1
|
Nguyen NM, Conrady CD. A Better Understanding of the Clinical and Pathological Changes in Viral Retinitis: Steps to Improve Visual Outcomes. Microorganisms 2024; 12:2513. [PMID: 39770716 PMCID: PMC11678148 DOI: 10.3390/microorganisms12122513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/03/2024] [Accepted: 12/03/2024] [Indexed: 01/11/2025] Open
Abstract
Infectious retinitis, though rare, poses a significant threat to vision, often leading to severe and irreversible damage. Various pathogens, including viruses, bacteria, tick-borne agents, parasites, and fungi, can cause this condition. Among these, necrotizing herpetic retinitis represents a critical spectrum of retinal infections primarily caused by herpes viruses such as varicella-zoster virus (VZV), herpes simplex virus (HSV), and cytomegalovirus (CMV). This review underscores the retina's susceptibility to viral infections, focusing on the molecular mechanisms through which herpetic viruses invade and damage retinal tissue, supported by clinical and preclinical evidence. We also identify existing knowledge gaps and propose future research directions to deepen our understanding and improve therapeutic outcomes.
Collapse
Affiliation(s)
- Nghi M. Nguyen
- Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Christopher D. Conrady
- Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
2
|
Letafati A, Jazayeri SM, Atwan H, Mahmoudi MK, Sarrafzadeh S, Ardekani OS, Norouzi M, Ghaziasadi A. Utilization of multiplex polymerase chain reaction for simultaneous and rapid detection of viral infections from different ocular structures. Sci Rep 2024; 14:17997. [PMID: 39097632 PMCID: PMC11297968 DOI: 10.1038/s41598-024-68171-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 07/22/2024] [Indexed: 08/05/2024] Open
Abstract
The impact of viral keratitis (VK) on individuals and society is notable. Early diagnosis and treatment are crucial in managing viral keratitis effectively. Timely intervention with antiviral medications and supportive care can help mitigate the severity of the infection and improve visual outcomes. We examined the prevalence of varicella-zoster virus (VZV), herpes simplex virus type 1 (HSV-1), adenovirus (AdV) and herpes simplex virus type 2 (HSV-2) in patients suspected for ocular infections. Patients included in the study exhibited various clinical manifestations indicative of ocular pathology, such as infectious keratitis, corneal scar, endogenous endophthalmitis, panuveitis, endothelitis, stromal edema, and other relevant conditions. Four different types of tear fluid, corneal samples epithelium, aqueous humor and vitreous humor were taken. After genome extraction, multiplex real-time PCR was used for diagnosis of viruses. 48 (29.6%) out of the total of 162 (100%) eye specimen were positive. The dominant prevalence was VZV (12.3%) and HSV-1 (11.7%) followed by AdV (4.9%) and HSV-2 (0.6%). There were 4 (8.3%) coinfections within the samples (HSV-1 and VZV). Aqueous humor samples demonstrated superior virus detection ability and our only HSV-2 positive sample was from aqueous humor. The utilization of multiplex real-time PCR assays in differential diagnosis of VK holds promise for expeditious diagnoses while also preventing unwarranted antibiotic prescriptions. Moreover, the aqueous humor appears to be a more sensitive site for detecting viral keratitis.
Collapse
Affiliation(s)
- Arash Letafati
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran
| | - Seyed Mohammad Jazayeri
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran.
| | - Hossein Atwan
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Masoud Karkhaneh Mahmoudi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Sheida Sarrafzadeh
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran
- Epidemiology and statistics Department, Tehran University of Medical Sciences, Tehran, Iran
| | - Omid Salahi Ardekani
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran
| | - Mehdi Norouzi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran
| | - Azam Ghaziasadi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran
| |
Collapse
|
3
|
Gevezova M, Ivanov Z, Pacheva I, Timova E, Kazakova M, Kovacheva E, Ivanov I, Sarafian V. Bioenergetic and Inflammatory Alterations in Regressed and Non-Regressed Patients with Autism Spectrum Disorder. Int J Mol Sci 2024; 25:8211. [PMID: 39125780 PMCID: PMC11311370 DOI: 10.3390/ijms25158211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/22/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
Autism spectrum disorder (ASD) is associated with multiple physiological abnormalities. Current laboratory and clinical evidence most commonly report mitochondrial dysfunction, oxidative stress, and immunological imbalance in almost every cell type of the body. The present work aims to evaluate oxygen consumption rate (OCR), extracellular acidification rate (ECAR), and inflammation-related molecules such as Cyclooxygenase-2 (COX-2), chitinase 3-like protein 1 (YKL-40), Interleukin-1 beta (IL-1β), Interleukin-9 (IL-9) in ASD children with and without regression compared to healthy controls. Children with ASD (n = 56) and typically developing children (TDC, n = 12) aged 1.11 to 11 years were studied. Mitochondrial activity was examined in peripheral blood mononuclear cells (PBMCs) isolated from children with ASD and from the control group, using a metabolic analyzer. Gene and protein levels of IL-1β, IL-9, COX-2, and YKL-40 were investigated in parallel. Our results showed that PBMCs of the ASD subgroup of regressed patients (ASD R(+), n = 21) had a specific pattern of mitochondrial activity with significantly increased maximal respiration, respiratory spare capacity, and proton leak compared to the non-regressed group (ASD R(-), n = 35) and TDC. Furthermore, we found an imbalance in the studied proinflammatory molecules and increased levels in ASD R(-) proving the involvement of inflammatory changes. The results of this study provide new evidence for specific bioenergetic profiles of immune cells and elevated inflammation-related molecules in ASD. For the first time, data on a unique metabolic profile in ASD R(+) and its comparison with a random group of children of similar age and sex are provided. Our data show that mitochondrial dysfunction is more significant in ASD R(+), while in ASD R(-) inflammation is more pronounced. Probably, in the group without regression, immune mechanisms (immune dysregulation, leading to inflammation) begin initially, and at a later stage mitochondrial activity is also affected under exogenous factors. On the other hand, in the regressed group, the initial damage is in the mitochondria, and perhaps at a later stage immune dysfunction is involved.
Collapse
Affiliation(s)
- Maria Gevezova
- Department of Medical Biology, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria; (M.G.); (Z.I.); (M.K.); (E.K.)
- Research Institute at MU-Plovdiv, 4002 Plovdiv, Bulgaria
| | - Zdravko Ivanov
- Department of Medical Biology, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria; (M.G.); (Z.I.); (M.K.); (E.K.)
| | - Iliana Pacheva
- Department of Pediatrics and Medical Genetics, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria; (I.P.); (I.I.)
- Pediatrics Clinic, St. George University Hospital, 4002 Plovdiv, Bulgaria;
| | - Elena Timova
- Pediatrics Clinic, St. George University Hospital, 4002 Plovdiv, Bulgaria;
| | - Maria Kazakova
- Department of Medical Biology, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria; (M.G.); (Z.I.); (M.K.); (E.K.)
- Research Institute at MU-Plovdiv, 4002 Plovdiv, Bulgaria
| | - Eleonora Kovacheva
- Department of Medical Biology, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria; (M.G.); (Z.I.); (M.K.); (E.K.)
- Research Institute at MU-Plovdiv, 4002 Plovdiv, Bulgaria
| | - Ivan Ivanov
- Department of Pediatrics and Medical Genetics, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria; (I.P.); (I.I.)
- Pediatrics Clinic, St. George University Hospital, 4002 Plovdiv, Bulgaria;
| | - Victoria Sarafian
- Department of Medical Biology, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria; (M.G.); (Z.I.); (M.K.); (E.K.)
- Research Institute at MU-Plovdiv, 4002 Plovdiv, Bulgaria
| |
Collapse
|
4
|
Feng S, Liu Y, Zhou Y, Shu Z, Cheng Z, Brenner C, Feng P. Mechanistic insights into the role of herpes simplex virus 1 in Alzheimer's disease. Front Aging Neurosci 2023; 15:1245904. [PMID: 37744399 PMCID: PMC10512732 DOI: 10.3389/fnagi.2023.1245904] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 08/21/2023] [Indexed: 09/26/2023] Open
Abstract
Alzheimer's Disease (AD) is an aging-associated neurodegenerative disorder, threatening millions of people worldwide. The onset and progression of AD can be accelerated by environmental risk factors, such as bacterial and viral infections. Human herpesviruses are ubiquitous infectious agents that underpin numerous inflammatory disorders including neurodegenerative diseases. Published studies concerning human herpesviruses in AD imply an active role HSV-1 in the pathogenesis of AD. This review will summarize the current understanding of HSV-1 infection in AD and highlight some barriers to advance this emerging field.
Collapse
Affiliation(s)
- Shu Feng
- Department of Diabetes and Cancer Metabolism, City of Hope National Medical Center, Duarte, CA, United States
| | - Yongzhen Liu
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, United States
| | - Yu Zhou
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, United States
| | - Zhenfeng Shu
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, United States
| | - Zhuxi Cheng
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, United States
- International Department, Beijing Bayi School, Beijing, China
| | - Charles Brenner
- Department of Diabetes and Cancer Metabolism, City of Hope National Medical Center, Duarte, CA, United States
| | - Pinghui Feng
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, United States
- Department of Molecular Microbiology and Immunology, Norris Comprehensive Cancer Center, Los Angeles, CA, United States
| |
Collapse
|
5
|
Dempsey MP, Conrady CD. The Host-Pathogen Interplay: A Tale of Two Stories within the Cornea and Posterior Segment. Microorganisms 2023; 11:2074. [PMID: 37630634 PMCID: PMC10460047 DOI: 10.3390/microorganisms11082074] [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: 06/05/2023] [Revised: 08/10/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
Ocular infectious diseases are an important cause of potentially preventable vision loss and blindness. In the following manuscript, we will review ocular immunology and the pathogenesis of herpesviruses and Pseudomonas aeruginosa infections of the cornea and posterior segment. We will highlight areas of future research and what is currently known to promote bench-to-bedside discoveries to improve clinical outcomes of these debilitating ocular diseases.
Collapse
Affiliation(s)
- Michael P. Dempsey
- Department of Ophthalmology and Visual Sciences, Truhlsen Eye Center, University of Nebraska Medical Center, Omaha, NE 68105, USA
| | - Christopher D. Conrady
- Department of Ophthalmology and Visual Sciences, Truhlsen Eye Center, University of Nebraska Medical Center, Omaha, NE 68105, USA
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
6
|
Lu H, Liao Y, Zhang C, Wen W, Du Y, Zhao M, Wang L. A case of herpes simplex virus induced peripheral neuropathy and encephalitis with positive GM3 and CASPR2 antibody. BMC Neurol 2023; 23:199. [PMID: 37210504 DOI: 10.1186/s12883-023-03238-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 05/04/2023] [Indexed: 05/22/2023] Open
Abstract
BACKGROUND We reported on a case involving an older patient with HSV-1 encephalitis who simultaneously experienced the onset of peripheral nerve symptoms associated with the presence of anti-GM3 immunoglobulin G (IgG). CASE PRESENTATION A 77-year-old male was admitted to hospital with high fever, weakness of both lower limbs, and an unstable gait. A CSF test revealed a strikingly increased protein level (1,002 mg/L, normative values: 150-450 mg/L) and MRI revealed hyper-signal lesions in the right temporal lobe, right hippocampus, right insula, and right cingulate gyrus. The CSF was positive for HSV PCR (HSV-1,17870). In addition, the serum samples were positive for CASPR2 antibodies (antibody titer: 1/10) and anti-GM3 immunoglobulin G (IgG) (+). The patient was diagnosed with HSV-1-induced peripheral nerve symptoms that were associated with encephalitis and the presence of anti-GM3 IgG and anti-CASPR2 antibodies. The patient had received included intravenous immunoglobulin, intravenous acyclovir, and corticosteroids therapy. At the one-year follow-up examination, he had regained the necessary skills associated with daily life. CONCLUSIONS Herpes simplex virus infection often induces encephalitis, and reaction to the virus may trigger an autoimmune response. Early diagnosis and treatment can avoid the progression of the disease to include autoimmune encephalitis.
Collapse
Affiliation(s)
- Hongji Lu
- The Neurological Intensive Care Unit of Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510120, China
- The Second Clinical School, Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Yingdi Liao
- The Rehabilitation Department, Kunming Municipal Hospital of Traditional Chinese Medicine, Kunming, 650000, China
| | - Changlin Zhang
- The Second Clinical School, Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
- Department of Stroke Center, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Wanxin Wen
- The Neurological Intensive Care Unit of Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510120, China
- The Second Clinical School, Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Yaming Du
- The Second Clinical School, Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Min Zhao
- The Second Clinical School, Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
- The Encephallopathy Department.1 of Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510120, China
| | - Lixin Wang
- The Neurological Intensive Care Unit of Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510120, China.
- The Second Clinical School, Guangzhou University of Chinese Medicine, Guangzhou, 510120, China.
| |
Collapse
|
7
|
Conrady CD. Infectious Eye Diseases and Prevention Control. Microorganisms 2023; 11:1286. [PMID: 37317260 DOI: 10.3390/microorganisms11051286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 05/08/2023] [Indexed: 06/16/2023] Open
Abstract
Ocular infections are rare but can be unfortunate, vision-threatening conditions that can affect any part of the eye, from the outer tissues including the episcleral, sclera, and cornea to inside the eye such as the anterior chamber, vitreous, optic nerve, and retina [...].
Collapse
Affiliation(s)
- Christopher D Conrady
- Department of Ophthalmology and Visual Sciences, Stanley M. Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, NE 68105, USA
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68105, USA
| |
Collapse
|
8
|
The Dilemma of HSV-1 Oncolytic Virus Delivery: The Method Choice and Hurdles. Int J Mol Sci 2023; 24:ijms24043681. [PMID: 36835091 PMCID: PMC9962028 DOI: 10.3390/ijms24043681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/03/2023] [Accepted: 02/08/2023] [Indexed: 02/15/2023] Open
Abstract
Oncolytic viruses (OVs) have emerged as effective gene therapy and immunotherapy drugs. As an important gene delivery platform, the integration of exogenous genes into OVs has become a novel path for the advancement of OV therapy, while the herpes simplex virus type 1 (HSV-1) is the most commonly used. However, the current mode of administration of HSV-1 oncolytic virus is mainly based on the tumor in situ injection, which limits the application of such OV drugs to a certain extent. Intravenous administration offers a solution to the systemic distribution of OV drugs but is ambiguous in terms of efficacy and safety. The main reason is the synergistic role of innate and adaptive immunity of the immune system in the response against the HSV-1 oncolytic virus, which is rapidly cleared by the body's immune system before it reaches the tumor, a process that is accompanied by side effects. This article reviews different administration methods of HSV-1 oncolytic virus in the process of tumor treatment, especially the research progress in intravenous administration. It also discusses immune constraints and solutions of intravenous administration with the intent to provide new insights into HSV-1 delivery for OV therapy.
Collapse
|
9
|
Fan S, Yoo JH, Park G, Yeh S, Conrady CD. Type I Interferon Signaling Is Critical During the Innate Immune Response to HSV-1 Retinal Infection. Invest Ophthalmol Vis Sci 2022; 63:28. [PMID: 36583876 PMCID: PMC9807183 DOI: 10.1167/iovs.63.13.28] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 12/10/2022] [Indexed: 12/31/2022] Open
Abstract
Purpose Acute retinal necrosis (ARN) is a herpesvirus infection of the retina with blinding complications. In this study, we sought to create a reproducible mouse model of ARN that mimics human disease to better understand innate immunity within the retina during virus infection. Methods C57Bl/6J wild type (WT) and type I interferon receptor-deficient (IFNAR-/-) mice were infected with varying amounts of herpes simplex virus type 1 (HSV-1) via subretinal injection. Viral titers, optical coherence tomography (OCT) and fundus photography, the development of encephalitis, and ocular histopathology were scored and compared between groups of WT and IFNAR-/- mice. Results The retina of WT mice could be readily infected with HSV-1 via subretinal injection resulting in retinal whitening and full-thickness necrosis as determined by in vivo imaging and histopathology. In IFNAR-/- mice, HSV-1-induced retinal pathology was significantly worse when compared with WT mice, and viral titers were significantly elevated within two days after infection and persisted to day 5 after infection within the retina. These results were also observed in the brain where there were significantly higher viral titers and frequency of encephalitis in IFNAR-/- when compared to WT mice. Conclusions Collectively, these findings show that our new mouse model of ARN mimics human disease and can be used to study innate immunity within the retina. We conclude that type I interferons are critical in containing HSV-1 locally within retinal tissues and prohibiting spread into the brain.
Collapse
Affiliation(s)
- Shan Fan
- Department of Ophthalmology and Visual Sciences, Truhlsen Eye Center, University of Nebraska Medical Center, Omaha, Nebraska, United States
| | - Jae Hyuk Yoo
- Department of Ophthalmology and Visual Sciences, Truhlsen Eye Center, University of Nebraska Medical Center, Omaha, Nebraska, United States
| | - Garam Park
- Department of Ophthalmology and Visual Sciences, Truhlsen Eye Center, University of Nebraska Medical Center, Omaha, Nebraska, United States
| | - Steven Yeh
- Department of Ophthalmology and Visual Sciences, Truhlsen Eye Center, University of Nebraska Medical Center, Omaha, Nebraska, United States
| | - Christopher D. Conrady
- Department of Ophthalmology and Visual Sciences, Truhlsen Eye Center, University of Nebraska Medical Center, Omaha, Nebraska, United States
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, United States
| |
Collapse
|
10
|
Abstract
After establishing latent infection, some viruses can be reactivated by the alteration of host immunological conditions. First, we reviewed viruses that can cause neuronal damage by reactivation. Then we focused on the herpes simplex virus (HSV). The reactivation leads to neuronal damages through two possible mechanisms; "reactivation of a latent herpes virus" by which viruses can cause direct virus neurotoxicity, and "post-infectious immune inflammatory response" by which a focal reactivation of HSV leads to an inflammatory reaction. The former is radiologically characterized by cortical lesions, the latter is characterized by subcortical white matter lesions. We experienced a female, who underwent the right posterior quadrantectomy and then developed recurrent herpes encephalitis caused by herpes simplex reactivation, which pathologically demonstrated inflammation in the white matter, suggesting a post-infectious immune inflammatory response. The patient was successfully treated with immunosuppressants. The reactivation of the HSV is extremely rare in Japan. Neurologists should recognize this condition because this disorder will increase as epilepsy surgery gains more popularity.
Collapse
Affiliation(s)
- Tomoyo Shimada
- Department of Neurology, Juntendo University School of Medicine
| | - Taiji Tsunemi
- Department of Neurology, Juntendo University School of Medicine
- Epilepsy Center, Juntendo University School of Medicine
| | - Yasushi Iimura
- Department of Neurosurgery, Juntendo University School of Medicine
- Epilepsy Center, Juntendo University School of Medicine
| | - Hidenori Sugano
- Department of Neurosurgery, Juntendo University School of Medicine
- Epilepsy Center, Juntendo University School of Medicine
| | | |
Collapse
|
11
|
Soltani Khaboushan A, Pahlevan-Fallahy MT, Shobeiri P, Teixeira AL, Rezaei N. Cytokines and chemokines profile in encephalitis patients: A meta-analysis. PLoS One 2022; 17:e0273920. [PMID: 36048783 PMCID: PMC9436077 DOI: 10.1371/journal.pone.0273920] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 08/17/2022] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Encephalitis is caused by autoimmune or infectious agents marked by brain inflammation. Investigations have reported altered concentrations of the cytokines in encephalitis. This study was conducted to determine the relationship between encephalitis and alterations of cytokine levels in cerebrospinal fluid (CSF) and serum. METHODS We found possibly suitable studies by searching PubMed, Embase, Scopus, and Web of Science, systematically from inception to August 2021. 23 articles were included in the meta-analysis. To investigate sources of heterogeneity, subgroup analysis and sensitivity analysis were conducted. The protocol of the study has been registered in PROSPERO with a registration ID of CRD42021289298. RESULTS A total of 23 met our eligibility criteria to be included in the meta-analysis. A total of 12 cytokines were included in the meta-analysis of CSF concentration. Moreover, 5 cytokines were also included in the serum/plasma concentration meta-analysis. According to the analyses, patients with encephalitis had higher CSF amounts of IL-6, IL-8, IL-10, CXCL10, and TNF-α than healthy controls. The alteration in the concentration of IL-2, IL-4, IL-17, CCL2, CXCL9, CXCL13, and IFN-γ was not significant. In addition, the serum/plasma levels of the TNF-α were increased in encephalitis patients, but serum/plasma concentration of the IL-6, IL-10, CXCL10, and CXCL13 remained unchanged. CONCLUSIONS This meta-analysis provides evidence for higher CSF concentrations of IL-6, IL-8, IL-10, CXCL10, and TNF-α in encephalitis patients compared to controls. The diagnostic and prognostic value of these cytokines and chemokines should be investigated in future studies.
Collapse
Affiliation(s)
- Alireza Soltani Khaboushan
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Students’ Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Systematic Review and Meta-Analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mohammad-Taha Pahlevan-Fallahy
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Students’ Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Systematic Review and Meta-Analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Parnian Shobeiri
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Systematic Review and Meta-Analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Non–Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Antônio L. Teixeira
- Neuropsychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States of America
| | - Nima Rezaei
- Systematic Review and Meta-Analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
Jeffries AM, Suptela AJ, Marriott I. Z-DNA binding protein 1 mediates necroptotic and apoptotic cell death pathways in murine astrocytes following herpes simplex virus-1 infection. J Neuroinflammation 2022; 19:109. [PMID: 35549723 PMCID: PMC9103380 DOI: 10.1186/s12974-022-02469-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 04/25/2022] [Indexed: 11/17/2022] Open
Abstract
Background The mechanisms by which glia respond to viral central nervous system (CNS) pathogens are now becoming apparent with the demonstration that microglia and astrocytes express an array of pattern recognition receptors that include intracellular RNA and DNA sensors. We have previously demonstrated that glia express Z-DNA binding protein 1 (ZBP1) and showed that this cytosolic nucleic acid sensor contributes to the inflammatory/neurotoxic responses of these cells to herpes simplex virus-1 (HSV-1). However, the relative contribution made by ZBP1- to HSV-1-mediated cell death in glia has not been determined. Methods We have investigated the relative contribution made by ZBP1- to HSV-1-mediated cell death in primary astrocytes derived from mice genetically deficient in this sensor. We have used capture ELISAs and immunoblot analysis to assess inflammatory cytokine production and ZBP1 and phosphorylated mixed lineage kinase domain-like protein (MLKL) expression levels, respectively, following HSV-1 challenge. Furthermore, we have used a commercially available cell viability assay to determine the proportion and rate of cell death in cells following infection with laboratory and neuroinvasive clinical strains of HSV-1, and pharmacological inhibitors of necroptotic and apoptotic pathway components to assess the relative role of each. Results We show that the loss of ZBP1 in astrocytes results in an increase in the number of viral particles released following HSV-1 infection. Importantly, we have confirmed that HSV-1 induces necroptosis in astrocytes and have established the ability of ZBP1 to mediate this cell death pathway. Interestingly, while ZBP1 is best known for its role in necroptotic signaling, our findings indicate that this sensor can also contribute to virally induced apoptosis in these glia. Conclusions Our findings indicate that ZBP1 serves as a restriction factor for HSV-1 infection and is associated with the induction of both necroptotic and apoptotic cell death pathways in primary murine astrocytes. While it remains to be seen whether ZBP1-mediated activation of cell death in astrocytes contributes significantly to host protection or, rather, exacerbates HSV-1 encephalitis pathology, the identification of such a role in resident CNS cells may represent a novel target for therapeutic intervention to reduce HSV encephalitis-associated morbidity and mortality. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02469-z.
Collapse
Affiliation(s)
- Austin M Jeffries
- Department of Biological Sciences, University of North Carolina at Charlotte, 9201 University City Blvd., Charlotte, NC, 28223, USA
| | - Alexander J Suptela
- Department of Biological Sciences, University of North Carolina at Charlotte, 9201 University City Blvd., Charlotte, NC, 28223, USA
| | - Ian Marriott
- Department of Biological Sciences, University of North Carolina at Charlotte, 9201 University City Blvd., Charlotte, NC, 28223, USA.
| |
Collapse
|
13
|
Hu X, Zeng Q, Xiao J, Qin S, Wang Y, Shan T, Hu D, Zhu Y, Liu K, Zheng K, Wang Y, Ren Z. Herpes Simplex Virus 1 Induces Microglia Gasdermin D-Dependent Pyroptosis Through Activating the NLR Family Pyrin Domain Containing 3 Inflammasome. Front Microbiol 2022; 13:838808. [PMID: 35387080 PMCID: PMC8978634 DOI: 10.3389/fmicb.2022.838808] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 01/24/2022] [Indexed: 12/13/2022] Open
Abstract
Herpes simplex virus type 1 (HSV-1) is a highly prevalent virus in humans and causes severe forms of inflammation, such as herpes simplex encephalitis (HSE). Pyroptosis is a new inflammatory cell death triggered by inflammasome and cysteine-requiring aspartate protease-1 (caspase-1) activation. Nonetheless, HSV-1 induces encephalitis, and cell death mechanisms are not understood. In this study, we confirmed for the first time that the DNA virus HSV-1 triggers Gasdermin D-dependent pyroptosis by activating NLR family pyrin domain containing 3 (NLRP3) inflammasomes in mouse microglia, leading to mature IL-1β production and active caspase-1 (p10) release. Inhibition of microglial NLRP3 inflammasome activation suppressed HSV-1-induced Gasdermin D-dependent pyroptosis. In addition, NLRP3 and IL-1β expression levels were significantly increased in the mouse model of herpes simplex encephalitis compared with normal mice without viral infection. Collectively, our data revealed that the activation of inflammasomes and GSDMD-dependent pyroptosis is the mechanism of HSV-1 inducing inflammation and provides treatment targets for viral inflammation.
Collapse
Affiliation(s)
- Xiao Hu
- Guangzhou Jinan Biomedical Research and Development Center, College of Life Science and Technology, Institute of Biomedicine, Jinan University, Guangzhou, China.,The Key Laboratory of Virology of Guangdong, Jinan University, Guangzhou, China.,Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou, China.,College of Pharmacy, Jinan University, Guangzhou, China.,Guangdong Province Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou, China.,National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou, China
| | - Qiongzhen Zeng
- Guangzhou Jinan Biomedical Research and Development Center, College of Life Science and Technology, Institute of Biomedicine, Jinan University, Guangzhou, China.,The Key Laboratory of Virology of Guangdong, Jinan University, Guangzhou, China.,Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou, China.,Guangdong Province Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou, China.,National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou, China
| | - Ji Xiao
- Guangzhou Jinan Biomedical Research and Development Center, College of Life Science and Technology, Institute of Biomedicine, Jinan University, Guangzhou, China.,The Key Laboratory of Virology of Guangdong, Jinan University, Guangzhou, China.,Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou, China.,Guangdong Province Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou, China.,National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou, China
| | - Shurong Qin
- Guangzhou Jinan Biomedical Research and Development Center, College of Life Science and Technology, Institute of Biomedicine, Jinan University, Guangzhou, China.,The Key Laboratory of Virology of Guangdong, Jinan University, Guangzhou, China.,Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou, China.,College of Pharmacy, Jinan University, Guangzhou, China.,Guangdong Province Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou, China.,National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou, China
| | - Yuan Wang
- Guangzhou Jinan Biomedical Research and Development Center, College of Life Science and Technology, Institute of Biomedicine, Jinan University, Guangzhou, China.,The Key Laboratory of Virology of Guangdong, Jinan University, Guangzhou, China.,Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou, China.,Guangdong Province Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou, China.,National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou, China
| | - Tianhao Shan
- Guangzhou Jinan Biomedical Research and Development Center, College of Life Science and Technology, Institute of Biomedicine, Jinan University, Guangzhou, China.,The Key Laboratory of Virology of Guangdong, Jinan University, Guangzhou, China.,Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou, China.,Guangdong Province Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou, China.,National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou, China
| | - Di Hu
- Guangzhou Jinan Biomedical Research and Development Center, College of Life Science and Technology, Institute of Biomedicine, Jinan University, Guangzhou, China.,The Key Laboratory of Virology of Guangdong, Jinan University, Guangzhou, China.,Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou, China.,College of Pharmacy, Jinan University, Guangzhou, China.,Guangdong Province Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou, China.,National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou, China
| | - Yexuan Zhu
- Guangzhou Jinan Biomedical Research and Development Center, College of Life Science and Technology, Institute of Biomedicine, Jinan University, Guangzhou, China.,The Key Laboratory of Virology of Guangdong, Jinan University, Guangzhou, China.,Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou, China.,Guangdong Province Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou, China.,National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou, China
| | - Kaisheng Liu
- Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Kai Zheng
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, China
| | - Yifei Wang
- Guangzhou Jinan Biomedical Research and Development Center, College of Life Science and Technology, Institute of Biomedicine, Jinan University, Guangzhou, China.,The Key Laboratory of Virology of Guangdong, Jinan University, Guangzhou, China.,Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou, China.,Guangdong Province Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou, China.,National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou, China
| | - Zhe Ren
- Guangzhou Jinan Biomedical Research and Development Center, College of Life Science and Technology, Institute of Biomedicine, Jinan University, Guangzhou, China.,The Key Laboratory of Virology of Guangdong, Jinan University, Guangzhou, China.,Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou, China.,Guangdong Province Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou, China.,National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou, China
| |
Collapse
|
14
|
Microglia activate early anti-viral responses upon HSV-1 entry into the brain to counteract development of encephalitis-like disease in mice. J Virol 2022; 96:e0131121. [PMID: 35045263 DOI: 10.1128/jvi.01311-21] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Spread of herpes simplex virus 1 (HSV1) from the periphery to the central nervous system (CNS) can lead to extensive infection and pathological inflammation in the brain, causing herpes simplex encephalitis (HSE). It has been shown that microglia, the CNS-resident macrophages, are involved in early sensing of HSV1 and an induction of antiviral responses. In addition, infiltration of peripheral immune cells may contribute to control of viral infection. In this study, we tested the effect of microglia depletion in a mouse model of HSE. Increased viral titers and increased disease severity were observed in microglia-depleted mice. The effect of microglia depletion was more pronounced in wild-type than in cGas-/- mice, revealing that this immune sensor contributes to the antiviral activity of microglia. Importantly, microglia depletion led to reduced production of type I interferon (IFN), pro-inflammatory cytokines and chemokines at early time points after viral entry into the CNS. In line with this, in vitro experiments on murine primary CNS cells demonstrated microglial presence to be essential for IFN RNA induction, and control of HSV1 replication. However, the effect of microglia depletion on expression of IFNs, and inflammatory cytokines was restricted to early time point of HSV1 entry into the CNS. There was no major alteration of infiltration of CD45-positive cells in microglia-depleted mice. Collectively, our data demonstrate a key role for microglia in controlling HSV1 replication early after viral entry into the CNS and highlight the importance of a prompt antiviral innate response to reduce the risk of HSE development. Importance One of the most devastating and acute neurological conditions is encephalitis, i.e. inflammation of brain tissue. Herpes simplex virus 1 (HSV1) is a highly prevalent pathogen in humans, and the most frequent cause of viral sporadic encephalitis, called herpes simplex encephalitis (HSE). HSV1 has the ability to infect peripheral neurons and reach the central nervous system (CNS) of humans, where it can be detected by brain resident cells and infiltrating immune cells, leading to protective and damaging immune responses. In this study, we investigated the effects of a depletion of microglia, the main brain-resident immune cell type. For this purpose, we used a mouse model of HSE. We found that viral levels increased and disease symptoms worsened in microglia-depleted mice. In addition, mice lacking a major sensor of viral DNA, cGAS, manifested more pronounced disease than wild-type mice, highlighting the importance of this immune sensor in the activity of microglia. Evidently, microglia depletion led to a reduced production of many known antiviral factors, most notably type I interferon (IFN). The importance of microglia in the early control of HSV1 spread and the generation of antiviral responses is further demonstrated by experiments on murine mixed glial cell cultures. Interestingly, mice with microglia depletion exhibited an unaltered activation of antiviral responses and recruitment of immune cells from the periphery at later time points of infection, but this did not prevent the development of the disease. Overall, the data highlight the importance of a rapid activation of the host defense, with microglia playing a critical role in controlling HSV1 infection, which eventually prevents damage to neurons and brain tissue.
Collapse
|
15
|
Feige L, Zaeck LM, Sehl-Ewert J, Finke S, Bourhy H. Innate Immune Signaling and Role of Glial Cells in Herpes Simplex Virus- and Rabies Virus-Induced Encephalitis. Viruses 2021; 13:2364. [PMID: 34960633 PMCID: PMC8708193 DOI: 10.3390/v13122364] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 11/12/2021] [Accepted: 11/18/2021] [Indexed: 12/19/2022] Open
Abstract
The environment of the central nervous system (CNS) represents a double-edged sword in the context of viral infections. On the one hand, the infectious route for viral pathogens is restricted via neuroprotective barriers; on the other hand, viruses benefit from the immunologically quiescent neural environment after CNS entry. Both the herpes simplex virus (HSV) and the rabies virus (RABV) bypass the neuroprotective blood-brain barrier (BBB) and successfully enter the CNS parenchyma via nerve endings. Despite the differences in the molecular nature of both viruses, each virus uses retrograde transport along peripheral nerves to reach the human CNS. Once inside the CNS parenchyma, HSV infection results in severe acute inflammation, necrosis, and hemorrhaging, while RABV preserves the intact neuronal network by inhibiting apoptosis and limiting inflammation. During RABV neuroinvasion, surveilling glial cells fail to generate a sufficient type I interferon (IFN) response, enabling RABV to replicate undetected, ultimately leading to its fatal outcome. To date, we do not fully understand the molecular mechanisms underlying the activation or suppression of the host inflammatory responses of surveilling glial cells, which present important pathways shaping viral pathogenesis and clinical outcome in viral encephalitis. Here, we compare the innate immune responses of glial cells in RABV- and HSV-infected CNS, highlighting different viral strategies of neuroprotection or Neuroinflamm. in the context of viral encephalitis.
Collapse
Affiliation(s)
- Lena Feige
- Institut Pasteur, Université de Paris, Lyssavirus Epidemiology and Neuropathology, 28 Rue Du Docteur Roux, 75015 Paris, France;
| | - Luca M. Zaeck
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut (FLI), Federal Institute of Animal Health, Südufer 10, 17493 Greifswald-Insel Riems, Germany; (L.M.Z.); (S.F.)
| | - Julia Sehl-Ewert
- Department of Experimental Animal Facilities and Biorisk Management, Friedrich-Loeffler-Institut (FLI), Federal Institute of Animal Health, Südufer 10, 17493 Greifswald-Insel Riems, Germany;
| | - Stefan Finke
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut (FLI), Federal Institute of Animal Health, Südufer 10, 17493 Greifswald-Insel Riems, Germany; (L.M.Z.); (S.F.)
| | - Hervé Bourhy
- Institut Pasteur, Université de Paris, Lyssavirus Epidemiology and Neuropathology, 28 Rue Du Docteur Roux, 75015 Paris, France;
| |
Collapse
|
16
|
Mesquita LP, Costa RC, Mesquita LLR, Lara MDCCSH, Villalobos EMC, Mori CMC, Mori E, Howerth EW, Maiorka PC. Pathogenesis of Equid Alphaherpesvirus 1 Infection in the Central Nervous System of Mice. Vet Pathol 2021; 58:1075-1085. [PMID: 34128432 DOI: 10.1177/03009858211020670] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Equid alphaherpesvirus 1 (EHV-1) causes myeloencephalopathy in horses and occasionally in non-equid species. Although mouse models have been developed to understand EHV-1 pathogenesis, few EHV-1 strains have been identified as highly neurovirulent to mice. The aim of this study was to evaluate the pathogenesis of 2 neurovirulent EHV-1 strains in mice, and to characterize the inflammatory cells and expression of chemokines and the apoptosis marker caspase-3 in the brain of infected mice. C57BL/6J mice were inoculated intranasally with EHV-1 strains A4/72 or A9/92 and evaluated on 1, 2, and 3 days post inoculation (DPI). EHV-1-infected mice showed severe neurological signs at 3 DPI. Ultrastructural analysis revealed numerous viral nucleocapsids and fewer enveloped virions within degenerated and necrotic neurons and in the surrounding neuropil. Histologically, at 3 DPI, there was severe diffuse neuronal degeneration and liquefactive necrosis, prominent microgliosis, and perivascular cuffing composed of CD3+ cells (T cells) and Iba-1+ cells (macrophages), mainly in the olfactory bulb and ventral portions of the brain. In these areas, moderate numbers of neuroglial cells expressed CCL5 and CCL2 chemokines. Numerous neurons, including those in less affected areas, were immunolabeled for cleaved caspase-3. In conclusion, neurovirulent EHV-1 strains induced a fulminant necrotizing lymphohistiocytic meningoencephalitis in mice, with microgliosis and expression of chemokines and caspase-3. This model will be useful for understanding the mechanisms underlying the extensive neuropathology induced by these viral infections.
Collapse
Affiliation(s)
- Leonardo P Mesquita
- 28133University of Sao Paulo, Sao Paulo, Brazil
- 1355University of Georgia, Athens, GA, USA
| | | | | | | | | | | | - Enio Mori
- 27058Pasteur Institute, Sao Paulo, Brazil
| | | | | |
Collapse
|
17
|
Beier KT. The Serendipity of Viral Trans-Neuronal Specificity: More Than Meets the Eye. Front Cell Neurosci 2021; 15:720807. [PMID: 34671244 PMCID: PMC8521040 DOI: 10.3389/fncel.2021.720807] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 09/07/2021] [Indexed: 12/25/2022] Open
Abstract
Trans-neuronal viruses are frequently used as neuroanatomical tools for mapping neuronal circuits. Specifically, recombinant one-step rabies viruses (RABV) have been instrumental in the widespread application of viral circuit mapping, as these viruses have enabled labs to map the direct inputs onto defined cell populations. Within the neuroscience community, it is widely believed that RABV spreads directly between neurons via synaptic connections, a hypothesis based principally on two observations. First, the virus labels neurons in a pattern consistent with known anatomical connectivity. Second, few glial cells appear to be infected following RABV injections, despite the fact that glial cells are abundant in the brain. However, there is no direct evidence that RABV can actually be transmitted through synaptic connections. Here we review the immunosubversive mechanisms that are critical to RABV’s success for infiltration of the central nervous system (CNS). These include interfering with and ultimately killing migratory T cells while maintaining levels of interferon (IFN) signaling in the brain parenchyma. Finally, we critically evaluate studies that support or are against synaptically-restricted RABV transmission and the implications of viral-host immune responses for RABV transmission in the brain.
Collapse
Affiliation(s)
- Kevin Thomas Beier
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
18
|
Krzyzowska M, Kowalczyk A, Skulska K, Thörn K, Eriksson K. Fas/FasL Contributes to HSV-1 Brain Infection and Neuroinflammation. Front Immunol 2021; 12:714821. [PMID: 34526992 PMCID: PMC8437342 DOI: 10.3389/fimmu.2021.714821] [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: 05/25/2021] [Accepted: 08/16/2021] [Indexed: 12/14/2022] Open
Abstract
The Fas/FasL pathway plays a key role in immune homeostasis and immune surveillance. In the central nervous system (CNS) Fas/FasL is involved in axonal outgrowth and adult neurogenesis. However, little is known about the role of the Fas/FasL pathway in herpes encephalitis. In this study, we used a neuropathogenic clinical strain of herpes simplex virus type 1 (HSV-1) to explore infection-induced inflammation and immune responses in the mouse brain and the role of Fas/FasL in antiviral CNS immunity. HSV-1 CNS infection induced the infiltration of Fas- FasL-bearing monocytes and T cells in the brain and also to an up-regulation of Fas and FasL expression on resident astrocytes and microglia within infected sites. Upon infection, Fas- and FasL-deficient mice (lpr and gld) were partially protected from encephalitis with a decreased morbidity and mortality compared to WT mice. Fas/FasL deficiency promoted cell-mediated immunity within the CNS. Fas receptor stimulation abrogated HSV-1 induced activation and inflammatory reactions in microglia from WT mice, while lack of Fas or FasL led to a more pronounced activation of monocytes and microglia and also to an enhanced differentiation of these cells into a pro-inflammatory M1 phenotype. Furthermore, the specific immune system was more efficient in Fas- and FasL-deficient mice with significantly higher numbers of infiltrating HSV-1-specific cytotoxic T cells in the brain. Our data indicate that the Fas/FasL pathway leads to excessive neuroinflammation during HSV-1 infection, which is associated with a diminished anti-viral response and an excessive neuroinflammation.
Collapse
Affiliation(s)
- Malgorzata Krzyzowska
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Virology and Cell Biology, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wroclaw, Poland.,Laboratory of Nanobiology and Biomaterials, Military Institute of Hygiene and Epidemiology, Warsaw, Poland
| | - Andrzej Kowalczyk
- Department of Virology and Cell Biology, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wroclaw, Poland
| | - Katarzyna Skulska
- Department of Virology and Cell Biology, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wroclaw, Poland
| | - Karolina Thörn
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Kristina Eriksson
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
19
|
Wang J, Cheng Y, Ma Y, Wu R, Xu Y, Yang S, Wang Y, Lin Y. Cytokines and chemokines expression pattern in herpes simplex virus type-1 encephalitis. Neurosci Lett 2021; 763:136170. [PMID: 34391869 DOI: 10.1016/j.neulet.2021.136170] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 08/08/2021] [Accepted: 08/10/2021] [Indexed: 01/20/2023]
Abstract
To explore the differently expressed cytokines and chemokines to understand the pathways that lead to herpes simplex encephalitis (HSE). Mice in the experimental group were inoculated intracranially with HSV-1. A high-throughput cytokine chip assay was employed to assess the expression of cytokines/chemokines in the mice brain. GO, KEGG, and PPIs analyses were used to investigate the biological process (BP), pathways and interaction network of the differently expressed proteins (DEPs) in HSE. 13 DEPs and various proteins-related signal pathways were identified in HSE, including three new factors (IL-1α, MIP-1γ, and sTNF RI). The proteins were mainly implicated in leukocyte activation and chemotaxis. Additionally, the DEPs constituted a pivotal protein interaction network where IL-6 might be a mediator. 13 DEPs and a series of related signal pathways were associated with the pathophysiological mechanisms responsible for HSE. IL-6 might be a key mediator in the inflammatory responses to the disease.
Collapse
Affiliation(s)
- Jiaojiao Wang
- School of Tropical Medicine and Laboratory Medicine, Hainan Medical University, Haikou, Hainan 571199, China
| | - Yi Cheng
- School of Tropical Medicine and Laboratory Medicine, Hainan Medical University, Haikou, Hainan 571199, China
| | - Yueting Ma
- School of Tropical Medicine and Laboratory Medicine, Hainan Medical University, Haikou, Hainan 571199, China
| | - Rihong Wu
- School of Tropical Medicine and Laboratory Medicine, Hainan Medical University, Haikou, Hainan 571199, China
| | - Yu Xu
- School of Tropical Medicine and Laboratory Medicine, Hainan Medical University, Haikou, Hainan 571199, China
| | - Shuling Yang
- School of Tropical Medicine and Laboratory Medicine, Hainan Medical University, Haikou, Hainan 571199, China
| | - Yongxia Wang
- School of Tropical Medicine and Laboratory Medicine, Hainan Medical University, Haikou, Hainan 571199, China.
| | - Yingzi Lin
- School of Tropical Medicine and Laboratory Medicine, Hainan Medical University, Haikou, Hainan 571199, China.
| |
Collapse
|
20
|
Duarte LF, Reyes A, Farías MA, Riedel CA, Bueno SM, Kalergis AM, González PA. Crosstalk Between Epithelial Cells, Neurons and Immune Mediators in HSV-1 Skin Infection. Front Immunol 2021; 12:662234. [PMID: 34012447 PMCID: PMC8126613 DOI: 10.3389/fimmu.2021.662234] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 04/06/2021] [Indexed: 12/12/2022] Open
Abstract
Herpes simplex virus type 1 (HSV-1) infection is highly prevalent in humans, with approximately two-thirds of the world population living with this virus. However, only a fraction of those carrying HSV-1, which elicits lifelong infections, are symptomatic. HSV-1 mainly causes lesions in the skin and mucosae but reaches the termini of sensory neurons innervating these tissues and travels in a retrograde manner to the neuron cell body where it establishes persistent infection and remains in a latent state until reactivated by different stimuli. When productive reactivations occur, the virus travels back along axons to the primary infection site, where new rounds of replication are initiated in the skin, in recurrent or secondary infections. During this process, new neuron infections occur. Noteworthy, the mechanisms underlying viral reactivations and the exit of latency are somewhat poorly understood and may be regulated by a crosstalk between the infected neurons and components of the immune system. Here, we review and discuss the immune responses that occur at the skin during primary and recurrent infections by HSV-1, as well as at the interphase of latently-infected neurons. Moreover, we discuss the implications of neuronal signals over the priming and migration of immune cells in the context of HSV-1 infection.
Collapse
Affiliation(s)
- Luisa F Duarte
- Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Antonia Reyes
- Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Mónica A Farías
- Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Claudia A Riedel
- Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Susan M Bueno
- Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Endocrinología, Facultad de Medicina, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A González
- Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
21
|
Campos EMN, Rodrigues LD, Oliveira LF, Dos Santos JCC. Dementia and cognitive impairment in adults as sequels of HSV-1-related encephalitis: a review. Dement Neuropsychol 2021; 15:164-172. [PMID: 34345357 PMCID: PMC8283880 DOI: 10.1590/1980-57642021dn15-020002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 02/01/2021] [Indexed: 11/24/2022] Open
Abstract
Considering the variety of mechanisms of Herpes simplex virus (HSV-1) contamination and its broad invasive potential of the nervous system, a life-long latent infection is established. Infected adult individuals may be susceptible to viral reactivation when under the influence of multiple stressors, especially regarding immunocompromised patients. This guides a series of neuroinflammatory events on the cerebral cortex, culminating, rarely, in encephalitis and cytotoxic / vasogenic brain edema. A sum of studies of such processes provides an explanation, even though not yet completely clarified, on how the clinical evolution to cognitive impairment and dementia might be enabled. In addition, it is of extreme importance to recognize the current dementia and cognitive deficit worldwide panorama. The aim of this literature review is to elucidate the available data upon the pathophysiology of HSV-1 infection as well as to describe the clinical panorama of the referred afflictions.
Collapse
Affiliation(s)
| | - Laís Damasceno Rodrigues
- Neuroscience Laboratory, Department of Neurology and Neurosurgery, Federal University of São Paulo, São Paulo, SP, Brazil
| | - Leandro Freitas Oliveira
- Neuroscience Laboratory, Department of Neurology and Neurosurgery, Federal University of São Paulo, São Paulo, SP, Brazil
| | - Júlio César Claudino Dos Santos
- Neuroscience Laboratory, Department of Neurology and Neurosurgery, Federal University of São Paulo, São Paulo, SP, Brazil.,Faculty of Medicine, Christus University Center, Fortaleza, CE, Brazil
| |
Collapse
|
22
|
Mesquita LP, Costa RC, Zanatto DA, Bruhn FRP, Mesquita LLR, Lara MCCSH, Villalobos EMC, Massoco CO, Mori CMC, Mori E, Maiorka PC. Equine herpesvirus 1 elicits a strong pro-inflammatory response in the brain of mice. J Gen Virol 2021; 102. [PMID: 33528354 DOI: 10.1099/jgv.0.001556] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Equine herpesvirus type 1 (EHV-1) is an emerging pathogen that causes encephalomyelitis in horses and non-equid species. Several aspects of the immune response in the central nervous system (CNS), mainly regarding the role of inflammatory mediators during EHV-1 encephalitis, remain unknown. Moreover, understanding the mechanisms underlying extensive neuropathology induced by viruses would be helpful to establish therapeutic strategies. Therefore, we aimed to evaluate some aspects of the innate immune response during highly neurovirulent EHV-1 infection. C57BL/6 mice infected intranasally with A4/72 and A9/92 EHV-1 strains developed a fulminant neurological disease at 3 days post-inoculation with high viral titres in the brain. These mice developed severe encephalitis with infiltration of monocytes and CD8+ T cells to the brain. The inflammatory infiltrate followed the detection of the chemokines CCL2, CCL3, CCL4, CCL5, CXCL2, CXCL9 and CXCL-10 in the brain. Notably, the levels of CCL3, CCL4, CCL5 and CXCL9 were higher in A4/72-infected mice, which presented higher numbers of inflammatory cells within the CNS. Pro-inflammatory cytokines, such as interleukins (ILs) IL-1α, IL-1β, IL-6, IL-12β, and tumour necrosis factor (TNF), were also detected in the CNS, and Toll-like receptor (TLR) TLR2, TLR3 and TLR9 genes were also upregulated within the brain of EHV-1-infected mice. However, no expression of interferon-γ (IFN-γ) and IL-12α, which are important for controlling the replication of other herpesviruses, was detected in EHV-1-infected mice. The results show that the activated innate immune mechanisms could not prevent EHV-1 replication within the CNS, but most likely contributed to the extensive neuropathology. The mouse model of viral encephalitis proposed here will also be useful to study the mechanisms underlying extensive neuropathology.
Collapse
Affiliation(s)
- Leonardo P Mesquita
- Department of Pathology, Faculty of Veterinary Medicine and Animal Sciences, University of São Paulo, Av. Professor Dr Orlando Marques de Paiva, 87, São Paulo, SP, 5508-010, Brazil
| | - Rafael C Costa
- Department of Pathology, Faculty of Veterinary Medicine and Animal Sciences, University of São Paulo, Av. Professor Dr Orlando Marques de Paiva, 87, São Paulo, SP, 5508-010, Brazil
| | - Dennis A Zanatto
- Department of Pathology, Faculty of Veterinary Medicine and Animal Sciences, University of São Paulo, Av. Professor Dr Orlando Marques de Paiva, 87, São Paulo, SP, 5508-010, Brazil
| | - Fábio R P Bruhn
- College of Veterinary Medicine, Federal University of Pelotas, Campus Universitário, Capão do Leão, Rio Grande do Sul, RS, 96160-000, Brazil
| | - Laís L R Mesquita
- Department of Pathology, Faculty of Veterinary Medicine and Animal Sciences, University of São Paulo, Av. Professor Dr Orlando Marques de Paiva, 87, São Paulo, SP, 5508-010, Brazil
| | - M C C S H Lara
- Biological Institute, Av. Conselheiro Rodrigues Alves, 1252, São Paulo, SP, 04014-002, Brazil
| | - E M C Villalobos
- Biological Institute, Av. Conselheiro Rodrigues Alves, 1252, São Paulo, SP, 04014-002, Brazil
| | - Cristina O Massoco
- Department of Pathology, Faculty of Veterinary Medicine and Animal Sciences, University of São Paulo, Av. Professor Dr Orlando Marques de Paiva, 87, São Paulo, SP, 5508-010, Brazil
| | - Claudia M C Mori
- Department of Pathology, Faculty of Veterinary Medicine and Animal Sciences, University of São Paulo, Av. Professor Dr Orlando Marques de Paiva, 87, São Paulo, SP, 5508-010, Brazil
| | - Enio Mori
- Pasteur Institute, Av. Paulista, 393, São Paulo, SP, 01311-000, Brazil
| | - Paulo C Maiorka
- Department of Pathology, Faculty of Veterinary Medicine and Animal Sciences, University of São Paulo, Av. Professor Dr Orlando Marques de Paiva, 87, São Paulo, SP, 5508-010, Brazil
| |
Collapse
|
23
|
Lian X, Yang X, Shao J, Hou F, Yang S, Pan D, Zhang Z. Prediction and analysis of human-herpes simplex virus type 1 protein-protein interactions by integrating multiple methods. QUANTITATIVE BIOLOGY 2020. [DOI: 10.1007/s40484-020-0222-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
24
|
Huang Y, Li X, Pan C, Cheng W, Wang X, Yang Z, Zheng L. The intervention mechanism of emodin on TLR3 pathway in the process of central nervous system injury caused by herpes virus infection. Neurol Res 2020; 43:307-313. [PMID: 33274693 DOI: 10.1080/01616412.2020.1853989] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background and purpose: To investigate the effect of Emodin on the inflammatory response of brain tissue and the expression of the TLR3 pathway in mice with herpes virus encephalitis.Method: Twenty male BALB/c mice were randomly divided into the NS group, HSV-1 group, HSV-1 + Emodin group and HSV-1 + ACV group. The histopathological features and the effect of TLR3 expression were observed by staining and immunohistochemistry (IHC) respectively. The gene expression of TLR3, trif, TRADD, TRAF6, traf3, p38, Nemo and IRF3 was detected by polymerase chain reaction (PCR). The protein production of TLR3 and its downstream molecules was detected by Western blot. The expression of IL-6, TNF-α and IFN-β in the brain tissues was detected by ELISA.Result: Compared to the HSV-1 group, the pathological changes (inflammatory cell infiltration, necrotic temporal lobe and massive hemorrhage) were not as obvious as those in the HSV-1+emodin and HSV-1+ACV groups. The TLR3 staining increased significantly in the HSV-1 groups and decreased in the HSV-1 + emodin group. Compared with the NS group, the mRNA expression of TLR3, TRIF, TRADD, TRAF6, traf3, p38, NEMO and IRF3 decreased by 20%-60% in the HSV-1 + emodin group and 30% in the HSV-1 + ACV group, respectively. The expression of IL-6, TNF-α and IFN-β decreased by 30%-50% in the HSV-1 + emodin group and showed no significant change in the HSV-1 + ACV group, respectively.Conclusion: Emodin could inhibit the inflammatory response in the brain of mice with herpes virus encephalitis. The inhibition of TLR3 expression may play an important role in this process.
Collapse
Affiliation(s)
- Yongqian Huang
- Department of Neurology, Puren Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Xing Li
- Department of Neurology, Puren Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Chunlian Pan
- Department of Neurology, Puren Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Wei Cheng
- Department of Neurology, Puren Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Xijia Wang
- Department of Neurology, Puren Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Zhigang Yang
- Department of Neurology, Puren Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Lifang Zheng
- Department of Neurology, Yantian Hospital of Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
25
|
An Early Microglial Response Is Needed To Efficiently Control Herpes Simplex Virus Encephalitis. J Virol 2020; 94:JVI.01428-20. [PMID: 32938766 DOI: 10.1128/jvi.01428-20] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 09/09/2020] [Indexed: 12/25/2022] Open
Abstract
The role of a signaling pathway through macrophage colony-stimulating factor (MCSF) and its receptor, macrophage colony-stimulating factor 1 receptor (CSF1R), during experimental herpes simplex virus 1 (HSV-1) encephalitis (HSE) was studied by two different approaches. First, we evaluated the effect of stimulation of the MCSF/CSF1R axis before infection. Exogenous MCSF (40 μg/kg of body weight intraperitoneally [i.p.]) was administered once daily to BALB/c mice on days 4 and 2 before intranasal infection with 2,500 PFU of HSV-1. MCSF treatment significantly increased mouse survival compared to saline (50% versus 10%; P = 0.0169). On day 6 postinfection (p.i.), brain viral titers were significantly decreased, whereas beta interferon (IFN-β) was significantly increased in mice treated with MCSF compared to mice treated with saline. The number of CD68+ (a phagocytosis marker) microglial cells was significantly increased in MCSF-treated mice compared to the saline-treated group. Secondly, we conditionally depleted CSF1R on microglial cells of CSF1R-loxP-CX3CR1-cre/ERT2 mice (in a C57BL/6 background) through induction with tamoxifen. The mice were then infected intranasally with 600,000 PFU of HSV-1. The survival rate of mice depleted of CSF1R (knockout [KO] mice) was significantly lower than that of wild-type (WT) mice (0% versus 67%). Brain viral titers and cytokine/chemokine levels were significantly higher in KO than in WT animals on day 6 p.i. Furthermore, increased infiltration of monocytes into the brains of WT mice was seen on day 6 p.i., but not in KO mice. Our results suggest that microglial cells are essential to control HSE at early stages of the disease and that the MCSF/CSF1R axis could be a therapeutic target to regulate their response to infection.IMPORTANCE Microglia appear to be one of the principal regulators of neuroinflammation in the central nervous system (CNS). An increasing number of studies have demonstrated that the activation of microglia could result in either beneficial or detrimental effects in different CNS disorders. Hence, the role of microglia during herpes simplex virus encephalitis (HSE) has not been fully characterized. Using experimental mouse models, we showed that an early activation of the MCSF/CSF1R axis improved the outcome of the disease, possibly by inducing a proliferation of microglia. In contrast, depletion of microglia before HSV-1 infection worsened the prognosis of HSE. Thus, an early microglial response followed by sustained infiltration of monocytes and T cells into the brain seem to be key components for a better clinical outcome. These data suggest that microglia could be a potential target for immunomodulatory strategies combined with antiviral therapy to better control the outcome of this devastating disease.
Collapse
|
26
|
Wong E, Montoya B, Stotesbury C, Ferez M, Xu RH, Sigal LJ. Langerhans Cells Orchestrate the Protective Antiviral Innate Immune Response in the Lymph Node. Cell Rep 2020; 29:3047-3059.e3. [PMID: 31801072 PMCID: PMC6927544 DOI: 10.1016/j.celrep.2019.10.118] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 09/17/2019] [Accepted: 10/29/2019] [Indexed: 12/22/2022] Open
Abstract
During disseminating viral infections, a swift innate immune response (IIR) in the draining lymph node (dLN) that restricts systemic viral spread is critical for optimal resistance to disease. However, it is unclear how this IIR is orchestrated. We show that after footpad infection of mice with ectromelia virus, dendritic cells (DCs) highly expressing major histocompatibility complex class II (MHC class IIhi DCs), including CD207+ epidermal Langerhans cells (LCs), CD103+CD207+ double-positive dermal DCs (DP-DCs), and CD103−CD207− double-negative dermal DCs (DN-DCs) migrate to the dLN from the skin carrying virus. MHC class IIhi DCs, predominantly LCs and DP-DCs, are the first cells upregulating IIR cytokines in the dLN. Preventing MHC class IIhi DC migration or depletion of LCs, but not DP-DC deficiency, suppresses the IIR in the dLN and results in high viral lethality. Therefore, LCs are the architects of an early IIR in the dLN that is critical for optimal resistance to a disseminating viral infection. Wong et al. show that by producing chemokines that recruit monocytes and by upregulating NKG2D ligands that activate ILCs, Langerhans cells are responsible for the innate immune cascade in the lymph node that is critical for survival of infection with a disseminating virus.
Collapse
Affiliation(s)
- Eric Wong
- Department of Microbiology and Immunology, Thomas Jefferson University, BLSB 709 233 South 10(th) Street, Philadelphia, PA 19107, USA
| | - Brian Montoya
- Department of Microbiology and Immunology, Thomas Jefferson University, BLSB 709 233 South 10(th) Street, Philadelphia, PA 19107, USA
| | - Colby Stotesbury
- Department of Microbiology and Immunology, Thomas Jefferson University, BLSB 709 233 South 10(th) Street, Philadelphia, PA 19107, USA
| | - Maria Ferez
- Department of Microbiology and Immunology, Thomas Jefferson University, BLSB 709 233 South 10(th) Street, Philadelphia, PA 19107, USA
| | - Ren-Huan Xu
- Immune Cell Development and Host Defense Program, Research Institute of Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA
| | - Luis J Sigal
- Department of Microbiology and Immunology, Thomas Jefferson University, BLSB 709 233 South 10(th) Street, Philadelphia, PA 19107, USA.
| |
Collapse
|
27
|
Pathophysiological roles and therapeutic potential of voltage-gated ion channels (VGICs) in pain associated with herpesvirus infection. Cell Biosci 2020; 10:70. [PMID: 32489585 PMCID: PMC7247163 DOI: 10.1186/s13578-020-00430-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 05/13/2020] [Indexed: 02/06/2023] Open
Abstract
Herpesvirus is ranked as one of the grand old members of all pathogens. Of all the viruses in the superfamily, Herpes simplex virus type 1 (HSV-1) is considered as a model virus for a variety of reasons. In a permissive non-neuronal cell culture, HSV-1 concludes the entire life cycle in approximately 18–20 h, encoding approximately 90 unique transcriptional units. In latency, the robust viral gene expression is suppressed in neurons by a group of noncoding RNA. Historically the lesions caused by the virus can date back to centuries ago. As a neurotropic pathogen, HSV-1 is associated with painful oral lesions, severe keratitis and lethal encephalitis. Transmission of pain signals is dependent on the generation and propagation of action potential in sensory neurons. T-type Ca2+ channels serve as a preamplifier of action potential generation. Voltage-gated Na+ channels are the main components for action potential production. This review summarizes not only the voltage-gated ion channels in neuropathic disorders but also provides the new insights into HSV-1 induced pain.
Collapse
|
28
|
Powell-Doherty RD, Abbott ARN, Nelson LA, Bertke AS. Amyloid-β and p-Tau Anti-Threat Response to Herpes Simplex Virus 1 Infection in Primary Adult Murine Hippocampal Neurons. J Virol 2020; 94:e01874-19. [PMID: 32075924 PMCID: PMC7163132 DOI: 10.1128/jvi.01874-19] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 02/12/2020] [Indexed: 12/18/2022] Open
Abstract
Alzheimer's Disease (AD) is the sixth leading cause of death in the United States. Recent studies have established a potential link between herpes simplex virus 1 (HSV-1) infection and the development of AD. HSV-1 DNA has been detected in AD amyloid plaques in human brains, and treatment with the antiviral acyclovir (ACV) was reported to block the accumulation of the AD-associated proteins beta-amyloid (Aβ) and hyper-phosphorylated tau (p-tau) in Vero and glioblastoma cells. Our goal was to determine whether the accumulation of AD-related proteins is attributable to acute and/or latent HSV-1 infection in mature hippocampal neurons, a region of the brain severely impacted by AD. Primary adult murine hippocampal neuronal cultures infected with HSV-1, with or without antivirals, were assessed for Aβ and p-tau expression over 7 days postinfection. P-tau expression was transiently elevated in HSV-1-infected neurons, as well as in the presence of antivirals alone. Infected neurons, as well as uninfected neurons treated with antivirals, had a greater accumulation of Aβ42 than uninfected untreated neurons. Furthermore, Aβ42 colocalized with HSV-1 latency-associated transcript (LAT) expression. These studies suggest that p-tau potentially acts as an acute response to any perceived danger-associated molecular pattern (DAMP) in primary adult hippocampal neurons, while Aβ aggregation is a long-term response to persistent threats, including HSV-1 infection.IMPORTANCE Growing evidence supports a link between HSV-1 infection and Alzheimer's disease (AD). Although AD is clearly a complex multifactorial disorder, an infectious disease etiology provides alternative therapy opportunities for this devastating disease. Understanding the impact that HSV-1 has on mature neurons and the proteins most strongly associated with AD pathology may identify specific mechanisms that could be manipulated to prevent progression of neurodegeneration and dementia.
Collapse
Affiliation(s)
- Rebecca D Powell-Doherty
- Department of Population Health Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, USA
| | - Amber R N Abbott
- Department of Biological Sciences, Virginia Tech, Blacksburg, Virginia, USA
| | - Laura A Nelson
- Department of Population Health Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, USA
| | - Andrea S Bertke
- Department of Population Health Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, USA
| |
Collapse
|
29
|
Modulation of Voltage-Gated Sodium Channel Activity in Human Dorsal Root Ganglion Neurons by Herpesvirus Quiescent Infection. J Virol 2020; 94:JVI.01823-19. [PMID: 31694955 DOI: 10.1128/jvi.01823-19] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 10/25/2019] [Indexed: 12/15/2022] Open
Abstract
The molecular mechanisms of pain associated with alphaherpesvirus latency are not clear. We hypothesize that the voltage-gated sodium channels (VGSC) on the dorsal root ganglion (DRG) neurons controlling electrical impulses may have abnormal activity during latent viral infection and reactivation. We used herpes simplex virus 1 (HSV-1) to infect the human DRG-derived neuronal cell line HD10.6 in order to study the establishment and maintenance of viral latency, viral reactivation, and changes in the functional expression of VGSCs. Differentiated cells exhibited robust tetrodotoxin (TTX)-sensitive sodium currents, and acute infection significantly reduced the functional expression of VGSCs within 24 h and completely abolished VGSC activity within 3 days. A quiescent state of infection mimicking latency can be achieved in the presence of acyclovir (ACV) for 7 days followed by 5 days of ACV washout, and then the viruses can remain dormant for another 3 weeks. It was noted that during the establishment of HSV-1 latency, the loss of VGSC activity caused by HSV-1 infection could not be blocked by ACV treatment. However, neurons with continued ACV treatment for another 4 days showed a gradual recovery of VGSC functional expression. Furthermore, the latently infected neurons exhibited higher VGSC activity than controls. The overall regulation of VGSCs by HSV-1 during quiescent infection was proved by increased transcription and possible translation of Nav1.7. Together, these observations demonstrated a very complex pattern of electrophysiological changes during HSV infection of DRG neurons, which may have implications for understanding of the mechanisms of virus-mediated pain linked to latency and reactivation.IMPORTANCE The reactivation of herpesviruses, most commonly varicella-zoster virus (VZV) and pseudorabies virus (PRV), may cause cranial nerve disorder and unbearable pain. Clinical studies have also reported that HSV-1 causes postherpetic neuralgia and chronic occipital neuralgia in humans. The current work meticulously studies the functional expression profile changes of VGSCs during the processes of HSV-1 latency establishment and reactivation using human dorsal root ganglion-derived neuronal HD10.6 cells as an in vitro model. Our results indicated that VGSC activity was eliminated upon infection but steadily recovered during latency establishment and that latent neurons exhibited even higher VGSC activity. This finding advances our knowledge of how ganglion neurons generate uncharacteristic electrical impulses due to abnormal VGSC functional expression influenced by the latent virus.
Collapse
|
30
|
Fahmy NM, Al-Sayed E, Moghannem S, Azam F, El-Shazly M, Singab AN. Breaking Down the Barriers to a Natural Antiviral Agent: Antiviral Activity and Molecular Docking of Erythrina speciosa Extract, Fractions, and the Major Compound. Chem Biodivers 2020; 17:e1900511. [PMID: 31800173 DOI: 10.1002/cbdv.201900511] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 11/27/2019] [Indexed: 12/18/2022]
Abstract
The in vitro cytotoxic activity in Vero cells and the antiviral activity of Erythrina speciosa methanol extract, fractions, and isolated vitexin were studied. The results revealed that E. speciosa leaves ethyl acetate soluble fraction of the methanol extract (ESLE) was the most active against herpes simplex virus type 1 (HSV-1). Bioactivity-guided fractionation was performed on ESLE to isolate the bioactive compounds responsible for this activity. One sub-fraction from ESLE (ESLE IV) showed the highest activity against HSV-1 and Hepatitis A HAV-H10 viruses. Vitexin isolated from ESLE VI exhibited a significant antiviral activity (EC50 =35±2.7 and 18±3.3 μg/mL against HAV-H10 and HSV-1 virus, respectively), which was notably greater than the activity of the extract and the fractions. Molecular docking studies were carried out to explore the molecular interactions of vitexin with different macromolecular targets. Analysis of the in silico data together with the in vitro studies validated the antiviral activity associated with vitexin. These outcomes indicated that vitexin is a potential candidate to be utilized commendably in lead optimization for the development of antiviral agents.
Collapse
Affiliation(s)
- Nouran M Fahmy
- Department of Pharmacognosy, Faculty of Pharmacy, Ain-Shams University, Abassia, 11566, Cairo, Egypt
| | - Eman Al-Sayed
- Department of Pharmacognosy, Faculty of Pharmacy, Ain-Shams University, Abassia, 11566, Cairo, Egypt
| | - Saad Moghannem
- Department of Botany and Microbiology, Faculty of Science, Al-Azhar University, 11884, Cairo, Egypt
| | - Faizul Azam
- Department of Pharmaceutical Chemistry and Pharmacognosy, Unaizah College of Pharmacy, Qassim University, 51911, Unaizah, Saudi Arabia
| | - Mohamed El-Shazly
- Department of Pharmacognosy, Faculty of Pharmacy, Ain-Shams University, Abassia, 11566, Cairo, Egypt.,Department of Pharmaceutical Biology, Faculty of Pharmacy and Biotechnology, German University in Cairo, 11835, Cairo, Egypt
| | - Abdel Nasser Singab
- Department of Pharmacognosy, Faculty of Pharmacy, Ain-Shams University, Abassia, 11566, Cairo, Egypt.,Center for Drug Discovery and Development Research, Faculty of Pharmacy, Ain Shams University, 11566, Cairo, Egypt
| |
Collapse
|
31
|
Amin I, Younas S, Afzal S, Shahid M, Idrees M. Herpes Simplex Virus Type 1 and Host Antiviral Immune Responses: An Update. Viral Immunol 2019; 32:424-429. [PMID: 31599707 DOI: 10.1089/vim.2019.0097] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Herpes simplex virus type 1 (HSV-1) infection activates a rapid stimulation of host innate immune responses and a delicate interplay between virus and host immune elements regulates the whole events. Although host immune elements play well in limiting the HSV-1 infection by interfering viral replication, they are still unable to remove the virus completely, because HSV-1 proteins are efficient enough to bypass the host antiviral immune responses and virus succeed to reactivate again from latency at opportune time. Type 1 interferon signaling pathway is the central point of innate immunity along with some of the activated neutrophils, monocytes, macrophages, and dendritic cells, and some natural killer cells play role, while the CD8+ T cells are crucial in adaptive immunity. In this review, the current knowledge of host and HSV-1 interaction has been described that how the host antiviral immune responses occur and what are the mechanisms of viral evasion adapted by virus to counteract with both arms of immunity.
Collapse
Affiliation(s)
- Iram Amin
- Division of Molecular Virology and Infectious Diseases, Centre of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
| | - Saima Younas
- Molecular Diagnostic Lab, Centre for Applied Molecular Biology (CAMB), University of the Punjab, Lahore, Pakistan
| | - Samia Afzal
- Division of Molecular Virology and Infectious Diseases, Centre of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
| | - Muhammad Shahid
- Division of Molecular Virology and Infectious Diseases, Centre of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
| | - Muhammad Idrees
- Division of Molecular Virology and Infectious Diseases, Centre of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
| |
Collapse
|
32
|
Wang Y, Jia J, Wang Y, Li F, Song X, Qin S, Wang Z, Kitazato K, Wang Y. Roles of HSV-1 infection-induced microglial immune responses in CNS diseases: friends or foes? Crit Rev Microbiol 2019; 45:581-594. [PMID: 31512533 DOI: 10.1080/1040841x.2019.1660615] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Microglia, as brain-resident macrophages, are the first line of defense against brain invading pathogens. Further, their dysfunction has been recognized to be closely associated with mounting CNS diseases. Of note, chronic HSV-1 infection leads to the persistent activation of microglia, which elicit a comprehensive response by generating certain factors with neurotoxic and neuroprotective effects. CNS infection with HSV-1 results in herpes simplex encephalitis and herpes simplex keratitis. Microglial immune response plays a crucial role in the development of these diseases. Moreover, HSV-1 infection is strongly associated with several CNS diseases, especially Alzheimer's disease and schizophrenia. These CNS diseases can be effectively ameliorated by eliciting an appropriate immune response, such as inhibition of microglial proliferation and activation. Therefore, it is crucial to reassess the positive and negative roles of microglia in HSV-1 CNS infection for a more comprehensive and detailed understanding of the relationship between microglia and CNS diseases. Hence, the present review focuses on the dual roles of microglia in mediating HSV-1 CNS infection, as well as on the strategy of targeting microglia to ameliorate CNS diseases. Further research in this field can help comprehensively elucidate the dual role of the microglial immune response in HSV-1 CNS infection, providing a theoretical basis for identifying therapeutic targets against overactive microglia in CNS diseases and HSV-1 infection.
Collapse
Affiliation(s)
- Yiliang Wang
- Guangzhou Jinan Biomedicine Research and Development Center, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, China.,Key Laboratory of Virology of Guangzhou, Jinan University, Guangzhou, China.,Key Laboratory of Bioengineering Medicine of Guangdong Province, Jinan University, Guangzhou, China
| | - Jiaoyan Jia
- Guangzhou Jinan Biomedicine Research and Development Center, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, China.,Key Laboratory of Virology of Guangzhou, Jinan University, Guangzhou, China.,Key Laboratory of Bioengineering Medicine of Guangdong Province, Jinan University, Guangzhou, China
| | - Yun Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Feng Li
- Guangzhou Jinan Biomedicine Research and Development Center, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, China.,Key Laboratory of Virology of Guangzhou, Jinan University, Guangzhou, China.,Key Laboratory of Bioengineering Medicine of Guangdong Province, Jinan University, Guangzhou, China
| | - Xiaowei Song
- Guangzhou Jinan Biomedicine Research and Development Center, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, China.,Key Laboratory of Virology of Guangzhou, Jinan University, Guangzhou, China.,Key Laboratory of Bioengineering Medicine of Guangdong Province, Jinan University, Guangzhou, China.,College of Pharmacy, Jinan University, Guangzhou, China
| | - Shurong Qin
- Guangzhou Jinan Biomedicine Research and Development Center, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, China.,Key Laboratory of Virology of Guangzhou, Jinan University, Guangzhou, China.,Key Laboratory of Bioengineering Medicine of Guangdong Province, Jinan University, Guangzhou, China.,College of Pharmacy, Jinan University, Guangzhou, China
| | - Zhaoyang Wang
- Guangzhou Jinan Biomedicine Research and Development Center, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, China.,Key Laboratory of Virology of Guangzhou, Jinan University, Guangzhou, China.,Key Laboratory of Bioengineering Medicine of Guangdong Province, Jinan University, Guangzhou, China
| | - Kaio Kitazato
- Division of Molecular Pharmacology of Infectious Agents, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Yifei Wang
- Guangzhou Jinan Biomedicine Research and Development Center, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, China.,Key Laboratory of Virology of Guangzhou, Jinan University, Guangzhou, China
| |
Collapse
|
33
|
Liu H, Huang CX, He Q, Li D, Luo MH, Zhao F, Lu W. Proteomics analysis of HSV-1-induced alterations in mouse brain microvascular endothelial cells. J Neurovirol 2019; 25:525-539. [PMID: 31144288 DOI: 10.1007/s13365-019-00752-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 03/01/2019] [Accepted: 04/08/2019] [Indexed: 02/08/2023]
Abstract
Herpes simplex virus 1 (HSV-1) is a predominant cause of herpes simplex encephalitis (HSE), leading to a high mortality rate and severe neurological sequelae worldwide. HSE is typically accompanied by the blood-brain barrier (BBB) disruption, but the underlying mechanisms are unclear. To explore the disruption mechanisms of the BBB, quantitative analysis of the cellular proteome was carried out to investigate the proteomic changes that occur after infection. In this study, bEnd.3 cells were infected with HSV-1, followed by liquid chromatography-tandem mass spectrometry. A total of 6761 proteins were identified in three independent mass spectrometry analyses. Compared to the uninfected cells, 386 and 293 differentially expressed proteins were markedly upregulated or downregulated, respectively. Bioinformatic analysis showed that the activator protein-1 factor, including Fos, Jun, and ATF family proteins and cell adhesion molecules were significantly changed. Further validation of the changes observed for these proteins was carried out by western blotting and quantitative real-time PCR. Transendothelial electrical resistance (TEER) studies were performed to explore the effects of ATF3, Fra1, or JunB overexpression on the function of bEnd.3 cells. Characterization of the differential expression of these proteins in bEnd.3 cells will facilitate further exploration of BBB disruption upon HSV-1 infection.
Collapse
Affiliation(s)
- Hui Liu
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Chu-Xin Huang
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Qiang He
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Dong Li
- State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430000, China
| | - Min-Hua Luo
- State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430000, China
| | - Fei Zhao
- State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430000, China.
| | - Wei Lu
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China.
| |
Collapse
|
34
|
Li LJ, Zhang SJ, Liu P, Wang YQ, Chen ZL, Wang YJ, Zhou JB, Guo YJ, Zhao L. Corilagin Interferes With Toll-Like Receptor 3-Mediated Immune Response in Herpes Simplex Encephalitis. Front Mol Neurosci 2019; 12:83. [PMID: 31080403 PMCID: PMC6497770 DOI: 10.3389/fnmol.2019.00083] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 03/18/2019] [Indexed: 12/21/2022] Open
Abstract
Herpes simplex encephalitis (HSE) is the most common infectious disease of the central nervous system worldwide. However, the pathogenesis of HSE is not clear. Research has shown that the immune response mediated by the toll-like receptor 3 (TLR3) signaling pathway is essential to protect the central nervous system against herpes simplex virus (HSV) infection. However, an excessive immune response may cause tissue damage accompanied by pathological changes. The aim of this study was to explore the molecular mechanism via which corilagin controls HSE through the TLR3 signaling pathway in vitro and in vivo. Cells and mice were pre-treated with polyriboinosinic polyribocytidylic acid [poly(I:C)] or HSV type 1, and then treated with corilagin. After treatment, the mRNA and protein levels of TLR3, TLR-like receptor-associated interferon factor (TRIF), tumor necrosis factor (TNF) receptor type 1-associated DEATH domain protein (TRADD), TNF receptor-associated factor (TRAF) 3 and 6, nuclear factor-kappa-B (NF-κB) essential modulator (NEMO), P38, and interferon regulatory factor 3 (IRF3) were decreased. Interleukin-6 (IL-6), TNF-α, and type 1 interferon-β were also decreased. When TLR3 expression was silenced or increased, corilagin still inhibited the expression of TLR3 and its downstream mediators. Hematoxylin-eosin (HE) staining and immunohistochemical examinations of mouse brain tissues revealed that corilagin lessened the degree of brain inflammation. Altogether, these results suggest that corilagin may regulate the immune response in HSE and relieve inflammatory injury by interfering with the TLR3 signaling pathway.
Collapse
Affiliation(s)
- Lu-Jun Li
- National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan, China
| | - Shao-Jun Zhang
- National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan, China
| | - Pan Liu
- School of Clinical Medicine, Hubei University of Chinese Medicine, Wuhan, China
| | - You-Qin Wang
- Renmin Hospital of Hubei University of Medicine, The Postgraduate Training Center of Jinzhou Medical University, Shiyan, China
| | - Zhi-Lin Chen
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu-Jie Wang
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia-Bin Zhou
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University and Shangdong Provincial Key Laboratory of Stem Cells and Neuro-Oncology, Jining, China
| | - Yuan-Jin Guo
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Zhao
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
35
|
Duarte LF, Farías MA, Álvarez DM, Bueno SM, Riedel CA, González PA. Herpes Simplex Virus Type 1 Infection of the Central Nervous System: Insights Into Proposed Interrelationships With Neurodegenerative Disorders. Front Cell Neurosci 2019; 13:46. [PMID: 30863282 PMCID: PMC6399123 DOI: 10.3389/fncel.2019.00046] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 01/30/2019] [Indexed: 12/21/2022] Open
Abstract
Herpes simplex virus type 1 (HSV-1) is highly prevalent in humans and can reach the brain without evident clinical symptoms. Once in the central nervous system (CNS), the virus can either reside in a quiescent latent state in this tissue, or eventually actively lead to severe acute necrotizing encephalitis, which is characterized by exacerbated neuroinflammation and prolonged neuroimmune activation producing a life-threatening disease. Although HSV-1 encephalitis can be treated with antivirals that limit virus replication, neurological sequelae are common and the virus will nevertheless remain for life in the neural tissue. Importantly, there is accumulating evidence that suggests that HSV-1 infection of the brain both, in symptomatic and asymptomatic individuals could lead to neuronal damage and eventually, neurodegenerative disorders. Here, we review and discuss acute and chronic infection of particular brain regions by HSV-1 and how this may affect neuron and cognitive functions in the host. We review potential cellular and molecular mechanisms leading to neurodegeneration, such as protein aggregation, dysregulation of autophagy, oxidative cell damage and apoptosis, among others. Furthermore, we discuss the impact of HSV-1 infection on brain inflammation and its potential relationship with neurodegenerative diseases.
Collapse
Affiliation(s)
- Luisa F Duarte
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Mónica A Farías
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Diana M Álvarez
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M Bueno
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Claudia A Riedel
- Millennium Institute on Immunology and Immunotherapy, Departamento de Biología Celular, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Pablo A González
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
36
|
The recruitment of peripheral blood leukocytes to the brain is delayed in susceptible BALB/c compared to resistant C57BL/6 mice during herpes simplex virus encephalitis. J Neurovirol 2019; 25:372-383. [PMID: 30758810 DOI: 10.1007/s13365-019-00730-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 01/11/2019] [Accepted: 01/25/2019] [Indexed: 02/05/2023]
Abstract
The cerebral immune response induced by herpes simplex virus (HSV) encephalitis (HSE) was evaluated in susceptible BALB/c and resistant C57BL/6 mice. BALB/c and C57BL/6 (named C57BL/6-high) mice were respectively infected intranasally with 1 × 103 and 5 × 105 plaque-forming units (PFUs) of HSV-1. C57BL/6 mice (named C57BL/6-low) infected with a low inoculum (1 × 103 PFUs) of HSV-1 were tested in parallel. Mice were monitored for weight loss, sickness signs, and survival for 21 days. The viral load, infectious titers, cytokine/chemokine levels, and peripheral leukocyte infiltration were determined in brain homogenates on days 0 (non-infected), 4, 6, and 8 post-infection (p.i.) by qPCR, plaque assay, ELISA/Luminex™, and flow cytometry, respectively. Our results showed that the mortality of BALB/c mice (67%) was higher compared to those of C57BL/6-low (0%; P ≤ 0.01) and C57BL/6-high (20%; P ≤ 0.05) animals. This higher mortality was associated with increased infectious titers and cytokine/chemokine levels in the brains of BALB/c compared to C57BL/6 mice. Recruitment of inflammatory monocytes, dendritic cells, natural killer, and natural killer T cells to the brain was higher in C57BL/6-high compared to BALB/c animals on day 4 p.i. Infiltration of inflammatory monocytes and T cells in the brain of BALB/c mice was seen on day 6 p.i. Our data suggest that a rapid, sustained, and coordinated recruitment of peripheral leukocytes to the brain of C57BL/6-high mice results in an effective control of viral replication and inflammation whereas the delayed infiltration of immune cells in the brain of BALB/c mice was associated with an exacerbated inflammatory response during HSE.
Collapse
|
37
|
Koujah L, Suryawanshi RK, Shukla D. Pathological processes activated by herpes simplex virus-1 (HSV-1) infection in the cornea. Cell Mol Life Sci 2019; 76:405-419. [PMID: 30327839 PMCID: PMC6349487 DOI: 10.1007/s00018-018-2938-1] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 10/03/2018] [Accepted: 10/08/2018] [Indexed: 12/13/2022]
Abstract
Herpes simplex virus type-1 (HSV-1) is a ubiquitous pathogen that infects a large majority of the human population worldwide. It is also a leading cause of infection-related blindness in the developed world. HSV-1 infection of the cornea begins with viral entry into resident cells via a multistep process that involves interaction of viral glycoproteins and host cell surface receptors. Once inside, HSV-1 infection induces a chronic immune-inflammatory response resulting in corneal scarring, thinning and neovascularization. This leads to development of various ocular diseases such as herpes stromal keratitis, resulting in visual impairment and eventual blindness. HSV-1 can also invade the central nervous system and lead to encephalitis, a relatively common cause of sporadic fetal encephalitis worldwide. In this review, we discuss the pathological processes activated by corneal HSV-1 infection and existing antiviral therapies as well as novel therapeutic options currently under development.
Collapse
Affiliation(s)
- Lulia Koujah
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, 1855 W. Taylor St, Chicago, IL, 60612, USA
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Rahul K Suryawanshi
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, 1855 W. Taylor St, Chicago, IL, 60612, USA
| | - Deepak Shukla
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, 1855 W. Taylor St, Chicago, IL, 60612, USA.
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
38
|
Acuña-Hinrichsen F, Muñoz M, Hott M, Martin C, Mancilla E, Salazar P, Leyton L, Zambrano A, Concha MI, Burgos PV, Otth C. Herpes Simplex Virus Type 1 Enhances Expression of the Synaptic Protein Arc for Its Own Benefit. Front Cell Neurosci 2019; 12:505. [PMID: 30692913 PMCID: PMC6340317 DOI: 10.3389/fncel.2018.00505] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 12/06/2018] [Indexed: 12/30/2022] Open
Abstract
Herpes simplex virus type 1 (HSV-1) is a neurotropic virus able to reach the central nervous system (CNS) after primary infection in oronasal mucosa. HSV-1 establishes latency inside neurons due the repression of its gene expression process, which is related to periodic reactivations in response to cellular stress conditions, constituting a risk factor for neurodegenerative diseases such as Alzheimer’s disease (AD). The immediate-early gene Arc plays an essential role in neuronal morphology, synaptic plasticity and memory formation. Arc acts as a hub protein, interacting with components of the endocytic machinery required for AMPA receptor (AMPAR) recycling as well as with proteins of the post-synaptic density and actin cytoskeleton. However, to date, no studies have evaluated whether persistent neurotropic HSV-1 infection modulates the expression or function of Arc protein in brain tissue. Here, we report that neuronal in vivo and in vitro infection of HSV-1 significantly increases Arc protein levels, showing a robust perinuclear distribution in neuronal cell lines, a process that is dependent on an active HSV-1 replication cycle. Finally, we found that silencing Arc protein caused a decrease in HSV-1 proteins and viral progeny, suggesting that Arc is involved in the lifecycle of HSV-1. Our studies strongly suggest that pathogenicity of HSV-1 neuronal reactivations in humans could be mediated in part by Arc neuronal upregulation and its potential role in endocytic trafficking and AMPA-neuronal function impairment. Further studies are necessary to define whether this phenomenon could have repercussions in cognition and learning processes in infected individuals.
Collapse
Affiliation(s)
| | - Mariela Muñoz
- Institute of Clinical Microbiology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile.,Centro de Excelencia en Estudios Morfológicos y Quirúrgicos (CEMyQ), Universidad de La Frontera, Temuco, Chile
| | - Melissa Hott
- Institute of Clinical Microbiology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Carolina Martin
- Institute of Clinical Microbiology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Evelyn Mancilla
- Institute of Clinical Microbiology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Paula Salazar
- Institute of Clinical Microbiology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Luis Leyton
- Institute of Clinical Microbiology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Angara Zambrano
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile.,Institute of Biochemistry and Microbiology, Faculty of Science, Universidad Austral de Chile, Valdivia, Chile
| | - Margarita I Concha
- Institute of Biochemistry and Microbiology, Faculty of Science, Universidad Austral de Chile, Valdivia, Chile
| | - Patricia V Burgos
- Institute of Physiology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile.,Centro de Biología Celular y Biomedicina, Facultad de Ciencia y Facultad de Medicina, Universidad San Sebastián, Santiago, Chile.,Center for Aging and Regeneration (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carola Otth
- Institute of Clinical Microbiology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile.,Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
| |
Collapse
|
39
|
Swartz AM, Shen SH, Salgado MA, Congdon KL, Sanchez-Perez L. Promising vaccines for treating glioblastoma. Expert Opin Biol Ther 2018; 18:1159-1170. [PMID: 30281978 DOI: 10.1080/14712598.2018.1531846] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Conventional therapies for glioblastoma (GBM) typically fail to provide lasting antitumor benefits, owing to their inability to specifically eliminate all malignant cells. Cancer vaccines are currently being evaluated as a means to direct the adaptive immune system to target residual GBM cells that remain following standard-of-care treatment. AREAS COVERED In this review, we provide an overview of the more noteworthy cancer vaccines that are under investigation for the treatment of GBM, as well as potential future directions that may enhance GBM-vaccine effectiveness. EXPERT OPINION To date, no cancer vaccines have been proven effective against GBM; however, only a few have reached phase III clinical testing. Clinical immunological monitoring data suggest that GBM vaccines are capable of stimulating immune responses reactive to GBM antigens, but whether these responses have an appreciable antitumor effect on GBM is still uncertain. Nevertheless, there have been several promising outcomes in early phase clinical trials, which lend encouragement to this area of study. Further studies with GBM vaccines are, therefore, warranted.
Collapse
Affiliation(s)
- Adam M Swartz
- a Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery , Duke University Medical Center , Durham , NC , USA.,b The Preston Robert Tisch Brain Tumor Center , Duke University Medical Center , Durham , NC , USA.,c Department of Pathology , Duke University Medical Center , Durham , NC , USA
| | - Steven H Shen
- a Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery , Duke University Medical Center , Durham , NC , USA.,b The Preston Robert Tisch Brain Tumor Center , Duke University Medical Center , Durham , NC , USA.,c Department of Pathology , Duke University Medical Center , Durham , NC , USA
| | - Miguel A Salgado
- a Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery , Duke University Medical Center , Durham , NC , USA.,b The Preston Robert Tisch Brain Tumor Center , Duke University Medical Center , Durham , NC , USA.,d Department of Neurosurgery , Duke University Medical Center , Durham , NC , USA
| | - Kendra L Congdon
- a Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery , Duke University Medical Center , Durham , NC , USA.,b The Preston Robert Tisch Brain Tumor Center , Duke University Medical Center , Durham , NC , USA.,d Department of Neurosurgery , Duke University Medical Center , Durham , NC , USA
| | - Luis Sanchez-Perez
- a Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery , Duke University Medical Center , Durham , NC , USA.,b The Preston Robert Tisch Brain Tumor Center , Duke University Medical Center , Durham , NC , USA.,d Department of Neurosurgery , Duke University Medical Center , Durham , NC , USA
| |
Collapse
|
40
|
Abstract
Endothelins were discovered more than thirty years ago as potent vasoactive compounds. Beyond their well-documented cardiovascular properties, however, the contributions of the endothelin pathway have been demonstrated in several neuroinflammatory processes and the peptides have been reported as clinically relevant biomarkers in neurodegenerative diseases. Several studies report that endothelin-1 significantly contributes to the progression of neuroinflammatory processes, particularly during infections in the central nervous system (CNS), and is associated with a loss of endothelial integrity at the blood brain barrier level. Because of the paucity of clinical trials with endothelin-1 antagonists in several infectious and non-infectious neuroinflammatory diseases, it remains an open question whether the 21 amino acid peptide is a mediator/modulator rather than a biomarker of the progression of neurodegeneration. This review focuses on the potential roles of endothelins in the pathology of neuroinflammatory processes, including infectious diseases of viral, bacterial or parasitic origin in which the synthesis of endothelins or its pharmacology have been investigated from the cell to the bedside in several cases, as well as in non-infectious inflammatory processes such as neurodegenerative disorders like Alzheimers Disease or central nervous system vasculitis.
Collapse
|
41
|
Ramirez KA, Choudhri AF, Patel A, Lenny NT, Thompson RE, Berkelhammer Greenberg L, Clanton Watson N, Kocak M, DeVincenzo JP. Comparing molecular quantification of herpes simplex virus (HSV) in cerebrospinal fluid (CSF) with quantitative structural and functional disease severity in patients with HSV encephalitis (HSVE): Implications for improved therapeutic approaches. J Clin Virol 2018; 107:29-37. [DOI: 10.1016/j.jcv.2018.08.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 08/13/2018] [Accepted: 08/16/2018] [Indexed: 11/25/2022]
|
42
|
Canivet C, Rhéaume C, Lebel M, Piret J, Gosselin J, Boivin G. Both IRF3 and especially IRF7 play a key role to orchestrate an effective cerebral inflammatory response in a mouse model of herpes simplex virus encephalitis. J Neurovirol 2018; 24:761-768. [PMID: 30094631 DOI: 10.1007/s13365-018-0666-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 07/03/2018] [Accepted: 07/12/2018] [Indexed: 11/26/2022]
Abstract
The impact of a deficiency in interferon regulatory factor (IRF)3 and IRF7 was evaluated in an herpes simplex virus encephalitis (HSE) model. Compared to wild type (WT), the mortality rates of infected IRF3-/- and IRF7-/- mice were higher and associated with increased brain viral titers. At a critical time post-infection, IRF7-/- mice exhibited a deficit in IFN-β production. At a later time point, levels of type I IFNs and cytokines were increased in brains of both deficient mice compared to WT. Our results suggest that IRF3, and especially IRF7, are important for an effective control of inflammatory responses during HSE.
Collapse
Affiliation(s)
- Coraline Canivet
- Research Center in Infectious Diseases of the CHU of Quebec and Laval University, 2705 Boul. Laurier, Quebec City, QC, G1V 4G2, Canada
| | - Chantal Rhéaume
- Research Center in Infectious Diseases of the CHU of Quebec and Laval University, 2705 Boul. Laurier, Quebec City, QC, G1V 4G2, Canada
| | - Manon Lebel
- Laboratory of Innate Immunology of the CHU of Quebec and Laval University, 2705 Boul. Laurier, Quebec City, QC, G1V 4G2, Canada
| | - Jocelyne Piret
- Research Center in Infectious Diseases of the CHU of Quebec and Laval University, 2705 Boul. Laurier, Quebec City, QC, G1V 4G2, Canada
| | - Jean Gosselin
- Laboratory of Innate Immunology of the CHU of Quebec and Laval University, 2705 Boul. Laurier, Quebec City, QC, G1V 4G2, Canada
| | - Guy Boivin
- Research Center in Infectious Diseases of the CHU of Quebec and Laval University, 2705 Boul. Laurier, Quebec City, QC, G1V 4G2, Canada.
| |
Collapse
|
43
|
Geng M, Xiao H, Liu J, Song Y, Fu P, Cheng X, Zhang J, Wang G. The diagnostic role and dynamic changes in cerebrospinal fluid neopterin during treatment of patients with primary central nervous system lymphoma. Cancer Med 2018; 7:3889-3898. [PMID: 29982995 PMCID: PMC6089159 DOI: 10.1002/cam4.1581] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 05/03/2018] [Accepted: 05/06/2018] [Indexed: 12/28/2022] Open
Abstract
This study aimed at evaluating the diagnostic and prognostic role of neopterin (Npt) concentration in the cerebrospinal fluid (CSF) of patients with primary central nervous system lymphoma (PCNSL). Ninety‐nine patients were enrolled in this retrospective study; these included patients with PCNSL (n = 21), other brain tumors (n = 44), and inflammatory diseases (n = 34). CSF Npt concentration was measured using ELISA. Receiver operating characteristic (ROC) curve analysis was performed to assess the discriminative ability of CSF Npt concentration for the diagnosis of PCNSL. CSF Npt concentration in patients with PCNSL was significantly higher than that in patients with other brain tumors and inflammatory diseases (P < .001). On ROC curve analysis, the optimal cutoff CSF Npt level of 10.77 ng/mL for the diagnosis of PCNSL and the diagnostic yield of MRI were increased when used in conjunction with CSF Npt concentration. The CSF Npt concentrations in PCNSL patients with multiple lesions were significantly higher than those in patients with a single lesion. Changes in CSF Npt concentration were consistent with post‐treatment changes in tumor sizes. The CSF Npt concentration may be a good biomarker for the diagnosis, for monitoring of disease course, and for prognostic evaluation of patients with PCNSL.
Collapse
Affiliation(s)
- Mingying Geng
- Cancer Center, Institute of Surgery Research, Third Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - He Xiao
- Cancer Center, Institute of Surgery Research, Third Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jiaqi Liu
- Department of Clinical Laboratory, Institute of Surgery Research, Third Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yang Song
- Cancer Center, Institute of Surgery Research, Third Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Ping Fu
- Department of Pathology, Institute of Surgery Research, Third Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqin, China
| | - Xing Cheng
- Department of Neurosurgery, Chongqing Cancer Hospital, Chongqing, China
| | - Jinwei Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Ge Wang
- Cancer Center, Institute of Surgery Research, Third Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
44
|
Bustamante HA, González AE, Cerda-Troncoso C, Shaughnessy R, Otth C, Soza A, Burgos PV. Interplay Between the Autophagy-Lysosomal Pathway and the Ubiquitin-Proteasome System: A Target for Therapeutic Development in Alzheimer's Disease. Front Cell Neurosci 2018; 12:126. [PMID: 29867359 PMCID: PMC5954036 DOI: 10.3389/fncel.2018.00126] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 04/20/2018] [Indexed: 12/14/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common cause of age-related dementia leading to severe irreversible cognitive decline and massive neurodegeneration. While therapeutic approaches for managing symptoms are available, AD currently has no cure. AD associates with a progressive decline of the two major catabolic pathways of eukaryotic cells—the autophagy-lysosomal pathway (ALP) and the ubiquitin-proteasome system (UPS)—that contributes to the accumulation of harmful molecules implicated in synaptic plasticity and long-term memory impairment. One protein recently highlighted as the earliest initiator of these disturbances is the amyloid precursor protein (APP) intracellular C-terminal membrane fragment β (CTFβ), a key toxic agent with deleterious effects on neuronal function that has become an important pathogenic factor for AD and a potential biomarker for AD patients. This review focuses on the involvement of regulatory molecules and specific post-translational modifications (PTMs) that operate in the UPS and ALP to control a single proteostasis network to achieve protein balance. We discuss how these aspects can contribute to the development of novel strategies to strengthen the balance of key pathogenic proteins associated with AD.
Collapse
Affiliation(s)
- Hianara A Bustamante
- Institute of Physiology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile.,Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
| | - Alexis E González
- Institute of Physiology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile.,Fundación Ciencia y Vida, Santiago, Chile
| | - Cristobal Cerda-Troncoso
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Ronan Shaughnessy
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile.,Center for Aging and Regeneration (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carola Otth
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile.,Institute of Clinical Microbiology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Andrea Soza
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile.,Center for Aging and Regeneration (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Patricia V Burgos
- Institute of Physiology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile.,Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile.,Center for Aging and Regeneration (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
45
|
Harris SA, Harris EA. Molecular Mechanisms for Herpes Simplex Virus Type 1 Pathogenesis in Alzheimer's Disease. Front Aging Neurosci 2018; 10:48. [PMID: 29559905 PMCID: PMC5845560 DOI: 10.3389/fnagi.2018.00048] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Accepted: 02/12/2018] [Indexed: 12/12/2022] Open
Abstract
This review focuses on research in the areas of epidemiology, neuropathology, molecular biology and genetics that implicates herpes simplex virus type 1 (HSV-1) as a causative agent in the pathogenesis of sporadic Alzheimer’s disease (AD). Molecular mechanisms whereby HSV-1 induces AD-related pathophysiology and pathology, including neuronal production and accumulation of amyloid beta (Aβ), hyperphosphorylation of tau proteins, dysregulation of calcium homeostasis, and impaired autophagy, are discussed. HSV-1 causes additional AD pathologies through mechanisms that promote neuroinflammation, oxidative stress, mitochondrial damage, synaptic dysfunction, and neuronal apoptosis. The AD susceptibility genes apolipoprotein E (APOE), phosphatidylinositol binding clathrin assembly protein (PICALM), complement receptor 1 (CR1) and clusterin (CLU) are involved in the HSV lifecycle. Polymorphisms in these genes may affect brain susceptibility to HSV-1 infection. APOE, for example, influences susceptibility to certain viral infections, HSV-1 viral load in the brain, and the innate immune response. The AD susceptibility gene cholesterol 25-hydroxylase (CH25H) is upregulated in the AD brain and is involved in the antiviral immune response. HSV-1 interacts with additional genes to affect cognition-related pathways and key enzymes involved in Aβ production, Aβ clearance, and hyperphosphorylation of tau proteins. Aβ itself functions as an antimicrobial peptide (AMP) against various pathogens including HSV-1. Evidence is presented supporting the hypothesis that Aβ is produced as an AMP in response to HSV-1 and other brain infections, leading to Aβ deposition and plaque formation in AD. Epidemiologic studies associating HSV-1 infection with AD and cognitive impairment are discussed. Studies are reviewed supporting subclinical chronic reactivation of latent HSV-1 in the brain as significant in the pathogenesis of AD. Finally, the rationale for and importance of clinical trials treating HSV-1-infected MCI and AD patients with antiviral medication is discussed.
Collapse
Affiliation(s)
- Steven A Harris
- St. Vincent Medical Group, Northside Internal Medicine, Indianapolis, IN, United States
| | - Elizabeth A Harris
- Department of Neurology, University of Chicago Medical Center, Chicago, IL, United States
| |
Collapse
|
46
|
Affiliation(s)
- Julian Scherer
- Department of Molecular Biology & Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA
| | - Lynn W Enquist
- Department of Molecular Biology & Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA
| |
Collapse
|
47
|
Baxter VK, Glowinski R, Braxton AM, Potter MC, Slusher BS, Griffin DE. Glutamine antagonist-mediated immune suppression decreases pathology but delays virus clearance in mice during nonfatal alphavirus encephalomyelitis. Virology 2017; 508:134-149. [PMID: 28531865 PMCID: PMC5510753 DOI: 10.1016/j.virol.2017.05.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 05/14/2017] [Accepted: 05/17/2017] [Indexed: 01/21/2023]
Abstract
Infection of weanling C57BL/6 mice with the TE strain of Sindbis virus (SINV) causes nonfatal encephalomyelitis associated with hippocampal-based memory impairment that is partially prevented by treatment with 6-diazo-5-oxo-l-norleucine (DON), a glutamine antagonist (Potter et al., J Neurovirol 21:159, 2015). To determine the mechanism(s) of protection, lymph node and central nervous system (CNS) tissues from SINV-infected mice treated daily for 1 week with low (0.3mg/kg) or high (0.6mg/kg) dose DON were examined. DON treatment suppressed lymphocyte proliferation in cervical lymph nodes resulting in reduced CNS immune cell infiltration, inflammation, and cell death compared to untreated SINV-infected mice. Production of SINV-specific antibody and interferon-gamma were also impaired by DON treatment with a delay in virus clearance. Cessation of treatment allowed activation of the antiviral immune response and viral clearance, but revived CNS pathology, demonstrating the ability of the immune response to mediate both CNS damage and virus clearance.
Collapse
Affiliation(s)
- Victoria K Baxter
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Rebecca Glowinski
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA.
| | - Alicia M Braxton
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA.
| | - Michelle C Potter
- Johns Hopkins Drug Discovery and Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Barbara S Slusher
- Johns Hopkins Drug Discovery and Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Diane E Griffin
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA.
| |
Collapse
|
48
|
Blank T, Prinz M. Type I interferon pathway in CNS homeostasis and neurological disorders. Glia 2017; 65:1397-1406. [PMID: 28519900 DOI: 10.1002/glia.23154] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 04/01/2017] [Accepted: 04/04/2017] [Indexed: 01/12/2023]
Abstract
Type I interferons (IFNs), IFN-α and IFN-β, represent the major effector cytokines of the host immune response against viruses and other intracellular pathogens. These cytokines are produced via activation of numerous pattern recognition receptors, including the Toll-like receptor signaling network, retinoic acid-inducible gene-1 (RIG-1), melanoma differentiation-associated protein-5 (MDA-5) and interferon gamma-inducible protein-16 (IFI-16). Whilst the contribution of type I IFNs to peripheral immunity is well documented, they can also be produced by almost every cell in the central nervous system (CNS). Furthermore, IFNs can reach the CNS from the periphery to modulate the function of not only microglia and astrocytes, but also neurons and oligodendrocytes, with major consequences for cognition and behavior. Given the pleiotropic nature of type I IFNs, it is critical to determine their exact cellular impact. Inappropriate upregulation of type I IFN signaling and interferon-stimulated gene expression have been linked to several CNS diseases termed "interferonopathies" including Aicardi-Goutieres syndrome and ubiquitin specific peptidase 18 (USP18)-deficiency. In contrast, in the CNS of mice with virus-induced neuroinflammation, type I IFNs can limit production of other cytokines to prevent potential damage associated with chronic cytokine expression. This capacity of type I IFNs could also explain the therapeutic benefits of exogenous type I IFN in chronic CNS autoimmune diseases such as multiple sclerosis. In this review we will highlight the importance of a well-balanced level of type I IFNs for healthy brain physiology, and to what extent dysregulation of this cytokine system can result in brain 'interferonopathies'.
Collapse
Affiliation(s)
- Thomas Blank
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| |
Collapse
|
49
|
Menasria R, Canivet C, Piret J, Gosselin J, Boivin G. Protective role of CX3CR1 signalling in resident cells of the central nervous system during experimental herpes simplex virus encephalitis. J Gen Virol 2017; 98:447-460. [PMID: 27902351 DOI: 10.1099/jgv.0.000667] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
CX3CR1 is an important chemokine receptor expressed on the surface of microglia and blood leukocytes, including monocytes. Signalling through this receptor influences the immune activity of microglia and monocyte trafficking into the central nervous system (CNS) in several neurological diseases. During experimental herpes simplex virus 1 (HSV-1) encephalitis (HSE), CX3CR1 deficiency has been reported to exacerbate the outcome of the disease. However, the precise contribution of CX3CR1 expressed in resident cells of the CNS or peripheral monocytes in protection against HSE remains unclear. To dissect the role of CX3CR1 during HSE, we reconstituted irradiated C57BL/6 WT and CX3CR1-/- mice with CX3CR1-/- (CX3CR1-/-→WT) and WT (WT→CX3CR1-/-) bone marrow cells, respectively. Our results showed that following intranasal infection with 1.2×106 p.f.u. of HSV-1, mortality rates were significantly higher in CX3CR1-/- (61.7 %) and WT→CX3CR1-/- (66.2 %) compared to WT (16.6 %; P=0.012 and P=0.016, respectively) and CX3CR1-/-→WT animals (20 %; P=0.013 and P=0.011, respectively). Higher mortality rates in CX3CR1-/- and WT→CX3CR1-/- mice were associated with increased infectious viral titres and wider HSV dissemination in brains, as well as an overproduction of inflammatory cytokines and chemokines including IL-1β, IL-6, IFN-γ, C-C motif ligand 2 and C-C motif ligand 5. Furthermore, CX3CR1 deficiency in resident cells of the CNS resulted in excessive and sustained Ly6Chi inflammatory monocyte and neutrophil infiltration into the brain. These data suggest that CX3CR1 deficiency in resident cells of the CNS affects mouse survival, HSV-1 replication control and cerebral inflammatory response whereas its deficiency in the haematopoietic system does not appear to influence the outcome of HSE.
Collapse
Affiliation(s)
- Rafik Menasria
- Research Center in Infectious Diseases of the CHU of Quebec and Laval University, 2705 Boul Laurier, Quebec City, QC G1V 4G2, Canada
| | - Coraline Canivet
- Research Center in Infectious Diseases of the CHU of Quebec and Laval University, 2705 Boul Laurier, Quebec City, QC G1V 4G2, Canada
| | - Jocelyne Piret
- Research Center in Infectious Diseases of the CHU of Quebec and Laval University, 2705 Boul Laurier, Quebec City, QC G1V 4G2, Canada
| | - Jean Gosselin
- Laboratory of Innate Immunology of the CHU of Quebec and Laval University, 2705 Boul Laurier, Quebec City, QC G1V 4G2, Canada
| | - Guy Boivin
- Research Center in Infectious Diseases of the CHU of Quebec and Laval University, 2705 Boul Laurier, Quebec City, QC G1V 4G2, Canada
| |
Collapse
|
50
|
Abstract
PURPOSE OF REVIEW The goal of this review is to provide an update on current thinking regarding herpes simplex encephalitis (HSE), emphasizing new information about pathogenesis, diagnosis, and immune responses. Specific questions to be addressed are the following: (1) Is there a genetic predisposition to HSE? (2) What clinical approaches have the greatest impact on improving the long-term outcomes in patients with HSE? And (3) are there immune-mediated mechanisms that may account for relapsing HSE? RECENT FINDINGS Toll-like receptor 3 (TLR 3) plays an important role in innate immune responses, including generation of interferons. Multiple single-gene errors in TLR 3 interferon pathways have recently been described in children that result in increased susceptibility to HSE. Conversely, studies in both animal models and humans indicate that both cytolytic viral replication and immune-mediated responses (including cytotoxic T lymphocytes and immune mechanisms mediated by TLR 2) contribute to the pathology of HSV, suggesting possible new therapeutic approaches. In terms of treatment, data clearly indicate that a longer duration between onset of symptoms and initiation of effective antiviral therapy correlates directly with less favorable clinical outcome. Recurrent or relapsing HSE may occasionally occur, but recent observations indicate that many instances of "relapsing HSE", especially in children, are more often anti-N-methyl-D-aspartate receptor (NMDAR) encephalitis triggered by the antecedent HSV infection. Innate immune responses are critical for defense against HSV; genetic defects in this system may predispose patients to HSE. During acute HSE, exuberant immune responses may contribute to the CNS pathology, suggesting that selective immunosuppressive therapy, coupled with potent antiviral drugs, may eventually play a role in the therapeutic management of HSV. While overall clinical outcomes of HSE remain suboptimal, the initiation of high-dose acyclovir therapy as early as possible in the course of the illness provides the best chance for a patient to survive with minimal neurologic damage. Distinguishing relapsing HSE from autoimmune anti-NMDAR antibody encephalitis is critically important because therapeutic approaches will be very different.
Collapse
Affiliation(s)
- John W Gnann
- Department of Medicine, Division of Infectious Diseases, Medical University of South Carolina, 135 Rutledge Avenue, MSC 752, Charleston, SC, 29425, USA.
| | - Richard J Whitley
- University of Alabama at Birmingham, 303 CHB, 1600 7th Ave. S, Birmingham, AL, 35233-1711, USA
| |
Collapse
|