of autophagy markers during the apoptosis inhibition/induction and the expression of apoptosis markers during autophagy inhibition/induction were evaluated to highlight any crosstalk between the two pathways. RESULTS FeHV-1 triggered apoptosis in a time- and dose-dependent manner. Caspase 3 cleavage was evident 48 h after infection, indicating the completeness of the process at this stage. While caspase 8 was not involved, caspase 9 cleavage started 24 h post-infection. The expression of other mitochondrial damage markers also changed, suggesting that apoptosis was induced via the intrinsic pathway. NF- κB was up-regulated at 12 h, followed by a gradual decrease in levels up to 72 h. The effects of apoptosis inhibitors and inducers on viral replication and autophagy were also investigated. Inhibition of caspases resulted in an increase in viral glycoprotein expression, higher titers, and enhanced autophagy, whereas induction of apoptosis resulted in a decrease in viral protein expression, lower viral titer, and attenuated autophagy. On the other hand, the induction of autophagy reduced the cleavage of caspase 3. CONCLUSIONS In this study, we established how FeHV-1 induces the apoptotic process, contributing to the understanding of the relationship between FeHV-1 and this pathway.
Collapse
Affiliation(s)
- Gianmarco Ferrara
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Via Federico Delpino n.1, Naples, 80137, Italy.
| | - Consiglia Longobardi
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Via Federico Delpino n.1, Naples, 80137, Italy
| | - Maria Francesca Sgadari
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Via Federico Delpino n.1, Naples, 80137, Italy
| | - Brunella Restucci
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Via Federico Delpino n.1, Naples, 80137, Italy
| | - Giuseppe Iovane
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Via Federico Delpino n.1, Naples, 80137, Italy
| | - Roberto Ciarcia
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Via Federico Delpino n.1, Naples, 80137, Italy
| | - Ugo Pagnini
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Via Federico Delpino n.1, Naples, 80137, Italy
| | - Serena Montagnaro
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Via Federico Delpino n.1, Naples, 80137, Italy
| |
Collapse
|
5
|
Li K, Geng Y, Lin B, Xi Z. Molecular mechanisms underlying mitochondrial damage, endoplasmic reticulum stress, and oxidative stress induced by environmental pollutants. Toxicol Res (Camb) 2023; 12:1014-1023. [PMID: 38145103 PMCID: PMC10734609 DOI: 10.1093/toxres/tfad094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 08/09/2023] [Accepted: 09/15/2023] [Indexed: 12/26/2023] Open
Abstract
Mitochondria and endoplasmic reticulum (ER) are essential organelles playing pivotal roles in the regulation of cellular metabolism, energy production, and protein synthesis. In addition, these organelles are important targets susceptible to external stimuli, such as environmental pollutants. Exposure to environmental pollutants can cause the mitochondrial damage, endoplasmic reticulum stress (ERS), and oxidative stress, leading to cellular dysfunction and death. Therefore, understanding the toxic effects and molecular mechanisms of environmental pollution underlying these processes is crucial for developing effective strategies to mitigate the adverse effects of environmental pollutants on human health. In the present study, we summarized and reviewed the toxic effects and molecular mechanisms of mitochondrial damage, ERS, and oxidative stress caused by exposure to environmental pollutants as well as interactions inducing the cell apoptosis and the roles in exposure to environmental pollutants.
Collapse
Affiliation(s)
- Kang Li
- Department of Health Toxicology, Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Yanpei Geng
- Department of Health Toxicology, Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Bencheng Lin
- Department of Health Toxicology, Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Zhuge Xi
- Department of Health Toxicology, Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| |
Collapse
|
6
|
Modulation of mitochondria by viral proteins. Life Sci 2023; 313:121271. [PMID: 36526048 DOI: 10.1016/j.lfs.2022.121271] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/24/2022] [Accepted: 12/03/2022] [Indexed: 12/15/2022]
Abstract
Mitochondria are dynamic cellular organelles with diverse functions including energy production, calcium homeostasis, apoptosis, host innate immune signaling, and disease progression. Several viral proteins specifically target mitochondria to subvert host defense as mitochondria stand out as the most suitable target for the invading viruses. They have acquired the capability to control apoptosis, metabolic state, and evade immune responses in host cells, by targeting mitochondria. In this way, the viruses successfully allow the spread of viral progeny and thus the infection. Viruses employ their proteins to alter mitochondrial dynamics and their specific functions by a modulation of membrane potential, reactive oxygen species, calcium homeostasis, and mitochondrial bioenergetics to help them achieve a state of persistent infection. A better understanding of such viral proteins and their impact on mitochondrial forms and functions is the main focus of this review. We also attempt to emphasize the importance of exploring the role of mitochondria in the context of SARS-CoV2 pathogenesis and identify host-virus protein interactions.
Collapse
|
7
|
Chaturvedi S, Pablo M, Wolf M, Rosas-Rivera D, Calia G, Kumar AJ, Vardi N, Du K, Glazier J, Ke R, Chan MF, Perelson AS, Weinberger LS. Disrupting autorepression circuitry generates "open-loop lethality" to yield escape-resistant antiviral agents. Cell 2022; 185:2086-2102.e22. [PMID: 35561685 PMCID: PMC9097017 DOI: 10.1016/j.cell.2022.04.022] [Citation(s) in RCA: 3] [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: 04/06/2021] [Revised: 03/01/2022] [Accepted: 04/14/2022] [Indexed: 12/27/2022]
Abstract
Across biological scales, gene-regulatory networks employ autorepression (negative feedback) to maintain homeostasis and minimize failure from aberrant expression. Here, we present a proof of concept that disrupting transcriptional negative feedback dysregulates viral gene expression to therapeutically inhibit replication and confers a high evolutionary barrier to resistance. We find that nucleic-acid decoys mimicking cis-regulatory sites act as "feedback disruptors," break homeostasis, and increase viral transcription factors to cytotoxic levels (termed "open-loop lethality"). Feedback disruptors against herpesviruses reduced viral replication >2-logs without activating innate immunity, showed sub-nM IC50, synergized with standard-of-care antivirals, and inhibited virus replication in mice. In contrast to approved antivirals where resistance rapidly emerged, no feedback-disruptor escape mutants evolved in long-term cultures. For SARS-CoV-2, disruption of a putative feedback circuit also generated open-loop lethality, reducing viral titers by >1-log. These results demonstrate that generating open-loop lethality, via negative-feedback disruption, may yield a class of antimicrobials with a high genetic barrier to resistance.
Collapse
Affiliation(s)
- Sonali Chaturvedi
- Gladstone/UCSF Center for Cell Circuitry, Gladstone Institutes, San Francisco, CA 94158, USA; Gladstone Institute of Virology, Gladstone Institutes, San Francisco, CA 94158, USA.
| | - Michael Pablo
- Gladstone/UCSF Center for Cell Circuitry, Gladstone Institutes, San Francisco, CA 94158, USA; Gladstone Institute of Virology, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Marie Wolf
- Gladstone/UCSF Center for Cell Circuitry, Gladstone Institutes, San Francisco, CA 94158, USA; Gladstone Institute of Virology, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Daniel Rosas-Rivera
- Gladstone/UCSF Center for Cell Circuitry, Gladstone Institutes, San Francisco, CA 94158, USA; Gladstone Institute of Virology, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Giuliana Calia
- Gladstone/UCSF Center for Cell Circuitry, Gladstone Institutes, San Francisco, CA 94158, USA; Gladstone Institute of Virology, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Arjun J Kumar
- Gladstone/UCSF Center for Cell Circuitry, Gladstone Institutes, San Francisco, CA 94158, USA; Gladstone Institute of Virology, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Noam Vardi
- Gladstone/UCSF Center for Cell Circuitry, Gladstone Institutes, San Francisco, CA 94158, USA; Gladstone Institute of Virology, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Kelvin Du
- Gladstone/UCSF Center for Cell Circuitry, Gladstone Institutes, San Francisco, CA 94158, USA; Gladstone Institute of Virology, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Joshua Glazier
- Gladstone/UCSF Center for Cell Circuitry, Gladstone Institutes, San Francisco, CA 94158, USA; Gladstone Institute of Virology, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Ruian Ke
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, Los Alamos, NM 87545, USA
| | - Matilda F Chan
- Francis I. Proctor Foundation, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Ophthalmology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Alan S Perelson
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, Los Alamos, NM 87545, USA
| | - Leor S Weinberger
- Gladstone/UCSF Center for Cell Circuitry, Gladstone Institutes, San Francisco, CA 94158, USA; Gladstone Institute of Virology, Gladstone Institutes, San Francisco, CA 94158, USA; Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
8
|
Oh DS, Park JH, Jung HE, Kim HJ, Lee HK. Autophagic protein ATG5 controls antiviral immunity via glycolytic reprogramming of dendritic cells against respiratory syncytial virus infection. Autophagy 2021; 17:2111-2127. [PMID: 32816604 PMCID: PMC8496528 DOI: 10.1080/15548627.2020.1812218] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 08/06/2020] [Accepted: 08/12/2020] [Indexed: 01/03/2023] Open
Abstract
Respiratory syncytial virus (RSV) is a leading cause of respiratory tract infections in infants. Macroautophagy/autophagy is a catalytic metabolic process required for cellular homeostasis. Although intracellular metabolism is important for immune responses in dendritic cells, the link between autophagy and immunometabolism remains unknown. Here, we show that the autophagy-related protein ATG5 regulates immunometabolism. Atg5-deficient mouse dendritic cells showed increased CD8A+ T-cell response and increased secretion of proinflammatory cytokines upon RSV infection. Transcriptome analysis showed that Atg5 deficiency alters the expression of metabolism-related genes. Atg5-deficient dendritic cells also showed increased activation of glycolysis and the AKT-MTOR-RPS6KB1 pathway and decreased mitochondrial activity, all of which are cellular signatures for metabolic activation. These cells also showed elevated CD8A+ T-cell priming and surface major histocompatibility complex (MHC) class I expression. Our results suggested that ATG5 regulated host immune responses by modulating dendritic cell metabolism. These findings may help develop potential antiviral therapies that alter host immunity by regulating autophagy and immunometabolism.Abbreviations : 2-DG: 2-deoxyglucose; AAK1: AP2 associated kinase 1; AKT: AKT serine/threonine kinase; AM: alveolar macrophage; ATG: autophagy; ATP: adenosine triphosphate; BAL: bronchoalveolar lavage; BMDC: bone marrow dendritic cell; CSF2/GM-CSF: colony-stimulating factor 2 (granulocyte-macrophage); CTL: cytotoxic T lymphocyte; ELISA: enzyme-linked immunosorbent assay; GFP: green fluorescent protein; GSEA: gene-set enrichment analysis; H-2Db: H-2 class I histocompatibility antigen, D-B alpha chain; H-2Kb: MHC class I H2-K-b; HIF1A: hypoxia-inducible factor 1 alpha; IFNG: interferon-gamma; IL: interleukin; ITGAX: integrin alpha X; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; MAP1LC3B/LC3B: microtubule-associated protein 1 light chain 3 beta; MHC: major histocompatibility complex; MTORC1: mammalian target of rapamycin kinase complex 1; PBS: phosphate-buffered saline; PFU: plaque-forming unit; RLR: retinoic acid-inducible-I-like receptor; ROS: reactive oxygen species; RPMI: Roswell Park Memorial Institute; RPS6KB1/S6K: ribosomal protein S6 kinase, polypeptide 1; RSV: respiratory syncytial virus; Th: T helper; TLR: toll-like receptor; Treg: regulatory T cells; UMAP: uniform manifold approximation and projection.
Collapse
Affiliation(s)
- Dong Sun Oh
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Jang Hyun Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Hi Eun Jung
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Hyun-Jin Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Heung Kyu Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- The Center for Epidemic Preparedness, KAIST Institute, KAIST, Daejeon, Republic of Korea
| |
Collapse
|
9
|
Valenzuela R, Rodriguez-Perez AI, Costa-Besada MA, Rivas-Santisteban R, Garrido-Gil P, Lopez-Lopez A, Navarro G, Lanciego JL, Franco R, Labandeira-Garcia JL. An ACE2/Mas-related receptor MrgE axis in dopaminergic neuron mitochondria. Redox Biol 2021; 46:102078. [PMID: 34333284 PMCID: PMC8346680 DOI: 10.1016/j.redox.2021.102078] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 07/17/2021] [Accepted: 07/19/2021] [Indexed: 02/06/2023] Open
Abstract
ACE2 plays a pivotal role in the balance between the pro-oxidative pro-inflammatory and the anti-oxidative anti-inflammatory arms of the renin-angiotensin system. Furthermore, ACE2 is the entry receptor for SARS-CoV-2. Clarification of ACE2-related mechanisms is crucial for the understanding of COVID-19 and other oxidative stress and inflammation-related processes. In rat and monkey brain, we discovered that the intracellular ACE2 and its products Ang 1–7 and alamandine are highly concentrated in the mitochondria and bind to a new mitochondrial Mas-related receptor MrgE (MrgE) to produce nitric oxide. We found MrgE expressed in neurons and glia of rodents and primates in the substantia nigra and different brain regions. In the mitochondria, ACE2 and MrgE expressions decreased and NOX4 increased with aging. This new ACE2/MrgE/NO axis may play a major role in mitochondrial regulation of oxidative stress in neurons, and possibly other cells. Therefore, dysregulation of the mitochondrial ACE2/MrgE/NO axis may play a major role in neurodegenerative processes of dopaminergic neurons, where mitochondrial dysfunction and oxidative stress play a crucial role. Since ACE2 binds SARS-CoV-2 spike protein, the mitochondrial ACE2/MrgE/NO axis may also play a role in SARS-CoV-2 cellular effects. ACE2 products Ang1-7 and alamandine (Ala) highly concentrate in brain mitochondria. Ang1-7 and Ala bind to mitochondrial Mas-related receptor MrgE producing nitric oxide. ACE2/MrgE may play a major role in mitochondrial function and oxidative stress. Clarification of ACE2-related mechanisms is also crucial for understanding COVID-19.
Collapse
Affiliation(s)
- Rita Valenzuela
- Cellular and Molecular Neurobiology of Parkinson's Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, Santiago de Compostela; Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Spain.
| | - Ana I Rodriguez-Perez
- Cellular and Molecular Neurobiology of Parkinson's Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, Santiago de Compostela; Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Spain
| | - Maria A Costa-Besada
- Cellular and Molecular Neurobiology of Parkinson's Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, Santiago de Compostela; Spain; Cell and Developmental Biology Department, University College London, London, UK
| | - Rafael Rivas-Santisteban
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Spain; Laboratory of Molecular Neurobiology, Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Pablo Garrido-Gil
- Cellular and Molecular Neurobiology of Parkinson's Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, Santiago de Compostela; Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Spain
| | - Andrea Lopez-Lopez
- Cellular and Molecular Neurobiology of Parkinson's Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, Santiago de Compostela; Spain
| | - Gemma Navarro
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Spain; Department of Biochemistry and Physiology, Faculty of Pharmacy, University of Barcelona, Barcelona, Spain
| | - Jose L Lanciego
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Spain; Neuroscience Department, Center for Applied Medical Research (CIMA, IdiSNA), University of Navarra, Pamplona, Spain
| | - Rafael Franco
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Spain; Laboratory of Molecular Neurobiology, Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Jose L Labandeira-Garcia
- Cellular and Molecular Neurobiology of Parkinson's Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, Santiago de Compostela; Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Spain.
| |
Collapse
|
10
|
Han T, Huang J, Gu J, Xie Q, Zhong Y, Huang T. Hepatitis B virus surface protein induces sperm dysfunction through the activation of a Bcl2/Bax signaling cascade triggering AIF/Endo G-mediated apoptosis. Andrology 2021; 9:944-955. [PMID: 33382193 PMCID: PMC8247882 DOI: 10.1111/andr.12965] [Citation(s) in RCA: 8] [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: 07/28/2020] [Revised: 12/16/2020] [Accepted: 12/29/2020] [Indexed: 02/05/2023]
Abstract
BACKGROUND Hepatitis B virus (HBV) was found to exist in semen and male germ cells of patients with chronic HBV infection. Our previous studies demonstrated that HBV surface protein (HBs) could induce sperm dysfunction by activating a calcium signaling cascade and triggering caspase-dependent apoptosis. However, the relationship between sperm dysfunction caused by HBs and caspase-independent apoptosis has not been investigated. OBJECTIVES To evaluate the effects of HBs exposure on sperm dysfunction by activating caspase-independent apoptosis. MATERIALS AND METHODS Spermatozoa were exposed to HBs at concentrations of 0, 25, 50, and 100 μg/mL for 3 h. Flow cytometry, qRT-PCR, immunofluorescence assay, ELISA, and zona-free hamster oocyte penetration assays were performed. RESULTS With increasing concentrations of HBs, various parameters of the spermatozoa changed. The number of Bcl2-positive cells declined and that of both Bax-positive cells and Apaf-1-positive cells increased. The transcription level of Bcl2 increased and that of both Bax and Apaf-1 declined. The average levels of AIF and Endo G declined in mitochondria and increased in the cytoplasm and nucleus. The sperm DNA fragmentation index increased. The mean percentages of live spermatozoa declined and that of both injured and dead spermatozoa increased; and the sperm penetration rate declined. For the aforementioned parameters, the differences between the test and the control groups were statistically significant. CONCLUSION HBs exposure can activate the Bax/Bcl2 signaling cascade that triggers AIF/Endo G-mediated apoptosis, resulting in sperm DNA fragmentation, sperm injury, and death, and a decrease in the sperm fertilizing capacity. This new knowledge will help to evaluate the negative impact of HBV on male fertility in HBV-infected patients.
Collapse
Affiliation(s)
- Ting‐Ting Han
- Research Center for Reproductive MedicineShantou University Medical CollegeShantouChina
| | - Ji‐Hua Huang
- Jinxin Research Institute for Reproductive Medicine and GeneticsChengdu Jinjiang Hospital for Maternal and Child Health CareChengduChina
| | - Jiang Gu
- Jinxin Research Institute for Reproductive Medicine and GeneticsChengdu Jinjiang Hospital for Maternal and Child Health CareChengduChina
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular ImmunopathologyCollaborative and Creative Center of Shantou UniversityShantouChina
| | - Qing‐Dong Xie
- Research Center for Reproductive MedicineShantou University Medical CollegeShantouChina
| | - Ying Zhong
- Jinxin Research Institute for Reproductive Medicine and GeneticsChengdu Jinjiang Hospital for Maternal and Child Health CareChengduChina
| | - Tian‐Hua Huang
- Research Center for Reproductive MedicineShantou University Medical CollegeShantouChina
- Jinxin Research Institute for Reproductive Medicine and GeneticsChengdu Jinjiang Hospital for Maternal and Child Health CareChengduChina
| |
Collapse
|
11
|
Wu X, Hua Y, Wei T, Ma C, Wang Z, Zhang L, Wang J. Effect and mechanism of action in vitroof cyclodextrin derivative nanoparticles loaded with tyroserleutide on hepatoma. NANOTECHNOLOGY 2021; 32:285101. [PMID: 33789260 DOI: 10.1088/1361-6528/abf3f2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 03/31/2021] [Indexed: 06/12/2023]
Abstract
In this study, a cyclodextrin derivative (R6RGD-CMβCD) nanoparticle with tumor targeting and cell penetration ability was successfully synthesized and loaded with tyroserleutide (YSL) to obtain YSL-loaded nanoparticles (YSL/R6RGD-CMβCD NPs). The characterization of these NPs revealed a smooth surfaces and an average diameter of approximately 170 nm. YSL/R6RGD-CMβCD NPs increased the NP uptake in Caco-2 cells. As regard the mechanism of action, the cell uptake was related to endocytosis mediated by reticulin and megacytosis. In addition, YSL/R6RGD-CMβCD NPs induced significantly higher cytotoxicity on tumor cells and better tumor targeting compared with the effect of CMβCD NPs. Most importantly, the good anti-cancer effect of YSL/R6RGD-CMβCD NPs might be due to the interference with the function of mitochondria. On the other hand, YSL/R6RGD-CMβCD NPs were not toxic for normal cells. Taken together, our results indicated that R6RGD-CMβCD could be considered as a nanopharmaceutical material with good tumor targeting abilities, and their combination with YSL could represent an effective anti-cancer system.
Collapse
Affiliation(s)
- Xiaoyan Wu
- Pharmacy, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, People's Republic of China
| | - Yingying Hua
- School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210046, People's Republic of China
| | - Tiantian Wei
- School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210046, People's Republic of China
| | - Chenjun Ma
- School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210046, People's Republic of China
| | - Zhongjie Wang
- School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210046, People's Republic of China
| | - Liefeng Zhang
- School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210046, People's Republic of China
| | - Jing Wang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, People's Republic of China
| |
Collapse
|
12
|
Mechanisms and consequences of Newcastle disease virus W protein subcellular localization in the nucleus or mitochondria. J Virol 2021; 95:JVI.02087-20. [PMID: 33441338 PMCID: PMC8092705 DOI: 10.1128/jvi.02087-20] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
We previously demonstrated that W proteins from different Newcastle disease virus (NDV) strains localize in either the cytoplasm (e.g., NDV strain SG10) or the nucleus (e.g., NDV strain La Sota). To clarify the mechanism behind these cell localization differences, we overexpressed W protein derived from four different NDV strains or W protein associated with different cellular regions in Vero cells. This revealed that the key region for determining W protein localization is 180-227aa. Further experiments found that there is a nuclear export signal (NES) motif in W protein 211-224aa. W protein could be transported into the nucleus via interaction with KPNA1, KPNA2, and KPNA6 in a nuclear localization signal-dependent manner, and W protein containing an NES was transported back to the cytoplasm in a CRM1-independent manner. Interestingly, we observed that the cytoplasm-localized W protein colocalizes with mitochondria. We rescued the NES-deletion W protein NDV strain rSG10-ΔWC/WΔNES using an NDV reverse genetics system and found that the replication ability, virulence, and pathogenicity of an NDV strain were all higher when the W protein cellular localization was in the nucleus rather than the mitochondria. Further experiments revealed that W protein nuclear localization reduced the expression of IFN-β otherwise stimulated by NDV. Our research reveals the mechanism by which NDV W protein becomes localized to different parts of the cell and demonstrates the outcomes of nuclear or cytoplasmic localization both in vitro and in vivo, laying a foundation for subsequent functional studies of the W protein in NDV and other paramyxoviruses.IMPORTANCE In Newcastle disease virus (NDV), the W protein, like the V protein, is a nonstructural protein encoded by the P gene via RNA editing. Compared with V protein, W protein has a common N-terminal domain but a unique C-terminal domain. V protein is known as a key virulence factor and an important interferon antagonist across the family Paramyxoviridae In contrast, very little is known about the function of NDV W protein, and this limited information is based on studies of the Nipah virus W protein. Here, we investigated the localization mechanism of NDV W protein and its subcellular distribution in mitochondria. We found that W protein localization differences impact IFN-β production, consequently affecting NDV virulence, replication, and pathogenicity. This work provides new insights on the differential localization mechanism of NDV W proteins, along with fundamental knowledge for understanding the functions of W proteins in NDV and other paramyxoviruses.
Collapse
|
13
|
Hagn M, Jahrsdörfer B. Why do human B cells secrete granzyme B? Insights into a novel B-cell differentiation pathway. Oncoimmunology 2021; 1:1368-1375. [PMID: 23243600 PMCID: PMC3518509 DOI: 10.4161/onci.22354] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
B cells are generally believed to operate as producers of high affinity antibodies to defend the body against microorganisms, whereas cellular cytotoxicity is considered as an exclusive prerogative of natural killer (NK) cells and cytotoxic T lymphocytes (CTLs). In conflict with this dogma, recent studies have demonstrated that the combination of interleukin-21 (IL-21) and B-cell receptor (BCR) stimulation enables B cells to produce and secrete the active form of the cytotoxic serine protease granzyme B (GrB). Although the production of GrB by B cells is not accompanied by that of perforin as in the case of many other GrB-secreting cells, recent findings suggest GrB secretion by B cells may play a significant role in early antiviral immune responses, in the regulation of autoimmune responses, and in cancer immunosurveillance. Here, we discuss in detail how GrB-secreting B cells may influence a variety of immune processes. A better understanding of the role that GrB-secreting B cells are playing in the immune system may allow for the development and improvement of novel immunotherapeutic approaches against infectious, autoimmune and malignant diseases.
Collapse
Affiliation(s)
- Magdalena Hagn
- Cancer Immunology Program; Peter MacCallum Cancer Centre; Melbourne, Australia
| | | |
Collapse
|
14
|
Saxena R, Saribas S, Jadiya P, Tomar D, Kaminski R, Elrod JW, Safak M. Human neurotropic polyomavirus, JC virus, agnoprotein targets mitochondrion and modulates its functions. Virology 2021; 553:135-153. [PMID: 33278736 PMCID: PMC7847276 DOI: 10.1016/j.virol.2020.11.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 11/12/2020] [Indexed: 01/18/2023]
Abstract
JC virus encodes an important regulatory protein, known as Agnoprotein (Agno). We have recently reported Agno's first protein-interactome with its cellular partners revealing that it targets various cellular networks and organelles, including mitochondria. Here, we report further characterization of the functional consequences of its mitochondrial targeting and demonstrated its co-localization with the mitochondrial networks and with the mitochondrial outer membrane. The mitochondrial targeting sequence (MTS) of Agno and its dimerization domain together play major roles in this targeting. Data also showed alterations in various mitochondrial functions in Agno-positive cells; including a significant reduction in mitochondrial membrane potential, respiration rates and ATP production. In contrast, a substantial increase in ROS production and Ca2+ uptake by the mitochondria were also observed. Finally, findings also revealed a significant decrease in viral replication when Agno MTS was deleted, highlighting a role for MTS in the function of Agno during the viral life cycle.
Collapse
Affiliation(s)
- Reshu Saxena
- Department of Neuroscience, Laboratory of Molecular Neurovirology, MERB-757, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Sami Saribas
- Department of Neuroscience, Laboratory of Molecular Neurovirology, MERB-757, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Pooja Jadiya
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, USA
| | - Dhanendra Tomar
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, USA
| | - Rafal Kaminski
- Department of Neuroscience, Laboratory of Molecular Neurovirology, MERB-757, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - John W Elrod
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, USA
| | - Mahmut Safak
- Department of Neuroscience, Laboratory of Molecular Neurovirology, MERB-757, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA, 19140, USA.
| |
Collapse
|
15
|
Labandeira-Garcia JL, Valenzuela R, Costa-Besada MA, Villar-Cheda B, Rodriguez-Perez AI. The intracellular renin-angiotensin system: Friend or foe. Some light from the dopaminergic neurons. Prog Neurobiol 2020; 199:101919. [PMID: 33039415 PMCID: PMC7543790 DOI: 10.1016/j.pneurobio.2020.101919] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 08/20/2020] [Accepted: 10/04/2020] [Indexed: 12/11/2022]
Abstract
The renin-angiotensin system (RAS) is one of the oldest hormone systems in vertebrate phylogeny. RAS was initially related to regulation of blood pressure and sodium and water homeostasis. However, local or paracrine RAS were later identified in many tissues, including brain, and play a major role in their physiology and pathophysiology. In addition, a major component, ACE2, is the entry receptor for SARS-CoV-2. Overactivation of tissue RAS leads several oxidative stress and inflammatory processes involved in aging-related degenerative changes. In addition, a third level of RAS, the intracellular or intracrine RAS (iRAS), with still unclear functions, has been observed. The possible interaction between the intracellular and extracellular RAS, and particularly the possible deleterious or beneficial effects of the iRAS activation are controversial. The dopaminergic system is particularly interesting to investigate the RAS as important functional interactions between dopamine and RAS have been observed in the brain and several peripheral tissues. Our recent observations in mitochondria and nucleus of dopaminergic neurons may clarify the role of the iRAS. This may be important for the developing of new therapeutic strategies, since the effects on both extracellular and intracellular RAS must be taken into account, and perhaps better understanding of COVID-19 cell mechanisms.
Collapse
Affiliation(s)
- Jose L Labandeira-Garcia
- Laboratory of Cellular and Molecular Neurobiology of Parkinson's Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Health Research Institute (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain; Networking Research Center on Neurodegenerative Diseases (CiberNed), Madrid, Spain.
| | - Rita Valenzuela
- Laboratory of Cellular and Molecular Neurobiology of Parkinson's Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Health Research Institute (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain; Networking Research Center on Neurodegenerative Diseases (CiberNed), Madrid, Spain
| | - Maria A Costa-Besada
- Laboratory of Cellular and Molecular Neurobiology of Parkinson's Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Health Research Institute (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain; Networking Research Center on Neurodegenerative Diseases (CiberNed), Madrid, Spain
| | - Begoña Villar-Cheda
- Laboratory of Cellular and Molecular Neurobiology of Parkinson's Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Health Research Institute (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain; Networking Research Center on Neurodegenerative Diseases (CiberNed), Madrid, Spain
| | - Ana I Rodriguez-Perez
- Laboratory of Cellular and Molecular Neurobiology of Parkinson's Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Health Research Institute (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain; Networking Research Center on Neurodegenerative Diseases (CiberNed), Madrid, Spain
| |
Collapse
|
16
|
Omsland M, Silic-Benussi M, Moles R, Sarkis S, Purcell DFJ, Yurick D, Khoury G, D'Agostino DM, Ciminale V, Franchini G. Functional properties and sequence variation of HTLV-1 p13. Retrovirology 2020; 17:11. [PMID: 32398094 PMCID: PMC7218495 DOI: 10.1186/s12977-020-00517-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 04/10/2020] [Indexed: 01/06/2023] Open
Abstract
Human T cell leukemia virus type-1 (HTLV-1) was the first retrovirus found to cause cancer in humans, but the mechanisms that drive the development of leukemia and other diseases associated with HTLV-1 infection remain to be fully understood. This review describes the functional properties of p13, an 87-amino acid protein coded by HTLV-1 open reading frame II (orf-II). p13 is mainly localized in the inner membrane of the mitochondria, where it induces potassium (K+) influx and reactive oxygen species (ROS) production, which can trigger either proliferation or apoptosis, depending on the ROS setpoint of the cell. Recent evidence indicates that p13 may influence the cell’s innate immune response to viral infection and the infected cell phenotype. Association of the HTLV-1 transcriptional activator, Tax, with p13 increases p13’s stability, leads to its partial co-localization with Tax in nuclear speckles, and reduces the ability of Tax to interact with the transcription cofactor CBP/p300. Comparison of p13 sequences isolated from HTLV-1-infected individuals revealed a small number of amino acid variations in the domains controlling the subcellular localization of the protein. Disruptive mutations of p13 were found in samples obtained from asymptomatic patients with low proviral load. p13 sequences of HTLV-1 subtype C isolates from indigenous Australian patients showed a high degree of identity among each other, with all samples containing a pattern of 5 amino acids that distinguished them from other subtypes. Further characterization of p13’s functional properties and sequence variants may lead to a deeper understanding of the impact of p13 as a contributor to the clinical manifestations of HTLV-1 infection.
Collapse
Affiliation(s)
- Maria Omsland
- Animal Models and Retroviral Vaccines Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | - Ramona Moles
- Animal Models and Retroviral Vaccines Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sarkis Sarkis
- Animal Models and Retroviral Vaccines Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Damian F J Purcell
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC, Australia
| | - David Yurick
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC, Australia
| | - Georges Khoury
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC, Australia.,Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | | | - Vincenzo Ciminale
- Veneto Institute of Oncology IOV-IRCCS, Padua, Italy.,Department of Surgery, Oncology, and Gastroenterology, University of Padua, Padua, Italy
| | - Genoveffa Franchini
- Animal Models and Retroviral Vaccines Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
17
|
Georgieva ER, Borbat PP, Fanouraki C, Freed JH. High-yield production in E. coli and characterization of full-length functional p13 II protein from human T-cell leukemia virus type 1. Protein Expr Purif 2020; 173:105659. [PMID: 32360379 DOI: 10.1016/j.pep.2020.105659] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 04/20/2020] [Accepted: 04/26/2020] [Indexed: 10/24/2022]
Abstract
Human T-cell leukemia virus type 1 is an oncovirus that causes aggressive adult T-cell leukemia but is also responsible for severe neurodegenerative and endocrine disorders. Combatting HTLV-1 infections requires a detailed understanding of the viral mechanisms in the host. Therefore, in vitro studies of important virus-encoded proteins would be critical. Our focus herein is on the HTLV-1-encoded regulatory protein p13II, which interacts with the inner mitochondrial membrane, increasing its permeability to cations (predominantly potassium, K+). Thereby, this protein affects mitochondrial homeostasis. We report on our progress in developing specific protocols for heterologous expression of p13II in E. coli, and methods for its purification and characterization. We succeeded in producing large quantities of highly-pure full-length p13II, deemed to be its fully functional form. Importantly, our particular approach based on the fusion of ubiquitin to the p13II C-terminus was instrumental in increasing the persistently low expression of soluble p13II in its native form. We subsequently developed approaches for protein spin labeling and a conformation study using double electron-electron resonance (DEER) spectroscopy and a fluorescence-based cation uptake assay for p13II in liposomes. Our DEER results point to large protein conformation changes occurring upon transition from the soluble to the membrane-bound state. The functional assay on p13II-assisted transport of thallium (Tl+) through the membrane, wherein Tl+ substituted for K+, suggests transmembrane potential involvement in p13II function. Our study lays the foundation for expansion of in vitro functional and structural investigations on p13II and would aid in the development of structure-based protein inhibitors and markers.
Collapse
Affiliation(s)
- Elka R Georgieva
- Department of Chemistry and Chemical Biology, Baker Laboratory, Cornell University, Ithaca, NY, 14853, USA.
| | - Peter P Borbat
- Department of Chemistry and Chemical Biology, Baker Laboratory, Cornell University, Ithaca, NY, 14853, USA; ACERT Center for Advanced ESR Technology, Cornell University, Ithaca, NY, 14853, USA
| | - Christina Fanouraki
- Department of Chemistry and Chemical Biology, Baker Laboratory, Cornell University, Ithaca, NY, 14853, USA
| | - Jack H Freed
- Department of Chemistry and Chemical Biology, Baker Laboratory, Cornell University, Ithaca, NY, 14853, USA; ACERT Center for Advanced ESR Technology, Cornell University, Ithaca, NY, 14853, USA
| |
Collapse
|
18
|
Gorwood J, Ejlalmanesh T, Bourgeois C, Mantecon M, Rose C, Atlan M, Desjardins D, Le Grand R, Fève B, Lambotte O, Capeau J, Béréziat V, Lagathu C. SIV Infection and the HIV Proteins Tat and Nef Induce Senescence in Adipose Tissue and Human Adipose Stem Cells, Resulting in Adipocyte Dysfunction. Cells 2020; 9:cells9040854. [PMID: 32244726 PMCID: PMC7226797 DOI: 10.3390/cells9040854] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 03/11/2020] [Accepted: 03/30/2020] [Indexed: 12/11/2022] Open
Abstract
Background: Aging is characterized by adipose tissue senescence, inflammation, and fibrosis, with trunk fat accumulation. Aging HIV-infected patients have a higher risk of trunk fat accumulation than uninfected individuals—suggesting that viral infection has a role in adipose tissue aging. We previously demonstrated that HIV/SIV infection and the Tat and Nef viral proteins were responsible for adipose tissue fibrosis and impaired adipogenesis. We hypothesized that SIV/HIV infection and viral proteins could induce adipose tissue senescence and thus lead to adipocyte dysfunctions. Methods: Features of tissue senescence were evaluated in subcutaneous and visceral adipose tissues of SIV-infected macaques and in human adipose stem cells (ASCs) exposed to Tat or Nef for up to 30 days. Results: p16 expression and p53 activation were higher in adipose tissue of SIV-infected macaques than in control macaques, indicating adipose tissue senescence. Tat and Nef induced higher senescence in ASCs, characterized by higher levels of senescence-associated beta-galactosidase activity, p16 expression, and p53 activation vs. control cells. Treatment with Tat and Nef also induced oxidative stress and mitochondrial dysfunction. Prevention of oxidative stress (using N-acetyl-cysteine) reduced senescence in ASCs. Adipocytes having differentiated from Nef-treated ASCs displayed alterations in adipogenesis with lower levels of triglyceride accumulation and adipocyte marker expression and secretion, and insulin resistance. Conclusion: HIV/SIV promotes adipose tissue senescence, which in turn may alter adipocyte function and contribute to insulin resistance.
Collapse
Affiliation(s)
- Jennifer Gorwood
- Lipodystrophies, Metabolic and Hormonal Adaptation, and Aging, UMR_S 938, Centre de Recherche Saint-Antoine-Institut Hospitalo-Universitaire de Cardiométabolisme et Nutrition (ICAN), INSERM, Sorbonne Université, F-75012 Paris, France; (J.G.); (T.E.); (M.M.); (C.R.); (M.A.); (B.F.); (J.C.)
| | - Tina Ejlalmanesh
- Lipodystrophies, Metabolic and Hormonal Adaptation, and Aging, UMR_S 938, Centre de Recherche Saint-Antoine-Institut Hospitalo-Universitaire de Cardiométabolisme et Nutrition (ICAN), INSERM, Sorbonne Université, F-75012 Paris, France; (J.G.); (T.E.); (M.M.); (C.R.); (M.A.); (B.F.); (J.C.)
| | - Christine Bourgeois
- Immunology of Viral infections and Autoimmune Diseases, IDMIT Department, IBFJ, U1184, INSERM-CEA-Université Paris Sud 11, F-92260 Fontenay-Aux-Roses and F-94270 Le Kremlin-Bicêtre, France; (C.B.); (O.L.)
| | - Matthieu Mantecon
- Lipodystrophies, Metabolic and Hormonal Adaptation, and Aging, UMR_S 938, Centre de Recherche Saint-Antoine-Institut Hospitalo-Universitaire de Cardiométabolisme et Nutrition (ICAN), INSERM, Sorbonne Université, F-75012 Paris, France; (J.G.); (T.E.); (M.M.); (C.R.); (M.A.); (B.F.); (J.C.)
| | - Cindy Rose
- Lipodystrophies, Metabolic and Hormonal Adaptation, and Aging, UMR_S 938, Centre de Recherche Saint-Antoine-Institut Hospitalo-Universitaire de Cardiométabolisme et Nutrition (ICAN), INSERM, Sorbonne Université, F-75012 Paris, France; (J.G.); (T.E.); (M.M.); (C.R.); (M.A.); (B.F.); (J.C.)
| | - Michael Atlan
- Lipodystrophies, Metabolic and Hormonal Adaptation, and Aging, UMR_S 938, Centre de Recherche Saint-Antoine-Institut Hospitalo-Universitaire de Cardiométabolisme et Nutrition (ICAN), INSERM, Sorbonne Université, F-75012 Paris, France; (J.G.); (T.E.); (M.M.); (C.R.); (M.A.); (B.F.); (J.C.)
- Plastic Surgery Department, Tenon Hospital, AP-HP, F-75020 Paris, France
| | - Delphine Desjardins
- IDMIT Department, Center for Immunology of Viral Infections and Autoimmune Diseases, Inserm, CEA, Université Paris Saclay, F-92260 Fontenay-aux-Roses, France; (D.D.); (R.L.G.)
| | - Roger Le Grand
- IDMIT Department, Center for Immunology of Viral Infections and Autoimmune Diseases, Inserm, CEA, Université Paris Saclay, F-92260 Fontenay-aux-Roses, France; (D.D.); (R.L.G.)
| | - Bruno Fève
- Lipodystrophies, Metabolic and Hormonal Adaptation, and Aging, UMR_S 938, Centre de Recherche Saint-Antoine-Institut Hospitalo-Universitaire de Cardiométabolisme et Nutrition (ICAN), INSERM, Sorbonne Université, F-75012 Paris, France; (J.G.); (T.E.); (M.M.); (C.R.); (M.A.); (B.F.); (J.C.)
- Diabétologie et Reproduction, PRISIS, Service d’Endocrinologie, Hôpital Saint-Antoine, AP-HP, F-75012 Paris, France
| | - Olivier Lambotte
- Immunology of Viral infections and Autoimmune Diseases, IDMIT Department, IBFJ, U1184, INSERM-CEA-Université Paris Sud 11, F-92260 Fontenay-Aux-Roses and F-94270 Le Kremlin-Bicêtre, France; (C.B.); (O.L.)
- Service de Médecine Interne et Immunologie Clinique, Groupe Hospitalier Universitaire Paris Sud, Hôpital Bicêtre, AP-HP, F-94270 Le Kremlin-Bicêtre, France
| | - Jacqueline Capeau
- Lipodystrophies, Metabolic and Hormonal Adaptation, and Aging, UMR_S 938, Centre de Recherche Saint-Antoine-Institut Hospitalo-Universitaire de Cardiométabolisme et Nutrition (ICAN), INSERM, Sorbonne Université, F-75012 Paris, France; (J.G.); (T.E.); (M.M.); (C.R.); (M.A.); (B.F.); (J.C.)
| | - Véronique Béréziat
- Lipodystrophies, Metabolic and Hormonal Adaptation, and Aging, UMR_S 938, Centre de Recherche Saint-Antoine-Institut Hospitalo-Universitaire de Cardiométabolisme et Nutrition (ICAN), INSERM, Sorbonne Université, F-75012 Paris, France; (J.G.); (T.E.); (M.M.); (C.R.); (M.A.); (B.F.); (J.C.)
- Correspondence: (V.B.); (C.L.); Tel.: +33140011321 (V.B.)
| | - Claire Lagathu
- Lipodystrophies, Metabolic and Hormonal Adaptation, and Aging, UMR_S 938, Centre de Recherche Saint-Antoine-Institut Hospitalo-Universitaire de Cardiométabolisme et Nutrition (ICAN), INSERM, Sorbonne Université, F-75012 Paris, France; (J.G.); (T.E.); (M.M.); (C.R.); (M.A.); (B.F.); (J.C.)
- Correspondence: (V.B.); (C.L.); Tel.: +33140011321 (V.B.)
| |
Collapse
|
19
|
Guzzi PH, Mercatelli D, Ceraolo C, Giorgi FM. Master Regulator Analysis of the SARS-CoV-2/Human Interactome. J Clin Med 2020; 9:E982. [PMID: 32244779 PMCID: PMC7230814 DOI: 10.3390/jcm9040982] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/27/2020] [Accepted: 03/28/2020] [Indexed: 12/20/2022] Open
Abstract
The recent epidemic outbreak of a novel human coronavirus called SARS-CoV-2 causing the respiratory tract disease COVID-19 has reached worldwide resonance and a global effort is being undertaken to characterize the molecular features and evolutionary origins of this virus. In this paper, we set out to shed light on the SARS-CoV-2/host receptor recognition, a crucial factor for successful virus infection. Based on the current knowledge of the interactome between SARS-CoV-2 and host cell proteins, we performed Master Regulator Analysis to detect which parts of the human interactome are most affected by the infection. We detected, amongst others, affected apoptotic and mitochondrial mechanisms, and a downregulation of the ACE2 protein receptor, notions that can be used to develop specific therapies against this new virus.
Collapse
Affiliation(s)
- Pietro H. Guzzi
- Department of Surgical and Medical Science, University of Catanzaro, 88100 Catanzaro, Italy;
| | - Daniele Mercatelli
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy; (D.M.); (C.C.)
| | - Carmine Ceraolo
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy; (D.M.); (C.C.)
| | - Federico M. Giorgi
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy; (D.M.); (C.C.)
| |
Collapse
|
20
|
Solà-Riera C, García M, Ljunggren HG, Klingström J. Hantavirus inhibits apoptosis by preventing mitochondrial membrane potential loss through up-regulation of the pro-survival factor BCL-2. PLoS Pathog 2020; 16:e1008297. [PMID: 32032391 PMCID: PMC7032725 DOI: 10.1371/journal.ppat.1008297] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 02/20/2020] [Accepted: 12/30/2019] [Indexed: 12/12/2022] Open
Abstract
Hantaviruses, zoonotic RNA viruses belonging to the order Bunyavirales, cause two severe acute diseases in humans, hemorrhagic fever with renal syndrome (HFRS) and hantavirus pulmonary syndrome (HPS). Hantavirus-infected patients show strong cytotoxic lymphocyte responses and hyperinflammation; however, infected cells remain mostly intact. Hantaviruses were recently shown to inhibit apoptosis in infected cells. By inhibiting granzyme B- and TRAIL-mediated apoptosis, hantaviruses specifically and efficiently inhibit cytotoxic lymphocyte-mediated killing of infected cells. Hantaviruses also strongly inhibit apoptosis triggered intrinsically; i.e., initiated through intracellular activation pathways different from those used by cytotoxic lymphocytes. However, insights into the latter mechanisms are currently largely unknown. Here, we dissected the mechanism behind how hantavirus infection, represented by the HFRS-causing Hantaan virus and the HPS-causing Andes virus, results in resistance to staurosporine-induced apoptosis. Less active caspase-8 and caspase-9, and consequently less active caspase-3, was observed in infected compared to uninfected staurosporine-exposed cells. While staurosporine-exposed uninfected cells showed massive release of pro-apoptotic cytochrome C into the cytosol, this was not observed in infected cells. Further, hantaviruses prevented activation of BAX and mitochondrial outer membrane permeabilization (MOMP). In parallel, a significant increase in levels of the pro-survival factor BCL-2 was observed in hantavirus-infected cells. Importantly, direct inhibition of BCL-2 by the inhibitor ABT-737, as well as silencing of BCL-2 by siRNA, resulted in apoptosis in staurosporine-exposed hantavirus-infected cells. Overall, we here provide a tentative mechanism by which hantaviruses protect infected cells from intrinsic apoptosis at the mitochondrial level by inducing an increased expression of the pro-survival factor BCL-2, thereby preventing MOMPs and subsequent activation of caspases. The variety of mechanisms used by hantaviruses to ensure survival of infected cells likely contribute to the persistent infection in natural hosts and may play a role in immunopathogenesis of HFRS and HPS in humans. Hantaviruses cause two severe, often fatal, diseases in humans: hemorrhagic fever with renal syndrome (HFRS) and hantavirus pulmonary syndrome (HPS; also called hantavirus cardiopulmonary syndrome (HCPS)). Two hallmarks of human hantavirus infection are robust immune cell activation and hyperinflammation. Despite these strong immune responses, hantavirus-infected cells do not succumb to cell death in patients. Recent studies have shown that hantaviruses hamper cytotoxic lymphocyte-mediated killing, by inhibiting cytotoxic granule-dependent induction of apoptosis and TRAIL-mediated apoptosis, as well as inhibiting intrinsic apoptosis. However, mechanisms behind hantavirus induced inhibition of intrinsic apoptosis have not been described. Here, we show that hantavirus infection leads to increased production of the anti-apoptotic protein BCL-2, hampering the permeabilization of mitochondria and thereby blocking downstream signaling and activation of caspases. Treatment of infected cells with a BCL-2 inhibitor, as well as silencing of BCL-2 with siRNA, both reverted the anti-apoptotic effect. Taken together, this study reveals new insights into the interactions between hantaviruses and infected cells and demonstrates novel mechanisms by which hantaviruses inhibit apoptosis by hampering the permeabilization of mitochondria.
Collapse
Affiliation(s)
- Carles Solà-Riera
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- * E-mail:
| | - Marina García
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Hans-Gustaf Ljunggren
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Jonas Klingström
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
21
|
Bourgeois C, Gorwood J, Barrail-Tran A, Lagathu C, Capeau J, Desjardins D, Le Grand R, Damouche A, Béréziat V, Lambotte O. Specific Biological Features of Adipose Tissue, and Their Impact on HIV Persistence. Front Microbiol 2019; 10:2837. [PMID: 31921023 PMCID: PMC6927940 DOI: 10.3389/fmicb.2019.02837] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 11/22/2019] [Indexed: 12/19/2022] Open
Abstract
Although white AT can contribute to anti-infectious immune responses, it can also be targeted and perturbed by pathogens. The AT's immune involvement is primarily due to strong pro-inflammatory responses (with both local and paracrine effects), and the large number of fat-resident macrophages. Adipocytes also exert direct antimicrobial responses. In recent years, it has been found that memory T cells accumulate in AT, where they provide efficient secondary responses against viral pathogens. These observations have prompted researchers to re-evaluate the links between obesity and susceptibility to infections. In contrast, AT serves as a reservoir for several persistence pathogens, such as human adenovirus Ad-36, Trypanosoma gondii, Mycobacterium tuberculosis, influenza A virus, and cytomegalovirus (CMV). The presence and persistence of bacterial DNA in AT has led to the concept of a tissue-specific microbiota. The unexpected coexistence of immune cells and pathogens within the specific AT environment is intriguing, and its impact on anti-infectious immune responses requires further evaluation. AT has been recently identified as a site of HIV persistence. In the context of HIV infection, AT is targeted by both the virus and the antiretroviral drugs. AT's intrinsic metabolic features, large overall mass, and wide distribution make it a major tissue reservoir, and one that may contribute to the pathophysiology of chronic HIV infections. Here, we review the immune, metabolic, viral, and pharmacological aspects that contribute to HIV persistence in AT. We also evaluate the respective impacts of both intrinsic and HIV-induced factors on AT's involvement as a viral reservoir. Lastly, we examine the potential consequences of HIV persistence on the metabolic and immune activities of AT.
Collapse
Affiliation(s)
- Christine Bourgeois
- Center for Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, CEA, Université Paris Sud, INSERM U1184, Fontenay-aux-Roses, France
| | - Jennifer Gorwood
- INSERM UMR_S 938, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire de Cardio-Métabolisme et Nutrition (ICAN), Sorbonne Université, Paris, France
| | - Aurélie Barrail-Tran
- Center for Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, CEA, Université Paris Sud, INSERM U1184, Fontenay-aux-Roses, France
- AP-HP, Service de Médecine Interne et Immunologie Clinique, Hôpital Bicêtre, Groupe Hospitalier Universitaire Paris Sud, Le Kremlin-Bicêtre, France
| | - Claire Lagathu
- INSERM UMR_S 938, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire de Cardio-Métabolisme et Nutrition (ICAN), Sorbonne Université, Paris, France
| | - Jacqueline Capeau
- INSERM UMR_S 938, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire de Cardio-Métabolisme et Nutrition (ICAN), Sorbonne Université, Paris, France
| | - Delphine Desjardins
- Center for Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, CEA, Université Paris Sud, INSERM U1184, Fontenay-aux-Roses, France
| | - Roger Le Grand
- Center for Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, CEA, Université Paris Sud, INSERM U1184, Fontenay-aux-Roses, France
| | - Abderaouf Damouche
- Center for Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, CEA, Université Paris Sud, INSERM U1184, Fontenay-aux-Roses, France
| | - Véronique Béréziat
- INSERM UMR_S 938, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire de Cardio-Métabolisme et Nutrition (ICAN), Sorbonne Université, Paris, France
| | - Olivier Lambotte
- Center for Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, CEA, Université Paris Sud, INSERM U1184, Fontenay-aux-Roses, France
- AP-HP, Service de Médecine Interne et Immunologie Clinique, Hôpital Bicêtre, Groupe Hospitalier Universitaire Paris Sud, Le Kremlin-Bicêtre, France
| |
Collapse
|
22
|
Saribas AS, Datta PK, Safak M. A comprehensive proteomics analysis of JC virus Agnoprotein-interacting proteins: Agnoprotein primarily targets the host proteins with coiled-coil motifs. Virology 2019; 540:104-118. [PMID: 31765920 DOI: 10.1016/j.virol.2019.10.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 10/13/2019] [Accepted: 10/16/2019] [Indexed: 11/29/2022]
Abstract
JC virus (JCV) Agnoprotein (Agno) plays critical roles in successful completion of the viral replication cycle. Understanding its regulatory roles requires a complete map of JCV-host protein interactions. Here, we report the first Agno interactome with host cellular targets utilizing "Two-Strep-Tag" affinity purification system coupled with mass spectroscopy (AP/MS). Proteomics data revealed that Agno primarily targets 501 cellular proteins, most of which contain "coiled-coil" motifs. Agno-host interactions occur in several cellular networks including those involved in protein synthesis and degradation; and cellular transport; and in organelles, including mitochondria, nucleus and ER-Golgi network. Among the Agno interactions, Rab11B, Importin and Crm-1 were first validated biochemically and further characterization was done for Crm-1, using a HIV-1 Rev-M10-like Agno mutant (L33D + E34L), revealing the critical roles of L33 and E34 residues in Crm-1 interaction. This comprehensive proteomics data provides new foundations to unravel the critical regulatory roles of Agno during the JCV life cycle.
Collapse
Affiliation(s)
- A Sami Saribas
- Department of Neuroscience, Laboratory of Molecular Neurovirology, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Prasun K Datta
- Department of Neuroscience, Laboratory of Molecular Neurovirology, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Mahmut Safak
- Department of Neuroscience, Laboratory of Molecular Neurovirology, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA, 19140, USA.
| |
Collapse
|
23
|
Abstract
The integration of drugs into nanocarriers favorably altered their pharmacodynamics and pharmacokinetics compared to free drugs, and increased their therapeutic index. However, selective cellular internalization in diseased tissues rather than normal tissues still presents a formidable challenge. In this chapter I will cover solutions involving environment-responsive cell-penetrating peptides (CPPs). I will discuss properties of CPPs as universal cellular uptake enhancers, and the modifications imparted to CPP-modified nanocarriers to confine CPP activation to diseased tissues.
Collapse
|
24
|
Acar DD, Stroobants VJE, Favoreel H, Saelens X, Nauwynck HJ. Identification of peptide domains involved in the subcellular localization of the feline coronavirus 3b protein. J Gen Virol 2019; 100:1417-1430. [PMID: 31483243 PMCID: PMC7079696 DOI: 10.1099/jgv.0.001321] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Feline coronavirus (FCoV) has been identified as the aetiological agent of feline infectious peritonitis (FIP), a highly fatal systemic disease in cats. FCoV open reading frame 3 (ORF3) encodes accessory proteins 3a, 3b and 3 c. The FCoV 3b accessory protein consists of 72 amino acid residues and localizes to nucleoli and mitochondria. The present work focused on peptide domains within FCoV 3b that drive its intracellular trafficking. Transfection of different cell types with FCoV 3b fused to enhanced green fluorescent protein (EGFP) or 3×FLAG confirmed localization of FCoV 3b in the mitochondria and nucleoli. Using serial truncated mutants, we showed that nucleolar accumulation is controlled by a joint nucleolar and nuclear localization signal (NoLS/NLS) in which the identified overlapping pat4 motifs (residues 53–57) play a critical role. Mutational analysis also revealed that mitochondrial translocation is mediated by N-terminal residues 10–35, in which a Tom20 recognition motif (residues 13–17) and two other overlapping hexamers (residues 24–30) associated with mitochondrial targeting were identified. In addition, a second Tom20 recognition motif was identified further downstream (residues 61–65), although the mitochondrial translocation evoked by these residues seemed less efficient as a diffuse cytoplasmic distribution was also observed. Assessing the spatiotemporal distribution of FCoV 3b did not provide convincing evidence of dynamic shuttling behaviour between the nucleoli and the mitochondria.
Collapse
Affiliation(s)
- Delphine D. Acar
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Veerle J. E. Stroobants
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Herman Favoreel
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Xavier Saelens
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
| | - Hans J. Nauwynck
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
- *Correspondence: Hans J. Nauwynck,
| |
Collapse
|
25
|
Orthohantaviruses belonging to three phylogroups all inhibit apoptosis in infected target cells. Sci Rep 2019; 9:834. [PMID: 30696898 PMCID: PMC6351540 DOI: 10.1038/s41598-018-37446-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 12/03/2018] [Indexed: 12/04/2022] Open
Abstract
Orthohantaviruses, previously known as hantaviruses, are zoonotic viruses that can cause hantavirus pulmonary syndrome (HPS) and hemorrhagic fever with renal syndrome (HFRS) in humans. The HPS-causing Andes virus (ANDV) and the HFRS-causing Hantaan virus (HTNV) have anti-apoptotic effects. To investigate if this represents a general feature of orthohantaviruses, we analysed the capacity of six different orthohantaviruses – belonging to three distinct phylogroups and representing both pathogenic and non-pathogenic viruses – to inhibit apoptosis in infected cells. Primary human endothelial cells were infected with ANDV, HTNV, the HFRS-causing Puumala virus (PUUV) and Seoul virus, as well as the putative non-pathogenic Prospect Hill virus and Tula virus. Infected cells were then exposed to the apoptosis-inducing chemical staurosporine or to activated human NK cells exhibiting a high cytotoxic potential. Strikingly, all orthohantaviruses inhibited apoptosis in both settings. Moreover, we show that the nucleocapsid (N) protein from all examined orthohantaviruses are potential targets for caspase-3 and granzyme B. Recombinant N protein from ANDV, PUUV and the HFRS-causing Dobrava virus strongly inhibited granzyme B activity and also, to certain extent, caspase-3 activity. Taken together, this study demonstrates that six different orthohantaviruses inhibit apoptosis, suggesting this to be a general feature of orthohantaviruses likely serving as a mechanism of viral immune evasion.
Collapse
|
26
|
Chen Q, Zheng L, Mao Q, Liu J, Wang H, Jia D, Chen H, Wu W, Wei T. Fibrillar structures induced by a plant reovirus target mitochondria to activate typical apoptotic response and promote viral infection in insect vectors. PLoS Pathog 2019; 15:e1007510. [PMID: 30653614 PMCID: PMC6353215 DOI: 10.1371/journal.ppat.1007510] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 01/30/2019] [Accepted: 12/07/2018] [Indexed: 11/19/2022] Open
Abstract
Numerous plant viruses that cause significant agricultural problems are persistently transmitted by insect vectors. We wanted to see if apoptosis was involved in viral infection process in the vector. We found that a plant reovirus (rice gall dwarf virus, RGDV) induced typical apoptotic response during viral replication in the leafhopper vector and cultured vector cells, as demonstrated by mitochondrial degeneration and membrane potential decrease. Fibrillar structures formed by nonstructural protein Pns11 of RGDV targeted the outer membrane of mitochondria, likely by interaction with an apoptosis-related mitochondrial protein in virus-infected leafhopper cells or nonvector insect cells. Such association of virus-induced fibrillar structures with mitochondria clearly led to mitochondrial degeneration and membrane potential decrease, suggesting that RGDV Pns11 was the inducer of apoptotic response in insect vectors. A caspase inhibitor treatment and knockdown of caspase gene expression using RNA interference each reduced apoptosis and viral accumulation, while the knockdown of gene expression for the inhibitor of apoptosis protein improved apoptosis and viral accumulation. Thus, RGDV exploited caspase-dependent apoptotic response to promote viral infection in insect vectors. For the first time, we directly confirmed that a nonstructural protein encoded by a persistent plant virus can induce the typical apoptotic response to benefit viral transmission by insect vectors.
Collapse
Affiliation(s)
- Qian Chen
- Fujian Province Key Laboratory of Plant Virology, Institute of Plant Virology, State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Vector-borne Virus Research Center, Fujian Agriculture and Forestry University, Fuzhou, Fujian, PR China
| | - Limin Zheng
- Fujian Province Key Laboratory of Plant Virology, Institute of Plant Virology, State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Vector-borne Virus Research Center, Fujian Agriculture and Forestry University, Fuzhou, Fujian, PR China
- Key Laboratory of Pest Management of Horticultural Crop of Hunan Province, Hunan Plant Protection Institute, Hunan Academy of Agricultural Science, Changsha, PR China
| | - Qianzhuo Mao
- Fujian Province Key Laboratory of Plant Virology, Institute of Plant Virology, State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Vector-borne Virus Research Center, Fujian Agriculture and Forestry University, Fuzhou, Fujian, PR China
| | - Jiejie Liu
- Fujian Province Key Laboratory of Plant Virology, Institute of Plant Virology, State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Vector-borne Virus Research Center, Fujian Agriculture and Forestry University, Fuzhou, Fujian, PR China
| | - Haitao Wang
- Fujian Province Key Laboratory of Plant Virology, Institute of Plant Virology, State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Vector-borne Virus Research Center, Fujian Agriculture and Forestry University, Fuzhou, Fujian, PR China
| | - Dongsheng Jia
- Fujian Province Key Laboratory of Plant Virology, Institute of Plant Virology, State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Vector-borne Virus Research Center, Fujian Agriculture and Forestry University, Fuzhou, Fujian, PR China
| | - Hongyan Chen
- Fujian Province Key Laboratory of Plant Virology, Institute of Plant Virology, State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Vector-borne Virus Research Center, Fujian Agriculture and Forestry University, Fuzhou, Fujian, PR China
| | - Wei Wu
- Fujian Province Key Laboratory of Plant Virology, Institute of Plant Virology, State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Vector-borne Virus Research Center, Fujian Agriculture and Forestry University, Fuzhou, Fujian, PR China
| | - Taiyun Wei
- Fujian Province Key Laboratory of Plant Virology, Institute of Plant Virology, State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Vector-borne Virus Research Center, Fujian Agriculture and Forestry University, Fuzhou, Fujian, PR China
| |
Collapse
|
27
|
Chasapis CT. Interactions between metal binding viral proteins and human targets as revealed by network-based bioinformatics. J Inorg Biochem 2018; 186:157-161. [DOI: 10.1016/j.jinorgbio.2018.06.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 05/24/2018] [Accepted: 06/14/2018] [Indexed: 12/13/2022]
|
28
|
Caspase-Dependent Apoptosis Induction via Viral Protein ORF4 of Porcine Circovirus 2 Binding to Mitochondrial Adenine Nucleotide Translocase 3. J Virol 2018; 92:JVI.00238-18. [PMID: 29491154 DOI: 10.1128/jvi.00238-18] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 02/17/2018] [Indexed: 12/31/2022] Open
Abstract
Apoptosis is an essential strategy of host defense responses and is used by viruses to maintain their life cycles. However, the apoptotic signals involved in virus replication are poorly known. In the present study, we report the molecular mechanism of apoptotic induction by the viral protein ORF4, a newly identified viral protein of porcine circovirus type 2 (PCV2). Apoptosis detection revealed not only that the activity of caspase-3 and -9 is increased in PCV2-infected and ORF4-transfected cells but also that cytochrome c release from the mitochondria to the cytosol is upregulated. Subsequently, ORF4 protein colocalization with adenine nucleotide translocase 3 (ANT3) was observed using structured illumination microscopy. Moreover, coimmunoprecipitation and pulldown analyses confirmed that the ORF4 protein interacts directly with mitochondrial ANT3 (mtANT3). Binding domain analysis further confirmed that N-terminal residues 1 to 30 of the ORF4 protein, comprising a mitochondrial targeting signal, are essential for the interaction with ANT3. Knockdown of ANT3 markedly inhibited the apoptotic induction of both ORF4 protein and PCV2, indicating that ANT3 plays an important role in ORF4 protein-induced apoptosis during PCV2 infection. Taken together, these data indicate that the ORF4 protein is a mitochondrial targeting protein that induces apoptosis by interacting with ANT3 through the mitochondrial pathway.IMPORTANCE The porcine circovirus type 2 (PCV2) protein ORF4 is a newly identified viral protein; however, little is known about its functions. Apoptosis is an essential strategy of the host defense response and is used by viruses to maintain their life cycles. In the present study, we report the molecular mechanism of the apoptosis induced by the ORF4 protein. The ORF4 protein contains a mitochondrial targeting signal and is an unstable protein that is degraded by the proteasome-dependent pathway. Viral protein ORF4 triggers caspase-3- and -9-dependent cellular apoptosis in mitochondria by directly binding to ANT3. We conclude that the ORF4 protein is a mitochondrial targeting protein and reveal a mechanism whereby circovirus recruits ANT3 to induce apoptosis.
Collapse
|
29
|
Huang XY, Li D, Chen ZX, Huang YH, Gao WY, Zheng BY, Wang XZ. Hepatitis B Virus X protein elevates Parkin-mediated mitophagy through Lon Peptidase in starvation. Exp Cell Res 2018; 368:75-83. [PMID: 29689279 DOI: 10.1016/j.yexcr.2018.04.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 04/05/2018] [Accepted: 04/15/2018] [Indexed: 01/13/2023]
Abstract
Hepatocellular Carcinoma (HCC) is the fifth most prevalent cancer worldwide. Specially, Hepatitis B viurs X protein (HBx) is a leading factor in the progression of Hepatitis B viurs-related HCC. Nutrient-deprived tumor microenvironment also contributes to tumor development. However, the role of HBx in nutrient-deprived HCC has received little investigation. Here, we show that HBx elevates PINK1-Parkin mediating mitophagy in starvation. HBx not only increases the PINK1/Parkin gene expression but also accelerates Parkin recruitment to partial mitochondria. Further analysis indicates that, HBx either promotes mitochondrial unfolded protein response, with remarkable mitochondrial LONP1 increases, or reduces LONP1 expression in cytosol inducing LONP1-Parkin pathway, both consequently enhancing mitophagy. Moreover, the enhanced mitophagy lowers mitochondrial apoptosis in starved hepatoma cells, and Bax is implied in the machinery. In addition, we define differential centrifuge, 3000 g or 12,000 g to pellet mitochondria, as an effective method to obtain distinct mitochondria. In collect, HBx regulates diverse aspects of LONP1 and Parkin, enhancing mitophagy in starvation. This study may shed new insights into the machinery development of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Xiao-Yun Huang
- Department of Gastroenterology, Fujian Medical University Union Hospital, 29, Xinquan Road, Gulou, Fuzhou, Fujian 350001, PR China.
| | - Dan Li
- Department of Gastroenterology, Fujian Medical University Union Hospital, 29, Xinquan Road, Gulou, Fuzhou, Fujian 350001, PR China.
| | - Zhi-Xin Chen
- Department of Gastroenterology, Fujian Medical University Union Hospital, 29, Xinquan Road, Gulou, Fuzhou, Fujian 350001, PR China.
| | - Yue-Hong Huang
- Department of Gastroenterology, Fujian Medical University Union Hospital, 29, Xinquan Road, Gulou, Fuzhou, Fujian 350001, PR China.
| | - Wen-Yu Gao
- Department of Gastroenterology, Fujian Medical University Union Hospital, 29, Xinquan Road, Gulou, Fuzhou, Fujian 350001, PR China.
| | - Bi-Yun Zheng
- Department of Gastroenterology, Fujian Medical University Union Hospital, 29, Xinquan Road, Gulou, Fuzhou, Fujian 350001, PR China.
| | - Xiao-Zhong Wang
- Department of Gastroenterology, Fujian Medical University Union Hospital, 29, Xinquan Road, Gulou, Fuzhou, Fujian 350001, PR China.
| |
Collapse
|
30
|
You DG, Lee HR, Kim WK, Kim HJ, Lee GY, Yoo YD. Hepatitis C virus p7 induces mitochondrial depolarization of isolated liver mitochondria. Mol Med Rep 2017; 16:9533-9538. [PMID: 29039530 DOI: 10.3892/mmr.2017.7809] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 03/20/2017] [Indexed: 11/06/2022] Open
Abstract
Hepatitis C virus (HCV)‑encoded protein p7 is a viroporin that acts as an ion channel and is indispensable for HCV particle production. Although the main target of HCV p7 is the endoplasmic reticulum, it also targets mitochondria. HCV‑infected cells show mitochondrial depolarization and ATP depletion; however, the function of HCV p7 in mitochondria is not fully understood. The present study demonstrated that treatment of isolated mouse liver mitochondria with the synthesized HCV p7 protein induced mitochondrial dysfunction. It also demonstrated that HCV p7 targeted isolated mouse liver mitochondria and induced mitochondrial depolarization. In addition, HCV p7 triggered matrix acidification and, ultimately, a decrease in ATP synthesis in isolated mitochondria. These findings indicate that targeting of mitochondria by HCV p7 in infected cells causes mitochondrial dysfunction to support HCV particle production. The present study provided evidence for the role of HCV p7 in mitochondria, and may lead to the development of novel strategies for HCV therapy.
Collapse
Affiliation(s)
- Deok-Gyun You
- Laboratory of Molecular Cell Biology, Graduate School of Medicine, College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Hye-Ra Lee
- Laboratory of Molecular Cell Biology, Graduate School of Medicine, College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Won-Ki Kim
- Department of Neuroscience, College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Hyung Jung Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Yonsei University Health System, Seoul 03722, Republic of Korea
| | - Gi Young Lee
- Laboratory of Molecular Cell Biology, Graduate School of Medicine, College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Young Do Yoo
- Laboratory of Molecular Cell Biology, Graduate School of Medicine, College of Medicine, Korea University, Seoul 02841, Republic of Korea
| |
Collapse
|
31
|
Voltage-Dependent Anion Channel 1 Interacts with Ribonucleoprotein Complexes To Enhance Infectious Bursal Disease Virus Polymerase Activity. J Virol 2017; 91:JVI.00584-17. [PMID: 28592532 DOI: 10.1128/jvi.00584-17] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 05/28/2017] [Indexed: 02/05/2023] Open
Abstract
Infectious bursal disease virus (IBDV) is a double-stranded RNA (dsRNA) virus. Segment A contains two overlapping open reading frames (ORFs), which encode viral proteins VP2, VP3, VP4, and VP5. Segment B contains one ORF and encodes the viral RNA-dependent RNA polymerase, VP1. IBDV ribonucleoprotein complexes are composed of VP1, VP3, and dsRNA and play a critical role in mediating viral replication and transcription during the virus life cycle. In the present study, we identified a cellular factor, VDAC1, which was upregulated during IBDV infection and found to mediate IBDV polymerase activity. VDAC1 senses IBDV infection by interacting with viral proteins VP1 and VP3. This association is caused by RNA bridging, and all three proteins colocalize in the cytoplasm. Furthermore, small interfering RNA (siRNA)-mediated downregulation of VDAC1 resulted in a reduction in viral polymerase activity and a subsequent decrease in viral yield. Moreover, overexpression of VDAC1 enhanced IBDV polymerase activity. We also found that the viral protein VP3 can replace segment A to execute polymerase activity. A previous study showed that mutations in the C terminus of VP3 directly influence the formation of VP1-VP3 complexes. Our immunoprecipitation experiments demonstrated that protein-protein interactions between VDAC1 and VP3 and between VDAC1 and VP1 play a role in stabilizing the interaction between VP3 and VP1, further promoting IBDV polymerase activity.IMPORTANCE The cellular factor VDAC1 controls the entry and exit of mitochondrial metabolites and plays a pivotal role during intrinsic apoptosis by mediating the release of many apoptogenic molecules. Here we identify a novel role of VDAC1, showing that VDAC1 interacts with IBDV ribonucleoproteins (RNPs) and facilitates IBDV replication by enhancing IBDV polymerase activity through its ability to stabilize interactions in RNP complexes. To our knowledge, this is the first report that VDAC1 is specifically involved in regulating IBDV RNA polymerase activity, providing novel insight into virus-host interactions.
Collapse
|
32
|
He L, Hu X, Zhu M, Liang Z, Chen F, Zhu L, Kuang S, Cao G, Xue R, Gong C. Identification and characterization of vp7 gene in Bombyx mori cytoplasmic polyhedrosis virus. Gene 2017; 627:343-350. [PMID: 28668346 PMCID: PMC7173298 DOI: 10.1016/j.gene.2017.06.048] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Revised: 05/31/2017] [Accepted: 06/27/2017] [Indexed: 01/15/2023]
Abstract
The genome of Bombyx mori cytoplasmic polyhedrosis virus (BmCPV) contains 10 double stranded RNA segments (S1-S10). The segment 7 (S7) encodes 50kDa protein which is considered as a structural protein. The expression pattern and function of p50 in the virus life cycle are still unclear. In this study, the viral structural protein 7 (VP7) polyclonal antibody was prepared with immunized mouse to explore the presence of small VP7 gene-encoded proteins in Bombyx mori cytoplasmic polyhedrosis virus. The expression pattern of vp7 gene was investigated by its overexpression in BmN cells. In addition to VP7, supplementary band was identified with western blotting technique. The virion, BmCPV infected cells and midguts were also examined using western blotting technique. 4, 2 and 5 bands were detected in the corresponding samples, respectively. The replication of BmCPV genome in the cultured cells and midgut of silkworm was decreased by reducing the expression level of vp7 gene using RNA interference. In immunoprecipitation experiments, using a polyclonal antiserum directed against the VP7, one additional shorter band in BmCPV infected midguts was detected, and then the band was analyzed with mass spectrum (MS), the MS results showed thatone candidate interacted protein (VP7 voltage-dependent anion-selective channel-like isoform, VDAC) was identified from silkworm. We concluded that the novel viral product was generated with a leaky scanning mechanism and the VDAC may be an interacted protein with VP7.
Collapse
Affiliation(s)
- Lei He
- School of Biology & Basic Medical Science, Soochow University, Suzhou 215123, China
| | - Xiaolong Hu
- School of Biology & Basic Medical Science, Soochow University, Suzhou 215123, China; National Engineering Laboratory for Modern Silk, Soochow University, Suzhou 215123, China
| | - Min Zhu
- School of Biology & Basic Medical Science, Soochow University, Suzhou 215123, China
| | - Zi Liang
- School of Biology & Basic Medical Science, Soochow University, Suzhou 215123, China
| | - Fei Chen
- School of Biology & Basic Medical Science, Soochow University, Suzhou 215123, China
| | - Liyuan Zhu
- School of Biology & Basic Medical Science, Soochow University, Suzhou 215123, China
| | - Sulan Kuang
- School of Biology & Basic Medical Science, Soochow University, Suzhou 215123, China
| | - Guangli Cao
- School of Biology & Basic Medical Science, Soochow University, Suzhou 215123, China; National Engineering Laboratory for Modern Silk, Soochow University, Suzhou 215123, China
| | - Renyu Xue
- School of Biology & Basic Medical Science, Soochow University, Suzhou 215123, China; National Engineering Laboratory for Modern Silk, Soochow University, Suzhou 215123, China
| | - Chengliang Gong
- School of Biology & Basic Medical Science, Soochow University, Suzhou 215123, China; National Engineering Laboratory for Modern Silk, Soochow University, Suzhou 215123, China.
| |
Collapse
|
33
|
Pickard A, Durzynska J, McCance DJ, Barton ER. The IGF axis in HPV associated cancers. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2017; 772:67-77. [PMID: 28528691 DOI: 10.1016/j.mrrev.2017.01.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 01/30/2017] [Accepted: 01/30/2017] [Indexed: 02/07/2023]
Abstract
Human papillomaviruses (HPV) infect and replicate in stratified epithelium at cutaneous and mucosal surfaces. The proliferation and maintenance of keratinocytes, the cells which make up this epithelium, are controlled by a number of growth factor receptors such as the keratinocyte growth factor receptor (KGFR, also called fibroblast growth factor receptor 2b (FGFR2b)), the epithelial growth factor receptor (EGFR) and the insulin-like growth factor receptors 1 and 2 (IGF1R and IGF2R). In this review, we will delineate the mutation, gene transcription, translation and processing of the IGF axis within HPV associated cancers. The IGFs are key for developmental and postnatal growth of almost all tissues; we explore whether this crucial axis has been hijacked by HPV.
Collapse
MESH Headings
- Cell Proliferation
- ErbB Receptors/genetics
- ErbB Receptors/metabolism
- Gene Expression Regulation, Neoplastic
- Humans
- Keratinocytes/cytology
- Keratinocytes/virology
- Neoplasms/genetics
- Neoplasms/virology
- Papillomaviridae/pathogenicity
- Receptor, Fibroblast Growth Factor, Type 2/genetics
- Receptor, Fibroblast Growth Factor, Type 2/metabolism
- Receptor, IGF Type 1
- Receptor, IGF Type 2/genetics
- Receptor, IGF Type 2/metabolism
- Receptors, Somatomedin/genetics
- Receptors, Somatomedin/metabolism
- Somatomedins/genetics
- Somatomedins/metabolism
Collapse
Affiliation(s)
- Adam Pickard
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, BT9 7AE, UK; Wellcome Centre for Cell Matrix Research, University of Manchester, M13 9PL, UK.
| | - Julia Durzynska
- Department of Molecular Virology, Institute of Experimental Biology, A. Mickiewicz University, ul. Umultowska 89, 61-614, Poznań, Poland; Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, FL, USA
| | - Dennis J McCance
- Department of Pathology, University of New Mexico, Albuquerque, NM, USA
| | - Elisabeth R Barton
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, FL, USA
| |
Collapse
|
34
|
Enterovirus 71 2B Induces Cell Apoptosis by Directly Inducing the Conformational Activation of the Proapoptotic Protein Bax. J Virol 2016; 90:9862-9877. [PMID: 27558414 DOI: 10.1128/jvi.01499-16] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 08/15/2016] [Indexed: 12/13/2022] Open
Abstract
To survive and replicate within a host, many viruses have evolved strategies that target crucial components within the apoptotic cascade, leading to either inhibition or induction of cell apoptosis. Enterovirus 71 (EV71) infections have been demonstrated to impact the mitochondrial apoptotic pathway and induce apoptosis in many cell lines. However, the detailed mechanism of EV71-induced apoptosis remains to be elucidated. In this study, we report that EV71 2B protein (2B) localized to the mitochondria and induced cell apoptosis by interacting directly with and activating the proapoptotic protein Bax. 2B recruited Bax to the mitochondria and induced Bax conformational activation. In addition, mitochondria isolated from 2B-expressing cells that were treated with a recombinant Bax showed increased Bax interaction and cytochrome c (Cyt c) release. Importantly, apoptosis in cells with either EV71 infection or 2B expression was dramatically reduced in Bax knockdown cells but not in Bak knockdown cells, suggesting that Bax played a pivotal role in EV71- or 2B-induced apoptosis. Further studies indicate that a hydrophobic region of 18 amino acids (aa) in the C-terminal region of 2B (aa 63 to 80) was responsible for the location of 2B in the mitochondria. A hydrophilic region of 14 aa in the N-terminal region of 2B was functional in Bax interaction and its subsequent activation. Moreover, overexpression of the antiapoptotic protein Bcl-XL abrogates 2B-induced release of Cyt c and caspase activation. Therefore, this study provides direct evidence that EV71 2B induces cell apoptosis and impacts the mitochondrial apoptotic pathway by directly modulating the redistribution and activation of proapoptotic protein Bax. IMPORTANCE EV71 infections are usually accompanied by severe neurological complications. It has also been postulated that the induction of cell apoptosis resulting from tissue damage is a possible process of EV71-related pathogenesis. In this study, we report that EV71 2B protein (2B) localized to the mitochondria and induced cell apoptosis by interacting directly with and activating the proapoptotic protein Bax. This study provides evidence that EV71 induces cell apoptosis by modulating Bax activation and reveals important clues regarding the mechanism of Cyt c release and mitochondrial permeabilization during EV71 infection.
Collapse
|
35
|
Thakur A, Rajput A, Kumar M. MSLVP: prediction of multiple subcellular localization of viral proteins using a support vector machine. MOLECULAR BIOSYSTEMS 2016; 12:2572-86. [DOI: 10.1039/c6mb00241b] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Knowledge of the subcellular location (SCL) of viral proteins in the host cell is important for understanding their function in depth.
Collapse
Affiliation(s)
- Anamika Thakur
- Bioinformatics Centre
- Institute of Microbial Technology
- Council of Scientific and Industrial Research
- Chandigarh-160036
- India
| | - Akanksha Rajput
- Bioinformatics Centre
- Institute of Microbial Technology
- Council of Scientific and Industrial Research
- Chandigarh-160036
- India
| | - Manoj Kumar
- Bioinformatics Centre
- Institute of Microbial Technology
- Council of Scientific and Industrial Research
- Chandigarh-160036
- India
| |
Collapse
|
36
|
Profiles of teleost DNA fragmentation factor alpha and beta from rock bream (Oplegnathus fasciatus): molecular characterization and genomic structure and gene expression in immune stress. Genes Genomics 2015. [DOI: 10.1007/s13258-015-0359-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
37
|
Beaupere C, Garcia M, Larghero J, Fève B, Capeau J, Lagathu C. The HIV proteins Tat and Nef promote human bone marrow mesenchymal stem cell senescence and alter osteoblastic differentiation. Aging Cell 2015; 14:534-46. [PMID: 25847297 PMCID: PMC4531068 DOI: 10.1111/acel.12308] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/14/2014] [Indexed: 12/28/2022] Open
Abstract
To maintain bone mass turnover and bone mineral density (BMD), bone marrow (BM) mesenchymal stem cells (MSCs) are constantly recruited and subsequently differentiated into osteoblasts. HIV-infected patients present lower BMD than non-HIV infected individuals and a higher prevalence of osteopenia/osteoporosis. In antiretroviral treatment (ART)-naive patients, encoded HIV proteins represent pathogenic candidates. They are released by infected cells within BM and can impact on neighbouring cells. In this study, we tested whether HIV proteins Tat and/or Nef could induce senescence of human BM-MSCs and reduce their capacity to differentiate into osteoblasts. When compared to nontreated cells, MSCs chronically treated with Tat and/or Nef up to 30 days reduced their proliferative activity and underwent early senescence, associated with increased oxidative stress and mitochondrial dysfunction. The antioxidant molecule N-acetyl- cysteine had no or minimal effects on Tat- or Nef-induced senescence. Tat but not Nef induced an early increase in NF-κB activity and cytokine/chemokine secretion. Tat-induced effects were prevented by the NF-κB inhibitor parthenolide, indicating that Tat triggered senescence via NF-κB activation leading to oxidative stress. Otherwise, Nef- but not Tat-treated cells displayed early inhibition of autophagy. Rapamycin, an autophagy inducer, reversed Nef-induced senescence and oxidative stress. Moreover, Tat+Nef had cumulative effects. Finally, Tat and/or Nef decreased the MSC potential of osteoblastic differentiation. In conclusion, our in vitro data show that Tat and Nef could reduce the number of available precursors by inducing MSC senescence, through either enhanced inflammation or reduced autophagy. These results offer new insights into the pathophysiological mechanisms of decreased BMD in HIV-infected patients.
Collapse
Affiliation(s)
- Carine Beaupere
- Sorbonne Universités; UPMC Univ Paris 06; UMR_S 938; CDR Saint-Antoine F-75012 Paris France
- INSERM; UMR_S 938; CDR Saint-Antoine F-75012 Paris France
- Institute of Cardiometabolism and Nutrition; Paris France
| | - Marie Garcia
- Sorbonne Universités; UPMC Univ Paris 06; UMR_S 938; CDR Saint-Antoine F-75012 Paris France
- INSERM; UMR_S 938; CDR Saint-Antoine F-75012 Paris France
- Institute of Cardiometabolism and Nutrition; Paris France
| | - Jerome Larghero
- Inserm; UMR1160; Institut Universitaire d'Hématologie; Hôpital Saint-Louis; 75010 Paris France
- AP-HP; Unité de Thérapie Cellulaire et CIC de Biothérapies; Hôpital Saint Louis; Paris France
- Univ Paris Diderot; Sorbonne Paris Cité F-75475 Paris France
| | - Bruno Fève
- Sorbonne Universités; UPMC Univ Paris 06; UMR_S 938; CDR Saint-Antoine F-75012 Paris France
- INSERM; UMR_S 938; CDR Saint-Antoine F-75012 Paris France
- Institute of Cardiometabolism and Nutrition; Paris France
- APHP; Hôpital Saint-Antoine; F-75012 Paris France
| | - Jacqueline Capeau
- Sorbonne Universités; UPMC Univ Paris 06; UMR_S 938; CDR Saint-Antoine F-75012 Paris France
- INSERM; UMR_S 938; CDR Saint-Antoine F-75012 Paris France
- Institute of Cardiometabolism and Nutrition; Paris France
- APHP; Hôpital Tenon; F-75020 Paris France
| | - Claire Lagathu
- Sorbonne Universités; UPMC Univ Paris 06; UMR_S 938; CDR Saint-Antoine F-75012 Paris France
- INSERM; UMR_S 938; CDR Saint-Antoine F-75012 Paris France
- Institute of Cardiometabolism and Nutrition; Paris France
| |
Collapse
|
38
|
Rabies virus phosphoprotein interacts with mitochondrial Complex I and induces mitochondrial dysfunction and oxidative stress. J Neurovirol 2015; 21:370-82. [PMID: 25698500 DOI: 10.1007/s13365-015-0320-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 01/09/2015] [Accepted: 01/16/2015] [Indexed: 12/25/2022]
Abstract
Our previous studies in an experimental model of rabies showed neuronal process degeneration in association with severe clinical disease. Cultured adult rodent dorsal root ganglion neurons infected with challenge virus standard (CVS)-11 strain of rabies virus (RABV) showed axonal swellings and reduced axonal growth with evidence of oxidative stress. We have shown that CVS infection alters a variety of mitochondrial parameters and increases reactive oxygen species (ROS) production and mitochondrial Complex I activity vs. mock infection. We have hypothesized that a RABV protein targets mitochondria and triggers dysfunction. Mitochondrial extracts of mouse neuroblastoma cells were analyzed with a proteomics approach. We have identified peptides belonging to the RABV nucleocapsid protein (N), phosphoprotein (P), and glycoprotein (G), and our data indicate that the extract was most highly enriched with P. P was also detected by immunoblotting in RABV-infected purified mitochondrial extracts and also in Complex I immunoprecipitates from the extracts but not in mock-infected extracts. A plasmid expressing P in cells increased Complex I activity and increased ROS generation, whereas expression of other RABV proteins did not. We have analyzed recombinant plasmids encoding various P gene segments. Expression of a peptide from amino acid 139-172 increased Complex I activity and ROS generation similar to expression of the entire P protein, whereas peptides that did not contain this region did not increase Complex I activity or induce ROS generation. These results indicate that a region of the RABV P interacts with Complex I in mitochondria causing mitochondrial dysfunction, increased generation of ROS, and oxidative stress.
Collapse
|
39
|
Zou J, Li W, Misra A, Yue F, Song K, Chen Q, Guo G, Yi J, Kimata JT, Liu L. The viral restriction factor tetherin prevents leucine-rich pentatricopeptide repeat-containing protein (LRPPRC) from association with beclin 1 and B-cell CLL/lymphoma 2 (Bcl-2) and enhances autophagy and mitophagy. J Biol Chem 2015; 290:7269-79. [PMID: 25631043 DOI: 10.1074/jbc.m114.627679] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Tetherin has been characterized as a key factor that restricts viral particles such as HIV and hepatitis C virus on plasma membranes, acts as a ligand of the immunoglobulin-like transcript 7 (ILT7) receptor in tumor cells, and suppresses antiviral innate immune responses mediated by human plasmacytoid dendritic cells. However, the normal cellular function of Tetherin without viral infection is unknown. Here we show that Tetherin not only serves as a substrate of autophagy but itself regulates the initiation of autophagy. Tetherin interacts with the autophagy/mitophagy suppressor LRPPRC and prevents LRPPRC from forming a ternary complex with Beclin 1 and Bcl-2 so that Beclin 1 is released to bind with PI3KCIII (class III PI3K) to activate the initiation of autophagy. Suppression of Tetherin leads to impairment of autophagy, whereas overexpression of Tetherin causes activation of autophagy. Under mitophagic stress, Tetherin is concentrated on mitochondria engulfed in autophagosomes. Tetherin plays a general role in the degradation of autophagosomes containing not only the symbiotic mitochondria but also, possibly, the infected virus. Therefore, Tetherin may enhance autophagy and mitophagy to suppress tumorigenesis, enhance innate immune responses, or prevent T cell apoptosis or pyroptosis.
Collapse
Affiliation(s)
- Jing Zou
- From the Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, 77030, the Jiangxi Research Institute of Ophthalmology and Visual Sciences, The Affiliated Eye Hospital of Nanchang University, Nanchang, 330006, China
| | - Wenjiao Li
- From the Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, 77030
| | - Anisha Misra
- the Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas 77030, and
| | - Fei Yue
- From the Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, 77030
| | - Kun Song
- From the Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, 77030
| | - Qi Chen
- From the Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, 77030
| | - Guanghua Guo
- the Jiangxi Research Institute of Ophthalmology and Visual Sciences, The Affiliated Eye Hospital of Nanchang University, Nanchang, 330006, China
| | - Jinglin Yi
- the Jiangxi Research Institute of Ophthalmology and Visual Sciences, The Affiliated Eye Hospital of Nanchang University, Nanchang, 330006, China
| | - Jason T Kimata
- the Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas 77030, and
| | - Leyuan Liu
- From the Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, 77030, the Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M Health Science Center, College Station, Texas 77843
| |
Collapse
|
40
|
Corporeau C, Tamayo D, Pernet F, Quéré C, Madec S. Proteomic signatures of the oyster metabolic response to herpesvirus OsHV-1 μVar infection. J Proteomics 2014; 109:176-87. [DOI: 10.1016/j.jprot.2014.06.030] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 06/18/2014] [Accepted: 06/28/2014] [Indexed: 10/25/2022]
|
41
|
Price AM, Luftig MA. Dynamic Epstein-Barr virus gene expression on the path to B-cell transformation. Adv Virus Res 2014; 88:279-313. [PMID: 24373315 DOI: 10.1016/b978-0-12-800098-4.00006-4] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Epstein-Barr virus (EBV) is an oncogenic human herpesvirus in the γ-herpesvirinae subfamily that contains a 170-180kb double-stranded DNA genome. In vivo, EBV commonly infects B and epithelial cells and persists for the life of the host in a latent state in the memory B-cell compartment of the peripheral blood. EBV can be reactivated from its latent state, leading to increased expression of lytic genes that primarily encode for enzymes necessary to replicate the viral genome and structural components of the virion. Lytic cycle proteins also aid in immune evasion, inhibition of apoptosis, and the modulation of other host responses to infection. In vitro, EBV has the potential to infect primary human B cells and induce cellular proliferation to yield effectively immortalized lymphoblastoid cell lines, or LCLs. EBV immortalization of B cells in vitro serves as a model system for studying EBV-mediated lymphomagenesis. While much is known about the steady-state viral gene expression within EBV-immortalized LCLs and other EBV-positive cell lines, relatively little is known about the early events after primary B-cell infection. It was previously thought that upon latent infection, EBV only expressed the well-characterized latency-associated transcripts found in LCLs. However, recent work has characterized the early, but transient, expression of lytic genes necessary for efficient transformation and delayed responses in the known latency genes. This chapter summarizes these recent findings that show how dynamic and controlled expression of multiple EBV genes can control the activation of B cells, entry into the cell cycle, the inhibition of apoptosis, and innate and adaptive immune responses.
Collapse
Affiliation(s)
- Alexander M Price
- Department of Molecular Genetics and Microbiology, Center for Virology, Duke University Medical Center, Durham, North Carolina, 27710 USA
| | - Micah A Luftig
- Department of Molecular Genetics and Microbiology, Center for Virology, Duke University Medical Center, Durham, North Carolina, 27710 USA.
| |
Collapse
|
42
|
Lacek K, Bauer B, Bieńkowska-Szewczyk K, Rziha HJ. Orf virus (ORFV) ANK-1 protein mitochondrial localization is mediated by ankyrin repeat motifs. Virus Genes 2014; 49:68-79. [PMID: 24743940 DOI: 10.1007/s11262-014-1069-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 04/03/2014] [Indexed: 10/25/2022]
Abstract
Orf virus (ORFV) strain D1701-V, a Parapoxvirus belonging to the family Poxviridae, became attractive as a novel virus vector system that we successfully used for the generation of recombinant vaccines. Therefore, the identification of viral genes involved in host tropisms or immune modulation is of great interest, as for instance the ORFV-encoded ankyrin-repeat (AR) containing proteins. The present study shows for the first time that the ANK-1 designated gene product of ORFV126 is targeted to mitochondria of ORFV-infected and in ANK-1 transiently expressing cells. Taking advantage of ANK-1 EGFP fusion proteins and confocal fluorescence microscopy mutational and deletion analyses indicated the importance of AR8 and AR9, which may contain a novel class of mitochondria-targeting sequence (MTS) in the central to C-terminal part of this AR-containing protein. The fluorescent findings were corroborated by cell fractionation and Western blotting experiments. The presented results open the avenue for more detailed investigations on cellular binding partners and the function of ANK-1 in viral replication or virulence.
Collapse
Affiliation(s)
- Krzysztof Lacek
- Laboratory of Virus Molecular Biology, University of Gdańsk, 80-822, Gdańsk, Poland
| | | | | | | |
Collapse
|
43
|
Mitochondrial dysfunction in rabies virus infection of neurons. J Neurovirol 2013; 19:537-49. [PMID: 24277436 DOI: 10.1007/s13365-013-0214-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Revised: 09/20/2013] [Accepted: 10/01/2013] [Indexed: 12/21/2022]
Abstract
Infection with the challenge virus standard-11 (CVS) strain of fixed rabies virus induces neuronal process degeneration in adult mice after hindlimb footpad inoculation. CVS-induced axonal swellings of primary rodent dorsal root ganglion neurons are associated with 4-hydroxy-2-nonenal protein adduct staining, indicating a critical role of oxidative stress. Mitochondrial dysfunction is the major cause of oxidative stress. We hypothesized that CVS infection induces mitochondrial dysfunction leading to oxidative stress. We investigated the effects of CVS infection on several mitochondrial parameters in different cell types. CVS infection significantly increased maximal uncoupled respiration and complex IV respiration and complex I and complex IV activities, but did not affect complex II-III or citrate synthase activities. Increases in complex I activity, but not complex IV activity, correlated with susceptibility of the cells to CVS infection. CVS infection maintained coupled respiration and rate of proton leak, indicating a tight mitochondrial coupling. Possibly as a result of enhanced complex activity and efficient coupling, a high mitochondrial membrane potential was generated. CVS infection reduced the intracellular ATP level and altered the cellular redox state as indicated by a high NADH/NAD+ ratio. The basal production of reactive oxygen species (ROS) was not affected in CVS-infected neurons. However, a higher rate of ROS generation occurred in CVS-infected neurons in the presence of mitochondrial substrates and inhibitors. We conclude that CVS infection induces mitochondrial dysfunction leading to ROS overgeneration and oxidative stress.
Collapse
|
44
|
Tovilovic G, Ristic B, Milenkovic M, Stanojevic M, Trajkovic V. The Role and Therapeutic Potential of Autophagy Modulation in Controlling Virus-Induced Cell Death. Med Res Rev 2013; 34:744-67. [DOI: 10.1002/med.21303] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Gordana Tovilovic
- Institute for Biological Research; University of Belgrade; Despot Stefan Boulevard 142 11000 Belgrade Serbia
| | - Biljana Ristic
- Institute of Microbiology and Immunology; School of Medicine; University of Belgrade; Dr. Subotica 1 11000 Belgrade Serbia
| | - Marina Milenkovic
- Institute of Microbiology and Immunology; School of Medicine; University of Belgrade; Dr. Subotica 1 11000 Belgrade Serbia
| | - Maja Stanojevic
- Institute of Microbiology and Immunology; School of Medicine; University of Belgrade; Dr. Subotica 1 11000 Belgrade Serbia
| | - Vladimir Trajkovic
- Institute of Microbiology and Immunology; School of Medicine; University of Belgrade; Dr. Subotica 1 11000 Belgrade Serbia
| |
Collapse
|
45
|
Yim SA, Lim YS, Kim JW, Hwang SB. Nonstructural 5A protein of hepatitis C virus interacts with pyruvate carboxylase and modulates viral propagation. PLoS One 2013; 8:e68170. [PMID: 23861867 PMCID: PMC3701667 DOI: 10.1371/journal.pone.0068170] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Accepted: 05/26/2013] [Indexed: 01/15/2023] Open
Abstract
Hepatitis C virus (HCV) is highly dependent on cellular factors for its own propagation. By employing tandem affinity purification method, we identified pyruvate carboxylase (PC) as a cellular partner for NS5A protein. NS5A interacted with PC through the N-terminal region of NS5A and the biotin carboxylase domain of PC. PC expression was decreased in cells expressing NS5A and HCV-infected cells. Promoter activity of PC was also decreased by NS5A protein. However, FAS expression was increased in cells expressing NS5A and cell culture grown HCV (HCVcc)-infected cells. Silencing of PC promoted fatty acid synthase (FAS) expression level. These data suggest HCV may modulate PC via NS5A protein for its own propagation.
Collapse
Affiliation(s)
- Seung-Ae Yim
- National Research Laboratory of Hepatitis C Virus, Ilsong Institute of Life Science, Hallym University, Anyang, Korea
| | - Yun-Sook Lim
- National Research Laboratory of Hepatitis C Virus, Ilsong Institute of Life Science, Hallym University, Anyang, Korea
| | - Jong-Wook Kim
- National Research Laboratory of Hepatitis C Virus, Ilsong Institute of Life Science, Hallym University, Anyang, Korea
| | - Soon B. Hwang
- National Research Laboratory of Hepatitis C Virus, Ilsong Institute of Life Science, Hallym University, Anyang, Korea
- * E-mail:
| |
Collapse
|
46
|
Kaiser WJ, Upton JW, Mocarski ES. Viral modulation of programmed necrosis. Curr Opin Virol 2013; 3:296-306. [PMID: 23773332 DOI: 10.1016/j.coviro.2013.05.019] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Revised: 05/21/2013] [Accepted: 05/22/2013] [Indexed: 01/16/2023]
Abstract
Apoptosis and programmed necrosis balance each other as alternate first line host defense pathways against which viruses have evolved countermeasures. Intrinsic apoptosis, the critical programmed cell death pathway that removes excess cells during embryonic development and tissue homeostasis, follows a caspase cascade triggered at mitochondria and modulated by virus-encoded anti-apoptotic B cell leukemia (BCL)2-like suppressors. Extrinsic apoptosis controlled by caspase 8 arose during evolution to trigger executioner caspases directly, circumventing viral suppressors of intrinsic (mitochondrial) apoptosis and providing the selective pressure for viruses to acquire caspase 8 suppressors. Programmed necrosis likely evolved most recently as a 'trap door' adaptation to extrinsic apoptosis. Receptor interacting protein (RIP)3 kinase (also called RIPK3) becomes active when either caspase 8 activity or polyubiquitylation of RIP1 is compromised. This evolutionary dialog implicates caspase 8 as a 'supersensor' alternatively activating and suppressing cell death pathways.
Collapse
Affiliation(s)
- William J Kaiser
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | |
Collapse
|
47
|
Hepatitis C virus-induced mitochondrial dysfunctions. Viruses 2013; 5:954-80. [PMID: 23518579 PMCID: PMC3705306 DOI: 10.3390/v5030954] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Revised: 03/15/2013] [Accepted: 03/20/2013] [Indexed: 12/15/2022] Open
Abstract
Chronic hepatitis C is characterized by metabolic disorders and a microenvironment in the liver dominated by oxidative stress, inflammation and regeneration processes that lead in the long term to hepatocellular carcinoma. Many lines of evidence suggest that mitochondrial dysfunctions, including modification of metabolic fluxes, generation and elimination of oxidative stress, Ca2+ signaling and apoptosis, play a central role in these processes. However, how these dysfunctions are induced by the virus and whether they play a role in disease progression and neoplastic transformation remains to be determined. Most in vitro studies performed so far have shown that several of the hepatitis C virus (HCV) proteins localize to mitochondria, but the consequences of these interactions on mitochondrial functions remain contradictory, probably due to the use of artificial expression and replication systems. In vivo studies are hampered by the fact that innate and adaptive immune responses will overlay mitochondrial dysfunctions induced directly in the hepatocyte by HCV. Thus, the molecular aspects underlying HCV-induced mitochondrial dysfunctions and their roles in viral replication and the associated pathology need yet to be confirmed in the context of productively replicating virus and physiologically relevant in vitro and in vivo model systems.
Collapse
|
48
|
Abstract
BCL-2 family proteins are the regulators of apoptosis, but also have other functions. This family of interacting partners includes inhibitors and inducers of cell death. Together they regulate and mediate the process by which mitochondria contribute to cell death known as the intrinsic apoptosis pathway. This pathway is required for normal embryonic development and for preventing cancer. However, before apoptosis is induced, BCL-2 proteins have critical roles in normal cell physiology related to neuronal activity, autophagy, calcium handling, mitochondrial dynamics and energetics, and other processes of normal healthy cells. The relative importance of these physiological functions compared to their apoptosis functions in overall organismal physiology is difficult to decipher. Apoptotic and noncanonical functions of these proteins may be intertwined to link cell growth to cell death. Disentanglement of these functions may require delineation of biochemical activities inherent to the characteristic three-dimensional shape shared by distantly related viral and cellular BCL-2 family members.
Collapse
|
49
|
Shoshan-Barmatz V, Mizrachi D. VDAC1: from structure to cancer therapy. Front Oncol 2012; 2:164. [PMID: 23233904 PMCID: PMC3516065 DOI: 10.3389/fonc.2012.00164] [Citation(s) in RCA: 148] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2012] [Accepted: 10/24/2012] [Indexed: 12/14/2022] Open
Abstract
Here, we review current evidence pointing to the function of VDAC1 in cell life and death, and highlight these functions in relation to cancer. Found at the outer mitochondrial membrane, VDAC1 assumes a crucial position in the cell, controlling the metabolic cross-talk between mitochondria and the rest of the cell. Moreover, its location at the boundary between the mitochondria and the cytosol enables VDAC1 to interact with proteins that mediate and regulate the integration of mitochondrial functions with other cellular activities. As a metabolite transporter, VDAC1 contributes to the metabolic phenotype of cancer cells. This is reflected by VDAC1 over-expression in many cancer types, and by inhibition of tumor development upon silencing VDAC1 expression. Along with regulating cellular energy production and metabolism, VDAC1 is also a key protein in mitochondria-mediated apoptosis, participating in the release of apoptotic proteins and interacting with anti-apoptotic proteins. The involvement of VDAC1 in the release of apoptotic proteins located in the inter-membranal space is discussed, as is VDAC1 oligomerization as an important step in apoptosis induction. VDAC also serves as an anchor point for mitochondria-interacting proteins, some of which are also highly expressed in many cancers, such as hexokinase (HK), Bcl2, and Bcl-xL. By binding to VDAC, HK provides both metabolic benefit and apoptosis-suppressive capacity that offers the cell a proliferative advantage and increases its resistance to chemotherapy. VDAC1-based peptides that bind specifically to HK, Bcl2, or Bcl-xL abolished the cell’s abilities to bypass the apoptotic pathway. Moreover, these peptides promote cell death in a panel of genetically characterized cell lines derived from different human cancers. These and other functions point to VDAC1 as a rational target for the development of a new generation of therapeutics.
Collapse
Affiliation(s)
- Varda Shoshan-Barmatz
- Department of Life Sciences, Ben-Gurion University of the Negev Beer-Sheva, Israel ; The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev Beer-Sheva, Israel
| | | |
Collapse
|
50
|
EBV up-regulates cytochrome c through VDAC1 regulations and decreases the release of cytoplasmic Ca2+ in the NPC cell line. Cell Biol Int 2012; 36:733-8. [PMID: 22497278 DOI: 10.1042/cbi20110368] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
EBV (Epstein-Barr virus) is considered to be a major factor that causes NPC (nasopharyngeal carcinoma), which is one of the sneakiest cancers frequently occurring in Southeast Asia and Southern China. Apoptosis and pro-apoptotic signals have been studied for decades; however, few have extended the prevailing view of EBV to its impact on NPC in perspective of apoptosis. One of the important proteins named VDAC1 (voltage-dependent anion protein 1) on the mitochondrial outer membrane controls the pro-apoptotic signals in mammalian cells. The impact of EBV infection on VDAC1 and related apoptotic signals remains unclear. In order to study the VDAC1's role in EBV-infected NPC cells, we employ siRNA (small interfering RNA) inhibition to analyse the release of Ca2+ and Cyto c (cytochrome c) signals in the cytoplasm, as they are important pro-apoptotic signals. The results show a decrease of Ca2+ release and up-regulation of Cyto c with EBV infection. After siRNA transfection, the dysregulation of Cyto c is neutralized, which is evidence that the level of Cyto c release in virus-infected NPC cells is the as same as that of non-infected NPC cells. This result indicates that EBV infection changes the cytoplasmic level of Cyto c through regulating VDAC1. In summary, this study reports that EBV changes the release of Ca2+ and Cyto c in the cytoplasm of NPC cells, and that Cyto c changes are mediated by VDAC1 regulation.
Collapse
|