1
|
Amiri M, Kaviari MA, Rostaminasab G, Barimani A, Rezakhani L. A novel cell-free therapy using exosomes in the inner ear regeneration. Tissue Cell 2024; 88:102373. [PMID: 38640600 DOI: 10.1016/j.tice.2024.102373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 03/01/2024] [Accepted: 04/03/2024] [Indexed: 04/21/2024]
Abstract
Cellular and molecular alterations associated with hearing loss are now better understood with advances in molecular biology. These changes indicate the participation of distinct damage and stress pathways that are unlikely to be fully addressed by conventional pharmaceutical treatment. Sensorineural hearing loss is a common and debilitating condition for which comprehensive pharmacologic intervention is not available. The complex and diverse molecular pathology that underlies hearing loss currently limits our ability to intervene with small molecules. The present review focuses on the potential for the use of extracellular vesicles in otology. It examines a variety of inner ear diseases and hearing loss that may be treatable using exosomes (EXOs). The role of EXOs as carriers for the treatment of diseases related to the inner ear as well as EXOs as biomarkers for the recognition of diseases related to the ear is discussed.
Collapse
Affiliation(s)
- Masoumeh Amiri
- Faculty of Medicine, Kermanshah University of Medical Science, Kermanshah, Iran
| | - Mohammad Amin Kaviari
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran; Universal Scientific Education and Research Network (USERN) Office, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Gelavizh Rostaminasab
- Clinical Research Development Center, Imam Khomeini and Mohammad Kermanshahi and Farabi Hospitals, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Amir Barimani
- Clinical Research Development Center, Imam Khomeini and Mohammad Kermanshahi and Farabi Hospitals, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Leila Rezakhani
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; Department of Tissue Engineering, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
2
|
Gunasegaran B, Ashley CL, Marsh-Wakefield F, Guillemin GJ, Heng B. Viruses in glioblastoma: an update on evidence and clinical trials. BJC REPORTS 2024; 2:33. [PMID: 39516641 PMCID: PMC11524015 DOI: 10.1038/s44276-024-00051-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/13/2024] [Accepted: 02/22/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Glioblastoma (GB) is a lethal and aggressive brain tumour. While molecular characteristics of GB is studied extensively, the aetiology of GB remains uncertain. The interest in exploring viruses as a potential contributor to the development of GB stems from the notion that viruses are known to play a key role in pathogenesis of other human cancers such as cervical cancer. Nevertheless, the role of viruses in GB remains controversial. METHODS This review delves into the current body of knowledge surrounding the presence of viruses in GB as well as provide updates on clinical trials examining the potential inclusion of antiviral therapies as part of the standard of care protocol. CONCLUSIONS The review summarises current evidences and important gaps in our knowledge related to the presence of viruses in GB.
Collapse
Affiliation(s)
- Bavani Gunasegaran
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Sydney, NSW, Australia
| | - Caroline L Ashley
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, Australia
- School of Medical Sciences Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Felix Marsh-Wakefield
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, Australia
- School of Medical Sciences Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
- Centenary Institute, Camperdown, NSW, Australia
| | | | - Benjamin Heng
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Sydney, NSW, Australia.
| |
Collapse
|
3
|
Heinz JL, Swagemakers SMA, von Hofsten J, Helleberg M, Thomsen MM, De Keukeleere K, de Boer JH, Ilginis T, Verjans GMGM, van Hagen PM, van der Spek PJ, Mogensen TH. Whole exome sequencing of patients with varicella-zoster virus and herpes simplex virus induced acute retinal necrosis reveals rare disease-associated genetic variants. Front Mol Neurosci 2023; 16:1253040. [PMID: 38025266 PMCID: PMC10630912 DOI: 10.3389/fnmol.2023.1253040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 10/09/2023] [Indexed: 12/01/2023] Open
Abstract
Purpose Herpes simplex virus (HSV) and varicella-zoster virus (VZV) are neurotropic human alphaherpesviruses endemic worldwide. Upon primary infection, both viruses establish lifelong latency in neurons and reactivate intermittently to cause a variety of mild to severe diseases. Acute retinal necrosis (ARN) is a rare, sight-threatening eye disease induced by ocular VZV or HSV infection. The virus and host factors involved in ARN pathogenesis remain incompletely described. We hypothesize an underlying genetic defect in at least part of ARN cases. Methods We collected blood from 17 patients with HSV-or VZV-induced ARN, isolated DNA and performed Whole Exome Sequencing by Illumina followed by analysis in Varseq with criteria of CADD score > 15 and frequency in GnomAD < 0.1% combined with biological filters. Gene modifications relative to healthy control genomes were filtered according to high quality and read-depth, low frequency, high deleteriousness predictions and biological relevance. Results We identified a total of 50 potentially disease-causing genetic variants, including missense, frameshift and splice site variants and on in-frame deletion in 16 of the 17 patients. The vast majority of these genes are involved in innate immunity, followed by adaptive immunity, autophagy, and apoptosis; in several instances variants within a given gene or pathway was identified in several patients. Discussion We propose that the identified variants may contribute to insufficient viral control and increased necrosis ocular disease presentation in the patients and serve as a knowledge base and starting point for the development of improved diagnostic, prophylactic, and therapeutic applications.
Collapse
Affiliation(s)
- Johanna L. Heinz
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Sigrid M. A. Swagemakers
- Department of Pathology and Clinical Bioinformatics, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Joanna von Hofsten
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Ophthalmology, Halland Hospital Halmstad, Halmstad, Sweden
| | - Marie Helleberg
- Department of Infectious Diseases, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Center of Excellence for Health, Immunity and Infections, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Michelle M. Thomsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Kerstin De Keukeleere
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Joke H. de Boer
- Department of Ophthalmology, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Tomas Ilginis
- Department of Ophthalmology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Georges M. G. M. Verjans
- HerpeslabNL, Department of Viroscience, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Peter M. van Hagen
- Department of Internal Medicine and Immunology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Peter J. van der Spek
- Department of Pathology and Clinical Bioinformatics, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Trine H. Mogensen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
4
|
Luo B, Ding L. Ion channels and ions as therapeutic targets and strategies for herpes simplex virus infection. Future Virol 2022. [DOI: 10.2217/fvl-2022-0052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Herpes simplex virus (HSV) is a highly contagious virus that cannot be completely cured currently. Existing treatment methods are mainly nucleoside antiviral drugs, and the emergence of drug-resistant strains severely limits their use. There is an urgent need to discover antiviral drugs that act on new targets. Ion channels, a class of cellular proteins with a wide range of functions, have become critical host factors for a wide variety of viral infections. Ion channel blockers have been shown to have antiviral activity. In this study, we discuss the role of ion channels and ions in the HSV life cycle, and the potential of targeting ion channels as a novel, pharmacologically safe and wide-range antiviral treatment option.
Collapse
Affiliation(s)
- Binhua Luo
- Department of Pharmaceutics, School of Pharmacy, Hubei University of Science & Technology, Xianning, 437100, China
- Hubei Engineering Research Center of Traditional Chinese Medicine of South Hubei Province, Xianning, 437100, China
| | - Liqiong Ding
- Department of Pharmaceutics, School of Pharmacy, Hubei University of Science & Technology, Xianning, 437100, China
- Hubei Engineering Research Center of Traditional Chinese Medicine of South Hubei Province, Xianning, 437100, China
| |
Collapse
|
5
|
A Single Herpes Simplex Virus 1 Genome Reactivates from Individual Cells. Microbiol Spectr 2022; 10:e0114422. [PMID: 35862979 PMCID: PMC9431706 DOI: 10.1128/spectrum.01144-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Latent infection is a characteristic feature of herpesviruses’ life cycle. Herpes simplex virus 1 is a common human pathogen that establishes lifelong latency in peripheral neurons. Symptomatic or asymptomatic periodic reactivations from the latent state allow the virus to replicate and spread among individuals. The latent viral genomes are found as several quiescent episomes inside the infected nuclei; however, it is not clear if and how many latent genomes are able to reactivate together. To address this question, we developed a quiescent infection assay, which provides a quantitative analysis of the number of genomes reactivating per cell, in cultured immortalized fibroblasts. We found that, almost always, only one viral genome reactivates per cell. We showed that different timing of entry to quiescence did not result in a significant change in the probability of reactivating. Reactivation from this quiescent state allowed only limited intergenomic recombination between two viral strains compared to lytic infection. Following coinfection with a mutant that is unable to reactivate, only coreactivation with a reactivation-proficient recombinant can provide the opportunity for the mutant to reactivate. We speculate that each individual quiescent viral genome has a low and stochastic chance to reactivate in each cell, an assumption that can explain the limited number of genomes reactivating per cell. IMPORTANCE Herpesviruses are highly prevalent and cause significant morbidity in the human and animal populations. Most individuals who are infected with herpes simplex virus (HSV-1), a common human pathogen, will become lifelong carriers of the virus, as HSV-1 establishes latent (quiescent) infections in the host cells. Reactivation from the latent state leads to many of the viral symptoms and to the spread of the virus among individuals. While many triggers for reactivation were identified, how many genomes reactivate from an individual cell and how are these genomes selected remain understudied. Here, we identify that, in most cases, only one genome per cell reactivates. Mutated HSV-1 genomes require coinfection with another strain to allow coreactivation. Our findings suggest that the decision to reactivate is determined for each quiescent genome separately and support the notion that reactivation preferences occur at the single-genome level.
Collapse
|
6
|
Jacob S, Kapadia R, Soule T, Luo H, Schellenberg KL, Douville RN, Pfeffer G. Neuromuscular Complications of SARS-CoV-2 and Other Viral Infections. Front Neurol 2022; 13:914411. [PMID: 35812094 PMCID: PMC9263266 DOI: 10.3389/fneur.2022.914411] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 05/25/2022] [Indexed: 12/15/2022] Open
Abstract
In this article we review complications to the peripheral nervous system that occur as a consequence of viral infections, with a special focus on complications of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). We discuss neuromuscular complications in three broad categories; the direct consequences of viral infection, autoimmune neuromuscular disorders provoked by viral infections, and chronic neurodegenerative conditions which have been associated with viral infections. We also include discussion of neuromuscular disorders that are treated by immunomodulatory therapies, and how this affects patient susceptibility in the current context of the coronavirus disease 2019 (COVID-19) pandemic. COVID-19 is associated with direct consequences to the peripheral nervous system via presumed direct viral injury (dysgeusia/anosmia, myalgias/rhabdomyolysis, and potentially mononeuritis multiplex) and autoimmunity (Guillain Barré syndrome and variants). It has important implications for people receiving immunomodulatory therapies who may be at greater risk of severe outcomes from COVID-19. Thus far, chronic post-COVID syndromes (a.k.a: long COVID) also include possible involvement of the neuromuscular system. Whether we may observe neuromuscular degenerative conditions in the longer term will be an important question to monitor in future studies.
Collapse
Affiliation(s)
- Sarah Jacob
- Hotchkiss Brain Institute, Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Ronak Kapadia
- Hotchkiss Brain Institute, Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Tyler Soule
- Hotchkiss Brain Institute, Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Honglin Luo
- Centre for Heart and Lung Innovation, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Kerri L. Schellenberg
- Division of Neurology, Department of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Renée N. Douville
- Division of Neurodegenerative Disorders, Department of Biology, Albrechtsen St. Boniface Research Centre, University of Winnipeg, Winnipeg, MB, Canada
| | - Gerald Pfeffer
- Hotchkiss Brain Institute, Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Medical Genetics, Alberta Child Health Research Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
7
|
Mehboob R, Kurdi M, Bamaga A, Aldardeir N, Nasief H, Moshref LH, Alsinani T, Rayes AO, Jabbad RH. Substance P/ Neurokinin-1 Receptor, Trigeminal Ganglion, Latency, and Coronavirus Infection-Is There Any Link? Front Med (Lausanne) 2021; 8:727593. [PMID: 34869423 PMCID: PMC8637107 DOI: 10.3389/fmed.2021.727593] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 10/01/2021] [Indexed: 11/13/2022] Open
Abstract
Novel Severe Acute Respiratory Syndrome-Corona Virus-2 infection (SARS-CoV-2) is an acute respiratory and infectious disease. This perspective aims to provide a basic understanding of the inflammation caused by SARS-CoV-2 and its relation to the trigeminal ganglion (TG). The virus enters through the mucous membranes of the orofacial region and reaches the TG, where it resides and takes control of its peptides including Substance P (SP). SP is the main neuropeptide, neuromodulator, and neuro-hormone of TG, associated with nociception and inflammation under noxious stimulus. SP release is triggered and, consequently, affects the immune cells and blood vessels to release the mediators for inflammation. Hence, cytokine storm is initiated and causes respiratory distress, bronchoconstriction, and death in complicated cases. Neurokinin-1 Receptor (NK-1R) is the receptor for SP and its antagonists, along with glucocorticoids, may be used to alleviate the symptoms and treat this infection by blocking this nociceptive pathway. SP seems to be the main culprit involved in the triggering of inflammatory pathways in SARS-CoV-2 infection. It may have a direct association with cardio-respiratory rhythm, sleep-wake cycle, nociception, and ventilatory responses and regulates many important physiological and pathological functions. Its over-secretion should be blocked by NK-1R antagonist. However, experimental work leading to clinical trials are mandatory for further confirmation. Here, it is further proposed that there is a possibility of latency in SARS-CoV-2 virus infection if it is acting through TG, which is the main site for other viruses that become latent.
Collapse
Affiliation(s)
- Riffat Mehboob
- Faculty of Allied Health Sciences, The University of Lahore, Lahore, Pakistan.,Lahore Medical Research Center, LLP, Lahore, Pakistan
| | - Maher Kurdi
- Department of Pathology, Faculty of Medicine, King Abdulaziz University, Rabigh, Saudi Arabia
| | - Ahmed Bamaga
- Neurology Division, Department of Pediatrics, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Njoud Aldardeir
- Faculty of Education, University of Ottawa, Ottawa, ON, Canada
| | - Hisham Nasief
- Department of Obstetrics and Gynecology, Faculty of Medicine, Jeddah, Saudi Arabia
| | - Leena H Moshref
- Department of Surgery, Doctor Soliman Fakeeh Hospital, Jeddah, Saudi Arabia
| | - Taghreed Alsinani
- Division of Neurosurgery, King Fahad General Hospital, Jeddah, Saudi Arabia
| | - Almotasimbellah O Rayes
- Department of Obstetrics and Gynecology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Reem H Jabbad
- Department of Medicine, King Fahad Armed Forces Hospital, Jeddah, Saudi Arabia
| |
Collapse
|
8
|
Napoletani G, Protto V, Marcocci ME, Nencioni L, Palamara AT, De Chiara G. Recurrent Herpes Simplex Virus Type 1 (HSV-1) Infection Modulates Neuronal Aging Marks in In Vitro and In Vivo Models. Int J Mol Sci 2021; 22:6279. [PMID: 34208020 PMCID: PMC8230621 DOI: 10.3390/ijms22126279] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/20/2021] [Accepted: 06/08/2021] [Indexed: 01/21/2023] Open
Abstract
Herpes simplex virus 1 (HSV-1) is a widespread neurotropic virus establishing a life-long latent infection in neurons with periodic reactivations. Recent studies linked HSV-1 to neurodegenerative processes related to age-related disorders such as Alzheimer's disease. Here, we explored whether recurrent HSV-1 infection might accelerate aging in neurons, focusing on peculiar marks of aged cells, such as the increase in histone H4 lysine (K) 16 acetylation (ac) (H4K16ac); the decrease of H3K56ac, and the modified expression of Sin3/HDAC1 and HIRA proteins. By exploiting both in vitro and in vivo models of recurrent HSV-1 infection, we found a significant increase in H4K16ac, Sin3, and HDAC1 levels, suggesting that the neuronal response to virus latency and reactivation includes the upregulation of these aging markers. On the contrary, we found a significant decrease in H3K56ac that was specifically linked to viral reactivation and apparently not related to aging-related markers. A complex modulation of HIRA expression and localization was found in the brain from HSV-1 infected mice suggesting a specific role of this protein in viral latency and reactivation. Overall, our results pointed out novel molecular mechanisms through which recurrent HSV-1 infection may affect neuronal aging, likely contributing to neurodegeneration.
Collapse
Affiliation(s)
- Giorgia Napoletani
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia–Fondazione Cenci Bolognetti, 00185 Rome, Italy; (G.N.); (V.P.); (M.E.M.); (L.N.); (A.T.P.)
| | - Virginia Protto
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia–Fondazione Cenci Bolognetti, 00185 Rome, Italy; (G.N.); (V.P.); (M.E.M.); (L.N.); (A.T.P.)
| | - Maria Elena Marcocci
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia–Fondazione Cenci Bolognetti, 00185 Rome, Italy; (G.N.); (V.P.); (M.E.M.); (L.N.); (A.T.P.)
| | - Lucia Nencioni
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia–Fondazione Cenci Bolognetti, 00185 Rome, Italy; (G.N.); (V.P.); (M.E.M.); (L.N.); (A.T.P.)
| | - Anna Teresa Palamara
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia–Fondazione Cenci Bolognetti, 00185 Rome, Italy; (G.N.); (V.P.); (M.E.M.); (L.N.); (A.T.P.)
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Giovanna De Chiara
- Institute of Translational Pharmacology, National Research Council (CNR), 00133 Rome, Italy
| |
Collapse
|
9
|
Duarte LF, Reyes A, Farías MA, Riedel CA, Bueno SM, Kalergis AM, González PA. Crosstalk Between Epithelial Cells, Neurons and Immune Mediators in HSV-1 Skin Infection. Front Immunol 2021; 12:662234. [PMID: 34012447 PMCID: PMC8126613 DOI: 10.3389/fimmu.2021.662234] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 04/06/2021] [Indexed: 12/12/2022] Open
Abstract
Herpes simplex virus type 1 (HSV-1) infection is highly prevalent in humans, with approximately two-thirds of the world population living with this virus. However, only a fraction of those carrying HSV-1, which elicits lifelong infections, are symptomatic. HSV-1 mainly causes lesions in the skin and mucosae but reaches the termini of sensory neurons innervating these tissues and travels in a retrograde manner to the neuron cell body where it establishes persistent infection and remains in a latent state until reactivated by different stimuli. When productive reactivations occur, the virus travels back along axons to the primary infection site, where new rounds of replication are initiated in the skin, in recurrent or secondary infections. During this process, new neuron infections occur. Noteworthy, the mechanisms underlying viral reactivations and the exit of latency are somewhat poorly understood and may be regulated by a crosstalk between the infected neurons and components of the immune system. Here, we review and discuss the immune responses that occur at the skin during primary and recurrent infections by HSV-1, as well as at the interphase of latently-infected neurons. Moreover, we discuss the implications of neuronal signals over the priming and migration of immune cells in the context of HSV-1 infection.
Collapse
Affiliation(s)
- Luisa F Duarte
- Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Antonia Reyes
- Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Mónica A Farías
- Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Claudia A Riedel
- Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Susan M Bueno
- Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Endocrinología, Facultad de Medicina, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A González
- Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
10
|
Pastor F, Shkreta L, Chabot B, Durantel D, Salvetti A. Interplay Between CMGC Kinases Targeting SR Proteins and Viral Replication: Splicing and Beyond. Front Microbiol 2021; 12:658721. [PMID: 33854493 PMCID: PMC8040976 DOI: 10.3389/fmicb.2021.658721] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 03/04/2021] [Indexed: 12/27/2022] Open
Abstract
Protein phosphorylation constitutes a major post-translational modification that critically regulates the half-life, intra-cellular distribution, and activity of proteins. Among the large number of kinases that compose the human kinome tree, those targeting RNA-binding proteins, in particular serine/arginine-rich (SR) proteins, play a major role in the regulation of gene expression by controlling constitutive and alternative splicing. In humans, these kinases belong to the CMGC [Cyclin-dependent kinases (CDKs), Mitogen-activated protein kinases (MAPKs), Glycogen synthase kinases (GSKs), and Cdc2-like kinases (CLKs)] group and several studies indicate that they also control viral replication via direct or indirect mechanisms. The aim of this review is to describe known and emerging activities of CMGC kinases that share the common property to phosphorylate SR proteins, as well as their interplay with different families of viruses, in order to advance toward a comprehensive knowledge of their pro- or anti-viral phenotype and better assess possible translational opportunities.
Collapse
Affiliation(s)
- Florentin Pastor
- International Center for Infectiology Research (CIRI), INSERM U1111, CNRS UMR5308, Université de Lyon (UCBL1), Lyon, France
| | - Lulzim Shkreta
- Department of Microbiology and Infectious Diseases, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Benoit Chabot
- Department of Microbiology and Infectious Diseases, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - David Durantel
- International Center for Infectiology Research (CIRI), INSERM U1111, CNRS UMR5308, Université de Lyon (UCBL1), Lyon, France
| | - Anna Salvetti
- International Center for Infectiology Research (CIRI), INSERM U1111, CNRS UMR5308, Université de Lyon (UCBL1), Lyon, France
| |
Collapse
|
11
|
Septins in Infections: Focus on Viruses. Pathogens 2021; 10:pathogens10030278. [PMID: 33801245 PMCID: PMC8001386 DOI: 10.3390/pathogens10030278] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/17/2021] [Accepted: 02/25/2021] [Indexed: 11/17/2022] Open
Abstract
Human septins comprise a family of 13 genes that encode conserved GTP-binding proteins. They form nonpolar complexes, which assemble into higher-order structures, such as bundles, scaffolding structures, or rings. Septins are counted among the cytoskeletal elements. They interact with the actin and microtubule networks and can bind to membranes. Many cellular functions with septin participation have been described in the literature, including cytokinesis, motility, forming of scaffolding platforms or lateral diffusion barriers, vesicle transport, exocytosis, and recognition of micron-scale curvature. Septin dysfunction has been implicated in diverse human pathologies, including neurodegeneration and tumorigenesis. Moreover, septins are thought to affect the outcome of host–microbe interactions. Implication of septins has been demonstrated in fungal, bacterial, and viral infections. Knowledge on the precise function of a particular septin in the different steps of the virus infection and replication cycle is still limited. Published data for vaccinia virus (VACV), hepatitis C virus (HCV), influenza A virus (H1N1 and H5N1), human herpesvirus 8 (HHV-8), and Zika virus (ZIKV), all of major concern for public health, will be discussed here.
Collapse
|
12
|
Mangold CA, Rathbun MM, Renner DW, Kuny CV, Szpara ML. Viral infection of human neurons triggers strain-specific differences in host neuronal and viral transcriptomes. PLoS Pathog 2021; 17:e1009441. [PMID: 33750985 PMCID: PMC8016332 DOI: 10.1371/journal.ppat.1009441] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 04/01/2021] [Accepted: 03/01/2021] [Indexed: 12/11/2022] Open
Abstract
Infection with herpes simplex virus 1 (HSV-1) occurs in over half the global population, causing recurrent orofacial and/or genital lesions. Individual strains of HSV-1 demonstrate differences in neurovirulence in vivo, suggesting that viral genetic differences may impact phenotype. Here differentiated SH-SY5Y human neuronal cells were infected with one of three HSV-1 strains known to differ in neurovirulence in vivo. Host and viral RNA were sequenced simultaneously, revealing strain-specific differences in both viral and host transcription in infected neurons. Neuronal morphology and immunofluorescence data highlight the pathological changes in neuronal cytoarchitecture induced by HSV-1 infection, which may reflect host transcriptional changes in pathways associated with adherens junctions, integrin signaling, and others. Comparison of viral protein levels in neurons and epithelial cells demonstrated that a number of differences were neuron-specific, suggesting that strain-to-strain variations in host and virus transcription are cell type-dependent. Together, these data demonstrate the importance of studying virus strain- and cell-type-specific factors that may contribute to neurovirulence in vivo, and highlight the specificity of HSV-1-host interactions.
Collapse
Affiliation(s)
- Colleen A. Mangold
- Departments of Biology, Biochemistry and Molecular Biology, Center for Infectious Disease Dynamics, and the Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, Pennsylvania, United States of America
- Department of Entomology, College of Agricultural Sciences, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Molly M. Rathbun
- Departments of Biology, Biochemistry and Molecular Biology, Center for Infectious Disease Dynamics, and the Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Daniel W. Renner
- Departments of Biology, Biochemistry and Molecular Biology, Center for Infectious Disease Dynamics, and the Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Chad V. Kuny
- Departments of Biology, Biochemistry and Molecular Biology, Center for Infectious Disease Dynamics, and the Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Moriah L. Szpara
- Departments of Biology, Biochemistry and Molecular Biology, Center for Infectious Disease Dynamics, and the Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, Pennsylvania, United States of America
| |
Collapse
|
13
|
Lee JH, Shim J, Kim SJ. Stunning symmetries involved in the self-assembly of the HSV-1 capsid. THE JOURNAL OF THE KOREAN PHYSICAL SOCIETY 2021; 78:357-364. [PMID: 33584000 PMCID: PMC7871024 DOI: 10.1007/s40042-020-00044-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 10/29/2020] [Accepted: 10/29/2020] [Indexed: 06/12/2023]
Abstract
Herpes simplex virus-1 (HSV-1) is an enveloped dsDNA virus, infecting ~ 67% of humans. Here, we present the essential components of the HSV-1, focusing on stunning symmetries on the capsid. However, little is known about how the symmetries are involved dynamically in the self-assembly process. We suggest small angle X-ray scattering as a suitable method to capture the dynamics of self-assembly. Furthermore, our understanding of the viruses can be expanded by using an integrative approach that combines heterogeneous types of data, thus promoting new diagnostic tools and a cure for viral infections.
Collapse
Affiliation(s)
- Joo-hyeon Lee
- Department of Physics, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141 Korea
| | - Jaehyu Shim
- Department of Physics, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141 Korea
| | - Seung Joong Kim
- Department of Physics, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141 Korea
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141 Korea
| |
Collapse
|
14
|
Lind L, Svensson A, Thörn K, Krzyzowska M, Eriksson K. CD8 + T cells in the central nervous system of mice with herpes simplex infection are highly activated and express high levels of CCR5 and CXCR3. J Neurovirol 2021; 27:145-153. [PMID: 33492607 PMCID: PMC7831625 DOI: 10.1007/s13365-020-00940-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 11/09/2020] [Accepted: 12/27/2020] [Indexed: 12/25/2022]
Abstract
Herpes simplex virus type 2 (HSV-2) is a neurotropic virus that can cause meningitis, an inflammation of the meninges in the central nervous system. T cells are key players in viral clearance, and these cells migrate from peripheral blood into the central nervous system upon infection. Several factors contribute to T cell migration, including the expression of chemokines in the inflamed tissue that attract T cells through their expression of chemokine receptors. Here we investigated CD8+ T cell profile in the spinal cord in a mouse model of herpes simplex virus type 2 neuroinflammation. Mice were infected with HSV-2 and sacrificed when showing signs of neuroinflammation. Cells and/or tissue from spinal cord, spleen, and blood were analyzed for expression of activation markers, chemokine receptors, and chemokines. High numbers of CD8+ T cells were present in the spinal cord following genital HSV-2-infection. CD8+ T cells were highly activated and HSV-2 glycoprotein B -specific effector cells, some of which showed signs of recent degranulation. They also expressed high levels of many chemokine receptors, in particular CCR2, CCR4, CCR5, and CXCR3. Investigating corresponding receptor ligands in spinal cord tissue revealed markedly increased expression of the cognate ligands CCL2, CCL5, CCL8, CCL12, and CXCL10. This study shows that during herpesvirus neuroinflammation anti-viral CD8+ T cells accumulate in the CNS. CD8+ T cells in the CNS also express chemotactic receptors cognate to the chemotactic gradients in the spinal cord. This indicates that anti-viral CD8+ T cells may migrate to infected areas in the spinal cord during herpesvirus neuroinflammation in response to chemotactic gradients.
Collapse
Affiliation(s)
- Liza Lind
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Alexandra Svensson
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Karolina Thörn
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Malgorzata Krzyzowska
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Military Institute of Hygiene and Epidemiology, Kozielska 4, 01-163, Warsaw, Poland
| | - Kristina Eriksson
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
15
|
Duarte LF, Farías MA, Álvarez DM, Bueno SM, Riedel CA, González PA. Herpes Simplex Virus Type 1 Infection of the Central Nervous System: Insights Into Proposed Interrelationships With Neurodegenerative Disorders. Front Cell Neurosci 2019; 13:46. [PMID: 30863282 PMCID: PMC6399123 DOI: 10.3389/fncel.2019.00046] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 01/30/2019] [Indexed: 12/21/2022] Open
Abstract
Herpes simplex virus type 1 (HSV-1) is highly prevalent in humans and can reach the brain without evident clinical symptoms. Once in the central nervous system (CNS), the virus can either reside in a quiescent latent state in this tissue, or eventually actively lead to severe acute necrotizing encephalitis, which is characterized by exacerbated neuroinflammation and prolonged neuroimmune activation producing a life-threatening disease. Although HSV-1 encephalitis can be treated with antivirals that limit virus replication, neurological sequelae are common and the virus will nevertheless remain for life in the neural tissue. Importantly, there is accumulating evidence that suggests that HSV-1 infection of the brain both, in symptomatic and asymptomatic individuals could lead to neuronal damage and eventually, neurodegenerative disorders. Here, we review and discuss acute and chronic infection of particular brain regions by HSV-1 and how this may affect neuron and cognitive functions in the host. We review potential cellular and molecular mechanisms leading to neurodegeneration, such as protein aggregation, dysregulation of autophagy, oxidative cell damage and apoptosis, among others. Furthermore, we discuss the impact of HSV-1 infection on brain inflammation and its potential relationship with neurodegenerative diseases.
Collapse
Affiliation(s)
- Luisa F Duarte
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Mónica A Farías
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Diana M Álvarez
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M Bueno
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Claudia A Riedel
- Millennium Institute on Immunology and Immunotherapy, Departamento de Biología Celular, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Pablo A González
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
16
|
Brun P, Qesari M, Marconi PC, Kotsafti A, Porzionato A, Macchi V, Schwendener RA, Scarpa M, Giron MC, Palù G, Calistri A, Castagliuolo I. Herpes Simplex Virus Type 1 Infects Enteric Neurons and Triggers Gut Dysfunction via Macrophage Recruitment. Front Cell Infect Microbiol 2018; 8:74. [PMID: 29600197 PMCID: PMC5862801 DOI: 10.3389/fcimb.2018.00074] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 02/27/2018] [Indexed: 12/27/2022] Open
Abstract
Herpes Simplex Virus type 1 (HSV-1), a neurotropic pathogen widespread in human population, infects the enteric nervous system (ENS) in humans and rodents and causes intestinal neuromuscular dysfunction in rats. Although infiltration of inflammatory cells in the myenteric plexus and neurodegeneration of enteric nerves are common features of patients suffering from functional intestinal disorders, the proof of a pathogenic link with HSV-1 is still unsettled mainly because the underlying mechanisms are largely unknown. In this study we demonstrated that following intragastrical administration HSV-1 infects neurons within the myenteric plexus resulting in functional and structural alterations of the ENS. By infecting mice with HSV-1 replication-defective strain we revealed that gastrointestinal neuromuscular anomalies were however independent of viral replication. Indeed, enteric neurons exposed to UV-inactivated HSV-1 produced monocyte chemoattractant protein-1 (MCP-1/CCL2) to recruit activated macrophages in the longitudinal muscle myenteric plexus. Infiltrating macrophages produced reactive oxygen and nitrogen species and directly harmed enteric neurons resulting in gastrointestinal dysmotility. In HSV-1 infected mice intestinal neuromuscular dysfunctions were ameliorated by in vivo administration of (i) liposomes containing dichloromethylene bisphosphonic acid (clodronate) to deplete tissue macrophages, (ii) CCR2 chemokine receptor antagonist RS504393 to block the CCL2/CCR2 pathway, (iii) Nω-Nitro-L-arginine methyl ester hydrochloride (L-NAME) and AR-C 102222 to quench production of nitrogen reactive species produced via iNOS. Overall these data demonstrate that HSV-1 infection makes enteric neurons recruit macrophages via production of a specific chemoattractant factor. The resulting inflammatory reaction is mandatory for intestinal dysmotility. These findings provide insights into the neuro-immune communication that occurs in the ENS following HSV-1 infection and allow recognition of an original pathophysiologic mechanism underlying gastrointestinal diseases as well as identification of novel therapeutic targets.
Collapse
Affiliation(s)
- Paola Brun
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Marsela Qesari
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Peggy C Marconi
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Andromachi Kotsafti
- Esophageal and Digestive Tract Surgery Unit, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | | | - Veronica Macchi
- Department of Neurosciences, University of Padova, Padova, Italy
| | - Reto A Schwendener
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | - Marco Scarpa
- Esophageal and Digestive Tract Surgery Unit, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Maria C Giron
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Giorgio Palù
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Arianna Calistri
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | | |
Collapse
|
17
|
Neuroinvasion and Inflammation in Viral Central Nervous System Infections. Mediators Inflamm 2016; 2016:8562805. [PMID: 27313404 PMCID: PMC4897715 DOI: 10.1155/2016/8562805] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 03/16/2016] [Accepted: 04/12/2016] [Indexed: 12/31/2022] Open
Abstract
Neurotropic viruses can cause devastating central nervous system (CNS) infections, especially in young children and the elderly. The blood-brain barrier (BBB) and the blood-cerebrospinal fluid barrier (BCSFB) have been described as relevant sites of entry for specific viruses as well as for leukocytes, which are recruited during the proinflammatory response in the course of CNS infection. In this review, we illustrate examples of established brain barrier models, in which the specific reaction patterns of different viral families can be analyzed. Furthermore, we highlight the pathogen specific array of cytokines and chemokines involved in immunological responses in viral CNS infections. We discuss in detail the link between specific cytokines and chemokines and leukocyte migration profiles. The thorough understanding of the complex and interrelated inflammatory mechanisms as well as identifying universal mediators promoting CNS inflammation is essential for the development of new diagnostic and treatment strategies.
Collapse
|
18
|
Abstract
UNLABELLED Latent infections by viruses usually involve minimizing viral protein expression so that the host immune system cannot recognize the infected cell through the viral peptides presented on its cell surface. Herpes simplex virus (HSV), for example, is thought to express noncoding RNAs such as latency-associated transcripts (LATs) and microRNAs (miRNAs) as the only abundant viral gene products during latent infection. Here we describe analysis of HSV-1 mutant viruses, providing strong genetic evidence that HSV-infected cell protein 0 (ICP0) is expressed during establishment and/or maintenance of latent infection in murine sensory neurons in vivo Studies of an ICP0 nonsense mutant virus showed that ICP0 promotes heterochromatin and latent and lytic transcription, arguing that ICP0 is expressed and functional. We propose that ICP0 promotes transcription of LATs during establishment or maintenance of HSV latent infection, much as it promotes lytic gene transcription. This report introduces the new concept that a lytic viral protein can be expressed during latent infection and can serve dual roles to regulate viral chromatin to optimize latent infection in addition to its role in epigenetic regulation during lytic infection. An additional implication of the results is that ICP0 might serve as a target for an antiviral therapeutic acting on lytic and latent infections. IMPORTANCE Latent infection by viruses usually involves minimizing viral protein synthesis so that the host immune system cannot recognize the infected cells and eliminate them. Herpes simplex virus has been thought to express only noncoding RNAs as abundant gene products during latency. In this study, we found genetic evidence that an HSV lytic protein is functional during latent infection, and this protein may provide a new target for antivirals that target both lytic and latent infections.
Collapse
|
19
|
Yah CS, Simate GS. Nanoparticles as potential new generation broad spectrum antimicrobial agents. Daru 2015; 23:43. [PMID: 26329777 PMCID: PMC4557602 DOI: 10.1186/s40199-015-0125-6] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 08/12/2015] [Indexed: 12/20/2022] Open
Abstract
The rapid emergence of antimicrobial resistant strains to conventional antimicrobial agents has complicated and prolonged infection treatment and increased mortality risk globally. Furthermore, some of the conventional antimicrobial agents are unable to cross certain cell membranes thus, restricting treatment of intracellular pathogens. Therefore, the disease-causing-organisms tend to persist in these cells. However, the emergence of nanoparticle (NP) technology has come with the promising broad spectrum NP-antimicrobial agents due to their vast physiochemical and functionalization properties. In fact, NP-antimicrobial agents are able to unlock the restrictions experienced by conventional antimicrobial agents. This review discusses the status quo of NP-antimicrobial agents as potent broad spectrum antimicrobial agents, sterilization and wound healing agents, and sustained inhibitors of intracellular pathogens. Indeed, the perspective of developing potent NP-antimicrobial agents that carry multiple-functionality will revolutionize clinical medicine and play a significant role in alleviating disease burden.
Collapse
Affiliation(s)
- Clarence S Yah
- Department of Biochemistry and Microbiology, Nelson Mandela Metropolitan University, Port Elizabeth, South Africa.
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, E7146, 615 N. Wolfe Street, Baltimore, 21205, , MD, USA.
| | - Geoffrey S Simate
- School of Chemical and Metallurgical Engineering, University of the Witwatersrand, P/Bag 3, Wits 2050, Johannesburg, South Africa.
| |
Collapse
|
20
|
Isaacs CJ, Shinnick JE, Schadt K, Lynch DR, Lin KY. Prospects of gene and cell therapy for managing cardiac complications in Friedreich ataxia. Expert Opin Orphan Drugs 2015. [DOI: 10.1517/21678707.2015.1083854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
21
|
Rousseau A, Nasser G, Chiquet C, Barreau E, Gendron G, Kaswin G, M’Garrech M, Benoudiba F, Ducreux D, Labetoulle M. Diffusion tensor magnetic resonance imaging of trigeminal nerves in relapsing herpetic keratouveitis. PLoS One 2015; 10:e0122186. [PMID: 25830672 PMCID: PMC4382307 DOI: 10.1371/journal.pone.0122186] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 02/09/2015] [Indexed: 01/27/2023] Open
Abstract
Background Corneal hypoesthesia is the landmark of HSV and VZV keratitis and can lead to neurotrophic keratitis. Diffusion tensor imaging (DTI) is a new magnetic resonance imaging (MRI) derived technique, which offers possibilities to study axonal architecture. We aimed at assessing the potential impact of recurrent HSV or VZV-related keratitis on the axonal architecture of trigeminal nerves using DTI. Design Prospective non-interventional study. Participants Twelve patients and 24 controls. Methods DTI using MRI of the trigeminal fibers and corneal esthesiometry using the Cochet-Bonnet esthesiometer were acquired for patients affected by unilateral and recurrent HSV or VZV-related keratitis (3 months after the last corneal inflammatory event), and control subjects with no history of ocular or neuronal disease affecting the trigeminal pathways. Main Outcome Measures Fractional anisotropy (FA) and apparent diffusion coefficient (ADC) were compared between the 2 eyes of both patients and controls, and correlated with corneal esthesiometry. Results FA was lower in the trigeminal fibers ipsilateral to the affected eye compared to the non-affected side (0.39±0.02 versus 0.46±0.04, P=0.03). This difference was more important than the intra-individual variability observed in controls. Concomitantly, the asymmetry in ADC results was significantly correlated with the loss of corneal sensitivity in the affected eye. Conclusions Corneal hypoesthesia related to HSV and VZV keratitis is associated with persistent modifications in the architecture and functionality of the trigeminal fibers. These results add further explanation to the pathogenesis of HSV and VZV-induced neurotrophic keratitis, which may occur despite an apparent quiescence of the disease.
Collapse
Affiliation(s)
- Antoine Rousseau
- Department of Ophthalmology, Bicêtre Hospital, Assistance Publique—Hôpitaux de Paris, Paris-Sud University, Le Kremlin-Bicêtre, France
| | - Ghaïdaa Nasser
- Department of Neuroradiology, Bicêtre Hospital, Assistance Publique—Hôpitaux de Paris, Paris-Sud University, Le Kremlin-Bicêtre, France
| | - Christophe Chiquet
- Department of Ophthalmology, Grenoble University Hospital, Grenoble, France
| | - Emmanuel Barreau
- Department of Ophthalmology, Bicêtre Hospital, Assistance Publique—Hôpitaux de Paris, Paris-Sud University, Le Kremlin-Bicêtre, France
| | - Gael Gendron
- Department of Ophthalmology, Bicêtre Hospital, Assistance Publique—Hôpitaux de Paris, Paris-Sud University, Le Kremlin-Bicêtre, France
| | - Godefroy Kaswin
- Department of Ophthalmology, Bicêtre Hospital, Assistance Publique—Hôpitaux de Paris, Paris-Sud University, Le Kremlin-Bicêtre, France
| | - Mohamed M’Garrech
- Department of Ophthalmology, Bicêtre Hospital, Assistance Publique—Hôpitaux de Paris, Paris-Sud University, Le Kremlin-Bicêtre, France
| | - Farida Benoudiba
- Department of Neuroradiology, Bicêtre Hospital, Assistance Publique—Hôpitaux de Paris, Paris-Sud University, Le Kremlin-Bicêtre, France
| | - Denis Ducreux
- Department of Neuroradiology, Bicêtre Hospital, Assistance Publique—Hôpitaux de Paris, Paris-Sud University, Le Kremlin-Bicêtre, France
- * E-mail: (ML); (DD)
| | - Marc Labetoulle
- Department of Ophthalmology, Bicêtre Hospital, Assistance Publique—Hôpitaux de Paris, Paris-Sud University, Le Kremlin-Bicêtre, France
- * E-mail: (ML); (DD)
| |
Collapse
|
22
|
de Oliveira A, Prince D, Lo CY, Lee LH, Chu TC. Antiviral activity of theaflavin digallate against herpes simplex virus type 1. Antiviral Res 2015; 118:56-67. [PMID: 25818500 PMCID: PMC7113870 DOI: 10.1016/j.antiviral.2015.03.009] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 03/17/2015] [Accepted: 03/19/2015] [Indexed: 12/12/2022]
Abstract
Black tea theaflavins show significant anti-herpes simplex virus type 1 (HSV-1) effect on both A549 and Vero cells. Theaflavin is able to inhibit the entry of HSV-1 to its host by blocking the adsorption and penetration processes. Application of these agents as natural topical remedy to prevent and treat HSV-1 infection and spreading is promising.
Tea is the second most consumed drink in the world. The beneficial effects of tea have been mostly attributed to its catechin content. Black tea is derived from the leaves of Camellia sinensis plant, and it is rich in theaflavin polyphenols, in particular theaflavin (TF1), theaflavin-3-monogallate (TF2A), theaflavin-3′-monogallate (TF2B), and theaflavin-3,3′-digallate (TF3). Vero and A549 cells were used to evaluate the effect of purified individual black tea theaflavins as anti-herpes simplex virus 1 agents. With the rise of HSV resistant strains, there is a critical need to develop novel antiherpesviral treatments. Results of the cytotoxicity assay tested by MTS [3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxy-phenyl)-2-(4-sulfophenyl)-2H-tetrazolium] showed that TF1, TF2, and TF3 are not toxic to Vero and A549 cells at a concentration up to 75 μM. The antiviral activity of the individual theaflavins was tested by plaque reduction assay, MTS assay, flow cytometric analysis and confocal microscopy observations. The results showed that TF1, TF2, and TF3 exhibit potent, dose-dependent anti-HSV-1 effect, with TF3 being the most efficient in both Vero and A549 cells. A concentration of 50 μM TF3 and above was sufficient to inhibit >99% of the production of HSV-1 viral particles. The anti-HSV-1 effect of TF3 is due to a direct effect on the virions, and treating Vero or A549 cells with TF3 for 1 h prior to infection, or treating the cells at different times post infection does not inhibit HSV-1 production. TF3 is stable at vaginal pH, indicating its potential to be a promising natural and affordable remedy against herpes simplex viral infections.
Collapse
Affiliation(s)
- Aline de Oliveira
- Department of Biological Sciences, Seton Hall University, South Orange, NJ, USA
| | - Derek Prince
- Department of Biological Sciences, Seton Hall University, South Orange, NJ, USA
| | - Chih-Yu Lo
- Department of Food Science, National Chiayi University, Chiayi City, Taiwan
| | - Lee H Lee
- Department of Biology & Molecular Biology, Montclair State University, Montclair, NJ, USA
| | - Tin-Chun Chu
- Department of Biological Sciences, Seton Hall University, South Orange, NJ, USA.
| |
Collapse
|
23
|
D’Aiuto L, Prasad KM, Upton CH, Viggiano L, Milosevic J, Raimondi G, McClain L, Chowdari K, Tischfield J, Sheldon M, Moore JC, Yolken RH, Kinchington PR, Nimgaonkar VL. Persistent infection by HSV-1 is associated with changes in functional architecture of iPSC-derived neurons and brain activation patterns underlying working memory performance. Schizophr Bull 2015; 41:123-32. [PMID: 24622295 PMCID: PMC4266288 DOI: 10.1093/schbul/sbu032] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Herpes simplex virus, type 1 (HSV-1) commonly produces lytic mucosal lesions. It invariably initiates latent infection in sensory ganglia enabling persistent, lifelong infection. Acute HSV-1 encephalitis is rare and definitive evidence of latent infection in the brain is lacking. However, exposure untraceable to encephalitis has been repeatedly associated with impaired working memory and executive functions, particularly among schizophrenia patients. METHODS Patterns of HSV-1 infection and gene expression changes were examined in human induced pluripotent stem cell (iPSC)-derived neurons. Separately, differences in blood oxygenation level-dependent (BOLD) responses to working memory challenges using letter n-back tests were investigated using functional magnetic resonance imaging (fMRI) among schizophrenia cases/controls. RESULTS HSV-1 induced lytic changes in iPSC-derived glutamatergic neurons and neuroprogenitor cells. In neurons, HSV-1 also entered a quiescent state following coincubation with antiviral drugs, with distinctive changes in gene expression related to functions such as glutamatergic signaling. In the fMRI studies, main effects of schizophrenia (P = .001) and HSV-1 exposure (1-back, P = 1.76 × 10(-4); 2-back, P = 1.39 × 10(-5)) on BOLD responses were observed. We also noted increased BOLD responses in the frontoparietal, thalamus, and midbrain regions among HSV-1 exposed schizophrenia cases and controls, compared with unexposed persons. CONCLUSIONS The lytic/quiescent cycles in iPSC-derived neurons indicate that persistent neuronal infection can occur, altering cellular function. The fMRI studies affirm the associations between nonencephalitic HSV-1 infection and functional brain changes linked with working memory impairment. The fMRI and iPSC studies together provide putative mechanisms for the cognitive impairments linked to HSV-1 exposure.
Collapse
Affiliation(s)
- Leonardo D’Aiuto
- Department of Psychiatry, WPIC, University of Pittsburgh, School of Medicine, Pittsburgh PA;,These authors contributed equally to the article
| | - Konasale M. Prasad
- Department of Psychiatry, WPIC, University of Pittsburgh, School of Medicine, Pittsburgh PA;,These authors contributed equally to the article
| | - Catherine H. Upton
- Department of Psychiatry, WPIC, University of Pittsburgh, School of Medicine, Pittsburgh PA
| | - Luigi Viggiano
- Department of Biology, University of Bari “Aldo Moro”, Bari, Italy
| | - Jadranka Milosevic
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Giorgio Raimondi
- Department of Plastic and Reconstructive Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Lora McClain
- Department of Psychiatry, WPIC, University of Pittsburgh, School of Medicine, Pittsburgh PA
| | - Kodavali Chowdari
- Department of Psychiatry, WPIC, University of Pittsburgh, School of Medicine, Pittsburgh PA
| | - Jay Tischfield
- Department of Genetics and The Human Genome Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ
| | - Michael Sheldon
- Department of Genetics and The Human Genome Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ
| | - Jennifer C. Moore
- Department of Genetics and The Human Genome Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ
| | - Robert H. Yolken
- Stanley Division of Neurovirology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Paul R. Kinchington
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA;,Department of Molecular Genetics & Biochemistry, University of Pittsburgh, Pittsburgh, PA
| | - Vishwajit L. Nimgaonkar
- Department of Psychiatry, WPIC, University of Pittsburgh, School of Medicine, Pittsburgh PA;,Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA,*To whom correspondence should be addressed; Western Psychiatric Institute and Clinic, TDH 441, 3811 O’Hara St, Pittsburgh, PA 15213, US; tel: 412-246-6353, fax: 412-246-6350, e-mail:
| |
Collapse
|
24
|
Abstract
By controlling gene expression at the level of mRNA translation, organisms temporally and spatially respond swiftly to an ever-changing array of environmental conditions. This capacity for rapid response is ideally suited for mobilizing host defenses and coordinating innate responses to infection. Not surprisingly, a growing list of pathogenic microbes target host mRNA translation for inhibition. Infection with bacteria, protozoa, viruses, and fungi has the capacity to interfere with ongoing host protein synthesis and thereby trigger and/or suppress powerful innate responses. This review discusses how diverse pathogens manipulate the host translation machinery and the impact of these interactions on infection biology and the immune response.
Collapse
Affiliation(s)
- Ian Mohr
- Department of Microbiology, NYU Cancer Institute, New York University School of Medicine, New York, NY 10016, USA
| | - Nahum Sonenberg
- Department of Biochemistry, Goodman Cancer Centre, McGill University, Montreal, QC H3A 1A3, Canada
| |
Collapse
|
25
|
Kramer MF, Jurak I, Pesola JM, Boissel S, Knipe DM, Coen DM. Herpes simplex virus 1 microRNAs expressed abundantly during latent infection are not essential for latency in mouse trigeminal ganglia. Virology 2011; 417:239-47. [PMID: 21782205 DOI: 10.1016/j.virol.2011.06.027] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2011] [Revised: 06/06/2011] [Accepted: 06/16/2011] [Indexed: 01/02/2023]
Abstract
Several herpes simplex virus 1 microRNAs are encoded within or near the latency associated transcript (LAT) locus, and are expressed abundantly during latency. Some of these microRNAs can repress the expression of important viral proteins and are hypothesized to play important roles in establishing and/or maintaining latent infections. We found that in lytically infected cells and in acutely infected mouse ganglia, expression of LAT-encoded microRNAs was weak and unaffected by a deletion that includes the LAT promoter. In mouse ganglia latently infected with wild type virus, the microRNAs accumulated to high levels, but deletions of the LAT promoter markedly reduced expression of LAT-encoded microRNAs and also miR-H6, which is encoded upstream of LAT and can repress expression of ICP4. Because these LAT deletion mutants establish and maintain latent infections, these microRNAs are not essential for latency, at least in mouse trigeminal ganglia, but may help promote it.
Collapse
Affiliation(s)
- Martha F Kramer
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | |
Collapse
|
26
|
Dosa S, Castellanos K, Bacsa S, Gagyi E, Kovacs SK, Valyi-Nagy K, Shukla D, Dermody TS, Valyi-Nagy T. Chronic progressive deficits in neuron size, density and number in the trigeminal ganglia of mice latently infected with herpes simplex virus. Brain Pathol 2011; 21:583-93. [PMID: 21371157 DOI: 10.1111/j.1750-3639.2011.00485.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Numerous epidemiological studies have proposed a link between herpes simplex virus (HSV) infection and several common chronic neuropsychiatric and neurodegenerative diseases. Experimental HSV infection of mice can lead to chronic behavioral and neurological deficits and chronic pain. While neuron injury and loss are well-documented consequences of the acute phase of infection, the pathologic consequences of latent HSV infection are poorly understood. To determine whether latent HSV infection can cause neuronal injury in mice, trigeminal ganglia (TG) derived from adult BALB/c mice 1, 12 and 31 weeks after corneal HSV type 1 (HSV-1) inoculation were analyzed for evidence of productive or latent HSV-1 infection, inflammation and changes in neuron size, density and number. We found that latent HSV-1 infection between 12 and 31 weeks after corneal virus inoculation was associated with inflammation and progressive deficits in mean neuron diameter, neuronal nucleus diameter, neuron density and neuron number in the TG relative to mock-infected controls. The extent of neuronal injury during latent infection correlated with the extent of inflammation. These studies demonstrate that latent HSV infection is associated with progressive neuronal pathology and may lead to a better understanding of the role of HSV infections in chronic neurological diseases.
Collapse
Affiliation(s)
- Sandor Dosa
- Department of Pathology, University of Illinois at Chicago, College of Medicine, Chicago, IL 60612, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Szpara ML, Kobiler O, Enquist LW. A common neuronal response to alphaherpesvirus infection. J Neuroimmune Pharmacol 2010; 5:418-27. [PMID: 20401540 PMCID: PMC2990883 DOI: 10.1007/s11481-010-9212-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2009] [Accepted: 03/12/2010] [Indexed: 12/11/2022]
Abstract
Alphaherpesviruses are a subfamily of the Herpesviridae that can invade the nervous system and establish either lytic or latent infections. The establishment of latent infection can occur only in neurons, indicating a unique virus-host interaction in these cells. Here, we compare results from seven microarray studies that focused on the host response of either neural tissue or isolated neurons to alphaherpesvirus infection. These studies utilized either herpes simplex virus type 1 or pseudorabies virus as the infectious agent. From these data, we have found common host responses spanning a variety of infection models in different species, with different herpesvirus strains, and during all phases of infection including lytic, latent, and reactivation. The repeated observation of transcriptional effects on these genes and gene families indicates their likely importance in host defenses or the viral infectious process. We discuss the possible role of these different genes and genes families in alphaherpesvirus infection.
Collapse
Affiliation(s)
- Moriah L. Szpara
- Department of Molecular Biology and Princeton Neuroscience Institute, Princeton University, 314 Schultz Laboratory, Princeton NJ 08544, USA,
| | - Oren Kobiler
- Department of Molecular Biology and Princeton Neuroscience Institute, Princeton University, 314 Schultz Laboratory, Princeton NJ 08544, USA,
| | - Lynn W. Enquist
- Department of Molecular Biology and Princeton Neuroscience Institute, Princeton University, 314 Schultz Laboratory, Princeton NJ 08544, USA,
| |
Collapse
|
28
|
Hukkanen V, Paavilainen H, Mattila RK. Host responses to herpes simplex virus and herpes simplex virus vectors. Future Virol 2010. [DOI: 10.2217/fvl.10.35] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Herpes simplex virus (HSV) is a well-known, ubiquitous pathogen of humans. Engineered mutants of HSV can also be exploited as vectors in gene therapy or for virotherapy of tumors. HSV has multiple abilities to evade and modulate the innate and adaptive responses of the host. The increasing knowledge on the mutual interactions of the invading HSV with the host defenses will contribute to our deeper understanding of the relationship between HSV and the host, and thereby lead to future development of more effective and specific HSV vectors for treatment of human diseases. The future advances of HSV vaccines and vaccine vectors are based on the knowlegde of the complex interplay between HSV and the host defenses.
Collapse
Affiliation(s)
| | - Henrik Paavilainen
- Department of Virology, University of Turku, Kiinamyllynkatu 13, FIN-20520 Turku, Finland
| | - Riikka K Mattila
- Institute of Diagnostics, University of Oulu, Aapistie 5A, FIN-90014, Finland
| |
Collapse
|
29
|
Zeier Z, Aguilar JS, Lopez CM, Devi-Rao GB, Watson ZL, Baker HV, Wagner EK, Bloom DC. A limited innate immune response is induced by a replication-defective herpes simplex virus vector following delivery to the murine central nervous system. J Neurovirol 2010; 15:411-24. [PMID: 20095947 DOI: 10.3109/13550280903473452] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Herpes simplex virus type 1 (HSV-1)-based vectors readily transduce neurons and have a large payload capacity, making them particularly amenable to gene therapy applications within the central nervous system (CNS). Because aspects of the host responses to HSV-1 vectors in the CNS are largely unknown, we compared the host response of a nonreplicating HSV-1 vector to that of a replication-competent HSV-1 virus using microarray analysis. In parallel, HSV-1 gene expression was tracked using HSV-specific oligonucleotide-based arrays in order to correlate viral gene expression with observed changes in host response. Microarray analysis was performed following stereotactic injection into the right hippocampal formation of mice with either a replication-competent HSV-1 or a nonreplicating recombinant of HSV-1, lacking the ICP4 gene (ICP4-). Genes that demonstrated a significant change (P < .001) in expression in response to the replicating HSV-1 outnumbered those that changed in response to mock or nonreplicating vector by approximately 3-fold. Pathway analysis revealed that both the replicating and nonreplicating vectors induced robust antigen presentation but only mild interferon, chemokine, and cytokine signaling responses. The ICP4- vector was restricted in several of the Toll-like receptor-signaling pathways, indicating reduced stimulation of the innate immune response. These array analyses suggest that although the nonreplicating vector induces detectable activation of immune response pathways, the number and magnitude of the induced response is dramatically restricted compared to the replicating vector, and with the exception of antigen presentation, host gene expression induced by the nonreplicating vector largely resembles mock infection.
Collapse
Affiliation(s)
- Zane Zeier
- Departments of Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, Florida 32610-0266, USA
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Brun P, Giron MC, Zoppellaro C, Bin A, Porzionato A, De Caro R, Barbara G, Stanghellini V, Corinaldesi R, Zaninotto G, Palù G, Gaion RM, Tonini M, De Giorgio R, Castagliuolo I. Herpes simplex virus type 1 infection of the rat enteric nervous system evokes small-bowel neuromuscular abnormalities. Gastroenterology 2010; 138:1790-801. [PMID: 20102717 DOI: 10.1053/j.gastro.2010.01.036] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2009] [Revised: 01/13/2010] [Accepted: 01/20/2010] [Indexed: 01/15/2023]
Abstract
BACKGROUND & AIMS Infectious agents, such as neurotropic viruses, are proposed to disrupt the enteric neuromuscular system, leading to dysmotility, although the mechanisms are unknown. Our purpose was to assess whether herpes simplex virus type-1 (HSV-1) establishes an enteric-neuronal infection and induces gut dysmotility. METHODS Rats were inoculated with HSV-1 intranasally and after 4 weeks intragastrically. After 1-10 weeks, infection was determined by molecular analysis whereas neuromuscular function was evaluated by pharmacologic/electrical stimulation of longitudinal ileal segments and by gastrointestinal transit and by [(3)H]acetylcholine release measurements. Inflammation in the neuromuscular layer was assessed by myeloperoxidase and cytokine levels and by anti-CD3(+) immunohistochemistry. RESULTS After 1-10 weeks of intragastric inoculation, HSV-1 latency-associated messenger RNA transcripts were detected in the brain and in ileal neurons with no signs of illness or histologic gut abnormalities. By using a recombinant HSV-1 carrying the lacZ gene, HSV-1 virions were localized in myenteric ganglia by in situ X-gal staining. Interleukin-2 and IFN-gamma levels were increased significantly 1 and 6 weeks after inoculation. CD3(+) cells were found around the myenteric ganglia 6 weeks after inoculation. Smooth muscle responses to carbachol, CaCl(2), and gut transit were increased significantly after 1 and 6 weeks, whereas KCl- and electrical field stimulation-mediated contractions were modified significantly only 1-2 weeks after HSV-1 administration. The release of [(3)H]acetylcholine was reduced significantly in ileum segments after 1 and 6 weeks. CONCLUSIONS After intragastric inoculation, HSV-1 establishes a latent infection in the rat myenteric ganglia, which leads to gut dysmotility.
Collapse
Affiliation(s)
- Paola Brun
- Department of Histology, Microbiology and Medical Biotechnologies, University of Padua, Padua, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Numerous conserved and divergent microRNAs expressed by herpes simplex viruses 1 and 2. J Virol 2010; 84:4659-72. [PMID: 20181707 DOI: 10.1128/jvi.02725-09] [Citation(s) in RCA: 127] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Certain viruses use microRNAs (miRNAs) to regulate the expression of their own genes, host genes, or both. Previous studies have identified a limited number of miRNAs expressed by herpes simplex viruses 1 and 2 (HSV-1 and -2), some of which are conserved between these two viruses. To more comprehensively analyze the miRNAs expressed by HSV-1 or HSV-2 during productive and latent infection, we applied a massively parallel sequencing approach. We were able to identify 16 and 17 miRNAs expressed by HSV-1 and HSV-2, respectively, including all previously known species, and a number of previously unidentified virus-encoded miRNAs. The genomic positions of most miRNAs encoded by these two viruses are within or proximal to the latency-associated transcript region. Nine miRNAs are conserved in position and/or sequence, particularly in the seed region, between these two viruses. Interestingly, we did not detect an HSV-2 miRNA homolog of HSV-1 miR-H1, which is highly expressed during productive infection, but we did detect abundant expression of miR-H6, whose seed region is conserved with HSV-1 miR-H1 and might represent a functional analog. We also identified a highly conserved miRNA family arising from the viral origins of replication. In addition, we detected several pairs of complementary miRNAs and we found miRNA-offset RNAs (moRs) arising from the precursors of HSV-1 and HSV-2 miR-H6 and HSV-2 miR-H4. Our results reveal elements of miRNA conservation and divergence that should aid in identifying miRNA functions.
Collapse
|
32
|
Mahller YY, Sakthivel B, Baird WH, Aronow BJ, Hsu YH, Cripe TP, Mehrian-Shai R. Molecular analysis of human cancer cells infected by an oncolytic HSV-1 reveals multiple upregulated cellular genes and a role for SOCS1 in virus replication. Cancer Gene Ther 2008; 15:733-41. [PMID: 18551144 DOI: 10.1038/cgt.2008.40] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Oncolytic herpes simplex viruses (oHSVs) are promising anticancer therapeutics. We sought to characterize the functional genomic response of human cancer cells to oHSV infection using G207, an oHSV previously evaluated in a phase I trial. Five human malignant peripheral nerve sheath tumor cell lines, with differing sensitivity to oHSV, were infected with G207 for 6 h. Functional genomic analysis of virus-infected cells demonstrated large clusters of downregulated cellular mRNAs and smaller clusters of those upregulated, including 21 genes commonly upregulated in all five lines. Of these, 7 are known to be HSV-1 induced and 14 represent novel virus-regulated genes. Gene ontology analysis revealed that a majority of G207-upregulated genes are involved in Janus kinase/signal transducer and activator of transcription signaling, transcriptional regulation, nucleic acid metabolism, protein synthesis and apoptosis. Ingenuity networks highlighted nodes for AP-1 subunits and interferon signaling via STAT1, suppressor of cytokine signaling-1 (SOCS1), SOCS3 and RANTES. As biological confirmation, we found that virus-mediated upregulation of SOCS1 correlated with sensitivity to G207 and that depletion of SOCS1 impaired virus replication by >10-fold. Further characterization of roles provided by oHSV-induced cellular genes during virus replication may be utilized to predict oncolytic efficacy and to provide rational strategies for designing next-generation oncolytic viruses.
Collapse
Affiliation(s)
- Y Y Mahller
- Division of Hematology and Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | | | | | | | | | | | | |
Collapse
|
33
|
Herpes simplex virus type 1 modulates cellular gene expression during quiescent infection of neuronal cells. Arch Virol 2008; 153:1335-45. [DOI: 10.1007/s00705-008-0122-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2008] [Accepted: 05/19/2008] [Indexed: 02/08/2023]
|
34
|
Pyykkö I, Zou J. Do Viruses Cause Inner Ear Disturbances? ORL J Otorhinolaryngol Relat Spec 2008; 70:32-40; discussion 40-1. [DOI: 10.1159/000111046] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
35
|
Hamza MA, Higgins DM, Ruyechan WT. Herpes simplex virus type-1 latency-associated transcript-induced immunoreactivity of substance P in trigeminal neurons is reversed by bone morphogenetic protein-7. Neurosci Lett 2006; 413:31-5. [PMID: 17239535 PMCID: PMC1821347 DOI: 10.1016/j.neulet.2006.11.063] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2006] [Revised: 10/10/2006] [Accepted: 11/07/2006] [Indexed: 11/23/2022]
Abstract
Herpes simplex virus type-1 (HSV-1) primarily infects mucoepithelial tissues of the eye and the orofacial region. Subsequently, the virus is retrogradely transported through the axons of the trigeminal sensory neurons to their nuclei, where the virus establishes a life-long latent infection. During this latency period, the viral genome is transcriptionally silent except for a single region encoding the latency-associated transcript (LAT). To understand how HSV-1 latency might affect the expression of substance P in sensory neurons, we transfected primary cultures of trigeminal neurons obtained from rat embryos, with LAT expressing plasmids. The expression of LAT increased the percentage of substance P-immunoreactive neurons by two thirds. To examine the effect of bone morphogenetic protein-7 (BMP7) on the LAT-induced increase in substance P expression in trigeminal neurons, cultures transfected with LAT were treated with BMP7. Treatment with BMP7 reversed the effects of LAT on substance P expression in trigeminal neurons. Our data show for the first time that LAT increases substance P expression in trigeminal neurons and BMP7 can reverse these effects of LAT.
Collapse
Affiliation(s)
- Mohamed A Hamza
- Departments of Pharmacology and Toxicology, University at Buffalo, SUNY, Buffalo, NY 14214, United States
| | | | | |
Collapse
|
36
|
Varaldi J, Ravallec M, Labrosse C, Lopez-Ferber M, Boulétreau M, Fleury F. Artifical transfer and morphological description of virus particles associated with superparasitism behaviour in a parasitoid wasp. JOURNAL OF INSECT PHYSIOLOGY 2006; 52:1202-12. [PMID: 17070831 DOI: 10.1016/j.jinsphys.2006.09.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2006] [Revised: 09/05/2006] [Accepted: 09/05/2006] [Indexed: 05/12/2023]
Abstract
In parasitoids, the adaptive significance of superparasitism (laying of egg(s) in already parasitized hosts) has been the subject of strong controversy. The current view is to interpret this behaviour as an adaptation to increased competition for hosts, because the supernumerary egg still has a chance to win possession for the host. However, we recently discovered that in the solitary parasitoid Leptopilina boulardi, superparasitism is rather caused by an unknown infectious element: stable non superparasitizing lineages (NS) are transformed into stable superparasitizing lineages (S) after eggs from both lineages have competed inside the same host (superparasitism). In this report, we investigate the nature and location of the causative agent. Involvement of bacteria is unlikely because antibiotic treatments do not affect wasp phenotype and because bacterial 16S ribosomal DNA was not detected using PCR. We report successful injection experiments showing that the causative agents are located in wasp poison gland and ovaries and are stably inherited. Electron microscopic studies demonstrate that long filamentous virus particles located in wasp oviducts are strongly associated with superparasitism behaviour, leading to reconsider the adaptive significance of this behaviour in parasitoids. Interestingly, parasitoids are often infected with similar viruses for which no phenotypic effect has been documented. This raises the possibility that they could induce the same behavioural manipulation.
Collapse
Affiliation(s)
- Julien Varaldi
- Laboratoire de Biométrie et Biologie Evolutive (UMR 5558);CNRS; Université Lyon 1, 43 bd 11 nov, 69622, Villeurbanne Cedex, France.
| | | | | | | | | | | |
Collapse
|
37
|
Pasieka TJ, Baas T, Carter VS, Proll SC, Katze MG, Leib DA. Functional genomic analysis of herpes simplex virus type 1 counteraction of the host innate response. J Virol 2006; 80:7600-12. [PMID: 16840339 PMCID: PMC1563739 DOI: 10.1128/jvi.00333-06] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2006] [Accepted: 05/15/2006] [Indexed: 12/15/2022] Open
Abstract
Herpes simplex virus type 1 (HSV-1) mutants lacking the ICP34.5 gene are severely attenuated in mouse models and have a significant growth defect in confluent mouse embryo fibroblasts. Previously, ICP34.5 was demonstrated to have a crucial role in evading the innate immune response to infection by mediating the dephosphorylation of eIF2alpha, a translation initiation factor phosphorylated by PKR during the antiviral response. To further understand the role of ICP34.5 in evasion of the antiviral response, we used transcriptional profiling to examine host cell gene expression in both wild-type and ICP34.5-null virus-infected mouse embryo fibroblasts over a time course of infection. Our study revealed that cells responded to infection within 3 h through PKR-dependent eIF2alpha phosphorylation and that the majority of up-regulated genes at 3 h postinfection were involved in the antiviral response. HSV-1 counters this response through early expression of ICP34.5 and dephosphorylation of eIF2alpha. By 12 h postinfection, the differences between the number and functional classification of genes differentially up- and down-regulated between wild-type and ICP34.5-null virus-infected cells were maximal. Specifically, in wild-type virus-infected cells, the majority of changed genes were involved in metabolic and biosynthetic processes, while in ICP34.5-null virus-infected cells, mostly antiviral genes were up-regulated. Further, ICP34.5-null virus-infected cells produced greater amounts of beta interferon than wild-type virus-infected cells. These results indicate that ICP34.5 expression and function at early times postinfection have a pivotal role in the ability of HSV-1 to gain control of the host cell and maintain an environment for successful viral replication.
Collapse
Affiliation(s)
- Tracy Jo Pasieka
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, 660 South Euclid Ave., Box 8096, St. Louis, MO 63110, USA
| | | | | | | | | | | |
Collapse
|
38
|
Blanchard Y, Le Meur N, Le Cunff M, Blanchard P, Léger J, Jestin A. Cellular gene expression survey of PseudoRabies Virus (PRV) infected Human Embryonic Kidney cells (HEK-293). Vet Res 2006; 37:705-23. [PMID: 16820135 DOI: 10.1051/vetres:2006027] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2005] [Accepted: 02/21/2006] [Indexed: 11/14/2022] Open
Abstract
Pseudorabies virus (PRV) is an alpha herpesvirus that causes Aujezsky disease in the pig. To characterize the impact of PRV infection on cellular expression, we used microarrays consisting of 9850 oligonucleotides corresponding to human genes and examined the expression levels of mRNA isolated 0.5, 3, 6, and 9 h post infection (hpi) from cultures of infected HEK-293 cells. Very few changes were observed during the first 3 h of infection but significant modifications in the cell expression of more than 1000 genes were clearly apparent by 6 hpi. More than 2400 genes were either up- or down-regulated during the 9 h experiment. These results were then analyzed using gene ontology and the MAPP and MAPPFinder software. This comprehensive analysis clearly shows that the down-regulated genes were mainly involved in macromolecular synthesis (DNA, RNA and proteins) and the cell cycle. The up-regulated genes primarily concerned the regulation of DNA transcription, developmental processes (central nervous system development, neurogenesis, angiogenesis), cell adhesion and potassium transport. This study is the first qualitative analysis of a gene expression survey in a human cell line following PRV infection. It demonstrates global changes in the cell expression profile, and identifies the main biological processes that are altered during virus replication.
Collapse
Affiliation(s)
- Yannick Blanchard
- Laboratoire de Génétique Virale et Biosécurité, Agence Française de Sécurité Sanitaire des Aliments, BP 53, 22440 Ploufragan, France.
| | | | | | | | | | | |
Collapse
|
39
|
Leong WF, Chow VTK. Transcriptomic and proteomic analyses of rhabdomyosarcoma cells reveal differential cellular gene expression in response to enterovirus 71 infection. Cell Microbiol 2006; 8:565-80. [PMID: 16548883 PMCID: PMC7162300 DOI: 10.1111/j.1462-5822.2005.00644.x] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Insights into the host antiviral strategies as well as viral disease manifestations can be achieved through the elucidation of host- and virus-mediated transcriptional responses. An oligo-based microarray was employed to analyse mRNAs from rhabdomyosarcoma cells infected with the MS/7423/87 strain of enterovirus 71 (EV71) at 20 h post infection. Using Acuity software and LOWESS normalization, 152 genes were found to be downregulated while 39 were upregulated by greater than twofold. Altered transcripts include those encoding components of cytoskeleton, protein translation and modification; cellular transport proteins; protein degradation mediators; cell death mediators; mitochondrial-related and metabolism proteins; cellular receptors and signal transducers. Changes in expression profiles of 15 representative genes were authenticated by real-time reverse transcription polymerase chain reaction (RT-PCR), which also compared the transcriptional responses of cells infected with EV71 strain 5865/Sin/000009 isolated from a fatal case during the Singapore outbreak in 2000. Western blot analyses of APOB, CLU, DCAMKL1 and ODC1 proteins correlated protein and transcript levels. Two-dimensional proteomic maps highlighted differences in expression of cellular proteins (CCT5, CFL1, ENO1, HSPB1, PSMA2 and STMN1) following EV71 infection. Expression of several apoptosis-associated genes was modified, coinciding with apoptosis attenuation observed in poliovirus infection. Interestingly, doublecortin and CaM kinase-like 1 (DCAMKL1) involved in brain development, was highly expressed during infection. Thus, microarray, real-time RT-PCR and proteomic analyses can elucidate the global view of the numerous and complex cellular responses that contribute towards EV71 pathogenesis.
Collapse
Affiliation(s)
- Wai Fook Leong
- Human Genome Laboratory, Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore 117597
| | | |
Collapse
|
40
|
Schmidt S, Pericak-Vance MA, Sawcer S, Barcellos LF, Hart J, Sims J, Prokop AM, van der Walt J, DeLoa C, Lincoln RR, Oksenberg JR, Compston A, Hauser SL, Haines JL, Gregory SG. Allelic association of sequence variants in the herpes virus entry mediator-B gene (PVRL2) with the severity of multiple sclerosis. Genes Immun 2006; 7:384-92. [PMID: 16738668 DOI: 10.1038/sj.gene.6364311] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Discrepant findings have been reported regarding an association of the apolipoprotein E (APOE) gene with the clinical course of multiple sclerosis (MS). To resolve these discrepancies, we examined common sequence variation in six candidate genes residing in a 380-kb genomic region surrounding and including the APOE locus for an association with MS severity. We genotyped at least three polymorphisms in each of six candidate genes in 1,540 Caucasian MS families (729 single-case and multiple-case families from the United States, 811 single-case families from the UK). By applying the quantitative transmission/disequilibrium test to a recently proposed MS severity score, the only statistically significant (P=0.003) association with MS severity was found for an intronic variant in the Herpes Virus Entry Mediator-B Gene PVRL2. Additional genotyping extended the association to a 16.6 kb block spanning intron 1 to intron 2 of the gene. Sequencing of PVRL2 failed to identify variants with an obvious functional role. In conclusion, the analysis of a very large data set suggests that genetic polymorphisms in PVRL2 may influence MS severity and supports the possibility that viral factors may contribute to the clinical course of MS, consistent with previous reports.
Collapse
Affiliation(s)
- S Schmidt
- Center for Human Genetics, Duke University Medical Center, Durham, NC 27710, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Surjit M, Liu B, Chow VTK, Lal SK. The nucleocapsid protein of severe acute respiratory syndrome-coronavirus inhibits the activity of cyclin-cyclin-dependent kinase complex and blocks S phase progression in mammalian cells. J Biol Chem 2006; 281:10669-81. [PMID: 16431923 PMCID: PMC7995956 DOI: 10.1074/jbc.m509233200] [Citation(s) in RCA: 152] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2005] [Revised: 01/17/2006] [Indexed: 11/15/2022] Open
Abstract
Deregulation of the cell cycle is a common strategy employed by many DNA and RNA viruses to trap and exploit the host cell machinery toward their own benefit. In many coronaviruses, the nucleocapsid protein (N protein) has been shown to inhibit cell cycle progression although the mechanism behind this is poorly understood. The N protein of severe acute respiratory syndrome-coronavirus (SARS-CoV) bears signature motifs for binding to cyclin and phosphorylation by cyclin-dependent kinase (CDK) and has recently been reported by us to get phosphorylated by the cyclin-CDK complex (Surjit, M., Kumar, R., Mishra, R. N., Reddy, M. K., Chow, V. T., and Lal, S. K. (2005) J. Virol. 79, 11476-11486). In the present study, we prove that the N protein of SARS-CoV can inhibit S phase progression in mammalian cell lines. N protein expression was found to directly inhibit the activity of the cyclin-CDK complex, resulting in hypophosphorylation of retinoblastoma protein with a concomitant down-regulation in E2F1-mediated transactivation. Coexpression of E2F1 under such conditions could restore the expression of S phase genes. Analysis of RXL and CDK phosphorylation mutant N protein identified the mechanism of inhibition of CDK4 and CDK2 activity to be different. Whereas N protein could directly bind to cyclin D and inhibit the activity of CDK4-cyclin D complex; inhibition of CDK2 activity appeared to be achieved in two different ways: indirectly by down-regulation of protein levels of CDK2, cyclin E, and cyclin A and by direct binding of N protein to CDK2-cyclin complex. Down-regulation of E2F1 targets was also observed in SARS-CoV-infected VeroE6 cells. These data suggest that the S phase inhibitory activity of the N protein may have major significance during viral pathogenesis.
Collapse
Affiliation(s)
- Milan Surjit
- Virology Group, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Rd., New Delhi 110067, India
| | | | | | | |
Collapse
|
42
|
Yuan J, Cahir-McFarland E, Zhao B, Kieff E. Virus and cell RNAs expressed during Epstein-Barr virus replication. J Virol 2006; 80:2548-65. [PMID: 16474161 PMCID: PMC1395376 DOI: 10.1128/jvi.80.5.2548-2565.2006] [Citation(s) in RCA: 129] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Changes in Epstein-Barr virus (EBV) and cell RNA levels were assayed following immunoglobulin G (IgG) cross-linking-induced replication in latency 1-infected Akata Burkitt B lymphoblasts. EBV replication as assayed by membrane gp350 expression was approximately 5% before IgG cross-linking and increased to more than 50% 48 h after induction. Seventy-two hours after IgG cross-linking, gp350-positive cells excluded propidium iodide as well as gp350-negative cells. EBV RNA levels changed temporally in parallel with previously defined sensitivity to inhibitors of protein or viral DNA synthesis. BZLF1 immediate-early RNA levels doubled by 2 h and reached a peak at 4 h, whereas BMLF1 doubled by 4 h with a peak at 8 h, and BRLF1 doubled by 8 h with peak at 12 h. Early RNAs peaked at 8 to 12 h, and late RNAs peaked at 24 h. Hybridization to intergenic sequences resulted in evidence for new EBV RNAs. Surprisingly, latency III (LTIII) RNAs for LMP1, LMP2, EBNALP, EBNA2, EBNA3A, EBNA3C, and BARTs were detected at 8 to 12 h and reached maxima at 24 to 48 h. EBNA2 and LMP1 were at full LTIII levels by 48 h and localized to gp350-positive cells. Thus, LTIII expression is a characteristic of late EBV replication in both B lymphoblasts and epithelial cells in immune-comprised people (J. Webster-Cyriaque, J. Middeldorp, and N. Raab-Traub, J. Virol. 74:7610-7618, 2000). EBV replication significantly altered levels of 401 Akata cell RNAs, of which 122 RNAs changed twofold or more relative to uninfected Akata cells. Mitogen-activated protein kinase levels were significantly affected. Late expression of LTIII was associated with induction of NF-kappaB responsive genes including IkappaBalpha and A20. The exclusion of propidium, expression of EBV LTIII RNAs and proteins, and up-regulation of specific cell RNAs are indicative of vital cell function late in EBV replication.
Collapse
Affiliation(s)
- Jing Yuan
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Channing Laboratory, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
43
|
Kent JR, Fraser NW. The cellular response to herpes simplex virus type 1 (HSV-1) during latency and reactivation. J Neurovirol 2005; 11:376-83. [PMID: 16162480 DOI: 10.1080/13550280591002405] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
In order to learn more about the cellular response to viral gene activity during latency and reactivation of herpes simplex virus type 1 (HSV-1), the authors have employed microarray analysis. On an array of about 1200 cellular genes, approximately 56 genes were found to be differentially regulated in infected trigeminal ganglia of mice, compared to uninfected mice, during latency and reactivation. Of these genes, 10 were examined more closely using quantitative real-time polymerase chain reaction (PCR) to confirm the microarray results. Genes involved in interferon and other signaling pathways appeared to predominate in response to a latent or reactivating HSV infection. Interestingly, some genes found to be differentially regulated in latently infected ganglia are neuronal-specific genes (pro-opiomelanocortinin; zinc finger proteins of the cerebellum 1 and 2). During reactivation, the involvement of several cell signaling molecules that may be important for the initiation of an HSV infection was observed, including various receptors and molecules involved in cell-cell spread.
Collapse
Affiliation(s)
- J R Kent
- Department of Microbiology, University of Pennsylvania, School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | | |
Collapse
|
44
|
Jones JO, Arvin AM. Viral and cellular gene transcription in fibroblasts infected with small plaque mutants of varicella-zoster virus. Antiviral Res 2005; 68:56-65. [PMID: 16118026 DOI: 10.1016/j.antiviral.2005.06.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2005] [Revised: 06/09/2005] [Accepted: 06/13/2005] [Indexed: 10/25/2022]
Abstract
Varicella-zoster virus (VZV) is an alphaherpesvirus that causes varicella and herpes zoster. In these experiments, cDNA corresponding to 69 VZV open reading frames was added to 42K human cDNA microarrays and used to examine viral as well as cellular gene transcription concurrently in fibroblasts infected with two genetically distinct small plaque VZV mutants, rOka/ORF63rev[T171] and rOkaDeltagI. rOka/ORF63rev[T171] has a point mutation in ORF63, which encodes the immediate early regulatory protein, IE63, and rOkaDeltagI has a deletion of ORF67, encoding glycoprotein I (gI). rOka/ORF63rev[T171] was deficient in the transcription of several viral genes compared to the recombinant rOka control virus. Deletion of ORF67 had minimal effects on viral gene transcription. Effects of rOka/ORF63rev[T171] and rOkaDeltagI on host cell gene transcription were similar to the rOka control, but a few host cell genes were regulated differently in rOkaDeltagI-infected cells. Infection of fibroblasts with intact or small plaque VZV mutants was associated with down-regulation of NF-kappaB and interferon responsive genes, down-regulation of TGF-beta responsive genes accompanied by reduced amounts of fibrotic/wound healing response genes (e.g. collagens, follistatin) and activation of cellular proliferation genes, and alteration of neuronal growth markers, as well as cellular genes encoding proteins important in protein and vesicle trafficking. These observations suggest that replication of VZV small plaque mutant viruses and intact VZV have similar consequences for host cell gene transcription in infected cells, and that the small plaque phenotype in these mutants reflects deficiencies in viral gene expression.
Collapse
Affiliation(s)
- Jeremy O Jones
- Department of Pediatrics, Stanford University, 300 Pasteur Drive, Rm G312, Stanford, CA, USA.
| | | |
Collapse
|
45
|
Jenner RG, Young RA. Insights into host responses against pathogens from transcriptional profiling. Nat Rev Microbiol 2005; 3:281-94. [PMID: 15806094 DOI: 10.1038/nrmicro1126] [Citation(s) in RCA: 432] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
DNA microarrays have allowed us to monitor the effects of pathogens on host-cell gene expression programmes in great depth and on a broad scale. The comparison of results that have been generated by these studies is complex, and such a comparison has not previously been attempted in a systematic manner. In this review, we have collated and compared published transcriptional-profiling data from 32 studies that involved 77 different host-pathogen interactions, and have defined a common host-transcriptional-response. We outline gene expression patterns in the context of Toll-like receptor and pathogen-mediated signalling pathways, and summarize the contributions that transcriptional-profiling studies have made to our understanding of the infectious disease process.
Collapse
Affiliation(s)
- Richard G Jenner
- Whitehead Institute for Biomedical Research, 9 Cambridge Center, Cambridge, Massachusetts 02142, USA.
| | | |
Collapse
|
46
|
Leong W, Tan H, Ooi E, Koh D, Chow VT. Microarray and real-time RT-PCR analyses of differential human gene expression patterns induced by severe acute respiratory syndrome (SARS) coronavirus infection of Vero cells. Microbes Infect 2005; 7:248-59. [PMID: 15777647 PMCID: PMC7110627 DOI: 10.1016/j.micinf.2004.11.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2004] [Revised: 11/03/2004] [Accepted: 11/03/2004] [Indexed: 12/02/2022]
Abstract
Vero E6 African green monkey kidney cells are highly susceptible to infection with the newly emerging severe acute respiratory syndrome coronavirus (SARS-CoV), and they are permissive for rapid viral replication, with resultant cytopathic effects. We employed cDNA microarray analysis to characterize the cellular transcriptional responses of homologous human genes at 12 h post-infection. Seventy mRNA transcripts belonging to various functional classes exhibited significant alterations in gene expression. There was considerable induction of heat shock proteins that are crucial to the immune response mechanism. Modified levels of several transcripts involved in pro-inflammatory and anti-inflammatory processes exemplified the balance between opposing forces during SARS pathogenesis. Other genes displaying altered transcription included those associated with host translation, cellular metabolism, cell cycle, signal transduction, transcriptional regulation, protein trafficking, protein modulators, and cytoskeletal proteins. Alterations in the levels of several novel transcripts encoding hypothetical proteins and expressed sequence tags were also identified. In addition, transcription of apoptosis-related genes DENN and hIAP1 was upregulated in contrast to FAIM. Elevated Mx1 expression signified a strong host response to mediate antiviral resistance. Also expressed in infected cells was the C-terminal alternative splice variant of the p53 tumor suppressor gene encoding a modified truncated protein that can influence the activity of wild-type p53. We observed the interplay between various mechanisms to favor virus multiplication before full-blown apoptosis and the triggering of several pathways in host cells in an attempt to eliminate the pathogen. Microarray analysis identifies the critical host–pathogen interactions during SARS-CoV infection and provides new insights into the pathophysiology of SARS.
Collapse
Affiliation(s)
- W.F. Leong
- Human Genome Laboratory, Department of Microbiology, Faculty of Medicine, National University of Singapore, Kent Ridge, Singapore 117597, Singapore
| | - H.C. Tan
- National Environment Agency, Singapore 228231, Singapore
| | - E.E. Ooi
- National Environment Agency, Singapore 228231, Singapore
| | - D.R. Koh
- Department of Physiology, Faculty of Medicine, National University of Singapore, Kent Ridge, Singapore 117597, Singapore
| | - Vincent T.K. Chow
- Human Genome Laboratory, Department of Microbiology, Faculty of Medicine, National University of Singapore, Kent Ridge, Singapore 117597, Singapore
- Corresponding author. Tel.: +65 6874 6200; fax: +65 6776 6872.
| |
Collapse
|
47
|
Hill JM, Steiner I, Matthews KE, Trahan SG, Foster TP, Ball MJ. Statins lower the risk of developing Alzheimer’s disease by limiting lipid raft endocytosis and decreasing the neuronal spread of Herpes simplex virus type 1. Med Hypotheses 2005; 64:53-8. [PMID: 15533611 DOI: 10.1016/j.mehy.2003.12.058] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2003] [Accepted: 12/05/2003] [Indexed: 02/08/2023]
Abstract
Many possible risk factors for Alzheimer's disease (AD) have been investigated, with only a very few showing positive associations and none defining the etiology of the neurodegenerative disease. The presence of herpes simplex virus type 1 (HSV-1) DNA in the brain, coupled with apolipoprotein E allele e4 (ApoE e4), has been suggested to confer an increased risk for AD. Studies have shown that pathogens, including viruses, utilize clathrin-independent endocytosis, i.e., lipid rafts that contain cholesterol, as part of their structure. Moreover, cholesterol-lowering statins have recently been linked with a reduced risk of developing Alzheimer's dementia. We, therefore, posit that long-term statin therapy protects individuals from AD by reducing the neuronal spread of HSV-1 via lipid raft domain pathways. Although the mechanism by which statins reduce AD risk is unknown, they reduce the amount of cholesterol in the plasma membrane and, thus, may decrease the availability of lipid raft pathways to spread HSV-1 within the brain.
Collapse
Affiliation(s)
- James M Hill
- Department of Ophthalmology, Louisiana State University Health Sciences Center, New Orleans, LA, USA.
| | | | | | | | | | | |
Collapse
|
48
|
Bello-Morales R, Fedetz M, Alcina A, Tabarés E, López-Guerrero JA. High susceptibility of a human oligodendroglial cell line to herpes simplex type 1 infection. J Neurovirol 2005; 11:190-8. [PMID: 16036797 DOI: 10.1080/13550280590924179] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
More than 20 infectious agents, ranging from retroviruses to mycobacteria, have been associated with multiple sclerosis onset or relapses in which oligodendrocytes, the myelin-forming cells of the central nervous system, are the initial target of the pathogenic status. In this work, the nature of the susceptibility of the human precursor oligodendroglial KG-1C cell line to herpes simplex virus type 1 (HSV-1) was investigated. Infection of KG-1C cells was characterized by a high level of virus production and a notable progression of the cytopathic effect. After infection, there was a significant shut-off of host mRNA translation, which was correlated with evident synthesis of viral proteins. An examination by electron microscopy of the infected cells revealed the presence of large clusters of mitochondria located in the proximity of intracellular HSV-1 particle groups. In addition, transmission electron microscopy and nuclear fluorescence analysis showed neither signs of chromatin condensation nor of apoptotic bodies. Furthermore, procaspase-3 remained uncleaved, suggesting that apoptosis does not take place, at least in this system. Finally, expression and localization of MAL2, a subpopulation of detergent-insoluble lipid raft protein, was studied. Detection of MAL2 significantly increased after infection and it was colocalized with HSV-1 proteins. From these findings the authors conclude that human oligodendrocyte-like cells are highly susceptible to HSV-1 infection. The implications of this for central nervous system viral infection are discussed.
Collapse
Affiliation(s)
- Raquel Bello-Morales
- Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid, Cantoblanco, Madrid, Spain
| | | | | | | | | |
Collapse
|
49
|
|
50
|
Cook WJ, Kramer MF, Walker RM, Burwell TJ, Holman HA, Coen DM, Knipe DM. Persistent expression of chemokine and chemokine receptor RNAs at primary and latent sites of herpes simplex virus 1 infection. Virol J 2004; 1:5. [PMID: 15507126 PMCID: PMC524517 DOI: 10.1186/1743-422x-1-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2004] [Accepted: 05/28/2004] [Indexed: 11/18/2022] Open
Abstract
Inflammatory cytokines and infiltrating T cells are readily detected in herpes simplex virus (HSV) infected mouse cornea and trigeminal ganglia (TG) during the acute phase of infection, and certain cytokines continue to be expressed at lower levels in infected TG during the subsequent latent phase. Recent results have shown that HSV infection activates Toll-like receptor signaling. Thus, we hypothesized that chemokines may be broadly expressed at both primary sites and latent sites of HSV infection for prolonged periods of time. Real-time reverse transcriptase-polymrease chain reaction (RT-PCR) to quantify expression levels of transcripts encoding chemokines and their receptors in cornea and TG following corneal infection. RNAs encoding the inflammatory-type chemokine receptors CCR1, CCR2, CCR5, and CXCR3, which are highly expressed on activated T cells, macrophages and most immature dendritic cells (DC), and the more broadly expressed CCR7, were highly expressed and strongly induced in infected cornea and TG at 3 and 10 days postinfection (dpi). Elevated levels of these RNAs persisted in both cornea and TG during the latent phase at 30 dpi. RNAs for the broadly expressed CXCR4 receptor was induced at 30 dpi but less so at 3 and 10 dpi in both cornea and TG. Transcripts for CCR3 and CCR6, receptors that are not highly expressed on activated T cells or macrophages, also appeared to be induced during acute and latent phases; however, their very low expression levels were near the limit of our detection. RNAs encoding the CCR1 and CCR5 chemokine ligands MIP-1α, MIP-1β and RANTES, and the CCR2 ligand MCP-1 were also strongly induced and persisted in cornea and TG during the latent phase. These and other recent results argue that HSV antigens or DNA can stimulate expression of chemokines, perhaps through activation of Toll-like receptors, for long periods of time at both primary and latent sites of HSV infection. These chemokines recruit activated T cells and other immune cells, including DC, that express chemokine receptors to primary and secondary sites of infection. Prolonged activation of chemokine expression could provide mechanistic explanations for certain aspects of HSV biology and pathogenesis.
Collapse
Affiliation(s)
- W James Cook
- Millennium Pharmaceuticals Inc., Cambridge, MA 02139, USA
- GlycoFi, Inc., 21 Lafayette Street, Suite 200, Lebanon, NH 03766, USA
| | - Martha F Kramer
- Department of Biological Chemistry and Molecular Pharmacology Harvard Medical School, Boston, MA 02115, USA
- Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, MA 02115, USA
| | | | | | - Holly A Holman
- Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Donald M Coen
- Department of Biological Chemistry and Molecular Pharmacology Harvard Medical School, Boston, MA 02115, USA
| | - David M Knipe
- Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|