1
|
Kasle G, Das Sarma J. The Role of Coronavirus Spike Protein in Inducing Optic Neuritis in Mice: Parallels to the SARS-CoV-2 Virus. J Neuroophthalmol 2024; 44:319-329. [PMID: 39164897 DOI: 10.1097/wno.0000000000002234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2024]
Abstract
BACKGROUND Optic neuritis (ON), one of the clinical manifestations of the human neurological disease multiple sclerosis (MS), was also reported in patients with COVID-19 infection, highlighting one potential neurological manifestation of SARS-CoV-2. However, the mechanism of ON in these patients is poorly understood. EVIDENCE ACQUISITION Insight may be gained by studying the neurotropic mouse hepatitis virus (MHV-A59), a β-coronavirus that belongs to the same family as SARS-CoV-2. RESULTS Mouse hepatitis virus-A59, or its isogenic spike protein recombinant strains, inoculation in mice provides an important experimental model to understand underpinning mechanisms of neuroinflammatory demyelination in association with acute stage optic nerve inflammation and chronic stage optic nerve demyelination concurrent with axonal loss. Spike is a surface protein that mediates viral binding and entry into host cells, as well as cell-cell fusion and viral spread. Studies have implicated spike-mediated mechanisms of virus-induced neuroinflammatory demyelination by comparing naturally occurring demyelinating (DM) and nondemyelinating (NDM) MHV strains. CONCLUSIONS Here, we summarize findings in MHV-induced experimental ON and myelitis, using natural DM and NDM strains as well as engineered recombinant strains of MHV to understand the role of spike protein in inducing ON and demyelinating disease pathology. Potential parallels in human coronavirus-mediated ON and demyelination, and insight into potential therapeutic strategies, are discussed.
Collapse
Affiliation(s)
- Grishma Kasle
- Department of Biological Sciences (GK, JDS), Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, India; and Department of Ophthalmology (JDS), University of Pennsylvania, Philadelphia, Pennsylvania
| | | |
Collapse
|
2
|
Hazra B, Das Sarma J. The CD40/CD40 ligand dyad and its downstream effector molecule ISG54 in relating acute neuroinflammation with persistent, progressive demyelination. IUBMB Life 2024; 76:313-331. [PMID: 38116887 DOI: 10.1002/iub.2798] [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: 02/08/2023] [Accepted: 11/14/2023] [Indexed: 12/21/2023]
Abstract
Although Multiple Sclerosis (MS) is primarily thought to be an autoimmune condition, its possible viral etiology must be taken into consideration. When mice are administered neurotropic viruses like mouse hepatitis virus MHV-A59, a murine coronavirus, or its isogenic recombinant strain RSA59, neuroinflammation along with demyelination are observed, which are some of the significant manifestations of MS. MHV-A59/RSA59 induced neuroinflammation is one of the best-studied experimental animal models to understand the viral-induced demyelination concurrent with axonal loss. In this experimental animal model, one of the major immune checkpoint regulators is the CD40-CD40L dyad, which helps in mediating both acute-innate, innate-adaptive, and chronic-adaptive immune responses. Hence, they are essential in reducing acute neuroinflammation and chronic progressive adaptive demyelination. While CD40 is expressed on antigen-presenting cells and endothelial cells, CD40L is expressed primarily on activated T cells and during severe inflammation on NK cells and mast cells. Experimental evidences revealed that genetic deficiency of both these proteins can lead to deleterious effects in an individual. On the other hand, interferon-stimulated genes (ISGs) possess potent antiviral properties and directly or indirectly alter acute neuroinflammation. In this review, we will discuss the role of an ISG, ISG54, and its tetratricopeptide repeat protein Ifit2; the genetic and experimental studies on the role of CD40 and CD40L in a virus-induced neuroinflammatory demyelination model.
Collapse
Affiliation(s)
- Bishal Hazra
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, India
| | - Jayasri Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, India
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
3
|
Balduz M, Fidancı H. Visual evoked potential abnormalities in patients with COVID-19. REVISTA DA ASSOCIACAO MEDICA BRASILEIRA (1992) 2024; 70:e20231061. [PMID: 38451579 PMCID: PMC10913780 DOI: 10.1590/1806-9282.20231061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 09/28/2023] [Indexed: 03/08/2024]
Abstract
OBJECTIVE It has been suggested that diseases that may cause visual evoked potential abnormality, such as optic neuritis, may be associated with the coronavirus disease 2019. This study aimed to find out whether there are visual evoked potential abnormalities in coronavirus disease 2019 patients using pattern reversal visual evoked potential and flash visual evoked potential. METHODS Patients with a history of coronavirus disease 2019 (coronavirus disease 2019 patients) and controls were included in this prospective case-control study. This study was conducted in the Clinical Neurophysiology Laboratory of Adana City Training and Research Hospital. Individuals without visual impairment were included. Coronavirus disease 2019 patients were required to have clinical features consistent with previous acute infection and a positive nose swab polymerase chain reaction test. Visual evoked potential was applied to coronavirus disease 2019 patients between July 2020 and July 2021. Controls consisted of patients without a history of chronic disease who underwent a visual evoked potential study between June 2017 and June 2018 due to headache or dizziness. Pattern reversal visual evoked potential and flash visual evoked potential were applied to all participants. N75, P100, and N135 waves obtained from pattern reversal visual evoked potential and P1, N1, P2, N2, P3, and N3 waves obtained from flash visual evoked potential were analyzed. RESULTS A total of 44 coronavirus disease 2019 patients and 40 controls were included in the study. Age and gender were not different between the two groups. Pattern reversal visual evoked potential parameters were not different between the two groups. Right P2 latency was 114.4±21.1 and 105.5±14.7 ms in coronavirus disease 2019 patients and controls, respectively (p=0.031). Patients with P100 and P2 wave abnormalities were 6 (13.6%) and 13 (29.6%), respectively. CONCLUSION This study showed that there may be visual evoked potential abnormalities in coronavirus disease 2019 patients.
Collapse
Affiliation(s)
- Metin Balduz
- Adana City Training and Research Hospital, Department of Neurology – Adana, Turkey
| | - Halit Fidancı
- Adana City Training and Research Hospital, Division of Clinical Neurophysiology, Department of Neurology – Adana, Turkey
| |
Collapse
|
4
|
Shi J, Danesh-Meyer HV. A review of neuro-ophthalmic sequelae following COVID-19 infection and vaccination. Front Cell Infect Microbiol 2024; 14:1345683. [PMID: 38299114 PMCID: PMC10827868 DOI: 10.3389/fcimb.2024.1345683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 01/03/2024] [Indexed: 02/02/2024] Open
Abstract
Background It has become increasingly clear that the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can affect most organs in the human body, including the neurologic and ophthalmic systems. Vaccination campaigns have been developed at rapid pace around the world to protect the population from the fast-mutating virus. This review seeks to summarise current knowledge of the neuro-ophthalmic manifestations of both COVID-19 infection and vaccination. Evidence acquisition Electronic searches for published literature were conducted using EMBASE and MEDLINE on the 30th of July 2023. The search strategy comprised of controlled vocabulary and free-text synonyms for the following terms in various combinations: "coronavirus, COVID-19, SARS-CoV-2, 2019-nCoV, vaccination, vaccine, immunisation and neuro-ophthalmology". No time range limits were set for the literature search. Published English abstracts for articles written in a different language were screened if available. Results A total of 54 case reports and case series were selected for use in the final report. 34 articles documenting neuro-ophthalmic manifestations following COVID-19 infection and 20 articles with neuro-ophthalmic complications following COVID-19 vaccination were included, comprising of 79 patients in total. The most commonly occurring condition was optic neuritis, with 25 cases following COVID-19 infection and 27 cases following vaccination against COVID-19. Conclusions The various COVID-19 vaccines that are currently available are part of the global effort to protect the most vulnerable of the human population. The incidence of neuro-ophthalmic consequences following infection with COVID-19 is hundred-folds higher and associated with more harrowing systemic effects than vaccination against the virus.
Collapse
Affiliation(s)
- Jane Shi
- Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Ophthalmology, Greenlane Clinical Centre, Te Whatu Ora – Health New Zealand, Auckland, New Zealand
| | - Helen V. Danesh-Meyer
- Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Ophthalmology, Greenlane Clinical Centre, Te Whatu Ora – Health New Zealand, Auckland, New Zealand
| |
Collapse
|
5
|
Curran CS, Cui X, Li Y, Jeakle M, Sun J, Demirkale CY, Minkove S, Hoffmann V, Dhamapurkar R, Chumbris S, Bolyard C, Iheanacho A, Eichacker PQ, Torabi-Parizi P. Anti-PD-L1 therapy altered inflammation but not survival in a lethal murine hepatitis virus-1 pneumonia model. Front Immunol 2024; 14:1308358. [PMID: 38259435 PMCID: PMC10801642 DOI: 10.3389/fimmu.2023.1308358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 12/05/2023] [Indexed: 01/24/2024] Open
Abstract
Introduction Because prior immune checkpoint inhibitor (ICI) therapy in cancer patients presenting with COVID-19 may affect outcomes, we investigated the beta-coronavirus, murine hepatitis virus (MHV)-1, in a lethal pneumonia model in the absence (Study 1) or presence of prior programmed cell death ligand-1 (PD-L1) antibody (PD-L1mAb) treatment (Study 2). Methods In Study 1, animals were inoculated intratracheally with MHV-1 or vehicle and evaluated at day 2, 5, and 10 after infection. In Study 2, uninfected or MHV-1-infected animals were pretreated intraperitoneally with control or PD-L1-blocking antibodies (PD-L1mAb) and evaluated at day 2 and 5 after infection. Each study examined survival, physiologic and histologic parameters, viral titers, lung immunophenotypes, and mediator production. Results Study 1 results recapitulated the pathogenesis of COVID-19 and revealed increased cell surface expression of checkpoint molecules (PD-L1, PD-1), higher expression of the immune activation marker angiotensin converting enzyme (ACE), but reduced detection of the MHV-1 receptor CD66a on immune cells in the lung, liver, and spleen. In addition to reduced detection of PD-L1 on all immune cells assayed, PD-L1 blockade was associated with increased cell surface expression of PD-1 and ACE, decreased cell surface detection of CD66a, and improved oxygen saturation despite reduced blood glucose levels and increased signs of tissue hypoxia. In the lung, PD-L1mAb promoted S100A9 but inhibited ACE2 production concomitantly with pAKT activation and reduced FOXO1 levels. PD-L1mAb promoted interferon-γ but inhibited IL-5 and granulocyte-macrophage colony-stimulating factor (GM-CSF) production, contributing to reduced bronchoalveolar lavage levels of eosinophils and neutrophils. In the liver, PD-L1mAb increased viral clearance in association with increased macrophage and lymphocyte recruitment and liver injury. PD-L1mAb increased the production of virally induced mediators of injury, angiogenesis, and neuronal activity that may play role in COVID-19 and ICI-related neurotoxicity. PD-L1mAb did not affect survival in this murine model. Discussion In Study 1 and Study 2, ACE was upregulated and CD66a and ACE2 were downregulated by either MHV-1 or PD-L1mAb. CD66a is not only the MHV-1 receptor but also an identified immune checkpoint and a negative regulator of ACE. Crosstalk between CD66a and PD-L1 or ACE/ACE2 may provide insight into ICI therapies. These networks may also play role in the increased production of S100A9 and neurological mediators in response to MHV-1 and/or PD-L1mAb, which warrant further study. Overall, these findings support observational data suggesting that prior ICI treatment does not alter survival in patients presenting with COVID-19.
Collapse
Affiliation(s)
- Colleen S. Curran
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Xizhong Cui
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Yan Li
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Mark Jeakle
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Junfeng Sun
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Cumhur Y. Demirkale
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Samuel Minkove
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Victoria Hoffmann
- Division of Veterinary Resources, National Institutes of Health, Bethesda, MD, United States
| | - Rhea Dhamapurkar
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Symya Chumbris
- Texcell North-America, Inc., Frederick, MD, United States
| | | | | | - Peter Q. Eichacker
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Parizad Torabi-Parizi
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
6
|
Kamble M, Saadi F, Kumar S, Saha B, Das Sarma J. Inducible nitric oxide synthase deficiency promotes murine-β-coronavirus induced demyelination. Virol J 2023; 20:51. [PMID: 36966345 PMCID: PMC10039690 DOI: 10.1186/s12985-023-02006-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 03/06/2023] [Indexed: 03/27/2023] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is characterized by neuroinflammation and demyelination orchestrated by activated neuroglial cells, CNS infiltrating leukocytes, and their reciprocal interactions through inflammatory signals. An inflammatory stimulus triggers inducible nitric oxide synthase (NOS2), a pro-inflammatory marker of microglia/macrophages (MG/Mφ) to catalyze sustained nitric oxide production. NOS2 during neuroinflammation, has been associated with MS disease pathology; however, studies dissecting its role in demyelination are limited. We studied the role of NOS2 in a recombinant β-coronavirus-MHV-RSA59 induced neuroinflammation, an experimental animal model mimicking the pathological hallmarks of MS: neuroinflammatory demyelination and axonal degeneration. OBJECTIVE Understanding the role of NOS2 in murine-β-coronavirus-MHV-RSA59 demyelination. METHODS Brain and spinal cords from mock and RSA59 infected 4-5-week-old MHV-free C57BL/6 mice (WT) and NOS2-/- mice were harvested at different disease phases post infection (p.i.) (day 5/6-acute, day 9/10-acute-adaptive and day 30-chronic phase) and compared for pathological outcomes. RESULTS NOS2 was upregulated at the acute phase of RSA59-induced disease in WT mice and its deficiency resulted in severe disease and reduced survival at the acute-adaptive transition phase. Low survival in NOS2-/- mice was attributed to (i) high neuroinflammation resulting from increased accumulation of macrophages and neutrophils and (ii) Iba1 + phagocytic MG/Mφ mediated-early demyelination as observed at this phase. The phagocytic phenotype of CNS MG/Mφ was confirmed by significantly higher mRNA transcripts of phagocyte markers-CD206, TREM2, and Arg1 and double immunolabelling of Iba1 with MBP and PLP. Further, NOS2 deficiency led to exacerbated demyelination at the chronic phase as well. CONCLUSION Taken together the results imply that the immune system failed to control the disease progression in the absence of NOS2. Thus, our observations highlight a protective role of NOS2 in murine-β-coronavirus induced demyelination.
Collapse
Affiliation(s)
- Mithila Kamble
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741246, India
| | - Fareeha Saadi
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741246, India
| | - Saurav Kumar
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741246, India
| | - Bhaskar Saha
- National Centre for Cell Science, Ganeshkhind, Pune, India
| | - Jayasri Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741246, India.
- Department of Ophthalmology, University of Pennsylvania Scheie Eye Institute, Philadelphia, PA, 19104, USA.
| |
Collapse
|
7
|
Safiriyu AA, Mulchandani V, Anakkacheri MN, Pal D, Das Sarma J. Proline-Proline Dyad in the Fusion Peptide of the Murine β-Coronavirus Spike Protein's S2 Domain Modulates Its Neuroglial Tropism. Viruses 2023; 15:215. [PMID: 36680255 PMCID: PMC9865228 DOI: 10.3390/v15010215] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 01/04/2023] [Accepted: 01/10/2023] [Indexed: 01/14/2023] Open
Abstract
The β-Coronavirus mouse hepatitis virus (MHV-A59)-RSA59 has a patent stretch of fusion peptide (FP) containing two consecutive central prolines (PP) in the S2 domain of the Spike protein. Our previous studies compared the PP-containing fusogenic-demyelinating strain RSA59(PP) to its one proline-deleted mutant strain RSA59(P) and one proline-containing non-fusogenic non-demyelinating parental strain RSMHV2(P) to its one proline inserted mutant strain RSMHV2(PP). These studies highlighted the crucial role of PP in fusogenicity, hepato-neuropathogenesis, and demyelination. Computational studies combined with biophysical data indicate that PP at the center of the FP provides local rigidity while imparting global fluctuation to the Spike protein that enhances the fusogenic properties of RSA59(PP) and RSMHV2(PP). To elaborate on the understanding of the role of PP in the FP of MHV, the differential neuroglial tropism of the PP and P mutant strains was investigated. Comparative studies demonstrated that PP significantly enhances the viral tropism for neurons, microglia, and oligodendrocytes. PP, however, is not essential for viral tropism for either astroglial or oligodendroglial precursors or the infection of meningeal fibroblasts in the blood-brain and blood-CSF barriers. PP in the fusion domain is critical for promoting gliopathy, making it a potential region for designing antivirals for neuro-COVID therapy.
Collapse
Affiliation(s)
- Abass Alao Safiriyu
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur 741246, India
| | - Vaishali Mulchandani
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur 741246, India
| | - Mohammed Nahaf Anakkacheri
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur 741246, India
| | - Debnath Pal
- Department of Computational and Data Sciences, Indian Institute of Science, Bengaluru 560012, India
| | - Jayasri Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur 741246, India
| |
Collapse
|
8
|
Kamalipour A, Ashraf MA, Moghimi S, Moattari A, Ashraf MJ, Abbasi F, Azodi F, Oboudi S, Pirbonyeh N, Mokhtaryan M, Roshanshad A, Do JL, Weinreb RN. Detection of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) RNA in the Human Eye. Ocul Immunol Inflamm 2023; 31:32-38. [PMID: 34637665 DOI: 10.1080/09273948.2021.1980810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
PURPOSE To determine the presence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) RNA in postmortem ocular specimens of patients with severe COVID-19 disease. PATIENTS AND METHODS Postmortem conjunctival (28 samples), aqueous humor (30 samples) and vitreous humor (30 samples) specimens were obtained bilaterally from the eyes of 15 deceased COVID-19 patients within one hour of death. The presence of viral RNA was evaluated in samples using Real-time reverse transcriptase-polymerase chain reaction (RT-PCR). RESULTS Positive RT-PCR SARS-COV-2 results were found in one conjunctival and 2 vitreous humor samples. All aqueous humor samples tested negative for the presence of SARS-COV-2 RNA. Of note, three positive samples were obtained from three different patients. The overall prevalence of positive RT-PCR ocular samples was 3.4% among all samples and 20% at the patient level. CONCLUSION SARS-CoV-2 RNA is detectable in postmortem conjunctival and vitreous humor samples of patients with severe COVID-19.
Collapse
Affiliation(s)
- Alireza Kamalipour
- Hamilton Glaucoma Center, Shiley Eye Institute, Viterbi Family Department of Ophthalmology, University of California, San Diego, California, USA
| | - Mohammad Ali Ashraf
- Poostchi Ophthalmology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sasan Moghimi
- Hamilton Glaucoma Center, Shiley Eye Institute, Viterbi Family Department of Ophthalmology, University of California, San Diego, California, USA
| | - Afagh Moattari
- Department of Virology and Bacteriology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.,Bioinformatics and Computational Biology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Javad Ashraf
- Department of Pathology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farhad Abbasi
- Department of Infectious Diseases, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Farzan Azodi
- Student Research Committee, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Shadi Oboudi
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Neda Pirbonyeh
- Department of Virology and Bacteriology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Microbiology, Burn and Wound Healing Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Mokhtaryan
- Department of Internal Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amirhossein Roshanshad
- Poostchi Ophthalmology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Jiun L Do
- Hamilton Glaucoma Center, Shiley Eye Institute, Viterbi Family Department of Ophthalmology, University of California, San Diego, California, USA
| | - Robert N Weinreb
- Hamilton Glaucoma Center, Shiley Eye Institute, Viterbi Family Department of Ophthalmology, University of California, San Diego, California, USA
| |
Collapse
|
9
|
Kanra AY, Altınel MG, Alparslan F. Evaluation of retinal and choroidal parameters as neurodegeneration biomarkers in patients with post-covid-19 syndrome. Photodiagnosis Photodyn Ther 2022; 40:103108. [PMID: 36075519 DOI: 10.1016/j.pdpdt.2022.103108] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/21/2022] [Accepted: 09/02/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND To investigate the neurodegenerative effects of post-COVID-19 syndrome. METHODS This comparative, cross-sectional study included patients who had post-COVID-19 prolonged neurologic symptoms. The control group was selected from volunteer participants with similar age, sex, and spherical equivalent characteristics. After detailed ophthalmic examinations, spectral-domain optic coherence tomography (SD-OCT) analysis of the macula and peripapillary retinal nerve fiber layer (m-RNFL, p-RNFL respectively) were obtained. We also evaluated the choroidal parameters with the ImageJ software. Post-COVID-19 symptoms and disease severity of the patients were also questioned. RESULTS Thirty-four eyes of 20 patients, and 39 eyes of 23 healthy individuals were included in the study. Thinning was found in the inner superior quadrant of mRNFL (p < 0.05). More prominent and common thinning of retinal layers was observed in the ganglion cell layer (GCL) and inner plexiform layer (IPL) segments in the patient group. There was no difference between the groups in the calculated choroidal parameters (p > 0.05). In the regression models created, lung involvement was the most prominent parameter associated with thinning of the OCT layers. The other factors were male sex, and the presence of loss of taste and smell also led to deterioration in some parameters. CONCLUSIONS We detected an overall thinning of the GCL and IPL layers with no significant change in CMT, pRNFL, and CVI in the post-COVID-19 period, indicating the direct or indirect effect of SARS-CoV-2 on these layers, rather than a long-term neurodegenerative effect.
Collapse
Affiliation(s)
- Ayşe Yağmur Kanra
- Department of Ophthalmology, Şişli Hamidiye Etfal Training and Research Hospital, Sarıyer, Istanbul, Turkey.
| | - Meltem Güzin Altınel
- Department of Ophthalmology, Fatih Sultan Mehmet Training and Research Hospital, Istanbul, Turkey
| | | |
Collapse
|
10
|
Ross AG, Chaqour B, McDougald DS, Dine KE, Duong TT, Shindler RE, Yue J, Liu T, Shindler KS. Selective Upregulation of SIRT1 Expression in Retinal Ganglion Cells by AAV-Mediated Gene Delivery Increases Neuronal Cell Survival and Alleviates Axon Demyelination Associated with Optic Neuritis. Biomolecules 2022; 12:830. [PMID: 35740955 PMCID: PMC9221096 DOI: 10.3390/biom12060830] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 06/07/2022] [Accepted: 06/11/2022] [Indexed: 11/16/2022] Open
Abstract
Optic neuritis (ON), the most common ocular manifestation of multiple sclerosis, is an autoimmune inflammatory demyelinating disease also characterized by degeneration of retinal ganglion cells (RGCs) and their axons, which commonly leads to visual impairment despite attempted treatments. Although ON disease etiology is not known, changes in the redox system and exacerbated optic nerve inflammation play a major role in the pathogenesis of the disease. Silent information regulator 1 (sirtuin-1/SIRT1) is a ubiquitously expressed NAD+-dependent deacetylase, which functions to reduce/prevent both oxidative stress and inflammation in various tissues. Non-specific upregulation of SIRT1 by pharmacologic and genetic approaches attenuates RGC loss in experimental ON. Herein, we hypothesized that targeted expression of SIRT1 selectively in RGCs using an adeno-associated virus (AAV) vector as a delivery vehicle is an effective approach to reducing neurodegeneration and preserving vision in ON. We tested this hypothesis through intravitreal injection of AAV7m8.SNCG.SIRT1, an AAV2-derived vector optimized for highly efficient SIRT1 transgene transfer and protein expression into RGCs in mice with experimental autoimmune encephalomyelitis (EAE), a model of multiple sclerosis that recapitulates optic neuritis RGC loss and axon demyelination. Our data show that EAE mice injected with a control vehicle exhibit progressive alteration of visual function reflected by decreasing optokinetic response (OKR) scores, whereas comparatively, AAV7m8.SNCG.SIRT1-injected EAE mice maintain higher OKR scores, suggesting that SIRT1 reduces the visual deficit imparted by EAE. Consistent with this, RGC survival determined by immunolabeling is increased and axon demyelination is decreased in the AAV7m8.SNCG.SIRT1 RGC-injected group of EAE mice compared to the mouse EAE counterpart injected with a vehicle or with control vector AAV7m8.SNCG.eGFP. However, immune cell infiltration of the optic nerve is not significantly different among all EAE groups of mice injected with either vehicle or AAV7m8.SNCG.SIRT1. We conclude that despite minimally affecting the inflammatory response in the optic nerve, AAV7m8-mediated SIRT1 transfer into RGCs has a neuroprotective potential against RGC loss, axon demyelination and vison deficits associated with EAE. Together, these data suggest that SIRT1 exerts direct effects on RGC survival and function.
Collapse
Affiliation(s)
- Ahmara G. Ross
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA 19104, USA; (A.G.R.); (B.C.); (D.S.M.); (K.E.D.); (T.T.D.); (R.E.S.); (J.Y.); (T.L.)
- F. M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
- Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Brahim Chaqour
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA 19104, USA; (A.G.R.); (B.C.); (D.S.M.); (K.E.D.); (T.T.D.); (R.E.S.); (J.Y.); (T.L.)
- F. M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Devin S. McDougald
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA 19104, USA; (A.G.R.); (B.C.); (D.S.M.); (K.E.D.); (T.T.D.); (R.E.S.); (J.Y.); (T.L.)
- F. M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Kimberly E. Dine
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA 19104, USA; (A.G.R.); (B.C.); (D.S.M.); (K.E.D.); (T.T.D.); (R.E.S.); (J.Y.); (T.L.)
- F. M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Thu T. Duong
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA 19104, USA; (A.G.R.); (B.C.); (D.S.M.); (K.E.D.); (T.T.D.); (R.E.S.); (J.Y.); (T.L.)
- F. M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Ryan E. Shindler
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA 19104, USA; (A.G.R.); (B.C.); (D.S.M.); (K.E.D.); (T.T.D.); (R.E.S.); (J.Y.); (T.L.)
- F. M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Jipeng Yue
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA 19104, USA; (A.G.R.); (B.C.); (D.S.M.); (K.E.D.); (T.T.D.); (R.E.S.); (J.Y.); (T.L.)
- F. M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Tehui Liu
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA 19104, USA; (A.G.R.); (B.C.); (D.S.M.); (K.E.D.); (T.T.D.); (R.E.S.); (J.Y.); (T.L.)
- F. M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Kenneth S. Shindler
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA 19104, USA; (A.G.R.); (B.C.); (D.S.M.); (K.E.D.); (T.T.D.); (R.E.S.); (J.Y.); (T.L.)
- F. M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
- Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
11
|
Safiriyu AA, Singh M, Kishore A, Mulchandani V, Maity D, Behera A, Sinha B, Pal D, Das Sarma J. Two Consecutive Prolines in the Fusion Peptide of Murine β-Coronavirus Spike Protein Predominantly Determine Fusogenicity and May Be Essential but Not Sufficient to Cause Demyelination. Viruses 2022; 14:v14040834. [PMID: 35458565 PMCID: PMC9031231 DOI: 10.3390/v14040834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/13/2022] [Accepted: 04/14/2022] [Indexed: 11/16/2022] Open
Abstract
Combined in silico, in vitro, and in vivo comparative studies between isogenic-recombinant Mouse-Hepatitis-Virus-RSA59 and its proline deletion mutant, revealed a remarkable contribution of centrally located two consecutive prolines (PP) from Spike protein fusion peptide (FP) in enhancing virus fusogenic and hepato-neuropathogenic potential. To deepen our understanding of the underlying factors, we extend our studies to a non-fusogenic parental virus strain RSMHV2 (P) with a single proline in the FP and its proline inserted mutant, RSMHV2 (PP). Comparative in vitro and in vivo studies between virus strains RSA59(PP), RSMHV2 (P), and RSMHV2 (PP) in the FP demonstrate that the insertion of one proline significantly resulted in enhancing the virus fusogenicity, spread, and consecutive neuropathogenesis. Computational studies suggest that the central PP in Spike FP induces a locally ordered, compact, and rigid structure of the Spike protein in RSMHV2 (PP) compared to RSMHV2 (P), but globally the Spike S2-domain is akin to the parental strain RSA59(PP), the latter being the most flexible showing two potential wells in the energy landscape as observed from the molecular dynamics studies. The critical location of two central prolines of the FP is essential for fusogenicity and pathogenesis making it a potential site for designing antiviral.
Collapse
Affiliation(s)
- Abass Alao Safiriyu
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur 741246, India; (A.A.S.); (M.S.); (A.K.); (V.M.); (A.B.); (B.S.)
| | - Manmeet Singh
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur 741246, India; (A.A.S.); (M.S.); (A.K.); (V.M.); (A.B.); (B.S.)
| | - Abhinoy Kishore
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur 741246, India; (A.A.S.); (M.S.); (A.K.); (V.M.); (A.B.); (B.S.)
| | - Vaishali Mulchandani
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur 741246, India; (A.A.S.); (M.S.); (A.K.); (V.M.); (A.B.); (B.S.)
| | - Dibyajyoti Maity
- Department of Computational and Data Sciences, Indian Institute of Science, Bengaluru 560012, India; (D.M.); (D.P.)
| | - Amrutamaya Behera
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur 741246, India; (A.A.S.); (M.S.); (A.K.); (V.M.); (A.B.); (B.S.)
| | - Bidisha Sinha
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur 741246, India; (A.A.S.); (M.S.); (A.K.); (V.M.); (A.B.); (B.S.)
| | - Debnath Pal
- Department of Computational and Data Sciences, Indian Institute of Science, Bengaluru 560012, India; (D.M.); (D.P.)
| | - Jayasri Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur 741246, India; (A.A.S.); (M.S.); (A.K.); (V.M.); (A.B.); (B.S.)
- Correspondence:
| |
Collapse
|
12
|
Sengupta S, Addya S, Biswas D, Banerjee P, Sarma JD. Matrix metalloproteinases and tissue inhibitors of metalloproteinases in murine β-coronavirus-induced neuroinflammation. Virology 2022; 566:122-135. [PMID: 34906793 PMCID: PMC8648396 DOI: 10.1016/j.virol.2021.11.012] [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: 07/05/2021] [Revised: 11/16/2021] [Accepted: 11/26/2021] [Indexed: 01/17/2023]
Abstract
Mouse hepatitis virus (MHV; m-β-CoV) serves as a useful model for studying the cellular factors involved in neuroinflammation. To understand the role of matrix metalloproteinases (MMPs) in neuroinflammation, brain tissues from m-β-CoV-infected mice were harvested at different days post-infection (d.p.i) and investigated for Mmp expression by RT-qPCR. Mmp-2, -3, -8, -12 showed significant mRNA upregulation peaking with viral replication between 5 and 6 d.p.i. Elevated levels of MMP regulator TIMP-1 are suggestive of a TIMP-1 mediated host antiviral response. Biological network assessment suggested a direct involvement of MMP-3, -8, -14 in facilitating peripheral leukocyte infiltrations. Flow cytometry confirmed the increased presence of NK cells, CD4+ and CD8+ T cells, neutrophils, and MHCII expressing cells in the m-β-CoV infected mice brain. Our study revealed that m-β-CoV upregulated Park7, RelA, Nrf2, and Hmox1 transcripts involved in ROS production and antioxidant pathways, describing the possible nexus between oxidative pathways, MMPs, and TIMP in m-β-CoV-induced neuroinflammation.
Collapse
Affiliation(s)
- Sourodip Sengupta
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata (IISER-K), Mohanpur, India
| | - Sankar Addya
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, USA
| | - Diptomit Biswas
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata (IISER-K), Mohanpur, India
| | - Paromita Banerjee
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata (IISER-K), Mohanpur, India
| | - Jayasri Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata (IISER-K), Mohanpur, India,Corresponding author
| |
Collapse
|
13
|
Saadi F, Chakravarty D, Kumar S, Kamble M, Saha B, Shindler KS, Das Sarma J. CD40L protects against mouse hepatitis virus-induced neuroinflammatory demyelination. PLoS Pathog 2021; 17:e1010059. [PMID: 34898656 PMCID: PMC8699621 DOI: 10.1371/journal.ppat.1010059] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 12/23/2021] [Accepted: 10/23/2021] [Indexed: 11/19/2022] Open
Abstract
Neurotropic mouse hepatitis virus (MHV-A59/RSA59) infection in mice induces acute neuroinflammation due to direct neural cell dystrophy, which proceeds with demyelination with or without axonal loss, the pathological hallmarks of human neurological disease, Multiple sclerosis (MS). Recent studies in the RSA59-induced neuroinflammation model of MS showed a protective role of CNS-infiltrating CD4+ T cells compared to their pathogenic role in the autoimmune model. The current study further investigated the molecular nexus between CD4+ T cell-expressed CD40Ligand and microglia/macrophage-expressed CD40 using CD40L-/- mice. Results demonstrate CD40L expression in the CNS is modulated upon RSA59 infection. We show evidence that CD40L-/- mice are more susceptible to RSA59 induced disease due to reduced microglia/macrophage activation and significantly dampened effector CD4+ T recruitment to the CNS on day 10 p.i. Additionally, CD40L-/- mice exhibited severe demyelination mediated by phagocytic microglia/macrophages, axonal loss, and persistent poliomyelitis during chronic infection, indicating CD40-CD40L as host-protective against RSA59-induced demyelination. This suggests a novel target in designing prophylaxis for virus-induced demyelination and axonal degeneration, in contrast to immunosuppression which holds only for autoimmune mechanisms of inflammatory demyelination. MS is primarily considered an autoimmune CNS disease, but its potential viral etiology cannot be ignored. Myelin-specific CD40L+CD4+ T cells migration into the CNS and resultant neuroinflammation is considered pathogenic in autoimmune MS. In contrast, CD40L+CD4+ T infiltration into the MHV-induced inflamed CNS and their interaction with CD40+ microglia/macrophages are shown to be protective in our study. Considering differential etiology but comparable demyelination and axonal loss, immunosuppressive treatments may not necessarily ameliorate MS in all patients. MHV-induced demyelination in this study indicates that the interaction between CD40L on CD4+T cells and CD40 on microglia/macrophage plays an important protective role against MHV-induced chronic progressive demyelination.
Collapse
Affiliation(s)
- Fareeha Saadi
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, India
| | - Debanjana Chakravarty
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, India
| | - Saurav Kumar
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, India
| | - Mithila Kamble
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, India
| | - Bhaskar Saha
- National Centre for Cell Science, Ganeshkhind, Pune, India
| | - Kenneth S. Shindler
- Departments of Ophthalmology and
- Neurology University of Pennsylvania Scheie Eye Institute, Philadelphia, Pennsylvania, United States of America
| | - Jayasri Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, India
- Departments of Ophthalmology and
- * E-mail:
| |
Collapse
|
14
|
Kundu S, Saadi F, Sengupta S, Antony GR, Raveendran VA, Kumar R, Kamble MA, Sarkar L, Burrows A, Pal D, Sen GC, Sarma JD. DJ-1-Nrf2 axis is activated upon murine β-coronavirus infection in the CNS. BRAIN DISORDERS 2021; 4:100021. [PMID: 34514445 PMCID: PMC8418700 DOI: 10.1016/j.dscb.2021.100021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 07/03/2021] [Accepted: 08/10/2021] [Indexed: 12/12/2022] Open
Abstract
Coronaviruses have emerged as alarming pathogens owing to their inherent ability of genetic variation and cross-species transmission. Coronavirus infection burdens the endoplasmic reticulum (ER.), causes reactive oxygen species production and induces host stress responses, including unfolded protein response (UPR) and antioxidant system. In this study, we have employed a neurotropic murine β-coronavirus (M-CoV) infection in the Central Nervous System (CNS) of experimental mice model to study the role of host stress responses mediated by interplay of DJ-1 and XBP1. DJ-1 is an antioxidant molecule with established functions in neurodegeneration. However, its regulation in virus-induced cellular stress response is less explored. Our study showed that M-CoV infection activated the glial cells and induced antioxidant and UPR genes during the acute stage when the viral titer peaks. As the virus particles decreased and acute neuroinflammation diminished at day ten p.i., a significant up-regulation in UPR responsive XBP1, antioxidant DJ-1, and downstream signaling molecules, including Nrf2, was recorded in the brain tissues. Additionally, preliminary in silico analysis of the binding between the DJ-1 promoter and a positively charged groove of XBP1 is also investigated, thus hinting at a mechanism behind the upregulation of DJ-1 during MHV-infection. The current study thus attempts to elucidate a novel interplay between the antioxidant system and UPR in the outcome of coronavirus infection.
Collapse
Affiliation(s)
- Soumya Kundu
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, West Bengal, India
| | - Fareeha Saadi
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, West Bengal, India
| | - Sourodip Sengupta
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, West Bengal, India
| | - Gisha Rose Antony
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, West Bengal, India
| | - Vineeth A Raveendran
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, West Bengal, India
| | - Rahul Kumar
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, West Bengal, India
| | - Mithila Ashok Kamble
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, West Bengal, India
| | - Lucky Sarkar
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, West Bengal, India
| | - Amy Burrows
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Ohio, USA
| | - Debnath Pal
- Department of Computational and Data Sciences, Indian Institute of Science, Bangalore, India
| | - Ganes C Sen
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Ohio, USA
| | - Jayasri Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, West Bengal, India
| |
Collapse
|
15
|
Ali L, Naeem M, Canibano B, John A, Iqrar A. Bilateral Acute Optic Perineuritis Associated With COVID-19 in a Patient With Seronegative Myelin Oligodendrocyte Glycoprotein (MOG) Antibody. Cureus 2021; 13:e18234. [PMID: 34712522 PMCID: PMC8542256 DOI: 10.7759/cureus.18234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/23/2021] [Indexed: 11/30/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-Cov-2) may cause various neuro-ophthalmologic manifestations including optic perineuritis. Optic perineuritis is a rare form of orbital inflammatory disease in which optic nerve sheath is inflamed and nonspecific fibrotic thickening with classic radiological finding is a perineural enhancement of optic nerve sheath. A 45-year-old gentleman with known diabetes mellitus, hypertension and dyslipidemia was admitted with a critically ill COVID-19 infection. During the recovery period, the patient developed sudden onset of painless loss of vision. MRI head and orbit with gadolinium was suggestive of optic perineuritis. Other secondary causes of autoimmune or vasculitis myelin oligodendrocyte glycoprotein (MOG) antibody disease and other common central nervous system (CNS) infection were excluded. The patient had dramatic response with steroids. This is the first rare case report of COVID-19-related optic perineuritis in critically ill COVID-19 patients with seronegative MOG antibody. Optic perineuritis is a rare orbital inflammatory disease and underlying mechanisms may arise from systemic response to COVID-19 infection as well as direct effects of the virus via angiotensin-converting enzyme 2 (ACE-2) receptors on ocular tissues. Optic perineuritis is a rare disease with inflammation restricted to the optic nerve sheath. Neuroimaging of the brain and orbit is the most important modality of choice for visualizing optic nerve sheath and optic nerve. Delay in the diagnosis of COVID-19-related optic perineuritis, may result in permanent optic nerve injury and irreversible vision loss.
Collapse
Affiliation(s)
- Liaquat Ali
- Neurology, Hamad General Hospital, Doha, QAT.,Neurology, Weill Cornell Medicine-Qatar, Doha, QAT
| | | | | | - Anju John
- Internal Medicine, Hamad General Hospital, Doha, QAT
| | | |
Collapse
|
16
|
Simões JLB, de Araújo JB, Bagatini MD. Anti-inflammatory Therapy by Cholinergic and Purinergic Modulation in Multiple Sclerosis Associated with SARS-CoV-2 Infection. Mol Neurobiol 2021; 58:5090-5111. [PMID: 34247339 PMCID: PMC8272687 DOI: 10.1007/s12035-021-02464-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 06/17/2021] [Indexed: 02/07/2023]
Abstract
The virus "acute respiratory syndrome coronavirus 2" (SARS-CoV-2) is the etiologic agent of coronavirus disease 2019 (COVID-19), initially responsible for an outbreak of pneumonia in Wuhan, China, which, due to the high level of contagion and dissemination, has become a pandemic. The clinical picture varies from mild to critical cases; however, all of these signs already show neurological problems, from sensory loss to neurological diseases. Thus, patients with multiple sclerosis (MS) infected with the new coronavirus are more likely to develop severe conditions; in addition to worsening the disease, this is due to the high level of pro-inflammatory cytokines, which is closely associated with increased mortality both in COVID-19 and MS. This increase is uncontrolled and exaggerated, characterizing the cytokine storm, so a possible therapy for this neuronal inflammation is the modulation of the cholinergic anti-inflammatory pathway, since acetylcholine (ACh) acts to reduce pro-inflammatory cytokines and acts directly on the brain for being released by cholinergic neurons, as well as acting on other cells such as immune and blood cells. In addition, due to tissue damage, there is an exacerbated release of adenosine triphosphate (ATP), potentiating the inflammatory process and activating purinergic receptors which act directly on neuroinflammation and positively modulate the inflammatory cycle. Associated with this, in neurological pathologies, there is greater expression of P2X7 in the cells of the microglia, which positively activates the immune inflammatory response. Thus, the administration of blockers of this receptor can act in conjunction with the action of ACh in the anticholinergic inflammatory pathway. Finally, there will be a reduction in the cytokine storm and triggered hyperinflammation, as well as the level of mortality in patients with multiple sclerosis infected with SARS-CoV-2 and the development of possible neurological damage.
Collapse
|
17
|
Hohberger B, Ganslmayer M, Lucio M, Kruse F, Hoffmanns J, Moritz M, Rogge L, Heltmann F, Szewczykowski C, Fürst J, Raftis M, Bergua A, Zenkel M, Gießl A, Schlötzer-Schrehardt U, Lehmann P, Strauß R, Mardin C, Herrmann M. Retinal Microcirculation as a Correlate of a Systemic Capillary Impairment After Severe Acute Respiratory Syndrome Coronavirus 2 Infection. Front Med (Lausanne) 2021; 8:676554. [PMID: 34307408 PMCID: PMC8299003 DOI: 10.3389/fmed.2021.676554] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 06/08/2021] [Indexed: 12/14/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which causes coronavirus disease 2019 (COVID-19), affects the pulmonary systems via angiotensin-converting enzyme-2 (ACE-2) receptor, being an entry to systemic infection. As COVID-19 disease features ACE-2 deficiency, a link to microcirculation is proposed. Optical coherence tomography angiography (OCT-A) enables non-invasive analysis of retinal microvasculature. Thus, an impaired systemic microcirculation might be mapped on retinal capillary system. As recent OCT-A studies, analyzing microcirculation in two subdivided layers, yielded contrary results, an increased subdivision of retinal microvasculature might offer an even more fine analysis. The aim of the study was to investigate retinal microcirculation by OCT-A after COVID-19 infection in three subdivided layers (I). In addition, short-term retinal affections were monitored during COVID-19 disease (II). Considering (I), a prospective study (33 patientspost-COVID and 28 controls) was done. Macula and peripapillary vessel density (VD) were scanned with the Spectralis II. Macula VD was measured in three layers: superficial vascular plexus (SVP), intermediate capillary plexus (ICP), and deep capillary plexus (DCP). Analysis was done by the EA-Tool, including an Anatomical Positioning System and an analysis of peripapillary VD by implementing Bruch's membrane opening (BMO) landmarks. Overall, circular (c1, c2, and c3) and sectorial VD (s1-s12) was analyzed. Considering (II), in a retrospective study, 29 patients with severe complications of COVID-19 infection, hospitalized at the intensive care unit, were monitored for retinal findings at bedside during hospitalization. (I) Overall (p = 0.0133) and circular (c1, p = 0.00257; c2, p = 0.0067; and c3, p = 0.0345). VD of the ICP was significantly reduced between patientspost-COVID and controls, respectively. Overall (p = 0.0179) and circular (c1, p = 0.0189) peripapillary VD was significantly reduced between both groups. Subgroup analysis of hospitalized vs. non-hospitalized patientspost-COVID yielded a significantly reduced VD of adjacent layers (DCP and SVP) with increased severity of COVID-19 disease. Clinical severity parameters showed a negative correlation with VD (ICP) and peripapillary VD. (II) Funduscopy yielded retinal hemorrhages and cotton wool spots in 17% of patients during SARS-CoV-2 infection. As VD of the ICP and peripapillary regions was significantly reduced after COVID-19 disease and showed a link to clinical severity markers, we assume that the severity of capillary impairment after COVID-19 infection is mapped on retinal microcirculation, visualized by non-invasive OCT-A.
Collapse
Affiliation(s)
- Bettina Hohberger
- Department of Ophthalmology, University of Erlangen-Nürnberg, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Marion Ganslmayer
- Department of Internal Medicine 1, University of Erlangen-Nürnberg, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Marianna Lucio
- Research Unit Analytical BioGeoChemistry, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Friedrich Kruse
- Department of Ophthalmology, University of Erlangen-Nürnberg, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Jakob Hoffmanns
- Department of Ophthalmology, University of Erlangen-Nürnberg, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Michael Moritz
- Department of Ophthalmology, University of Erlangen-Nürnberg, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Lennart Rogge
- Department of Ophthalmology, University of Erlangen-Nürnberg, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Felix Heltmann
- Department of Ophthalmology, University of Erlangen-Nürnberg, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Charlotte Szewczykowski
- Department of Ophthalmology, University of Erlangen-Nürnberg, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Julia Fürst
- Department of Internal Medicine 1, University of Erlangen-Nürnberg, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Maximilian Raftis
- Department of Ophthalmology, University of Erlangen-Nürnberg, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Antonio Bergua
- Department of Ophthalmology, University of Erlangen-Nürnberg, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Matthias Zenkel
- Department of Ophthalmology, University of Erlangen-Nürnberg, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Andreas Gießl
- Department of Ophthalmology, University of Erlangen-Nürnberg, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Ursula Schlötzer-Schrehardt
- Department of Ophthalmology, University of Erlangen-Nürnberg, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Paul Lehmann
- Department of Ophthalmology, University of Erlangen-Nürnberg, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Richard Strauß
- Department of Internal Medicine 1, University of Erlangen-Nürnberg, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Christian Mardin
- Department of Ophthalmology, University of Erlangen-Nürnberg, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Martin Herrmann
- Department of Internal Medicine 3, University of Erlangen-Nürnberg, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
18
|
Deane K, Sarfraz A, Sarfraz Z, Valentine D, Idowu AR, Sanchez V. Unilateral Optic Neuritis Associated with SARS-CoV-2 Infection: A Rare Complication. AMERICAN JOURNAL OF CASE REPORTS 2021; 22:e931665. [PMID: 34120138 PMCID: PMC8212842 DOI: 10.12659/ajcr.931665] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Patient: Female, 21-year-old Final Diagnosis: Optic neuritis Symptoms: Vision changes Medication:— Clinical Procedure: — Specialty: Infectious Diseases • Neurology
Collapse
Affiliation(s)
- Kitson Deane
- Department of Medicine, Woodhull Medical Center, New York City, NY, USA
| | - Azza Sarfraz
- Division of Women and Child Health, Aga Khan University, Karachi, Pakistan
| | - Zouina Sarfraz
- Department of Medicine, Fatima Jinnah Medical University, Lahore, Pakistan
| | | | | | - Vincent Sanchez
- Department of Medicine, St. George's University School of Medicine, St. Georges, Grenada
| |
Collapse
|
19
|
Žorić L, Rajović-Mrkić I, Čolak E, Mirić D, Kisić B. Optic Neuritis in a Patient with Seropositive Myelin Oligodendrocyte Glycoprotein Antibody During the Post-COVID-19 Period. Int Med Case Rep J 2021; 14:349-355. [PMID: 34079389 PMCID: PMC8165557 DOI: 10.2147/imcrj.s315103] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 05/10/2021] [Indexed: 12/25/2022] Open
Abstract
Reports of neuro-ophthalmological manifestations and complications in patients with coronavirus disease 19 (COVID-19) are still scarce. The aim of this article is to present optic neuritis, as possible post-infectious manifestation of COVID-19. Four weeks after hospitalization for seropositive coronavirus disease 19 (COVID-19), presented as bilateral bronchopneumonia, with radiology and laboratory findings also pointed to high clinical suspicion to COVID-19, a 63-year-old man developed a headache and subacute and profound visual loss on his right eye. The disease presentation was the right eye papillitis. Inflammatory parameters were normal at the time of hospitalization, and IgM and IgG for SARS-CoV-2 were still positive. After corticosteroid pulse therapy, his vision improved significantly and on follow-up visits returned to normal. All laboratory and radiology findings were unremarkable, except for antibodies against SARS-CoV-2 and myelin oligodendrocyte glycoprotein (MOG). We discuss about capacity of SARS-CoV-2 to cause optic neuritis and possible significance of MOG antibodies in similar cases.
Collapse
Affiliation(s)
- Lepša Žorić
- Faculty of Medicine, University of Pristina Settled in Kosovska Mitrovica (UPKM), Kosovska Mitrovica, Serbia.,Institute of Ophthalmology, University Clinical Center of Serbia, Belgrade, Serbia
| | - Isidora Rajović-Mrkić
- Ophthalmology Department, Clinical Center of Kosovska Mitrovica, Kosovska Mitrovica, Serbia
| | - Emina Čolak
- Institute of Medical Biochemistry, Clinical Center of Serbia, Belgrade, Serbia
| | - Dijana Mirić
- Institute of Medical Biochemistry, Faculty of Medicine, University of Priština Settled in Kosovska Mitrovica (UPKM), Kosovska Mitrovica, Serbia
| | - Bojana Kisić
- Institute of Medical Biochemistry, Faculty of Medicine, University of Priština Settled in Kosovska Mitrovica (UPKM), Kosovska Mitrovica, Serbia
| |
Collapse
|
20
|
Tisdale AK, Dinkin M, Chwalisz BK. Afferent and Efferent Neuro-Ophthalmic Complications of Coronavirus Disease 19. J Neuroophthalmol 2021; 41:154-165. [PMID: 33935220 DOI: 10.1097/wno.0000000000001276] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
PURPOSE To provide a summary of the neuro-ophthalmic manifestations of coronavirus disease 19 (COVID-19) documented in the literature thus far. METHODS The PubMed and Google Scholar databases were searched using the keywords: Neuro-Ophthalmology, COVID-19, SARS-CoV-2, and coronavirus. A manual search through reference lists of relevant articles was also performed. RESULTS/CONCLUSIONS The literature on COVID-associated neuro-ophthalmic disease continues to grow. Afferent neuro-ophthalmic complications associated with COVID-19 include optic neuritis, papillophlebitis, papilledema, visual disturbance associated with posterior reversible encephalopathy syndrome, and vision loss caused by stroke. Efferent neuro-ophthalmic complications associated with COVID-19 include cranial neuropathies, Miller Fisher syndrome, Adie's pupils, ocular myasthenia gravis, nystagmus and eye movement disorders. Proposed mechanisms of neurologic disease include immunologic upregulation, vasodilation and vascular permeability, endothelial dysfunction, coagulopathy, and direct viral neurotropism. When patients present to medical centers with new onset neuro-ophthalmic conditions during the pandemic, COVID-19 infection should be kept on the differential.
Collapse
Affiliation(s)
- Alanna K Tisdale
- Department of Ophthalmology and Visual Sciences (AKT), University of Iowa Hospital and Clinics, University of Iowa Carver College of Medicine, Iowa City, Iowa; Departments of Ophthalmology and Neurology (MD), New York- Presbyterian Hospital, Weill Cornell Medical College, New York City, New York; Department of Ophthalmology (BKC), Massachusetts Eye & Ear; and Department of Neurology (BKC), Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | | | | |
Collapse
|
21
|
Oren B, Aksoy Aydemır G, Aydemır E, Atesoglu HI, Goker YS, Kızıltoprak H, Ozcelık KC. Quantitative assessment of retinal changes in COVID-19 patients. Clin Exp Optom 2021; 104:717-722. [PMID: 34016010 DOI: 10.1080/08164622.2021.1916389] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
CLINICAL RELEVANCE The SARS-COV 2 virus, which is responsible for the COVID-19 pandemic, acts on the angiotensin converting enzyme 2 (ACE-2) receptor in the host cell. Ocular effects may occur because of the ACE-2 receptor in the retina. BACKGROUND To investigate the impact of COVID-19 on the retinal layers and optic disc parameters in previously confirmed COVID-19 patients using spectral domain optical coherence tomography (SD-OCT). METHODS This study included 60 eyes of 60 subjects; 35 of them were in the COVID-19 group and the remaining 25 were in the control group. Patients with the diagnosis of COVID-19 that had a negative result after treatment were included in the study. Macular and peripapillary retinal nerve fiber layer (RNFL) thickness measurements, each retinal layer thickness of all participants were done 14-30 days after COVID-19 symptom onset, following the negative result of real time reverse transcriptase-polymerase chain reaction test using SD-OCT. RESULTS The mean value of central macular thickness was significantly higher in the COVID-19 group than the control group (p = 0.02). The mean values of the ganglion cell layer and inner nuclear layer thickness in the COVID-19 group were significantly thinner than control group (p = 0.04 and p = 0.04, respectively). Even though mean RNFL thickness measurements in all sections in the COVID-19 group was thinner than controls, there were no significant differences between groups (p > 0.05 for all). CONCLUSION In the early recovery phase, changes in the macula, ganglion cell layer and inner nuclear layer could be seen. These patients should be followed up closely for the recognition of new pathologies that could be seen in the late recovery phase.
Collapse
Affiliation(s)
- Burak Oren
- Ophthalmology Department, Adıyaman University Training and Research Hospital, Adıyaman, Turkey
| | - Gozde Aksoy Aydemır
- Ophthalmology Department, Adıyaman University Training and Research Hospital, Adıyaman, Turkey
| | - Emre Aydemır
- Ophthalmology Department, Adıyaman University Training and Research Hospital, Adıyaman, Turkey
| | | | - Yasin Sakir Goker
- Ophthalmology Department, Ulucanlar Eye Training and Research Hospital, Ankara, Turkey
| | - Hasan Kızıltoprak
- Ophthalmology Department, Adıyaman University Training and Research Hospital, Adıyaman, Turkey
| | | |
Collapse
|
22
|
Bypareddy R, Rathod BLS, Shilpa YD, Hithashree HR, Nagaraj KB, Hemalatha BC, Basumatary J, Bekal D, Niranjan R, Anusha PG. Fundus evaluation in COVID-19 positives with non-severe disease. Indian J Ophthalmol 2021; 69:1271-1274. [PMID: 33913875 PMCID: PMC8186568 DOI: 10.4103/ijo.ijo_3227_20] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Purpose: The aim of this work was to study and document retinal changes in coronavirus disease-2019 (COVID-19) positive patients with nonsevere disease using a nonmydriatic handheld fundus camera. Methods: A cross-sectional observational study was conducted on patients affected by COVID-19 who were admitted at our center. Our study included patients with no, mild, and moderate symptoms (nonsevere cases). Intensive care unit (ICU)-admitted patients were excluded considering the difficulty in procuring the fundus image by the handheld camera due to patients positioning. Patients with systemic conditions (diabetes, hypertension, and severe anemia) known to cause retinopathy were also excluded. Bedside anterior segment examination, fundus examination using indirect ophthalmoscopy and fundus imaging of each patient using a nonmydriatic handheld fundus camera was done by a trained ophthalmologist posted for COVID duty. Results: In a cohort of 138 patients, 94 (68.1%) were men and 44 (31.9%) were women. A total of 276 eyes were evaluated. The mean age of the patients was 38.51 ± 14.4 years. Anterior segment evaluation showed no abnormality in any of the eyes. On fundus screening using nonmydriatic handheld camera, a single streak of superficial retinal hemorrhage was noted at the posterior pole of the fundus in the left eye of one patient (0.72%), which was away from fovea. Laboratory tests revealed low hemoglobin (between 10 and 10.9 g/dL falling under mild Anemia) in 12 patients, elevated total leucocyte count in 6 patients, raised LDH in majority of patients (323 ± 101 Units/L) and elevated CRP (14.6 ± 30.99 mg/L). Rest of the lab parameters were within the normal range. Conclusion: In our study, COVID patients with mild-to-moderate symptoms did not show any inflammatory/infective or vaso-occlusive lesions in the retina attributable to COVID-19 infection, except one patient who had a single streak hemorrhage in the macula away from fovea, probably incidental.
Collapse
Affiliation(s)
- Ravi Bypareddy
- Department of Vitreo-Retina, Regional Institute of Ophthalmology, Minto Ophthalmic Hospital, BMCRI, Bengaluru, Karnataka, India
| | - B L Sujatha Rathod
- Department of Vitreo-Retina, Regional Institute of Ophthalmology, Minto Ophthalmic Hospital, BMCRI, Bengaluru, Karnataka, India
| | - Y D Shilpa
- Department of Vitreo-Retina, Regional Institute of Ophthalmology, Minto Ophthalmic Hospital, BMCRI, Bengaluru, Karnataka, India
| | - H R Hithashree
- Department of Vitreo-Retina, Regional Institute of Ophthalmology, Minto Ophthalmic Hospital, BMCRI, Bengaluru, Karnataka, India
| | - Kalpana Badami Nagaraj
- Department of Vitreo-Retina, Regional Institute of Ophthalmology, Minto Ophthalmic Hospital, BMCRI, Bengaluru, Karnataka, India
| | - B C Hemalatha
- Department of Vitreo-Retina, Regional Institute of Ophthalmology, Minto Ophthalmic Hospital, BMCRI, Bengaluru, Karnataka, India
| | - Jessica Basumatary
- Department of Vitreo-Retina, Regional Institute of Ophthalmology, Minto Ophthalmic Hospital, BMCRI, Bengaluru, Karnataka, India
| | - Deeksha Bekal
- Department of Vitreo-Retina, Regional Institute of Ophthalmology, Minto Ophthalmic Hospital, BMCRI, Bengaluru, Karnataka, India
| | - R Niranjan
- Department of Vitreo-Retina, Regional Institute of Ophthalmology, Minto Ophthalmic Hospital, BMCRI, Bengaluru, Karnataka, India
| | - P G Anusha
- Department of Vitreo-Retina, Regional Institute of Ophthalmology, Minto Ophthalmic Hospital, BMCRI, Bengaluru, Karnataka, India
| |
Collapse
|
23
|
Sopp NM, Sharda V. An Eye on COVID-19: A Meta-analysis of Positive Conjunctival Reverse Transcriptase-Polymerase Chain Reaction and SARS-CoV-2 Conjunctivitis Prevalence. Optom Vis Sci 2021; 98:429-436. [PMID: 33973912 PMCID: PMC8136458 DOI: 10.1097/opx.0000000000001687] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 01/30/2021] [Indexed: 01/08/2023] Open
Abstract
SIGNIFICANCE This analysis and review demonstrate that, although emerging data indicate that the prevalence of severe acute respiratory coronavirus 2 (SARS-CoV-2) on the ocular surface and coronavirus disease 2019 (COVID-19) conjunctivitis is rare, the ocular surface remains of interest as a potential inoculation and transmission site for SARS-CoV-2. Continued safety precautions should be taken as more data become available.COVID-19, caused by SARS-CoV-2, is a novel, global pandemic that has infected millions and, up to this point, caused more than two million fatalities worldwide. The ocular surface has become of interest as a possible vector for transmission by acting as a direct inoculation site, being a conduit for the virus into the respiratory system or as a method of transmission from potentially infected conjunctiva or tears. The components necessary for SARS-CoV-2 to theoretically infect ocular tissues are present: binding receptors (angiotensin-converting enzyme 2 and cluster of differentiation 147) and mechanisms for cell entry (transmembrane protease serine 2 and cathepsin L). This meta-analysis of COVID-19 prevalence data indicates that SARS-CoV-2 RNA has been infrequently found in conjunctival samples when tested with reverse transcriptase-polymerase chain reaction. This review estimates the prevalence of SARS-CoV-2 on the ocular surface and prevalence of conjunctivitis in patients with laboratory-confirmed COVID-19. There is much to be learned regarding ocular tropism of SARS-CoV-2.
Collapse
|
24
|
Chakravarty D, Das Sarma J. Murine-β-coronavirus-induced neuropathogenesis sheds light on CNS pathobiology of SARS-CoV2. J Neurovirol 2021; 27:197-216. [PMID: 33547593 PMCID: PMC7864135 DOI: 10.1007/s13365-021-00945-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 12/29/2020] [Accepted: 01/12/2021] [Indexed: 02/06/2023]
Abstract
The pandemic caused by SARS-CoV-2 has caused widespread infection and significant mortality across the globe. Combined virology perspective of SARS-CoV-2 with a deep-rooted understanding of pathophysiological and immunological processes underlying the clinical manifestations of COVID-19 is of prime importance. The characteristic symptom of COVID-19 is respiratory distress with diffused alveolar damage, but emerging evidence suggests COVID-19 might also have neurologic consequences. Dysregulated homeostasis in the lungs has proven to be fatal, but one cannot ignore that the inability to breathe might be due to defects in the respiratory control center of the brainstem. While the mechanism of pulmonary distress has been documented in the literature, awareness of neurological features and their pathophysiology is still in the nascent state. This review makes references to the neuro-immune axis and neuro-invasive potential of SARS-CoV and SARS-CoV2, as well as the prototypic H-CoV strains in human brains. Simultaneously, considerable discussion on relevant experimental evidence of mild to severe neurological manifestations of fellow neurotropic murine-β-CoVs (m-CoVs) in the mouse model will help understand the underpinning mechanisms of Neuro-COVID. In this review, we have highlighted the neuroimmunopathological processes in murine CoVs. While MHV infection in mice and SARS-CoV-2 infection in humans share numerous parallels, there are critical differences in viral recognition and viral entry. These similarities are highlighted in this review, while differences have also been emphasized. Though CoV-2 Spike does not favorably interact with murine ACE2 receptor, modification of murine SARS-CoV2 binding domain or development of transgenic ACE-2 knock-in mice might help in mediating consequential infection and understanding human CoV2 pathogenesis in murine models. While a global animal model that can replicate all aspects of the human disease remains elusive, prior insights and further experiments with fellow m-β-CoV-induced cause-effect experimental models and current human COVID-19 patients data may help to mitigate the SARS-CoV-2-induced multifactorial multi-organ failure.
Collapse
Affiliation(s)
- Debanjana Chakravarty
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Haringhata, 741246, Mohanpur, India
| | - Jayasri Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Haringhata, 741246, Mohanpur, India.
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
25
|
Colina SE, Serena MS, Echeverría MG, Metz GE. Clinical and molecular aspects of veterinary coronaviruses. Virus Res 2021; 297:198382. [PMID: 33705799 PMCID: PMC7938195 DOI: 10.1016/j.virusres.2021.198382] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 12/20/2020] [Accepted: 03/04/2021] [Indexed: 12/12/2022]
Abstract
Coronaviruses are a large group of RNA viruses that infect a wide range of animal species. The replication strategy of coronaviruses involves recombination and mutation events that lead to the possibility of cross-species transmission. The high plasticity of the viral receptor due to a continuous modification of the host species habitat may be the cause of cross-species transmission that can turn into a threat to other species including the human population. The successive emergence of highly pathogenic coronaviruses such as the Severe Acute Respiratory Syndrome (SARS) in 2003, the Middle East Respiratory Syndrome Coronavirus in 2012, and the recent SARS-CoV-2 has incentivized a number of studies on the molecular basis of the coronavirus and its pathogenesis. The high degree of interrelatedness between humans and wild and domestic animals and the modification of animal habitats by human urbanization, has favored new viral spreads. Hence, knowledge on the main clinical signs of coronavirus infection in the different hosts and the distinctive molecular characteristics of each coronavirus is essential to prevent the emergence of new coronavirus diseases. The coronavirus infections routinely studied in veterinary medicine must be properly recognized and diagnosed not only to prevent animal disease but also to promote public health.
Collapse
Affiliation(s)
- Santiago Emanuel Colina
- Virology, Faculty of Veterinary Sciences, National University of La Plata, La Plata, Argentina; CONICET (National Scientific and Technical Research Council), CCT La Plata, Argentina
| | - María Soledad Serena
- Virology, Faculty of Veterinary Sciences, National University of La Plata, La Plata, Argentina; CONICET (National Scientific and Technical Research Council), CCT La Plata, Argentina
| | - María Gabriela Echeverría
- Virology, Faculty of Veterinary Sciences, National University of La Plata, La Plata, Argentina; CONICET (National Scientific and Technical Research Council), CCT La Plata, Argentina
| | - Germán Ernesto Metz
- Virology, Faculty of Veterinary Sciences, National University of La Plata, La Plata, Argentina; CONICET (National Scientific and Technical Research Council), CCT La Plata, Argentina.
| |
Collapse
|
26
|
Papazoglou A, Conen A, Haubitz S, Tschopp M, Guignard VJ, Menke MN, Enz TJ. Ophthalmic Screening in Patients with Coronavirus Disease 2019: A Prospective Cohort Study. J Clin Med 2021; 10:jcm10050896. [PMID: 33668256 PMCID: PMC7956798 DOI: 10.3390/jcm10050896] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/18/2021] [Accepted: 02/19/2021] [Indexed: 12/18/2022] Open
Abstract
Postmortem pathological examinations, animal studies, and anecdotal reports suggest that coronavirus disease 2019 (COVID-19) could potentially affect intraocular tissue. However, published evidence is scarce and conflicting. In our study, we screened 100 eyes of 50 patients hospitalized for COVID-19. Relevant medical and ophthalmological history was assessed as well as symptoms, laboratory results, specific treatments, clinical course, and outcome. Ophthalmic exams including assessment of best corrected visual acuity (BCVA), intraocular pressure (IOP), color perception, ocular motility, ophthalmoscopy as well as optical coherence tomography (OCT) of the macula and the optic disc was performed at hospital admission and 29 to 192 days later. Of the 50 patients included, 14 (28%) were female. Median age was 64.5 (range 29–90) years. COVID-19 severity was mild in 15 (30%), severe in 30 (60%), and critical in five cases (10%). At baseline, median BCVA was 0.1 (0–1.8) Logarithm of the Minimum Angle of Resolution (LogMAR) and median IOP was 16 (8–22) mmHg. At follow-up, no relevant changes in BCVA and IOP were documented. No signs of active intraocular inflammation or optic nerve affection were found and OCT findings were widely stable during the observation period. Our findings suggest that COVID-19 does not regularly affect intraocular tissue.
Collapse
Affiliation(s)
- Anthia Papazoglou
- Department of Ophthalmology, Cantonal Hospital Aarau, 5000 Aarau, Switzerland; (M.T.); (V.J.G.); (M.N.M.)
- Correspondence: (A.P.); (T.J.E.)
| | - Anna Conen
- Department of Infectious Diseases and Hospital Hygiene, Cantonal Hospital Aarau, 5000 Aarau, Switzerland; (A.C.); (S.H.)
| | - Sebastian Haubitz
- Department of Infectious Diseases and Hospital Hygiene, Cantonal Hospital Aarau, 5000 Aarau, Switzerland; (A.C.); (S.H.)
- Department of Internal and Emergency Medicine, Cantonal Hospital Aarau, 5000 Aarau, Switzerland
| | - Markus Tschopp
- Department of Ophthalmology, Cantonal Hospital Aarau, 5000 Aarau, Switzerland; (M.T.); (V.J.G.); (M.N.M.)
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
| | - Viviane J. Guignard
- Department of Ophthalmology, Cantonal Hospital Aarau, 5000 Aarau, Switzerland; (M.T.); (V.J.G.); (M.N.M.)
| | - Marcel N. Menke
- Department of Ophthalmology, Cantonal Hospital Aarau, 5000 Aarau, Switzerland; (M.T.); (V.J.G.); (M.N.M.)
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
| | - Tim J. Enz
- Department of Ophthalmology, Cantonal Hospital Aarau, 5000 Aarau, Switzerland; (M.T.); (V.J.G.); (M.N.M.)
- Department of Clinical Neuroscience, Section of Ophthalmology and Vision, St. Erik Eye Hospital, Karolinska Institutet, 171 64 Stockholm, Sweden
- Correspondence: (A.P.); (T.J.E.)
| |
Collapse
|
27
|
Mohammadi S, Moosaie F, Aarabi MH. Understanding the Immunologic Characteristics of Neurologic Manifestations of SARS-CoV-2 and Potential Immunological Mechanisms. Mol Neurobiol 2020; 57:5263-5275. [PMID: 32869183 PMCID: PMC7458880 DOI: 10.1007/s12035-020-02094-y] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 08/25/2020] [Indexed: 12/12/2022]
Abstract
Similar to its predecessors, coronavirus disease 2019 (COVID-19) exhibits neurotrophic properties, which lead to progression of neurologic sequelae. Besides direct viral invasion to the central nervous system (CNS), indirect CNS involvement through viral-mediated immune response is plausible. Aberrant immune pathways such as extreme release of cytokines (cytokine storm), autoimmunity mediated by cross-reactivity between CNS components and viral particles, and microglial activation propagate CNS damage in these patients. Here, we review the currently available evidence to discuss the plausible immunologic pathways that may contribute to the development of COVID-19 neurological complications, namely Alzheimer's disease, Parkinson's disease, stroke, multiple sclerosis, Guillain-Barre syndrome, seizure, and brainstem involvement.
Collapse
Affiliation(s)
- Soheil Mohammadi
- Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- NeuroImaging Network (NIN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Fatemeh Moosaie
- Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- NeuroImaging Network (NIN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | | |
Collapse
|
28
|
Das Sarma J, Burrows A, Rayman P, Hwang MH, Kundu S, Sharma N, Bergmann C, Sen GC. Ifit2 deficiency restricts microglial activation and leukocyte migration following murine coronavirus (m-CoV) CNS infection. PLoS Pathog 2020; 16:e1009034. [PMID: 33253295 PMCID: PMC7738193 DOI: 10.1371/journal.ppat.1009034] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 12/15/2020] [Accepted: 10/02/2020] [Indexed: 02/07/2023] Open
Abstract
The interferon-induced tetratricopeptide repeat protein (Ifit2) protects mice from lethal neurotropic viruses. Neurotropic coronavirus MHV-RSA59 infection of Ifit2-/- mice caused pronounced morbidity and mortality accompanied by rampant virus replication and spread throughout the brain. In spite of the higher virus load, induction of many cytokines and chemokines in the brains of infected Ifit2-/- mice were similar to that in wild-type mice. In contrast, infected Ifit2-/- mice revealed significantly impaired microglial activation as well as reduced recruitment of NK1.1 T cells and CD4 T cells to the brain, possibly contributing to the lack of viral clearance. These two deficiencies were associated with a lower level of microglial expression of CX3CR1, the receptor of the CX3CL1 (Fractalkine) chemokine, which plays a critical role in both microglial activation and leukocyte recruitment. The above results uncovered a new potential role of an interferon-induced protein in immune protection. Interferons (IFNs) are known to protect from virus dissemination and pathogenesis. Several IFN stimulated genes (ISG) regulate neuropathogenesis but the mechanisms underlying the antiviral effects are not clearly understood. IFN induced tetratricopeptide repeats (Ifit) are a class of ISGs. Among the Ifits, Ifit2 is known to play a beneficial role in restricting neurotropic viral replication. To provide better cellular insights into the protective mechanisms of Ifit2 functions, using a neurotropic coronavirus infection in Ifit2 depleted mice we report that in the absence of Ifit2, viral replication is dramatically increased and mice develop severe clinical signs and symptoms of neurological deficit. Despite the enormous viral load, Ifit2 deficient mice are impaired in microglial activation and recruitment of peripheral leukocytes into the CNS. This impaired leuocyte infiltration in Ifit2 deficient mice was also associated with reduced expression of a novel chemokine receptor CX3CR1,which is important for viral induced microglial activation and maintaining tissue homeostasis.
Collapse
Affiliation(s)
- Jayasri Das Sarma
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Ohio, United States of America
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, India
- * E-mail:
| | - Amy Burrows
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Ohio, United States of America
| | - Patricia Rayman
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Ohio, United States of America
| | - Mi-Hyun Hwang
- Department of Neurosciences, Cleveland Clinic, Ohio, United States of America
| | - Soumya Kundu
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, India
| | - Nikhil Sharma
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Ohio, United States of America
| | - Cornelia Bergmann
- Department of Neurosciences, Cleveland Clinic, Ohio, United States of America
| | - Ganes C. Sen
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Ohio, United States of America
| |
Collapse
|
29
|
Abdul-Kadir MA, Lim LT. Human coronaviruses: ophthalmic manifestations. BMJ Open Ophthalmol 2020; 5:e000630. [PMID: 33195813 PMCID: PMC7607601 DOI: 10.1136/bmjophth-2020-000630] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/17/2020] [Accepted: 10/25/2020] [Indexed: 01/08/2023] Open
Abstract
The 2019 novel coronavirus which causes severe acute respiratory syndrome (SARS) known as SARS-CoV-2 still remains as a global pandemic since its discovery and continues to spread across the world, given how highly contagious the virus is. We reviewed various articles that explore eye involvement in COVID-19 and other human coronaviruses, its human manifestations in comparison to animal studies and potential mechanism of viral entry into the eye surface. Evidence of animal studies depicted various complications of coronaviruses infection into the eyes, in both anterior and posterior segments of the eye. Conjunctival inflammation remains uncommon in association with COVID-19, with other ophthalmic findings. The risk of transmission via the ocular surface remains likely low, though it is inarguably present based on preliminary finding of viral load in ocular samples and expression of ACE2 on the ocular surface. Testing the tears sample for diagnosing SARS-CoV-2 was unreliable due to limitations of the testing kits and conflicting evidence of the viral titre in the ocular samples. Further larger, more precise and specific studies are required to allow us to better understand the pattern of virulence underlying the associations of SARS-CoV-2 in the eye despite its rare occurrence. This review article aims to enhance better awareness among clinicians regarding ocular manifestations associated with COVID-19 and necessary precautions should be implemented to minimise the risk of person-to-person especially in the nosocomial setting.
Collapse
Affiliation(s)
- Mohd-Asyraaf Abdul-Kadir
- Ophthalmology Department, Universiti Malaysia Sarawak (UNIMAS), Kota Samarahan, Sarawak, Malaysia
| | - Lik Thai Lim
- Ophthalmology Department, Universiti Malaysia Sarawak (UNIMAS), Kota Samarahan, Sarawak, Malaysia
| |
Collapse
|
30
|
Abstract
PURPOSE OF REVIEW To provide a summary of the neuro-ophthalmic manifestations of coronavirus disease 19 (COVID-19), documented in the literature thus far. RECENT FINDINGS A small but growing literature documents cases of new onset neuro-ophthalmic disease, in the setting of COVID-19 infection. Patients with COVID-19 have experienced acute onset vision loss, optic neuritis, cranial neuropathies, and Miller Fisher syndrome. In addition, COVID-19 increases the risk of cerebrovascular diseases that can impact the visual system. SUMMARY The literature on COVID-19 continues to evolve. Although COVID-19 primarily impacts the respiratory system, there are several reports of new onset neuro-ophthalmic conditions in COVID-infected patients. When patients present with new onset neuro-ophthalmic issues, COVID-19 should be kept on the differential. Testing for COVID-19 should be considered, especially when fever or respiratory symptoms are also present. When screening general patients for COVID-19-associated symptoms, frontline physicians can consider including questions about diplopia, eye pain, pain with extraocular movements, decreased vision, gait issues, and other neurologic symptoms. The presence of these symptoms may increase the overall probability of viral infection, especially when fever or respiratory symptoms are present. More research is needed to establish a causal relationship between COVID-19 and neuro-ophthalmic disease, and better understand pathogenesis.
Collapse
Affiliation(s)
- Alanna K Tisdale
- Department of Ophthalmology and Visual Sciences, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Bart K Chwalisz
- Department of Ophthalmology, Massachusetts Eye & Ear
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
31
|
Retinal involvement and ocular findings in COVID-19 pneumonia patients. Sci Rep 2020; 10:17419. [PMID: 33060700 PMCID: PMC7566835 DOI: 10.1038/s41598-020-74446-6] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 09/30/2020] [Indexed: 01/01/2023] Open
Abstract
Changes in immune and coagulation systems and possible viral spread through the blood–brain barrier have been described in SARS-CoV-2 infection. In this study, we evaluated the possible retinal involvement and ocular findings in severe COVID-19 pneumonia patients. A cross-sectional study was conducted on 46 patients affected by severe COVID-19 who were hospitalized in one intensive care unit (ICU) and in two infectious disease wards, including bedside eye screening, corneal sensitivity assessment and retinography. A total of 43 SARS-CoV-2-positive pneumonia patients affected with COVID-19 pneumonia were included, including 25 males and 18 females, with a median age of 70 years [IQR 59–78]. Except for one patient with unilateral posterior chorioretinitis of opportunistic origin, of whom aqueous tap was negative for SARS-CoV-2, no further retinal manifestation related to COVID-19 infection was found in our cohort. We found 3 patients (7%) with bilateral conjunctivitis in whom PCR analysis on conjunctival swabs provided negative results for SARS-CoV-2. No alterations in corneal sensitivity were found. We demonstrated the absence of retinal involvement in SARS-CoV-2 pneumonia patients. Ophthalmologic evaluation in COVID-19, particularly in patients hospitalized in an ICU setting, may be useful to reveal systemic co-infections by opportunistic pathogens.
Collapse
|
32
|
Amesty MA, Alió Del Barrio JL, Alió JL. COVID-19 Disease and Ophthalmology: An Update. Ophthalmol Ther 2020; 9:1-12. [PMID: 32445134 PMCID: PMC7243216 DOI: 10.1007/s40123-020-00260-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Indexed: 12/20/2022] Open
Abstract
The worldwide outbreak of the severe and acute respiratory coronavirus disease (COVID-19) caused by the coronavirus strain SARS-CoV-2 is currently the focal point of discussion due to the suffering this syndrome is causing to humanity. However, the ophthalmological implications of this syndrome has not yet been well described. Both eyes and tears as portals of entry and sources of contagion have been the subject of debate by many authors. The purpose of this review is to summarize the evidence currently available on COVID-19 and its ocular implications and manifestations, in both animals and humans, with the aim to facilitate prevention and educate the ophthalmological community on this subject. A review of the literature revealed that the results of some studies suggest that ocular symptoms commonly appear in patients with severe COVID-19 pneumonia and that it is possible to isolate the virus from the conjunctival sac of these patients. Conjunctivitis is not a common manifestation of the disease, but contact with infected eyes could be one route of transmission. Consequently, ophthalmologists need to have correct prevention strategies in place. Some guidelines regarding the prevention and management of ophthalmology clinics are reviewed. However, well-designed trials should be conducted to rule out other ocular manifestations that may result from COVID-19 infection and to understand the transmission of the virus through the eyes.
Collapse
Affiliation(s)
- María A Amesty
- Oculoplastic Department, Vissum (Grupo Miranza), Alicante, Spain
| | - Jorge L Alió Del Barrio
- Cornea, Cataract and Refractive Surgery Unit, Vissum (Grupo Miranza), Alicante, Spain
- Division of Ophthalmology, School of Medicine, Universidad Miguel Hernández, Alicante, Spain
| | - Jorge L Alió
- Cornea, Cataract and Refractive Surgery Unit, Vissum (Grupo Miranza), Alicante, Spain.
- Division of Ophthalmology, School of Medicine, Universidad Miguel Hernández, Alicante, Spain.
| |
Collapse
|
33
|
Abstract
Based on the available publications, the article systematizes the forms of damage to the central nervous system in СOVID-19patients. Along with a description of clinical manifestations, pathogenesis, methods of laboratory, instrumental and radiological diagnosis with the discussion of the nosological forms. An interdisciplinary approach and international cooperation are required to study the problems pathogenesis, course, outcomes, and the development of new methods of diagnosis and treatment.
Collapse
|
34
|
Palao M, Fernández-Díaz E, Gracia-Gil J, Romero-Sánchez CM, Díaz-Maroto I, Segura T. Multiple sclerosis following SARS-CoV-2 infection. Mult Scler Relat Disord 2020; 45:102377. [PMID: 32698095 PMCID: PMC7340057 DOI: 10.1016/j.msard.2020.102377] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 07/02/2020] [Accepted: 07/04/2020] [Indexed: 12/21/2022]
Abstract
The first case of multiple sclerosis (MS) shortly after COVID-19 is presented. Viral infections strongly relate to MS onset and its relapses. SARS-CoV-2 can play a role in the triggering of demyelinating diseases. Systemic immune response against SARS-CoV-2 could reach CNS leading to demyelination.
SARS-CoV-2 infection can produce neurological features. The most common are headache, anosmia and dysgeusia but patients may also develop other central nervous system (CNS) injuries. We present a patient affected by Covid-19 who initially consulted for decreased visual acuity. The MRI showed inflammation in the right optic nerve and demyelinating lesions in the CNS. We speculate that an immune mechanism induced by SARS-CoV-2, which can activate lymphocytes and an inflammatory response, plays a role in the clinical onset of the disease. This pathogen may be associated with either the triggering or the exacerbation of inflammatory/demyelinating disease.
Collapse
Affiliation(s)
- M Palao
- Neurology Department, Complejo Hospitalario Universitario de Albacete, Castilla- La Mancha, Spain.
| | - E Fernández-Díaz
- Neurology Department, Complejo Hospitalario Universitario de Albacete, Castilla- La Mancha, Spain
| | - J Gracia-Gil
- Neurology Department, Complejo Hospitalario Universitario de Albacete, Castilla- La Mancha, Spain
| | - C M Romero-Sánchez
- Neurology Department, Complejo Hospitalario Universitario de Albacete, Castilla- La Mancha, Spain
| | - I Díaz-Maroto
- Neurology Department, Complejo Hospitalario Universitario de Albacete, Castilla- La Mancha, Spain
| | - T Segura
- Neurology Department, Complejo Hospitalario Universitario de Albacete, Castilla- La Mancha, Spain
| |
Collapse
|
35
|
Chakravarty D, Saadi F, Kundu S, Bose A, Khan R, Dine K, Kenyon LC, Shindler KS, Das Sarma J. CD4 Deficiency Causes Poliomyelitis and Axonal Blebbing in Murine Coronavirus-Induced Neuroinflammation. J Virol 2020; 94:e00548-20. [PMID: 32404525 PMCID: PMC7343199 DOI: 10.1128/jvi.00548-20] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 05/07/2020] [Indexed: 12/20/2022] Open
Abstract
Mouse hepatitis virus (MHV) is a murine betacoronavirus (m-CoV) that causes a wide range of diseases in mice and rats, including hepatitis, enteritis, respiratory diseases, and encephalomyelitis in the central nervous system (CNS). MHV infection in mice provides an efficient cause-effect experimental model to understand the mechanisms of direct virus-induced neural-cell damage leading to demyelination and axonal loss, which are pathological features of multiple sclerosis (MS), the most common disabling neurological disease in young adults. Infiltration of T lymphocytes, activation of microglia, and their interplay are the primary pathophysiological events leading to disruption of the myelin sheath in MS. However, there is emerging evidence supporting gray matter involvement and degeneration in MS. The investigation of T cell function in the pathogenesis of deep gray matter damage is necessary. Here, we employed RSA59 (an isogenic recombinant strain of MHV-A59)-induced experimental neuroinflammation model to compare the disease in CD4-/- mice with that in CD4+/+ mice at days 5, 10, 15, and 30 postinfection (p.i.). Viral titer estimation, nucleocapsid gene amplification, and viral antinucleocapsid staining confirmed enhanced replication of the virions in the absence of functional CD4+ T cells in the brain. Histopathological analyses showed elevated susceptibility of CD4-/- mice to axonal degeneration in the CNS, with augmented progression of acute poliomyelitis and dorsal root ganglionic inflammation rarely observed in CD4+/+ mice. Depletion of CD4+ T cells showed unique pathological bulbar vacuolation in the brain parenchyma of infected mice with persistent CD11b+ microglia/macrophages in the inflamed regions on day 30 p.i. In summary, the current study suggests that CD4+ T cells are critical for controlling acute-stage poliomyelitis (gray matter inflammation), chronic axonal degeneration, and inflammatory demyelination due to loss of protective antiviral host immunity.IMPORTANCE The current trend in CNS disease biology is to attempt to understand the neural-cell-immune interaction to investigate the underlying mechanism of neuroinflammation, rather than focusing on peripheral immune activation. Most studies in MS are targeted toward understanding the involvement of CNS white matter. However, the importance of gray matter damage has become critical in understanding the long-term progressive neurological disorder. Our study highlights the importance of CD4+ T cells in safeguarding neurons against axonal blebbing and poliomyelitis from murine betacoronavirus-induced neuroinflammation. Current knowledge of the mechanisms that lead to gray matter damage in MS is limited, because the most widely used animal model, experimental autoimmune encephalomyelitis (EAE), does not present this aspect of the disease. Our results, therefore, add to the existing limited knowledge in the field. We also show that the microglia, though important for the initiation of neuroinflammation, cannot establish a protective host immune response without the help of CD4+ T cells.
Collapse
Affiliation(s)
- Debanjana Chakravarty
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, India
| | - Fareeha Saadi
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, India
| | - Soumya Kundu
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, India
| | - Abhishek Bose
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, India
| | - Reas Khan
- Department of Ophthalmology, University of Pennsylvania Scheie Eye Institute, Philadelphia, Pennsylvania, USA
| | - Kimberly Dine
- Department of Ophthalmology, University of Pennsylvania Scheie Eye Institute, Philadelphia, Pennsylvania, USA
| | - Lawrence C Kenyon
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Kenneth S Shindler
- Department of Ophthalmology, University of Pennsylvania Scheie Eye Institute, Philadelphia, Pennsylvania, USA
- Department of Neurology, University of Pennsylvania Scheie Eye Institute, Philadelphia, Pennsylvania, USA
| | - Jayasri Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, India
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
36
|
Rout SS, Singh M, Shindler KS, Das Sarma J. One proline deletion in the fusion peptide of neurotropic mouse hepatitis virus (MHV) restricts retrograde axonal transport and neurodegeneration. J Biol Chem 2020; 295:6926-6935. [PMID: 32249210 DOI: 10.1074/jbc.ra119.011918] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 04/02/2020] [Indexed: 01/06/2023] Open
Abstract
Mouse hepatitis virus (MHV; murine coronavirus) causes meningoencephalitis, myelitis, and optic neuritis followed by axonal loss and demyelination. This murine virus is used as a common model to study acute and chronic virus-induced demyelination in the central nervous system. Studies with recombinant MHV strains that differ in the gene encoding the spike protein have demonstrated that the spike has a role in MHV pathogenesis and retrograde axonal transport. Fusion peptides (FPs) in the spike protein play a key role in MHV pathogenesis. In a previous study of the effect of deleting a single proline residue in the FP of a demyelinating MHV strain, we found that two central, consecutive prolines are important for cell-cell fusion and pathogenesis. The dihedral fluctuation of the FP was shown to be repressed whenever two consecutive prolines were present, in contrast to the presence of a single proline in the chain. Using this proline-deleted MHV strain, here we investigated whether intracranial injection of this strain can induce optic neuritis by retrograde axonal transport from the brain to the retina through the optic nerve. We observed that the proline-deleted recombinant MHV strain is restricted to the optic nerve, is unable to translocate to the retina, and causes only minimal demyelination and no neuronal death. We conclude that an intact proline dyad in the FP of the recombinant demyelinating MHV strain plays a crucial role in translocation of the virus through axons and subsequent neurodegeneration.
Collapse
Affiliation(s)
- Saurav Saswat Rout
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur 741246, India
| | - Manmeet Singh
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur 741246, India
| | - Kenneth S Shindler
- Scheie Eye Institute and F. M. Kirby Center for Molecular Ophthalmology, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Jayasri Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur 741246, India
| |
Collapse
|
37
|
Seah I, Agrawal R. Can the Coronavirus Disease 2019 (COVID-19) Affect the Eyes? A Review of Coronaviruses and Ocular Implications in Humans and Animals. Ocul Immunol Inflamm 2020; 28:391-395. [PMID: 32175797 PMCID: PMC7103678 DOI: 10.1080/09273948.2020.1738501] [Citation(s) in RCA: 428] [Impact Index Per Article: 85.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In December 2019, a novel coronavirus (CoV) epidemic, caused by the severe acute respiratory syndrome coronavirus - 2 (SARS-CoV-2) emerged from China. This virus causes the coronavirus disease 2019 (COVID-19). Since then, there have been anecdotal reports of ocular infection. The ocular implications of human CoV infections have not been widely studied. However, CoVs have been known to cause various ocular infections in animals. Clinical entities such as conjunctivitis, anterior uveitis, retinitis, and optic neuritis have been documented in feline and murine models. In this article, the current evidence suggesting possible human CoV infection of ocular tissue is reviewed. The review article will also highlight animal CoVs and their associated ocular infections. We hope that this article will serve as a start for further research into the ocular implications of human CoV infections.
Collapse
Affiliation(s)
- Ivan Seah
- Department of Ophthalmology, National University Hospital, Singapore, Singapore
| | - Rupesh Agrawal
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,NHS Foundation Trust, Moorfields Eye Hospital, London, UK.,National Healthcare Group Eye Institute, Tan Tock Seng Hospital, Singapore, Singapore
| |
Collapse
|
38
|
Sanabria-Castro A, Flores-Díaz M, Alape-Girón A. Biological models in multiple sclerosis. J Neurosci Res 2019; 98:491-508. [PMID: 31571267 DOI: 10.1002/jnr.24528] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 09/05/2019] [Accepted: 09/05/2019] [Indexed: 12/13/2022]
Abstract
Considering the etiology of multiple sclerosis (MS) is still unknown, experimental models resembling specific aspects of this immune-mediated demyelinating human disease have been developed to increase the understanding of processes related to pathogenesis, disease evolution, evaluation of therapeutic interventions, and demyelination and remyelination mechanisms. Based on the nature of the investigation, biological models may include in vitro, in vivo, and ex vivo assessments. Even though these approaches have disclosed valuable information, every disease animal model has limitations and can only replicate specific features of MS. In vitro and ex vivo models generally do not reflect what occurs in the organism, and in vivo animal models are more likely used; nevertheless, they are able to reproduce only certain stages of the disease. In vivo MS disease animal models in mammals include: experimental autoimmune encephalomyelitis, viral encephalomyelitis, and induced demyelination. This review examines and describes the most common biological disease animal models for the study of MS, their specific characteristics and limitations.
Collapse
Affiliation(s)
- Alfredo Sanabria-Castro
- Research Unit, San Juan de Dios Hospital CCSS, San José, Costa Rica.,School of Pharmacy, University of Costa Rica, San José, Costa Rica
| | | | | |
Collapse
|
39
|
Singh M, Khan RS, Dine K, Das Sarma J, Shindler KS. Intracranial Inoculation Is More Potent Than Intranasal Inoculation for Inducing Optic Neuritis in the Mouse Hepatitis Virus-Induced Model of Multiple Sclerosis. Front Cell Infect Microbiol 2018; 8:311. [PMID: 30234031 PMCID: PMC6132074 DOI: 10.3389/fcimb.2018.00311] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 08/14/2018] [Indexed: 01/30/2023] Open
Abstract
Neurotropic strains of mouse hepatitis virus (MHV) induce acute inflammation and chronic demyelination in the spinal cord and optic nerves mediated by axonal spread following intracranial inoculation in mice, with pathologic features similar to the human demyelinating disease multiple sclerosis. Spinal cord demyelination is also induced following intranasal inoculation with neurotropic MHV strains, however much higher viral doses are required as compared to intracranial inoculation. Recently, it was shown that intranasal administration of low concentrations of proteins leads to significant, rapid accumulation of protein in the optic nerve and in the eye, with only low levels reaching spinal cord and other brain regions. Thus, we examined whether intranasal inoculation with MHV at doses equivalent to those given intracranially could induce optic neuritis—inflammation, demyelination and loss of retinal ganglion cells (RGCs) in the optic nerve with or without inducing spinal cord demyelination. Four week old male C57BL/6J mice were inoculated intracranially with the recombinant demyelinating strain RSA59, or intranasally with RSA59 or the non-demyelinating strain RSMHV2 as control. One month post-inoculation, mice inoculated intracranially with RSA59 had significant myelin loss in both spinal cord and optic nerves, with significant loss of RGCs as well, consistent with prior studies. As expected, intranasal inoculation with RSA59 failed to induce demyelination in spinal cord; however, it also did not induce optic nerve demyelination. No acute inflammation was found, and no viral antigen was detected, in the optic nerve or retina 1 day after inoculation. Results confirm the neurotropic effects of RSA59 following intracranial inoculation, and suggest that direct infection with axonal transport of virus from brain to spinal cord and optic nerve is required to induce demyelinating disease. These studies suggest that MHV does not selectively concentrate in optic nerve and retina to sufficient levels to induce demyelination following intranasal inoculation. Intracranial inoculation should continue to be considered a preferred method for studies of MHV-induced optic neuritis and central nervous system (CNS) demyelinating disease.
Collapse
Affiliation(s)
- Manmeet Singh
- Department of Biological Science, Indian Institute of Science Education and Research Kolkata, Mohanpur, India
| | - Reas S Khan
- FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, United States
| | - Kimberly Dine
- FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, United States
| | - Jayasri Das Sarma
- Department of Biological Science, Indian Institute of Science Education and Research Kolkata, Mohanpur, India
| | - Kenneth S Shindler
- FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
40
|
Mentis AFA, Dardiotis E, Grigoriadis N, Petinaki E, Hadjigeorgiou GM. Viruses and Multiple Sclerosis: From Mechanisms and Pathways to Translational Research Opportunities. Mol Neurobiol 2017; 54:3911-3923. [PMID: 28455696 DOI: 10.1007/s12035-017-0530-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 04/06/2017] [Indexed: 12/26/2022]
Abstract
Viruses are directly or indirectly implicated in multiple sclerosis (MS). Here, we review the evidence on the virus-related pathophysiology of MS, introduce common experimental models, and explore the ways in which viruses cause demyelination. By emphasizing knowledge gaps, we highlight future research directions for effective MS diagnostics and therapies: (i) identifying biomarkers for at-risk individuals, (ii) searching for direct evidence of specific causative viruses, (iii) establishing the contribution of host genetic factors and viruses, and (iv) investigating the contribution of immune regulation at extra-CNS sites. Research in these areas is likely to be facilitated by the application of high-throughput technologies, the development of systems-based bioinformatic approaches, careful selection of experimental models, and the acquisition of high-quality clinical material for tissue-based research.
Collapse
Affiliation(s)
- Alexios-Fotios A Mentis
- Department of Microbiology, University Hospital of Larissa, University of Thessaly, Larissa, Greece. .,The Johns Hopkins University, AAP, Baltimore, MD, USA.
| | - Efthimios Dardiotis
- Department of Neurology, University Hospital of Larissa, University of Thessaly, Larissa, Greece
| | - Nikolaos Grigoriadis
- B' Department of Neurology, Laboratory of Experimental Neurology and Neuroimmunology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Efthimia Petinaki
- Department of Microbiology, University Hospital of Larissa, University of Thessaly, Larissa, Greece
| | - Georgios M Hadjigeorgiou
- Department of Neurology, University Hospital of Larissa, University of Thessaly, Larissa, Greece
| |
Collapse
|
41
|
Biswas K, Chatterjee D, Addya S, Khan RS, Kenyon LC, Choe A, Cohrs RJ, Shindler KS, Das Sarma J. Demyelinating strain of mouse hepatitis virus infection bridging innate and adaptive immune response in the induction of demyelination. Clin Immunol 2016; 170:9-19. [PMID: 27394164 PMCID: PMC7106046 DOI: 10.1016/j.clim.2016.07.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 05/23/2016] [Accepted: 07/05/2016] [Indexed: 02/06/2023]
Abstract
The presence of immunoglobulin oligoclonal bands in the cerebrospinal fluid of Multiple Sclerosis (MS) patients supports the hypothesis of an infectious etiology, although the antigenic targets remain elusive. Neurotropic mouse hepatitis virus (MHV) infection in mice provides a useful tool for studying mechanisms of demyelination in a virus-induced experimental model of MS. This study uses Affymetrix microarray analysis to compare differential spinal cord mRNA levels between mice infected with demyelinating and non-demyelinating strains of MHV to identify host immune genes expressed in this demyelinating disease model. The study reveals that during the acute stage of infection, both strains induce inflammatory innate immune response genes, whereas upregulation of several immunoglobulin genes during chronic stage infection is unique to infection with the demyelinating strain. Results suggest that the demyelinating strain induced an innate-immune response during acute infection that may promote switching of Ig isotype genes during chronic infection, potentially playing a role in antibody-mediated progressive demyelination even after viral clearance.
Collapse
Affiliation(s)
- Kaushiki Biswas
- Department of Biological Sciences, Indian Institute of Science Education and ResearchKolkata (IISER-K), India
| | - Dhriti Chatterjee
- Department of Biological Sciences, Indian Institute of Science Education and ResearchKolkata (IISER-K), India
| | - Sankar Addya
- Kimmel Cancer Centre, Thomas Jefferson University, Philadelphia, PA, USA
| | - Reas S Khan
- Scheie Eye Institute and FM Kirby Centre for Molecular Ophthalmology, University of Pennsylvania, Philadelphia, PA, USA
| | - Lawrence C Kenyon
- Departments of Anatomy, Pathology and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Alexander Choe
- Department of Neurology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Randall J Cohrs
- Department of Neurology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Kenneth S Shindler
- Scheie Eye Institute and FM Kirby Centre for Molecular Ophthalmology, University of Pennsylvania, Philadelphia, PA, USA
| | - Jayasri Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and ResearchKolkata (IISER-K), India.
| |
Collapse
|
42
|
Wu GF, Parker Harp CR, Shindler KS. Optic Neuritis: A Model for the Immuno-pathogenesis of Central Nervous System Inflammatory Demyelinating Diseases. ACTA ACUST UNITED AC 2015; 11:85-92. [PMID: 29399010 DOI: 10.2174/1573395511666150707181644] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Evidence for the tenuous regulation between the immune system and central nervous system (CNS) can be found with examples of interaction between these organ systems gone awry. Multiple sclerosis (MS) is the prototypical inflammatory disease of the CNS and is characterized by widely distributed inflammatory demyelinating plaques that can involve the brain, spinal cord and/or optic nerves. Optic neuritis (ON), inflammatory injury of the optic nerve that frequently occurs in patients with MS, has been the focus of intense study in part given the readily accessible nature of clinical outcome measures. Exploring the clinical and pathological features of ON in relation to other inflammatory demyelinating conditions of the CNS, namely MS and neuromyelitis optica, provides an opportunity to glean common and distinct mechanisms of disease. Emerging data from clinical studies along with various animal models involving ON implicate innate and adaptive immune responses directed at glial targets, including myelin oligodendrocyte glycoprotein and aquaporin 4. Resolution of inflammation in ON is commonly observed both clinically and experimentally, but persistent nerve injury is also one emerging hallmark of ON. One hypothesis seeking evaluation is that, in comparison to other sites targeted in MS, the optic nerve is a highly specialized target within the CNS predisposing to unique immunologic processes that generate ON. Overall, ON serves as a highly relevant entity for understanding the pathogenesis of other CNS demyelinating conditions, most notably MS.
Collapse
Affiliation(s)
- Gregory F Wu
- Department of Neurology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA.,Department of Pathology & Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Chelsea R Parker Harp
- Department of Pathology & Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Kenneth S Shindler
- Department of Ophthalmology, University of Pennsylvania School of Medicine, Philadelphia, PA 19004, USA
| |
Collapse
|
43
|
Vogel I, Kasran A, Cremer J, Kim YJ, Boon L, Van Gool SW, Ceuppens JL. CD28/CTLA-4/B7 costimulatory pathway blockade affects regulatory T-cell function in autoimmunity. Eur J Immunol 2015; 45:1832-41. [PMID: 25727069 DOI: 10.1002/eji.201445190] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 02/06/2015] [Accepted: 02/25/2015] [Indexed: 11/06/2022]
Abstract
Naïve T cells require B7/CD28 costimulation in order to be fully activated. Attempts to block this pathway have been effective in preventing unwanted immune reactions. As B7 blockade might also affect Treg cells and interfere with negative signaling through membrane CTLA-4 on effector T (Teff) cells, its immune-modulatory effects are potentially more complex. Here, we used the mouse model of multiple sclerosis (MS), EAE, to study the effect of B7 blockade. An effective therapy for MS patients has to interfere with ongoing inflammation, and therefore we injected CTLA-4Ig at day 7 and 9 after immunization, when myelin-reactive T cells have been primed and start migrating toward the CNS. Surprisingly, B7 blockade exacerbated disease signs and resulted in more severe CNS inflammation and demyelination, and was associated with an enhanced production of the inflammatory cytokines IL-17 and IFN-γ. Importantly, CTLA-4Ig treatment resulted in a transient reduction of Ki67 and CTLA-4 expression and function of peripheral Treg cells. Taken together, B7 blockade at a particular stage of the autoimmune response can result in the suppression of Treg cells, leading to a more severe disease.
Collapse
Affiliation(s)
- Isabel Vogel
- Laboratory of Clinical Immunology, KU Leuven, University Hospital Gasthuisberg, Leuven, Belgium
| | - Ahmad Kasran
- Laboratory of Clinical Immunology, KU Leuven, University Hospital Gasthuisberg, Leuven, Belgium
| | - Jonathan Cremer
- Laboratory of Clinical Immunology, KU Leuven, University Hospital Gasthuisberg, Leuven, Belgium
| | - Yoo-Jin Kim
- Department of Pathology, Saarland University Hospital, Homburg, Germany
| | | | - Stefaan W Van Gool
- Laboratory of Clinical Immunology, KU Leuven, University Hospital Gasthuisberg, Leuven, Belgium.,Childhood Immunology, KU Leuven, University Hospital Gasthuisberg, Leuven, Belgium
| | - Jan L Ceuppens
- Laboratory of Clinical Immunology, KU Leuven, University Hospital Gasthuisberg, Leuven, Belgium
| |
Collapse
|
44
|
Chatterjee D, Addya S, Khan RS, Kenyon LC, Choe A, Cohrs RJ, Shindler KS, Sarma JD. Mouse hepatitis virus infection upregulates genes involved in innate immune responses. PLoS One 2014; 9:e111351. [PMID: 25360880 PMCID: PMC4216085 DOI: 10.1371/journal.pone.0111351] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 09/24/2014] [Indexed: 11/19/2022] Open
Abstract
Neurotropic recombinant strain of Mouse Hepatitis Virus, RSA59, induces meningo-encephalitis, myelitis and demyelination following intracranial inoculation. RSA59 induced neuropathology is partially caused by activation of CNS resident microglia, as demonstrated by changes in cellular morphology and increased expression of a microglia/macrophage specific calcium ion binding factor, Iba1. Affymetrix Microarray analysis for mRNA expression data reveals expression of inflammatory mediators that are known to be released by activated microglia. Microglia-specific cell surface molecules, including CD11b, CD74, CD52 and CD68, are significantly upregulated in contrast to CD4, CD8 and CD19. Protein analysis of spinal cord extracts taken from mice 6 days post-inoculation, the time of peak inflammation, reveals robust expression of IFN-γ, IL-12 and mKC. Data suggest that activated microglia and inflammatory mediators contribute to a local CNS microenvironment that regulates viral replication and IFN-γ production during the acute phase of infection, which in turn can cause phagolysosome maturation and phagocytosis of the myelin sheath, leading to demyelination.
Collapse
Affiliation(s)
- Dhriti Chatterjee
- Department of Biological Sciences, Indian Institute of Science Education and Research-Kolkata (IISER-K), Mohanpur, West Bengal, India
| | - Sankar Addya
- Kimmel Cancer Centre, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Reas S. Khan
- Scheie Eye Institute and FM Kirby Centre for Molecular Ophthalmology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Lawrence C. Kenyon
- Departments of Anatomy, Pathology and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Alexander Choe
- Departments of Neurology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Randall J. Cohrs
- Departments of Neurology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Kenneth S. Shindler
- Scheie Eye Institute and FM Kirby Centre for Molecular Ophthalmology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail: (KS); (JDS)
| | - Jayasri Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research-Kolkata (IISER-K), Mohanpur, West Bengal, India
- * E-mail: (KS); (JDS)
| |
Collapse
|
45
|
Schürch AC, Schipper D, Bijl MA, Dau J, Beckmen KB, Schapendonk CME, Raj VS, Osterhaus ADME, Haagmans BL, Tryland M, Smits SL. Metagenomic survey for viruses in Western Arctic caribou, Alaska, through iterative assembly of taxonomic units. PLoS One 2014; 9:e105227. [PMID: 25140520 PMCID: PMC4139337 DOI: 10.1371/journal.pone.0105227] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Accepted: 07/18/2014] [Indexed: 12/16/2022] Open
Abstract
Pathogen surveillance in animals does not provide a sufficient level of vigilance because it is generally confined to surveillance of pathogens with known economic impact in domestic animals and practically nonexistent in wildlife species. As most (re-)emerging viral infections originate from animal sources, it is important to obtain insight into viral pathogens present in the wildlife reservoir from a public health perspective. When monitoring living, free-ranging wildlife for viruses, sample collection can be challenging and availability of nucleic acids isolated from samples is often limited. The development of viral metagenomics platforms allows a more comprehensive inventory of viruses present in wildlife. We report a metagenomic viral survey of the Western Arctic herd of barren ground caribou (Rangifer tarandus granti) in Alaska, USA. The presence of mammalian viruses in eye and nose swabs of 39 free-ranging caribou was investigated by random amplification combined with a metagenomic analysis approach that applied exhaustive iterative assembly of sequencing results to define taxonomic units of each metagenome. Through homology search methods we identified the presence of several mammalian viruses, including different papillomaviruses, a novel parvovirus, polyomavirus, and a virus that potentially represents a member of a novel genus in the family Coronaviridae.
Collapse
Affiliation(s)
- Anita C. Schürch
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Debby Schipper
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Maarten A. Bijl
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Jim Dau
- Alaska Department of Fish and Game, Kotzebue, Alaska, United States of America
| | - Kimberlee B. Beckmen
- Alaska Department of Fish and Game, Division of Wildlife Conservation, Fairbanks, Alaska, United States of America
| | | | - V. Stalin Raj
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Albert D. M. E. Osterhaus
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
- Viroclinics Biosciences, Rotterdam, The Netherlands
| | - Bart L. Haagmans
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Morten Tryland
- Research Group for Arctic Infection Biology, Department of Arctic and Marine Biology, UiT - the Arctic University of Norway, Tromsø, Norway
- Genøk - Centre for Biosafety, Tromsø, Norway
| | - Saskia L. Smits
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
- Viroclinics Biosciences, Rotterdam, The Netherlands
| |
Collapse
|
46
|
Khan RS, Dine K, Das Sarma J, Shindler KS. SIRT1 activating compounds reduce oxidative stress mediated neuronal loss in viral induced CNS demyelinating disease. Acta Neuropathol Commun 2014; 2:3. [PMID: 24383546 PMCID: PMC3892130 DOI: 10.1186/2051-5960-2-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Accepted: 12/21/2013] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is characterized by central nervous system inflammation and demyelination, and increasing evidence demonstrates significant neuronal damage also occurs and is associated with permanent functional impairment. Current MS therapies have limited ability to prevent neuronal damage, suggesting additional neuroprotective therapies are needed. Compounds that activate the NAD+-dependent SIRT1 deacetylase prevent neuronal loss in an autoimmune-mediated MS model, but the mechanism of this effect is unknown, and it is unclear whether SIRT1 activating compounds exert similar effects in demyelinating disease induced by other etiologies. We measured neuronal loss in C57BL/6 mice inoculated with a neurotropic strain of mouse hepatitis virus, MHV-A59, that induces an MS-like disease. RESULTS Oral treatment with the SIRT1 activating compound SRTAW04 significantly increased SIRT1 activity within optic nerves and prevented neuronal loss during optic neuritis, an inflammatory demyelinating optic nerve lesion that occurs in MS and its animal models. MHV-A59 induced neuronal loss was associated with reactive oxygen species (ROS) accumulation, and SRTAW04 treatment significantly reduced ROS levels while promoting increased expression of enzymes involved in mitochondrial function and reduction of ROS. SRTAW04 exerted similar protective effects in EAE spinal cords, with decreased demyelination. CONCLUSIONS Results demonstrate that SIRT1 activating compounds prevent neuronal loss in viral-induced demyelinating disease similar to their effects in autoimmune-mediated disease. One mechanism of this neuroprotective effect involves increasing mitochondrial biogenesis with reduction of oxidative stress. SIRT1 activators represent a potential neuroprotective therapy for MS. Understanding common mechanisms of these effects in distinct disease models will help identify targets for more specific therapies.
Collapse
Affiliation(s)
- Reas S Khan
- Department of Ophthalmology, Scheie Eye Institute and FM Kirby Center for Molecular Ophthalmology, Stellar-Chance Laboratories, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kimberly Dine
- Department of Ophthalmology, Scheie Eye Institute and FM Kirby Center for Molecular Ophthalmology, Stellar-Chance Laboratories, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jayasri Das Sarma
- Department of Biological Science, Indian Institute of Science Education and Research-Kolkata (IISER-K), Mohanpur Campus Mohanpur, Nadia, West Bengal 741252, India
| | - Kenneth S Shindler
- Department of Ophthalmology, Scheie Eye Institute and FM Kirby Center for Molecular Ophthalmology, Stellar-Chance Laboratories, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
47
|
Different mechanisms of inflammation induced in virus and autoimmune-mediated models of multiple sclerosis in C57BL6 mice. BIOMED RESEARCH INTERNATIONAL 2013; 2013:589048. [PMID: 24083230 PMCID: PMC3780522 DOI: 10.1155/2013/589048] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2013] [Accepted: 07/01/2013] [Indexed: 01/17/2023]
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the human central nervous system (CNS). Neurotropic demyelinating strain of MHV (MHV-A59 or its isogenic recombinant strain RSA59) induces MS-like disease in mice mediated by microglia, along with a small population of T cells. The mechanism of demyelination is at least in part due to microglia-mediated myelin stripping, with some direct axonal injury. Immunization with myelin oligodendrocyte glycoprotein (MOG) induces experimental autoimmune encephalomyelitis (EAE), a mainly CD4+ T-cell-mediated disease, although CD8+ T cells may play a significant role in demyelination. It is possible that both autoimmune and nonimmune mechanisms such as direct viral toxicity may induce MS. Our study directly compares CNS pathology in autoimmune and viral-induced MS models. Mice with viral-induced and EAE demyelinating diseases demonstrated similar patterns and distributions of demyelination that accumulated over the course of the disease. However, significant differences in acute inflammation were noted. Inflammation was restricted mainly to white matter at all times in EAE, whereas inflammation initially largely involved gray matter in acute MHV-induced disease and then is subsequently localized only in white matter in the chronic disease phase. The presence of dual mechanisms of demyelination may be responsible for the failure of immunosuppression to promote long-term remission in many MS patients.
Collapse
|
48
|
Das Sarma J. Microglia-mediated neuroinflammation is an amplifier of virus-induced neuropathology. J Neurovirol 2013; 20:122-36. [PMID: 23979705 DOI: 10.1007/s13365-013-0188-4] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Revised: 06/14/2013] [Accepted: 07/03/2013] [Indexed: 02/07/2023]
Abstract
Microglia, the major resident immune cells in the central nervous system (CNS) are considered as the key cellular mediators of neuroinflammatory processes. In the past few years, microglial research has become a main focus in cellular neuroimmunology and neuroinflammation. Chronic/remitting neurological disease such as multiple sclerosis (MS) has long been considered an inflammatory autoimmune disease with the infiltration of peripheral myelin-specific T cells into the CNS. With the rapid advancement in the field of microglia and astrocytic neurobiology, the term neuroinflammation progressively started to denote chronic CNS cell-specific inflammation in MS. The direct glial responses in MS are different from conventional peripheral immune responses. This review attempts to summarize current findings of neuroinflammatory responses within the CNS by direct infection of neural cells by mouse hepatitis virus (MHV) and the mechanisms by which glial cell responses ultimately contribute to the neuropathology on demyelination. Microglia can be persistently infected by MHV. Microglial activation and phagocytosis are recognized to be critically important in the pathogenesis of demyelination. Emerging evidence for the pathogenic role of microglia and the activation of inflammatory pathways in these cells in MHV infection supports the concept that microglia induced neuroinflammation is an amplifier of virus-induced neuropathology.
Collapse
Affiliation(s)
- Jayasri Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research, Kolkata, India,
| |
Collapse
|
49
|
Microglia play a major role in direct viral-induced demyelination. Clin Dev Immunol 2013; 2013:510396. [PMID: 23864878 PMCID: PMC3705805 DOI: 10.1155/2013/510396] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Revised: 05/14/2013] [Accepted: 05/15/2013] [Indexed: 11/17/2022]
Abstract
Microglia are the resident macrophage-like populations in the central nervous system (CNS). Microglia remain quiescent, unable to perform effector and antigen presentation (APC) functions until activated by injury or infection, and have been suggested to represent the first line of defence for the CNS. Previous studies demonstrated that microglia can be persistently infected by neurotropic mouse hepatitis virus (MHV) which causes meningoencephalitis, myelitis with subsequent axonal loss, and demyelination and serve as a virus-induced model of human neurological disease multiple sclerosis (MS). Current studies revealed that MHV infection is associated with the pronounced activation of microglia during acute inflammation, as evidenced by characteristic changes in cellular morphology and increased expression of microglia-specific proteins, Iba1 (ionized calcium-binding adaptor molecule 1), which is a macrophage/microglia-specific novel calcium-binding protein and involved in membrane ruffling and phagocytosis. During chronic inflammation (day 30 postinfection), microglia were still present within areas of demyelination. Experiments performed in ex vivo spinal cord slice culture and in vitro neonatal microglial culture confirmed direct microglial infection. Our results suggest that MHV can directly infect and activate microglia during acute inflammation, which in turn during chronic inflammation stage causes phagocytosis of myelin sheath leading to chronic inflammatory demyelination.
Collapse
|
50
|
Kipp M, van der Star B, Vogel DYS, Puentes F, van der Valk P, Baker D, Amor S. Experimental in vivo and in vitro models of multiple sclerosis: EAE and beyond. Mult Scler Relat Disord 2011; 1:15-28. [PMID: 25876447 DOI: 10.1016/j.msard.2011.09.002] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Accepted: 09/05/2011] [Indexed: 12/16/2022]
Abstract
Although the primary cause of multiple sclerosis (MS) is unknown, the widely accepted view is that aberrant (auto)immune responses possibly arising following infection(s) are responsible for the destructive inflammatory demyelination and neurodegeneration in the central nervous system (CNS). This notion, and the limited access of human brain tissue early in the course of MS, has led to the development of autoimmune, viral and toxin-induced demyelination animal models as well as the development of human CNS cell and organotypic brain slice cultures in an attempt to understand events in MS. The autoimmune models, collectively known as experimental autoimmune encephalomyelitis (EAE), and viral models have shaped ideas of how environmental factors may trigger inflammation, demyelination and neurodegeneration in the CNS. Understandably, these models have also heavily influenced the development of therapies targeting the inflammatory aspect of MS. Demyelination and remyelination in the absence of overt inflammation are better studied in toxin-induced demyelination models using cuprizone and lysolecithin. The paradigm shift of MS as an autoimmune disease of myelin to a neurodegenerative disease has required more appropriate models reflecting the axonal and neuronal damage. Thus, secondary progressive EAE and spastic models have been crucial to develop neuroprotective approaches. In this review the current in vivo and in vitro experimental models to examine pathological mechanisms involved in inflammation, demyelination and neuronal degeneration, as well as remyelination and repair in MS are discussed. Since this knowledge is the basis for the development of new therapeutic approaches for MS, we particularly address whether the currently available models truly reflect the human disease, and discuss perspectives to further optimise and develop more suitable experimental models to study MS.
Collapse
Affiliation(s)
- Markus Kipp
- Department of Pathology, VU University Medical Centre, PO Box 7057, 1007 MB Amsterdam, The Netherlands; Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Baukje van der Star
- Department of Pathology, VU University Medical Centre, PO Box 7057, 1007 MB Amsterdam, The Netherlands
| | - Daphne Y S Vogel
- Department of Pathology, VU University Medical Centre, PO Box 7057, 1007 MB Amsterdam, The Netherlands; Department of Molecular Cell Biology and Immunology, VU University Medical Centre, Amsterdam, The Netherlands
| | - Fabìola Puentes
- Neuroimmunology Unit, Blizard Institute, Queen Mary University of London, Barts and The London School of Medicine and Dentistry, London, UK
| | - Paul van der Valk
- Department of Pathology, VU University Medical Centre, PO Box 7057, 1007 MB Amsterdam, The Netherlands
| | - David Baker
- Neuroimmunology Unit, Blizard Institute, Queen Mary University of London, Barts and The London School of Medicine and Dentistry, London, UK
| | - Sandra Amor
- Department of Pathology, VU University Medical Centre, PO Box 7057, 1007 MB Amsterdam, The Netherlands; Neuroimmunology Unit, Blizard Institute, Queen Mary University of London, Barts and The London School of Medicine and Dentistry, London, UK.
| |
Collapse
|