1
|
Cao C, Fu G, Xu R, Li N. Coupling of Alzheimer's Disease Genetic Risk Factors with Viral Susceptibility and Inflammation. Aging Dis 2024; 15:2028-2050. [PMID: 37962454 PMCID: PMC11346407 DOI: 10.14336/ad.2023.1017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 10/17/2023] [Indexed: 11/15/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by persistent cognitive decline. Amyloid plaque deposition and neurofibrillary tangles are the main pathological features of AD brain, though mechanisms leading to the formation of lesions remain to be understood. Genetic efforts through genome-wide association studies (GWAS) have identified dozens of risk genes influencing the pathogenesis and progression of AD, some of which have been revealed in close association with increased viral susceptibilities and abnormal inflammatory responses in AD patients. In the present study, we try to present a list of AD candidate genes that have been shown to affect viral infection and inflammatory responses. Understanding of how AD susceptibility genes interact with the viral life cycle and potential inflammatory pathways would provide possible therapeutic targets for both AD and infectious diseases.
Collapse
Affiliation(s)
| | | | - Ruodan Xu
- Department of Biomedical Engineering and Technology, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Ning Li
- Department of Biomedical Engineering and Technology, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| |
Collapse
|
2
|
Canova PN, Charron AJ, Leib DA. Models of Herpes Simplex Virus Latency. Viruses 2024; 16:747. [PMID: 38793628 PMCID: PMC11125678 DOI: 10.3390/v16050747] [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: 04/05/2024] [Revised: 04/29/2024] [Accepted: 05/03/2024] [Indexed: 05/26/2024] Open
Abstract
Our current understanding of HSV latency is based on a variety of clinical observations, and in vivo, ex vivo, and in vitro model systems, each with unique advantages and drawbacks. The criteria for authentically modeling HSV latency include the ability to easily manipulate host genetics and biological pathways, as well as mimicking the immune response and viral pathogenesis in human infections. Although realistically modeling HSV latency is necessary when choosing a model, the cost, time requirement, ethical constraints, and reagent availability are also equally important. Presently, there remains a pressing need for in vivo models that more closely recapitulate human HSV infection. While the current in vivo, ex vivo, and in vitro models used to study HSV latency have limitations, they provide further insights that add to our understanding of latency. In vivo models have shed light on natural infection routes and the interplay between the host immune response and the virus during latency, while in vitro models have been invaluable in elucidating molecular pathways involved in latency. Below, we review the relative advantages and disadvantages of current HSV models and highlight insights gained through each.
Collapse
Affiliation(s)
- Paige N. Canova
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA;
- Guarini School of Graduate and Advanced Studies at Dartmouth, Hanover, NH 03755, USA;
| | - Audra J. Charron
- Guarini School of Graduate and Advanced Studies at Dartmouth, Hanover, NH 03755, USA;
| | - David A. Leib
- Guarini School of Graduate and Advanced Studies at Dartmouth, Hanover, NH 03755, USA;
| |
Collapse
|
3
|
De Francesco MA. Herpesviridae, Neurodegenerative Disorders and Autoimmune Diseases: What Is the Relationship between Them? Viruses 2024; 16:133. [PMID: 38257833 PMCID: PMC10818483 DOI: 10.3390/v16010133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/06/2023] [Accepted: 01/15/2024] [Indexed: 01/24/2024] Open
Abstract
Alzheimer's disease and Parkinson's disease represent the most common forms of cognitive impairment. Multiple sclerosis is a chronic inflammatory disease of the central nervous system responsible for severe disability. An aberrant immune response is the cause of myelin destruction that covers axons in the brain, spinal cord, and optic nerves. Systemic lupus erythematosus is an autoimmune disease characterized by alteration of B cell activation, while Sjögren's syndrome is a heterogeneous autoimmune disease characterized by altered immune responses. The etiology of all these diseases is very complex, including an interrelationship between genetic factors, principally immune associated genes, and environmental factors such as infectious agents. However, neurodegenerative and autoimmune diseases share proinflammatory signatures and a perturbation of adaptive immunity that might be influenced by herpesviruses. Therefore, they might play a critical role in the disease pathogenesis. The aim of this review was to summarize the principal findings that link herpesviruses to both neurodegenerative and autoimmune diseases; moreover, briefly underlining the potential therapeutic approach of virus vaccination and antivirals.
Collapse
Affiliation(s)
- Maria Antonia De Francesco
- Department of Molecular and Translational Medicine, Institute of Microbiology, University of Brescia-ASST Spedali Civili, 25123 Brescia, Italy
| |
Collapse
|
4
|
Olivera E, Sáez A, Carniglia L, Caruso C, Lasaga M, Durand D. Alzheimer's disease risk after COVID-19: a view from the perspective of the infectious hypothesis of neurodegeneration. Neural Regen Res 2023; 18:1404-1410. [PMID: 36571334 PMCID: PMC10075115 DOI: 10.4103/1673-5374.360273] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
In light of the rising evidence of the association between viral and bacterial infections and neurodegeneration, we aimed at revisiting the infectious hypothesis of Alzheimer's disease and analyzing the possible implications of COVID-19 neurological sequelae in long-term neurodegeneration. We wondered how SARS-CoV-2 could be related to the amyloid-β cascade and how it could lead to the pathological hallmarks of the disease. We also predict a paradigm change in clinical medicine, which now has a great opportunity to conduct prospective surveillance of cognitive sequelae and progression to dementia in people who suffered severe infections together with other risk factors for Alzheimer's disease.
Collapse
Affiliation(s)
- Eugenia Olivera
- Instituto de Investigaciones Biomédicas INBIOMED UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | - Albany Sáez
- Instituto de Investigaciones Biomédicas INBIOMED UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | - Lila Carniglia
- Instituto de Investigaciones Biomédicas INBIOMED UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | - Carla Caruso
- Instituto de Investigaciones Biomédicas INBIOMED UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | - Mercedes Lasaga
- Instituto de Investigaciones Biomédicas INBIOMED UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | - Daniela Durand
- Instituto de Investigaciones Biomédicas INBIOMED UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Argentina
| |
Collapse
|
5
|
Bourgade K, Frost EH, Dupuis G, Witkowski JM, Laurent B, Calmettes C, Ramassamy C, Desroches M, Rodrigues S, Fülöp T. Interaction Mechanism Between the HSV-1 Glycoprotein B and the Antimicrobial Peptide Amyloid-β. J Alzheimers Dis Rep 2022; 6:599-606. [DOI: 10.3233/adr-220061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 08/20/2022] [Indexed: 11/15/2022] Open
Abstract
Background: Unravelling the mystery of Alzheimer’s disease (AD) requires urgent resolution given the worldwide increase of the aging population. There is a growing concern that the current leading AD hypothesis, the amyloid cascade hypothesis, does not stand up to validation with respect to emerging new data. Indeed, several paradoxes are being discussed in the literature, for instance, both the deposition of the amyloid-β peptide (Aβ) and the intracellular neurofibrillary tangles could occur within the brain without any cognitive pathology. Thus, these paradoxes suggest that something more fundamental is at play in the onset of the disease and other key and related pathomechanisms must be investigated. Objective: The present study follows our previous investigations on the infectious hypothesis, which posits that some pathogens are linked to late onset AD. Our studies also build upon the finding that Aβ is a powerful antimicrobial agent, produced by neurons in response to viral infection, capable of inhibiting pathogens as observed in in vitro experiments. Herein, we ask what are the molecular mechanisms in play when Aβ neutralizes infectious pathogens? Methods: To answer this question, we probed at nanoscale lengths with FRET (Förster Resonance Energy Transfer), the interaction between Aβ peptides and glycoprotein B (responsible of virus-cell binding) within the HSV-1 virion Results: The experiments show an energy transfer between Aβ peptides and glycoprotein B when membrane is intact. No energy transfer occurs after membrane disruption or treatment with blocking antibody. Conclusion: We concluded that Aβ insert into viral membrane, close to glycoprotein B, and participate in virus neutralization.
Collapse
Affiliation(s)
- Karine Bourgade
- Research Center on Aging, Geriatric Division, Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Eric H. Frost
- Department of Microbiology and Infectious diseases, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Gilles Dupuis
- Department of Biochemistry, Graduate Program in Immunology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Jacek M. Witkowski
- Department of Pathophysiology, Medical University of Gdansk, Gdansk, Poland
| | - Benoit Laurent
- Research Center on Aging, Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | | | | | - Mathieu Desroches
- MathNeuro Team, Inria Sophia Antipolis Méditerranée, France
- Université Côte d’Azur, Nice, France
| | - Serafim Rodrigues
- Ikerbasque, The Basque Foundation for Science, Bilbao, Spain
- BCAM - The Basque Center for Applied Mathematics, Bilbao, Spain
| | - Tamás Fülöp
- Research Center on Aging, Geriatric Division, Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
6
|
New Insights into the Molecular Interplay between Human Herpesviruses and Alzheimer’s Disease—A Narrative Review. Brain Sci 2022; 12:brainsci12081010. [PMID: 36009073 PMCID: PMC9406069 DOI: 10.3390/brainsci12081010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 07/24/2022] [Accepted: 07/28/2022] [Indexed: 12/15/2022] Open
Abstract
Human herpesviruses (HHVs) have been implicated as possible risk factors in Alzheimer’s disease (AD) pathogenesis. Persistent lifelong HHVs infections may directly or indirectly contribute to the generation of AD hallmarks: amyloid beta (Aβ) plaques, neurofibrillary tangles composed of hyperphosphorylated tau proteins, and synaptic loss. The present review focuses on summarizing current knowledge on the molecular mechanistic links between HHVs and AD that include processes involved in Aβ accumulation, tau protein hyperphosphorylation, autophagy, oxidative stress, and neuroinflammation. A PubMed search was performed to collect all the available research data regarding the above mentioned mechanistic links between HHVs and AD pathology. The vast majority of research articles referred to the different pathways exploited by Herpes Simplex Virus 1 that could lead to AD pathology, while a few studies highlighted the emerging role of HHV 6, cytomegalovirus, and Epstein–Barr Virus. The elucidation of such potential links may guide the development of novel diagnostics and therapeutics to counter this devastating neurological disorder that until now remains incurable.
Collapse
|
7
|
Uselman TW, Medina CS, Gray HB, Jacobs RE, Bearer EL. Longitudinal manganese-enhanced magnetic resonance imaging of neural projections and activity. NMR IN BIOMEDICINE 2022; 35:e4675. [PMID: 35253280 PMCID: PMC11064873 DOI: 10.1002/nbm.4675] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 10/19/2021] [Accepted: 12/07/2021] [Indexed: 06/14/2023]
Abstract
Manganese-enhanced magnetic resonance imaging (MEMRI) holds exceptional promise for preclinical studies of brain-wide physiology in awake-behaving animals. The objectives of this review are to update the current information regarding MEMRI and to inform new investigators as to its potential. Mn(II) is a powerful contrast agent for two main reasons: (1) high signal intensity at low doses; and (2) biological interactions, such as projection tracing and neural activity mapping via entry into electrically active neurons in the living brain. High-spin Mn(II) reduces the relaxation time of water protons: at Mn(II) concentrations typically encountered in MEMRI, robust hyperintensity is obtained without adverse effects. By selectively entering neurons through voltage-gated calcium channels, Mn(II) highlights active neurons. Safe doses may be repeated over weeks to allow for longitudinal imaging of brain-wide dynamics in the same individual across time. When delivered by stereotactic intracerebral injection, Mn(II) enters active neurons at the injection site and then travels inside axons for long distances, tracing neuronal projection anatomy. Rates of axonal transport within the brain were measured for the first time in "time-lapse" MEMRI. When delivered systemically, Mn(II) enters active neurons throughout the brain via voltage-sensitive calcium channels and clears slowly. Thus behavior can be monitored during Mn(II) uptake and hyperintense signals due to Mn(II) uptake captured retrospectively, allowing pairing of behavior with neural activity maps for the first time. Here we review critical information gained from MEMRI projection mapping about human neuropsychological disorders. We then discuss results from neural activity mapping from systemic Mn(II) imaged longitudinally that have illuminated development of the tonotopic map in the inferior colliculus as well as brain-wide responses to acute threat and how it evolves over time. MEMRI posed specific challenges for image data analysis that have recently been transcended. We predict a bright future for longitudinal MEMRI in pursuit of solutions to the brain-behavior mystery.
Collapse
Affiliation(s)
- Taylor W. Uselman
- University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | | | - Harry B. Gray
- Beckman Institute, California Institute of Technology, Pasadena, California, USA
| | - Russell E. Jacobs
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Elaine L. Bearer
- University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
- Beckman Institute, California Institute of Technology, Pasadena, California, USA
| |
Collapse
|
8
|
Mielcarska MB, Skowrońska K, Wyżewski Z, Toka FN. Disrupting Neurons and Glial Cells Oneness in the Brain-The Possible Causal Role of Herpes Simplex Virus Type 1 (HSV-1) in Alzheimer's Disease. Int J Mol Sci 2021; 23:ijms23010242. [PMID: 35008671 PMCID: PMC8745046 DOI: 10.3390/ijms23010242] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 12/21/2021] [Accepted: 12/22/2021] [Indexed: 12/15/2022] Open
Abstract
Current data strongly suggest herpes simplex virus type 1 (HSV-1) infection in the brain as a contributing factor to Alzheimer's disease (AD). The consequences of HSV-1 brain infection are multilateral, not only are neurons and glial cells damaged, but modifications also occur in their environment, preventing the transmission of signals and fulfillment of homeostatic and immune functions, which can greatly contribute to the development of disease. In this review, we discuss the pathological alterations in the central nervous system (CNS) cells that occur, following HSV-1 infection. We describe the changes in neurons, astrocytes, microglia, and oligodendrocytes related to the production of inflammatory factors, transition of glial cells into a reactive state, oxidative damage, Aβ secretion, tau hyperphosphorylation, apoptosis, and autophagy. Further, HSV-1 infection can affect processes observed during brain aging, and advanced age favors HSV-1 reactivation as well as the entry of the virus into the brain. The host activates pattern recognition receptors (PRRs) for an effective antiviral response during HSV-1 brain infection, which primarily engages type I interferons (IFNs). Future studies regarding the influence of innate immune deficits on AD development, as well as supporting the neuroprotective properties of glial cells, would reveal valuable information on how to harness cytotoxic inflammatory milieu to counter AD initiation and progression.
Collapse
Affiliation(s)
- Matylda Barbara Mielcarska
- Department of Preclinical Sciences, Institute of Veterinary Sciences, Warsaw University of Life Sciences–SGGW, Jana Ciszewskiego 8, 02-786 Warsaw, Poland;
- Correspondence: ; Tel.: +48-22-59-36063
| | - Katarzyna Skowrońska
- Department of Neurotoxicology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Adolfa Pawińskiego 5, 02-106 Warsaw, Poland;
| | - Zbigniew Wyżewski
- Institute of Biological Sciences, Cardinal Stefan Wyszyński University in Warsaw, Dewajtis 5, 01-815 Warsaw, Poland;
| | - Felix Ngosa Toka
- Department of Preclinical Sciences, Institute of Veterinary Sciences, Warsaw University of Life Sciences–SGGW, Jana Ciszewskiego 8, 02-786 Warsaw, Poland;
- Department of Biomedical Sciences, Ross University School of Veterinary Medicine, Basseterre 42123, Saint Kitts and Nevis
| |
Collapse
|
9
|
Yong SJ, Yong MH, Teoh SL, Soga T, Parhar I, Chew J, Lim WL. The Hippocampal Vulnerability to Herpes Simplex Virus Type I Infection: Relevance to Alzheimer's Disease and Memory Impairment. Front Cell Neurosci 2021; 15:695738. [PMID: 34483839 PMCID: PMC8414573 DOI: 10.3389/fncel.2021.695738] [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: 04/15/2021] [Accepted: 07/20/2021] [Indexed: 12/24/2022] Open
Abstract
Herpes simplex virus type 1 (HSV-1) as a possible infectious etiology in Alzheimer’s disease (AD) has been proposed since the 1980s. The accumulating research thus far continues to support the association and a possible causal role of HSV-1 in the development of AD. HSV-1 has been shown to induce neuropathological and behavioral changes of AD, such as amyloid-beta accumulation, tau hyperphosphorylation, as well as memory and learning impairments in experimental settings. However, a neuroanatomical standpoint of HSV-1 tropism in the brain has not been emphasized in detail. In this review, we propose that the hippocampal vulnerability to HSV-1 infection plays a part in the development of AD and amnestic mild cognitive impairment (aMCI). Henceforth, this review draws on human studies to bridge HSV-1 to hippocampal-related brain disorders, namely AD and aMCI/MCI. Next, experimental models and clinical observations supporting the neurotropism or predilection of HSV-1 to infect the hippocampus are examined. Following this, factors and mechanisms predisposing the hippocampus to HSV-1 infection are discussed. In brief, the hippocampus has high levels of viral cellular receptors, neural stem or progenitor cells (NSCs/NPCs), glucocorticoid receptors (GRs) and amyloid precursor protein (APP) that support HSV-1 infectivity, as well as inadequate antiviral immunity against HSV-1. Currently, the established diseases HSV-1 causes are mucocutaneous lesions and encephalitis; however, this review revises that HSV-1 may also induce and/or contribute to hippocampal-related brain disorders, especially AD and aMCI/MCI.
Collapse
Affiliation(s)
- Shin Jie Yong
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Petaling Jaya, Malaysia
| | - Min Hooi Yong
- Department of Psychology, School of Medical and Life Sciences, Sunway University, Petaling Jaya, Malaysia.,Aging Health and Well-being Research Centre, School of Medical and Life Sciences, Sunway University, Petaling Jaya, Malaysia
| | - Seong Lin Teoh
- Department of Anatomy, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Tomoko Soga
- Jeffrey Cheah School of Medicine and Health Sciences, Brain Research Institute Monash Sunway, Monash University Malaysia, Subang Jaya, Malaysia
| | - Ishwar Parhar
- Jeffrey Cheah School of Medicine and Health Sciences, Brain Research Institute Monash Sunway, Monash University Malaysia, Subang Jaya, Malaysia
| | - Jactty Chew
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Petaling Jaya, Malaysia
| | - Wei Ling Lim
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Petaling Jaya, Malaysia.,Aging Health and Well-being Research Centre, School of Medical and Life Sciences, Sunway University, Petaling Jaya, Malaysia
| |
Collapse
|
10
|
DuRaine G, Johnson DC. Anterograde transport of α-herpesviruses in neuronal axons. Virology 2021; 559:65-73. [PMID: 33836340 DOI: 10.1016/j.virol.2021.02.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/08/2021] [Accepted: 02/19/2021] [Indexed: 02/04/2023]
Abstract
α-herpesviruses have been very successful, principally because they establish lifelong latency in sensory ganglia. An essential piece of the lifecycle of α-herpesviruses involves the capacity to travel from sensory neurons to epithelial tissues following virus reactivation from latency, a process known as anterograde transport. Virus particles formed in neuron cell bodies hitchhike on kinesin motors that run along microtubules, the length of axons. Herpes simplex virus (HSV) and pseudorabies virus (PRV) have been intensely studied to elucidate anterograde axonal transport. Both viruses use similar strategies for anterograde transport, although there are significant differences in the form of virus particles transported in axons, the identity of the kinesins that transport viruses, and how certain viral membrane proteins, gE/gI and US9, participate in this process. This review compares the older models for HSV and PRV anterograde transport with recent results, which are casting a new light on several aspects of this process.
Collapse
Affiliation(s)
- Grayson DuRaine
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR, 97239, USA
| | - David C Johnson
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR, 97239, USA.
| |
Collapse
|
11
|
Iqbal UH, Zeng E, Pasinetti GM. The Use of Antimicrobial and Antiviral Drugs in Alzheimer's Disease. Int J Mol Sci 2020; 21:E4920. [PMID: 32664669 PMCID: PMC7404195 DOI: 10.3390/ijms21144920] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/09/2020] [Accepted: 07/10/2020] [Indexed: 12/15/2022] Open
Abstract
The aggregation and accumulation of amyloid-β plaques and tau proteins in the brain have been central characteristics in the pathophysiology of Alzheimer's disease (AD), making them the focus of most of the research exploring potential therapeutics for this neurodegenerative disease. With success in interventions aimed at depleting amyloid-β peptides being limited at best, a greater understanding of the physiological role of amyloid-β peptides is needed. The development of amyloid-β plaques has been determined to occur 10-20 years prior to AD symptom manifestation, hence earlier interventions might be necessary to address presymptomatic AD. Furthermore, recent studies have suggested that amyloid-β peptides may play a role in innate immunity as an antimicrobial peptide. These findings, coupled with the evidence of pathogens such as viruses and bacteria in AD brains, suggests that the buildup of amyloid-β plaques could be a response to the presence of viruses and bacteria. This has led to the foundation of the antimicrobial hypothesis for AD. The present review will highlight the current understanding of amyloid-β, and the role of bacteria and viruses in AD, and will also explore the therapeutic potential of antimicrobial and antiviral drugs in Alzheimer's disease.
Collapse
Affiliation(s)
| | | | - Giulio M. Pasinetti
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (U.H.I.); (E.Z.)
| |
Collapse
|
12
|
Diwaker D, Murray JW, Barnes J, Wolkoff AW, Wilson DW. Deletion of the Pseudorabies Virus gE/gI-US9p complex disrupts kinesin KIF1A and KIF5C recruitment during egress, and alters the properties of microtubule-dependent transport in vitro. PLoS Pathog 2020; 16:e1008597. [PMID: 32511265 PMCID: PMC7302734 DOI: 10.1371/journal.ppat.1008597] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 06/18/2020] [Accepted: 05/04/2020] [Indexed: 01/18/2023] Open
Abstract
During infection of neurons by alphaherpesviruses including Pseudorabies virus (PRV) and Herpes simplex virus type 1 (HSV-1) viral nucleocapsids assemble in the cell nucleus, become enveloped in the cell body then traffic into and down axons to nerve termini for spread to adjacent epithelia. The viral membrane protein US9p and the membrane glycoprotein heterodimer gE/gI play critical roles in anterograde spread of both HSV-1 and PRV, and several models exist to explain their function. Biochemical studies suggest that PRV US9p associates with the kinesin-3 motor KIF1A in a gE/gI-stimulated manner, and the gE/gI-US9p complex has been proposed to recruit KIF1A to PRV for microtubule-mediated anterograde trafficking into or along the axon. However, as loss of gE/gI-US9p essentially abolishes delivery of alphaherpesviruses to the axon it is difficult to determine the microtubule-dependent trafficking properties and motor-composition of Δ(gE/gI-US9p) particles. Alternatively, studies in HSV-1 have suggested that gE/gI and US9p are required for the appearance of virions in the axon because they act upstream, to help assemble enveloped virions in the cell body. We prepared Δ(gE/gI-US9p) mutant, and control parental PRV particles from differentiated cultured neuronal or porcine kidney epithelial cells and quantitated the efficiency of virion assembly, the properties of microtubule-dependent transport and the ability of viral particles to recruit kinesin motors. We find that loss of gE/gI-US9p has no significant effect upon PRV particle assembly but leads to greatly diminished plus end-directed traffic, and enhanced minus end-directed and bidirectional movement along microtubules. PRV particles prepared from infected differentiated mouse CAD neurons were found to be associated with either kinesin KIF1A or kinesin KIF5C, but not both. Loss of gE/gI-US9p resulted in failure to recruit KIF1A and KF5C, but did not affect dynein binding. Unexpectedly, while KIF5C was expressed in undifferentiated and differentiated CAD neurons it was only found associated with PRV particles prepared from differentiated cells.
Collapse
Affiliation(s)
- Drishya Diwaker
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
| | - John W. Murray
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
- Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
| | - Jenna Barnes
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
| | - Allan W. Wolkoff
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
- Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
| | - Duncan W. Wilson
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
13
|
Rizzo R, Bortolotti D, Gentili V, Rotola A, Bolzani S, Caselli E, Tola MR, Di Luca D. KIR2DS2/KIR2DL2/HLA-C1 Haplotype Is Associated with Alzheimer's Disease: Implication for the Role of Herpesvirus Infections. J Alzheimers Dis 2020; 67:1379-1389. [PMID: 30689576 DOI: 10.3233/jad-180777] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder, where neuroinflammation and immune cells are key pathological factors. Recently, it was suggested a possible association between AD and human herpesvirus 6 (HHV-6) infection. Since we recently observed that multiple sclerosis patients with KIR2DL2 expression on natural killer (NK) cells are more susceptible to herpesvirus infection, we tested the possible implication of KIR/HLA genetic for HHV-6A infection. We identified, for the first time, a possible implication of a specific KIR/HLA subset in AD. The combination KIR2DS2/KIR2DL2/C1 correlated with a lower MMSEDi score, representative of a severe AD status and an increased susceptibility to HHV-6A infection. Therefore, the results seem to converge on the hypothesis that herpesvirus infection might play a role in AD. If this hypothesis finds experimental confirmation, a new therapeutic strategy, modulating KIR2DL2 expression on NK cells, for AD might be envisaged.
Collapse
Affiliation(s)
- Roberta Rizzo
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Daria Bortolotti
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Valentina Gentili
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Antonella Rotola
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Silvia Bolzani
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | | | - Maria Rosaria Tola
- Department of Neurosciences and Rehabilitation, University Hospital, Arcispedale S. Anna, Ferrara, Italy
| | - Dario Di Luca
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| |
Collapse
|
14
|
Lathe R, Darlix JL. Prion protein PrP nucleic acid binding and mobilization implicates retroelements as the replicative component of transmissible spongiform encephalopathy. Arch Virol 2020; 165:535-556. [PMID: 32025859 PMCID: PMC7024060 DOI: 10.1007/s00705-020-04529-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 12/13/2019] [Indexed: 12/21/2022]
Abstract
The existence of more than 30 strains of transmissible spongiform encephalopathy (TSE) and the paucity of infectivity of purified PrPSc, as well as considerations of PrP structure, are inconsistent with the protein-only (prion) theory of TSE. Nucleic acid is a strong contender as a second component. We juxtapose two key findings: (i) PrP is a nucleic-acid-binding antimicrobial protein that is similar to retroviral Gag proteins in its ability to trigger reverse transcription. (ii) Retroelement mobilization is widely seen in TSE disease. Given further evidence that PrP also mediates nucleic acid transport into and out of the cell, a strong case is to be made that a second element – retroelement nucleic acid – bound to PrP constitutes the second component necessary to explain the multiple strains of TSE.
Collapse
Affiliation(s)
- Richard Lathe
- Division of Infection Medicine, University of Edinburgh School of Medicine, Edinburgh, UK. .,Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Pushchino, Moscow, Moscow Region, Russia.
| | - Jean-Luc Darlix
- Faculté de Pharmacie, Centre Nationale de la Recherche Scientifique (CNRS) Laboratory of Bioimaging and Pathologies (Unité Mixte de Recherche 7021), Université de Strasbourg, Illkirch, France.
| |
Collapse
|
15
|
Peterson CT. Dysfunction of the Microbiota-Gut-Brain Axis in Neurodegenerative Disease: The Promise of Therapeutic Modulation With Prebiotics, Medicinal Herbs, Probiotics, and Synbiotics. J Evid Based Integr Med 2020; 25:2515690X20957225. [PMID: 33092396 PMCID: PMC7586271 DOI: 10.1177/2515690x20957225] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 07/12/2020] [Accepted: 08/15/2020] [Indexed: 12/19/2022] Open
Abstract
Recent data suggest gut microbiota dysbiosis as a contributing factor in neurodegenerative diseases, such as Parkinson's Disease (PD) and Alzheimer's Disease (AD), and these pathologies may manifest via the microbiota-gut-brain-axis, which comprises bidirectional communication through neuroimmune, neuroendocrine, and direct neural pathways such as the vagus nerve. Preclinical and human clinical trial data reveal exciting potential for novel treatment targets and therapeutic modulation with prebiotics, medicinal herbs, probiotics, and synbiotics in health, aging, and neurodegeneration and are reviewed here. While greater insights and characterization of the microbiota-gut-brain axis have been revealed over the past decade, salient questions related to the pathology, pathogenesis and clinical treatment of the axis in the context of both health and neurodegenerative disease remain and are discussed in this review. Future directions such as additional well-controlled, large scale, longitudinal human clinical trials are urgently needed to further elucidate both mechanism and therapeutic opportunity in health, neurological disease, and disease subpopulations to ensure that the next decade ushers the dawn of targeted therapeutic modulation of the microbiota-gut-brain axis.
Collapse
Affiliation(s)
- Christine Tara Peterson
- Department of Family Medicine and Public Health, Center of Excellence for Research and Training in Integrative Health, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
16
|
Bathini P, Brai E, Auber LA. Olfactory dysfunction in the pathophysiological continuum of dementia. Ageing Res Rev 2019; 55:100956. [PMID: 31479764 DOI: 10.1016/j.arr.2019.100956] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 07/29/2019] [Accepted: 08/26/2019] [Indexed: 12/21/2022]
Abstract
Sensory capacities like smell, taste, hearing, vision decline with aging, but increasing evidence show that sensory dysfunctions are one of the early signs diagnosing the conversion from physiological to pathological brain state. Smell loss represents the best characterized sense in clinical practice and is considered as one of the first preclinical signs of Alzheimer's and Parkinson's disease, occurring a decade or more before the onset of cognitive and motor symptoms. Despite the numerous scientific reports and the adoption in clinical practice, the etiology of sensory damage as prodromal of dementia remains largely unexplored and more studies are needed to resolve the mechanisms underlying sensory network dysfunction. Although both cognitive and sensory domains are progressively affected, loss of sensory experience in early stages plays a major role in reducing the autonomy of demented people in their daily tasks or even possibly contributing to their cognitive decline. Interestingly, the chemosensory circuitry is devoid of a blood brain barrier, representing a vulnerable port of entry for neurotoxic species that can spread to the brain. Furthermore, the exposure of the olfactory system to the external environment make it more susceptible to mechanical injury and trauma, which can cause degenerative neuroinflammation. In this review, we will summarize several findings about chemosensory impairment signing the conversion from healthy to pathological brain aging and we will try to connect those observations to the promising research linking environmental influences to sporadic dementia. The scientific body of knowledge will support the use of chemosensory diagnostics in the presymptomatic stages of AD and other biomarkers with the scope of finding treatment strategies before the onset of the disease.
Collapse
Affiliation(s)
- Praveen Bathini
- Department of Medicine, University of Fribourg, Fribourg, Switzerland
| | - Emanuele Brai
- VIB-KU Leuven Center for Brain & Disease Research, Laboratory for the Research of Neurodegenerative Diseases, Leuven, Belgium
| | - Lavinia Alberi Auber
- Department of Medicine, University of Fribourg, Fribourg, Switzerland; Swiss Integrative Center of Human Health, Fribourg, Switzerland.
| |
Collapse
|
17
|
Persistent Infection with Herpes Simplex Virus 1 and Alzheimer's Disease-A Call to Study How Variability in Both Virus and Host may Impact Disease. Viruses 2019; 11:v11100966. [PMID: 31635156 PMCID: PMC6833100 DOI: 10.3390/v11100966] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 10/14/2019] [Accepted: 10/14/2019] [Indexed: 02/06/2023] Open
Abstract
Increasing attention has focused on the contributions of persistent microbial infections with the manifestation of disease later in life, including neurodegenerative conditions such as Alzheimer’s disease (AD). Current data has shown the presence of herpes simplex virus 1 (HSV-1) in regions of the brain that are impacted by AD in elderly individuals. Additionally, neuronal infection with HSV-1 triggers the accumulation of amyloid beta deposits and hyperphosphorylated tau, and results in oxidative stress and synaptic dysfunction. All of these factors are implicated in the development of AD. These data highlight the fact that persistent viral infection is likely a contributing factor, rather than a sole cause of disease. Details of the correlations between HSV-1 infection and AD development are still just beginning to emerge. Future research should investigate the relative impacts of virus strain- and host-specific factors on the induction of neurodegenerative processes over time, using models such as infected neurons in vitro, and animal models in vivo, to begin to understand their relationship with cognitive dysfunction.
Collapse
|
18
|
La Rosa F, Agostini S, Bianchi A, Nemni R, Piancone F, Marventano I, Mancuso R, Saresella M, Clerici M. Herpes simplex virus-1 (HSV-1) infection induces a potent but ineffective IFN-λ production in immune cells of AD and PD patients. J Transl Med 2019; 17:286. [PMID: 31455413 PMCID: PMC6712644 DOI: 10.1186/s12967-019-2034-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 08/18/2019] [Indexed: 11/23/2022] Open
Abstract
Background The sequential activation of immediate early (IE), early (E) and late (L) genes is required to allow productive herpes simplex virus type 1 (HSV-1) infection. Several evidences suggest that, together with inflammation, an immunological response incapable to counteract HSV-1 reactivation plays a role in the pathogenesis of Alzheimer’s (AD) and Parkinson’s (PD) diseases. IFN-lambda (IFN-λ), a cytokine endowed with a robust antiviral activity, contains HSV-1 reactivation. HSV-1-induced IFN-λ, IL-10 and IL-1β as well as the expression of viral IE, E and L genes were analyzed in vitro in peripheral blood mononuclear cells (PBMC) of AD and PD patients as well as of healthy controls (HC). Methods PBMC of AD, PD and HC were in vitro infected with one multiplicity of infection (1 MOI) HSV-1. IE, E, and L viral genes transcription as well as IFN-λ, IL-10 and IL-1β production were analyzed. Results In HSV-1-infected cells of AD and PD patients compared to HC: (1) transcription of IE (ICP0, ICP27) genes was reduced whereas that of E (UL41, UL29) and L (UL48, LAT) genes was increased; (2) IFN-λ mRNA expression was increased. IL-1β was augmented and IL-10 was reduced in unstimulated cells of AD and PD compared to HC; HSV-1 infection significantly increased IL-10 production in HC alone. Conclusions Data herein show that a proinflammatory condition is present in AD and PD, in whom attempts to obstacle viral replication via an initial, possibly more potent IFN-λ-mediated control of IE viral genes is unsuccessful.
Collapse
Affiliation(s)
- Francesca La Rosa
- Laboratory of Molecular Medicine and Biotechnology, IRCCS Fondazione Don Carlo Gnocchi, via Capecelatro, 66, 20148, Milan, Italy.
| | - Simone Agostini
- Laboratory of Molecular Medicine and Biotechnology, IRCCS Fondazione Don Carlo Gnocchi, via Capecelatro, 66, 20148, Milan, Italy
| | - Anna Bianchi
- Laboratory of Molecular Medicine and Biotechnology, IRCCS Fondazione Don Carlo Gnocchi, via Capecelatro, 66, 20148, Milan, Italy
| | - Raffaello Nemni
- Laboratory of Molecular Medicine and Biotechnology, IRCCS Fondazione Don Carlo Gnocchi, via Capecelatro, 66, 20148, Milan, Italy
| | - Federica Piancone
- Laboratory of Molecular Medicine and Biotechnology, IRCCS Fondazione Don Carlo Gnocchi, via Capecelatro, 66, 20148, Milan, Italy
| | - Ivana Marventano
- Laboratory of Molecular Medicine and Biotechnology, IRCCS Fondazione Don Carlo Gnocchi, via Capecelatro, 66, 20148, Milan, Italy
| | - Roberta Mancuso
- Laboratory of Molecular Medicine and Biotechnology, IRCCS Fondazione Don Carlo Gnocchi, via Capecelatro, 66, 20148, Milan, Italy
| | - Marina Saresella
- Laboratory of Molecular Medicine and Biotechnology, IRCCS Fondazione Don Carlo Gnocchi, via Capecelatro, 66, 20148, Milan, Italy
| | - Mario Clerici
- Laboratory of Molecular Medicine and Biotechnology, IRCCS Fondazione Don Carlo Gnocchi, via Capecelatro, 66, 20148, Milan, Italy.,Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| |
Collapse
|
19
|
Bearer EL, Wu C. Herpes Simplex Virus, Alzheimer's Disease and a Possible Role for Rab GTPases. Front Cell Dev Biol 2019; 7:134. [PMID: 31448273 PMCID: PMC6692634 DOI: 10.3389/fcell.2019.00134] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 07/04/2019] [Indexed: 12/17/2022] Open
Abstract
Herpes simplex virus (HSV) is a common pathogen, infecting 85% of adults in the United States. After reaching the nucleus of the long-lived neuron, HSV may enter latency to persist throughout the life span. Re-activation of latent herpesviruses is associated with progressive cognitive impairment and Alzheimer's disease (AD). As an enveloped DNA virus, HSV exploits cellular membrane systems for its life cycle, and thereby comes in contact with the Rab family of GTPases, master regulators of intracellular membrane dynamics. Knock-down and overexpression of specific Rabs reduce HSV production. Disheveled membrane compartments could lead to AD because membrane sorting and trafficking are crucial for synaptic vesicle formation, neuronal survival signaling and Abeta production. Amyloid precursor protein (APP), a transmembrane glycoprotein, is the parent of Abeta, the major component of senile plaques in AD. Up-regulation of APP expression due to HSV is significant since excess APP interferes with Rab5 endocytic trafficking in neurons. Here, we show that purified PC12-cell endosomes transport both anterograde and retrograde when injected into the squid giant axon at rates similar to isolated HSV. Intracellular HSV co-fractionates with these endosomes, contains APP, Rab5 and TrkA, and displays a second membrane. HSV infected PC12 cells up-regulate APP expression. Whether interference with Rabs has a specific effect on HSV or indirectly affects membrane compartment dynamics co-opted by virus needs further study. Ultimately Rabs, their effectors or their membrane-binding partners may serve as handles to reduce the impact of viral re-activation on cognitive function, or even as more general-purpose anti-microbial therapies.
Collapse
Affiliation(s)
- Elaine L. Bearer
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Chengbiao Wu
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
20
|
Infectious Herpes Simplex Virus in the Brain Stem Is Correlated with Reactivation in the Trigeminal Ganglia. J Virol 2019; 93:JVI.02209-18. [PMID: 30728262 PMCID: PMC6450102 DOI: 10.1128/jvi.02209-18] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 01/23/2019] [Indexed: 12/14/2022] Open
Abstract
Latent herpes simplex virus (HSV) DNA has been detected in the central nervous systems (CNS) of humans postmortem, and infection with HSV has been correlated with the development of neurodegenerative diseases. However, whether HSV can directly reactivate in the CNS and/or infectious virus can be transported to the CNS following reactivation in peripheral ganglia has been unclear. In this study, infectious virus was recovered from both the trigeminal ganglia and the brain stem of latently infected mice following a reactivation stimulus, but a higher frequency of reactivation and increased titers of infectious virus were recovered from the trigeminal ganglia. Viral proteins were detected in neurons of the trigeminal ganglia, but a cellular source of infectious virus could not be identified in the brain stem. These results suggest that infectious virus is transported from the ganglia to the CNS following reactivation but do not exclude the potential for direct reactivation in the CNS. Herpes simplex virus (HSV) establishes latency in neurons of the peripheral and central nervous systems (CNS). Evidence is mounting that HSV latency and reactivation in the nervous system has the potential to promote neurodegenerative processes. Understanding how this occurs is an important human health goal. In the mouse model, in vivo viral reactivation in the peripheral nervous system, triggered by hyperthermic stress, has been well characterized with respect to frequency and cell type. However, characterization of in vivo reactivation in the CNS is extremely limited. Further, it remains unclear whether virus reactivated in the peripheral nervous system is transported to the CNS in an infectious form, how often this occurs, and what parameters underlie the efficiency and outcomes of this process. In this study, reactivation was quantified in the trigeminal ganglia (TG) and the brain stem from the same latently infected animal using direct assays of equivalent sensitivity. Reactivation was detected more frequently in the TG than in the brain stem and, in all but one case, the amount of virus recovered was greater in the TG than that detected in the brain stem. Viral protein positive neurons were observed in the TG, but a cellular source for reactivation in the brain stem was not identified, despite serially sectioning and examining the entire tissue (0/6 brain stems). These findings suggest that infectious virus detected in the brain stem is primarily the result of transport of reactivated virus from the TG into the brain stem. IMPORTANCE Latent herpes simplex virus (HSV) DNA has been detected in the central nervous systems (CNS) of humans postmortem, and infection with HSV has been correlated with the development of neurodegenerative diseases. However, whether HSV can directly reactivate in the CNS and/or infectious virus can be transported to the CNS following reactivation in peripheral ganglia has been unclear. In this study, infectious virus was recovered from both the trigeminal ganglia and the brain stem of latently infected mice following a reactivation stimulus, but a higher frequency of reactivation and increased titers of infectious virus were recovered from the trigeminal ganglia. Viral proteins were detected in neurons of the trigeminal ganglia, but a cellular source of infectious virus could not be identified in the brain stem. These results suggest that infectious virus is transported from the ganglia to the CNS following reactivation but do not exclude the potential for direct reactivation in the CNS.
Collapse
|
21
|
Fanni AM, Monge FA, Lin CY, Thapa A, Bhaskar K, Whitten DG, Chi EY. High Selectivity and Sensitivity of Oligomeric p-Phenylene Ethynylenes for Detecting Fibrillar and Prefibrillar Amyloid Protein Aggregates. ACS Chem Neurosci 2019; 10:1813-1825. [PMID: 30657326 DOI: 10.1021/acschemneuro.8b00719] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Misfolding and aggregation of amyloid proteins into fibrillar aggregates is a central pathogenic event in neurodegenerative disorders such as Alzheimer's (AD) and Parkinson's diseases (PD). Currently, there is a lack of reliable sensors for detecting the range of protein aggregates involved in disease etiology, particularly the prefibrillar aggregate conformations that are more neurotoxic. In this study, the fluorescent sensing of two novel oligomeric p-phenylene ethynylenes (OPEs), anionic OPE1- and cationic OPE2+, for detecting prefibrillar and fibrillar aggregates of AD-associated amyloid-β (Aβ40 and Aβ42) and PD-associated α-synuclein proteins (wildtype, and single mutants A30P, E35K, and A53T) over their monomeric counterparts, were tested. Furthermore, the performance of OPEs was evaluated and compared to thioflavin T (ThT), the most widely used fibril dye. Our results show that OPE1- and OPE2+ exhibited aggregate-specific binding inducing large fluorescence turn-on and spectral shifts based on a combination of backbone planarization, hydrophobic unquenching, and superluminescent OPE complex formation sensing modes. OPEs exhibited higher selectivity, higher binding affinity, and comparable limits of detection for Aβ40 fibrils compared to ThT. OPE2+ exhibited the largest fluorescence turn-on and highest sensitivity. Significantly, OPEs detected prefibrillar aggregates of Aβ42 and α-synuclein that ThT failed to detect. The superior sensing performance, the nonprotein specific detection, and the ability to selectively detect fibrillar and prefibrillar amyloid protein aggregates point to the potential of OPEs to overcome the limitations of existing probes and promise significant advancement in the detection of the myriad of protein aggregates involved in the early stages of AD and PD.
Collapse
|
22
|
Duarte LF, Farías MA, Álvarez DM, Bueno SM, Riedel CA, González PA. Herpes Simplex Virus Type 1 Infection of the Central Nervous System: Insights Into Proposed Interrelationships With Neurodegenerative Disorders. Front Cell Neurosci 2019; 13:46. [PMID: 30863282 PMCID: PMC6399123 DOI: 10.3389/fncel.2019.00046] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 01/30/2019] [Indexed: 12/21/2022] Open
Abstract
Herpes simplex virus type 1 (HSV-1) is highly prevalent in humans and can reach the brain without evident clinical symptoms. Once in the central nervous system (CNS), the virus can either reside in a quiescent latent state in this tissue, or eventually actively lead to severe acute necrotizing encephalitis, which is characterized by exacerbated neuroinflammation and prolonged neuroimmune activation producing a life-threatening disease. Although HSV-1 encephalitis can be treated with antivirals that limit virus replication, neurological sequelae are common and the virus will nevertheless remain for life in the neural tissue. Importantly, there is accumulating evidence that suggests that HSV-1 infection of the brain both, in symptomatic and asymptomatic individuals could lead to neuronal damage and eventually, neurodegenerative disorders. Here, we review and discuss acute and chronic infection of particular brain regions by HSV-1 and how this may affect neuron and cognitive functions in the host. We review potential cellular and molecular mechanisms leading to neurodegeneration, such as protein aggregation, dysregulation of autophagy, oxidative cell damage and apoptosis, among others. Furthermore, we discuss the impact of HSV-1 infection on brain inflammation and its potential relationship with neurodegenerative diseases.
Collapse
Affiliation(s)
- Luisa F Duarte
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Mónica A Farías
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Diana M Álvarez
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M Bueno
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Claudia A Riedel
- Millennium Institute on Immunology and Immunotherapy, Departamento de Biología Celular, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Pablo A González
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
23
|
Romeo MA, Faggioni A, Cirone M. Could autophagy dysregulation link neurotropic viruses to Alzheimer's disease? Neural Regen Res 2019; 14:1503-1506. [PMID: 31089040 PMCID: PMC6557098 DOI: 10.4103/1673-5374.253508] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Neurotropic herpesviruses have been associated with the onset and progression of Alzheimer’s disease, a common form of dementia that afflicts a large percentage of elderly individuals. Interestingly, among the neurotropic herpesviruses, herpes simplex virus-1, human herpesvirus-6A, and human herpesvirus-6B have been reported to infect several cell types present in the central nervous system and to dysregulate autophagy, a process required for homeostasis of cells, especially neurons. Indeed autophagosome accumulation, indicating an unbalance between autophagosome formation and autophagosome degradation, has been observed in neurons of Alzheimer’s disease patients and may play a role in the intracellular and extracellular accumulation of amyloid β and in the altered protein tau metabolism. Moreover, herpesvirus infection of central nervous system cells such as glia and microglia can increase the production of oxidant species through the alteration of mitochondrial dynamics and promote inflammation, another hallmark of Alzheimer’s disease. This evidence suggests that it is worth further investigating the role of neurotropic herpesviruses, particularly human herpesvirus-6A/B, in the etiopathogenesis of Alzheimer’s disease.
Collapse
Affiliation(s)
- Maria Anele Romeo
- Department of Experimental Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Alberto Faggioni
- Department of Experimental Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Mara Cirone
- Department of Experimental Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| |
Collapse
|
24
|
Fisher-Owens SA, Mertz E. Preventing Oral Disease: Alternative Providers and Places to Address This Commonplace Condition. Pediatr Clin North Am 2018; 65:1063-1072. [PMID: 30213349 DOI: 10.1016/j.pcl.2018.05.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Oral health disease in young children has not decreased, despite adequate modalities for treatment and prevention. Because many children may not see a dentist before oral disease has begun, disease progression can be expected, affecting short-term and long-term oral health. However, most children are seen by other health professionals frequently in their youngest years, providing a unique opportunity to help weave a safety net of oral health care until they are established in a dental home. This article details ways primary care providers can promote oral health, including ways to integrate ancillary dental professionals into the primary care home.
Collapse
Affiliation(s)
- Susan A Fisher-Owens
- Department of Pediatrics, UCSF School of Medicine, Zuckerberg San Francisco General Hospital, 1001 Potrero Avenue/MS6E37, San Francisco, CA 94110, USA; Department of Preventive and Restorative Dental Sciences, UCSF School of Dentistry, 505 Parnassus Avenue, San Francisco, CA 94143, USA.
| | - Elizabeth Mertz
- Faculty, Healthforce Center for Research and Leadership Development, 3333 California Street, Suite 410, San Francisco, CA 94143, USA
| |
Collapse
|
25
|
Fülöp T, Itzhaki RF, Balin BJ, Miklossy J, Barron AE. Role of Microbes in the Development of Alzheimer's Disease: State of the Art - An International Symposium Presented at the 2017 IAGG Congress in San Francisco. Front Genet 2018; 9:362. [PMID: 30250480 PMCID: PMC6139345 DOI: 10.3389/fgene.2018.00362] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 08/21/2018] [Indexed: 12/17/2022] Open
Abstract
This article reviews research results and ideas presented at a special symposium at the International Association of Gerontology and Geriatrics (IAGG) Congress held in July 2017 in San Francisco. Five researchers presented their results related to infection and Alzheimer's disease (AD). Prof. Itzhaki presented her work on the role of viruses, specifically HSV-1, in the pathogenesis of AD. She maintains that although it is true that most people harbor HSV-1 infection, either latent or active, nonetheless aspects of herpes infection can play a role in the pathogenesis of AD, based on extensive experimental evidence from AD brains and infected cell cultures. Dr. Miklossy presented research on the high prevalence of bacterial infections that correlate with AD, specifically spirochete infections, which have been known for a century to be a significant cause of dementia (e.g., in syphilis). She demonstrated how spirochetes drive senile plaque formation, which are in fact biofilms. Prof. Balin then described the involvement of brain tissue infection by the Chlamydia pneumoniae bacterium, with its potential to use the innate immune system in its spread, and its initiation of tissue damage characteristic of AD. Prof. Fülöp described the role of AD-associated amyloid beta (Aβ) peptide as an antibacterial, antifungal and antiviral innate immune effector produced in reaction to microorganisms that attack the brain. Prof. Barron put forward the novel hypothesis that, according to her experiments, there is strong sequence-specific binding between the AD-associated Aβ and another ubiquitous and important human innate immune effector, the cathelicidin peptide LL-37. Given this binding, LL-37 expression in the brain will decrease Aβ deposition via formation of non-toxic, soluble Aβ/LL-37 complexes. Therefore, a chronic underexpression of LL-37 could be the factor that simultaneously permits chronic infections in brain tissue and allows for pathological accumulation of Aβ. This first-of-its-kind symposium opened the way for a paradigm shift in studying the pathogenesis of AD, from the "amyloid cascade hypothesis," which so far has been quite unsuccessful, to a new "infection hypothesis," or perhaps more broadly, "innate immune system dysregulation hypothesis," which may well permit and lead to the discovery of new treatments for AD patients.
Collapse
Affiliation(s)
- Tamàs Fülöp
- Department of Medicine, Division of Geriatrics, Research Center on Aging, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Ruth F. Itzhaki
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Brian J. Balin
- Department of Bio-Medical Sciences, Center for Chronic Disorders of Aging, Philadelphia College of Osteopathic Medicine, Philadelphia, PA, United States
| | - Judith Miklossy
- International Alzheimer Research Centre, Prevention Alzheimer International Foundation, Martigny-Croix, Switzerland
| | - Annelise E. Barron
- Department of Bioengineering, Stanford University, Stanford, CA, United States
| |
Collapse
|
26
|
Abstract
PURPOSE OF REVIEW Viruses, particularly herpes simplex virus (HSV), may be a cause of Alzheimer's disease (AD). The evidence supporting the viral hypothesis suggests that antiviral treatment trials, which have not been conducted, are warranted. RECENT FINDINGS HSV1 (oral herpes) and HSV2 (genital herpes) can trigger amyloid aggregation, and their DNA is common in amyloid plaques. HSV1 reactivation is associated with tau hyperphosphorylation and possibly tau propagation. Anti-HSV drugs reduce Aβ and p-tau accumulation in infected mouse brains. Clinically, after the initial oral infection, herpes simplex virus-1 (HSV1) becomes latent in the trigeminal ganglion and recurrent reactivation may produce neuronal damage and AD pathology. Clinical studies show cognitive impairment in HSV seropositive patients, and antiviral drugs show strong efficacy against HSV. An antiviral treatment trial in AD is clearly warranted. A phase II treatment trial with valacyclovir, an anti-HSV drug, recently began with evaluation of clinical and biomarker outcomes.
Collapse
|
27
|
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: 8.8] [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
|
28
|
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: 16.5] [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
|
29
|
Hogestyn JM, Mock DJ, Mayer-Proschel M. Contributions of neurotropic human herpesviruses herpes simplex virus 1 and human herpesvirus 6 to neurodegenerative disease pathology. Neural Regen Res 2018; 13:211-221. [PMID: 29557362 PMCID: PMC5879884 DOI: 10.4103/1673-5374.226380] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Human herpesviruses (HVs) have developed ingenious mechanisms that enable them to traverse the defenses of the central nervous system (CNS). The ability of HVs to enter a state of latency, a defining characteristic of this viral family, allows them to persist in the human host indefinitely. As such, HVs represent the most frequently detected pathogens in the brain. Under constant immune pressure, these infections are largely asymptomatic in healthy hosts. However, many neurotropic HVs have been directly connected with CNS pathology in the context of other stressors and genetic risk factors. In this review, we discuss the potential mechanisms by which neurotropic HVs contribute to neurodegenerative disease (NDD) pathology by highlighting two prominent members of the HV family, herpes simplex virus 1 (HSV-1) and human herpesvirus 6 (HHV-6). We (i) introduce the infectious pathways and replicative cycles of HSV-1 and HHV-6 and then (ii) review the clinical evidence supporting associations between these viruses and the NDDs Alzheimer's disease (AD) and multiple sclerosis (MS), respectively. We then (iii) highlight and discuss potential mechanisms by which these viruses exert negative effects on neurons and glia. Finally, we (iv) discuss how these viruses could interact with other disease-modifying factors to contribute to the initiation and/or progression of NDDs.
Collapse
Affiliation(s)
| | - David J Mock
- Department of Biomedical Genetics, University of Rochester, Rochester, NY, USA
| | - Margot Mayer-Proschel
- Department of Neuroscience, University of Rochester; Department of Biomedical Genetics, University of Rochester, Rochester, NY, USA
| |
Collapse
|
30
|
Buch A, Müller O, Ivanova L, Döhner K, Bialy D, Bosse JB, Pohlmann A, Binz A, Hegemann M, Nagel CH, Koltzenburg M, Viejo-Borbolla A, Rosenhahn B, Bauerfeind R, Sodeik B. Inner tegument proteins of Herpes Simplex Virus are sufficient for intracellular capsid motility in neurons but not for axonal targeting. PLoS Pathog 2017; 13:e1006813. [PMID: 29284065 PMCID: PMC5761964 DOI: 10.1371/journal.ppat.1006813] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 01/10/2018] [Accepted: 12/14/2017] [Indexed: 02/07/2023] Open
Abstract
Upon reactivation from latency and during lytic infections in neurons, alphaherpesviruses assemble cytosolic capsids, capsids associated with enveloping membranes, and transport vesicles harboring fully enveloped capsids. It is debated whether capsid envelopment of herpes simplex virus (HSV) is completed in the soma prior to axonal targeting or later, and whether the mechanisms are the same in neurons derived from embryos or from adult hosts. We used HSV mutants impaired in capsid envelopment to test whether the inner tegument proteins pUL36 or pUL37 necessary for microtubule-mediated capsid transport were sufficient for axonal capsid targeting in neurons derived from the dorsal root ganglia of adult mice. Such neurons were infected with HSV1-ΔUL20 whose capsids recruited pUL36 and pUL37, with HSV1-ΔUL37 whose capsids associate only with pUL36, or with HSV1-ΔUL36 that assembles capsids lacking both proteins. While capsids of HSV1-ΔUL20 were actively transported along microtubules in epithelial cells and in the somata of neurons, those of HSV1-ΔUL36 and -ΔUL37 could only diffuse in the cytoplasm. Employing a novel image analysis algorithm to quantify capsid targeting to axons, we show that only a few capsids of HSV1-ΔUL20 entered axons, while vesicles transporting gD utilized axonal transport efficiently and independently of pUL36, pUL37, or pUL20. Our data indicate that capsid motility in the somata of neurons mediated by pUL36 and pUL37 does not suffice for targeting capsids to axons, and suggest that capsid envelopment needs to be completed in the soma prior to targeting of herpes simplex virus to the axons, and to spreading from neurons to neighboring cells.
Collapse
Affiliation(s)
- Anna Buch
- Institute of Virology, Hannover Medical School, Hannover, Germany
- NRENNT–Niedersachsen Research Network on Neuroinfectiology, Hannover, Germany
- DZIF—German Center for Infection Research, Hannover, Germany
| | - Oliver Müller
- Institute for Information Processing, Leibniz University, Hannover, Germany
- REBIRTH—From Regenerative Biology to Reconstructive Therapy, Hannover, Germany
| | - Lyudmila Ivanova
- Institute of Virology, Hannover Medical School, Hannover, Germany
- NRENNT–Niedersachsen Research Network on Neuroinfectiology, Hannover, Germany
- REBIRTH—From Regenerative Biology to Reconstructive Therapy, Hannover, Germany
| | - Katinka Döhner
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Dagmara Bialy
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Jens B. Bosse
- Heinrich-Pette-Institute, Leibniz-Institute for Experimental Virology, Hamburg, Germany
| | - Anja Pohlmann
- Institute of Virology, Hannover Medical School, Hannover, Germany
- REBIRTH—From Regenerative Biology to Reconstructive Therapy, Hannover, Germany
| | - Anne Binz
- Institute of Virology, Hannover Medical School, Hannover, Germany
- REBIRTH—From Regenerative Biology to Reconstructive Therapy, Hannover, Germany
| | - Maike Hegemann
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | | | | | - Abel Viejo-Borbolla
- Institute of Virology, Hannover Medical School, Hannover, Germany
- NRENNT–Niedersachsen Research Network on Neuroinfectiology, Hannover, Germany
| | - Bodo Rosenhahn
- Institute for Information Processing, Leibniz University, Hannover, Germany
- REBIRTH—From Regenerative Biology to Reconstructive Therapy, Hannover, Germany
| | - Rudolf Bauerfeind
- Research Core Unit Laser Microscopy, Hannover Medical School, Hannover, Germany
| | - Beate Sodeik
- Institute of Virology, Hannover Medical School, Hannover, Germany
- NRENNT–Niedersachsen Research Network on Neuroinfectiology, Hannover, Germany
- DZIF—German Center for Infection Research, Hannover, Germany
- REBIRTH—From Regenerative Biology to Reconstructive Therapy, Hannover, Germany
| |
Collapse
|
31
|
Itzhaki RF, Lathe R, Balin BJ, Ball MJ, Bearer EL, Braak H, Bullido MJ, Carter C, Clerici M, Cosby SL, Del Tredici K, Field H, Fulop T, Grassi C, Griffin WST, Haas J, Hudson AP, Kamer AR, Kell DB, Licastro F, Letenneur L, Lövheim H, Mancuso R, Miklossy J, Otth C, Palamara AT, Perry G, Preston C, Pretorius E, Strandberg T, Tabet N, Taylor-Robinson SD, Whittum-Hudson JA. Microbes and Alzheimer's Disease. J Alzheimers Dis 2016; 51:979-84. [PMID: 26967229 DOI: 10.3233/jad-160152] [Citation(s) in RCA: 375] [Impact Index Per Article: 46.9] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Ruth F Itzhaki
- Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester, UK
| | - Richard Lathe
- Division of Infection and Pathway Medicine, University of Edinburgh, Little France, Edinburgh, UK
| | - Brian J Balin
- Center for Chronic Disorders of Aging, Philadelphia College of Osteopathic Medicine, Philadelphia, USA
| | - Melvyn J Ball
- Department of Pathology (Neuropathology), Oregon Health and Science University, Portland, OR, USA
| | - Elaine L Bearer
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Heiko Braak
- Clinical Neuroanatomy Section, Department of Neurology, Center for Biomedical Research, University of Ulm, Ulm, Germany
| | - Maria J Bullido
- Centro de Biologia Molecular 'Severo Ochoa' (CSIC-UAM), Universidad Autonoma de Madrid, and Centro de Investigacion en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | | | - Mario Clerici
- University of Milano and IRCCS SM Nascente, Don C Gnocchi Foundation, Milan, Italy
| | - S Louise Cosby
- Centre for Infection and Immunity, Medical Biology Centre, Queen's University, Belfast, UK
| | - Kelly Del Tredici
- Clinical Neuroanatomy Section, Department of Neurology, Center for Biomedical Research, University of Ulm, Ulm, Germany
| | | | - Tamas Fulop
- Department of Medicine, Division of Geriatrics, Université de Sherbrooke, Sherbrooke, PQ, Canada
| | - Claudio Grassi
- Institute of Human Physiology, Medical School, Universitá Cattolica, Rome; San Raffaele Pisana Scientific Institute for Research, Hospitalization, and Health Care, Rome, Italy
| | - W Sue T Griffin
- Department of Geriatrics, University of Arkansas for Medical Sciences, and Geriatric Research, Education, and Clinical Center, Little Rock, AR, USA
| | - Jürgen Haas
- Division of Infection and Pathway Medicine, University of Edinburgh, Little France, Edinburgh, UK
| | - Alan P Hudson
- Department of Immunology and Microbiology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Angela R Kamer
- NYU College of Dentistry, Department of Periodontology and Implant Dentistry, New York, NY, USA
| | - Douglas B Kell
- School of Chemistry, Manchester Institute of Biotechnology, University of Manchester, Manchester, UK
| | - Federico Licastro
- Department of Experimental, Diagnostic, and Specialty Medicine, School of Medicine, University of Bologna, Bologna, Italy
| | | | - Hugo Lövheim
- Department of Community Medicine and Rehabilitation, Geriatric Medicine, Umeå University, Umeå, Sweden
| | | | - Judith Miklossy
- Prevention Alzheimer International Foundation, International Alzheimer Research Center, Martigny-Croix, Switzerland
| | - Carola Otth
- Institute of Clinical Microbiology, Faculty of Medicine, Austral University of Chile, Valdivia, Chile
| | - Anna Teresa Palamara
- Department of Public Health and Infectious Diseases, Institute Pasteur Cenci Bolognetti Foundation, Sapienza University of Rome; San Raffaele Pisana Scientific Institute for Research, Hospitalization, and Health Care, Rome, Italy
| | - George Perry
- College of Sciences, University of Texas at San Antonio, San Antonio, TX, USA
| | | | - Etheresia Pretorius
- Applied Morphology Research Centre, Department of Physiology, Faculty of Health Sciences, University of Pretoria, Arcadia, South Africa
| | - Timo Strandberg
- Helsinki University Hospital and University of Helsinki; University of Oulu, Centre of Life Course Health Research, Oulu, Finland
| | - Naji Tabet
- Division of Old Age Psychiatry, Brighton and Sussex Medical School, Brighton, UK
| | | | - Judith A Whittum-Hudson
- Departments of Immunology and Microbiology, Internal Medicine (Rheumatology), and Ophthalmology, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
32
|
Agostini S, Mancuso R, Baglio F, Clerici M. A protective role for herpes simplex virus type-1-specific humoral immunity in Alzheimer’s Disease. Expert Rev Anti Infect Ther 2016; 15:89-91. [DOI: 10.1080/14787210.2017.1264271] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
| | | | | | - Mario Clerici
- Don C. Gnocchi Foundation IRCCS – ONLUS, Milano, Italy
- Department of Pathophysiology and Transplantation, University of Milano, Milano, Italy
| |
Collapse
|
33
|
Agostini S, Mancuso R, Baglio F, Cabinio M, Hernis A, Costa AS, Calabrese E, Nemni R, Clerici M. High avidity HSV-1 antibodies correlate with absence of amnestic Mild Cognitive Impairment conversion to Alzheimer's disease. Brain Behav Immun 2016; 58:254-260. [PMID: 27470229 DOI: 10.1016/j.bbi.2016.07.153] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 07/11/2016] [Accepted: 07/22/2016] [Indexed: 12/12/2022] Open
Abstract
Amnestic Mild Cognitive Impairment (aMCI) is an alteration in cognitive abilities that can progress to Alzheimer's disease (AD), a condition in which herpes simplex type 1 (HSV-1) infection might play a pathogenetic role. Prognostic indexes capable of predicting aMCI conversion to AD are only partially understood. The objective of the present work is to verify whether HSV-1 immune responses is involved in conversion of aMCI to AD and correlate with grey matter brain morphometry. Two homogeneous groups of individuals who did or did not convert to AD over a 24-months period were selected after retrospective analysis of a cohort of patients with a diagnosis of aMCI. The selection of subjects was based on: a) clinical follow-up; b) neurocognitive evaluation at baseline and after 24months; c) availability of serum and DNA samples at baseline. 36 aMCI individuals, 21 of whom did (aMCI-converters) and 15 of whom did not (aMCI-non-converters) convert to AD, were included in the study. HSV-1 antibody (Ab) titers, avidity index and APOE genotyping were performed in all the enrolled individuals at baseline. Brain magnetic resonance imaging (MRI) by 1.5T scanner results at baseline were available as well in most (29/36) of these individuals. HSV-1-specific Ab titers were increased at baseline in aMCI-non-converters, and the avidity of these Ab was significantly higher in aMCI-non-converter compared to aMCI-converter (p=0.0018). Receiver operating characteristics analysis showed that HSV-1 avidity had a predictive value in distinguishing between aMCI-non-converters and aMCI-converters (p<0.0001). Notably, a positive correlation was detected as well between HSV-1 antibody titers and MRI-evaluated cortical volumes in the left hippocampus and amigdala (pcorr<0.05). In conclusion, stronger HSV-1-specific humoral responses associate with protection against AD conversion and better-preserved cortical volumes. These results reinforce the hypothesis for a role for HSV-1 in the pathogenesis of AD.
Collapse
Affiliation(s)
- Simone Agostini
- Don C. Gnocchi Foundation IRCCS - ONLUS, Piazza Morandi 3, 20121 Milano, Italy.
| | - Roberta Mancuso
- Don C. Gnocchi Foundation IRCCS - ONLUS, Piazza Morandi 3, 20121 Milano, Italy
| | - Francesca Baglio
- Don C. Gnocchi Foundation IRCCS - ONLUS, Piazza Morandi 3, 20121 Milano, Italy
| | - Monia Cabinio
- Don C. Gnocchi Foundation IRCCS - ONLUS, Piazza Morandi 3, 20121 Milano, Italy
| | - Ambra Hernis
- Don C. Gnocchi Foundation IRCCS - ONLUS, Piazza Morandi 3, 20121 Milano, Italy
| | - Andrea Saul Costa
- Don C. Gnocchi Foundation IRCCS - ONLUS, Piazza Morandi 3, 20121 Milano, Italy
| | - Elena Calabrese
- Don C. Gnocchi Foundation IRCCS - ONLUS, Piazza Morandi 3, 20121 Milano, Italy
| | - Raffaello Nemni
- Don C. Gnocchi Foundation IRCCS - ONLUS, Piazza Morandi 3, 20121 Milano, Italy; Department of Physiopathology and Transplantation, University of Milano, via Fratelli Cervi 93, 20090 Milano, Italy
| | - Mario Clerici
- Don C. Gnocchi Foundation IRCCS - ONLUS, Piazza Morandi 3, 20121 Milano, Italy; Department of Physiopathology and Transplantation, University of Milano, via Fratelli Cervi 93, 20090 Milano, Italy
| |
Collapse
|
34
|
Harris SA, Harris EA. Herpes Simplex Virus Type 1 and Other Pathogens are Key Causative Factors in Sporadic Alzheimer's Disease. J Alzheimers Dis 2016; 48:319-53. [PMID: 26401998 PMCID: PMC4923765 DOI: 10.3233/jad-142853] [Citation(s) in RCA: 162] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
This review focuses on research in epidemiology, neuropathology, molecular biology, and genetics regarding the hypothesis that pathogens interact with susceptibility genes and are causative in sporadic Alzheimer's disease (AD). Sporadic AD is a complex multifactorial neurodegenerative disease with evidence indicating coexisting multi-pathogen and inflammatory etiologies. There are significant associations between AD and various pathogens, including Herpes simplex virus type 1 (HSV-1), Cytomegalovirus, and other Herpesviridae, Chlamydophila pneumoniae, spirochetes, Helicobacter pylori, and various periodontal pathogens. These pathogens are able to evade destruction by the host immune system, leading to persistent infection. Bacterial and viral DNA and RNA and bacterial ligands increase the expression of pro-inflammatory molecules and activate the innate and adaptive immune systems. Evidence demonstrates that pathogens directly and indirectly induce AD pathology, including amyloid-β (Aβ) accumulation, phosphorylation of tau protein, neuronal injury, and apoptosis. Chronic brain infection with HSV-1, Chlamydophila pneumoniae, and spirochetes results in complex processes that interact to cause a vicious cycle of uncontrolled neuroinflammation and neurodegeneration. Infections such as Cytomegalovirus, Helicobacter pylori, and periodontal pathogens induce production of systemic pro-inflammatory cytokines that may cross the blood-brain barrier to promote neurodegeneration. Pathogen-induced inflammation and central nervous system accumulation of Aβ damages the blood-brain barrier, which contributes to the pathophysiology of AD. Apolipoprotein E4 (ApoE4) enhances brain infiltration by pathogens including HSV-1 and Chlamydophila pneumoniae. ApoE4 is also associated with an increased pro-inflammatory response by the immune system. Potential antimicrobial treatments for AD are discussed, including the rationale for antiviral and antibiotic clinical trials.
Collapse
Affiliation(s)
- Steven A Harris
- St. Vincent Medical Group, Northside Internal Medicine, Indianapolis, IN, USA
| | | |
Collapse
|
35
|
Owen DJ, Crump CM, Graham SC. Tegument Assembly and Secondary Envelopment of Alphaherpesviruses. Viruses 2015; 7:5084-114. [PMID: 26393641 PMCID: PMC4584305 DOI: 10.3390/v7092861] [Citation(s) in RCA: 134] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 08/22/2015] [Accepted: 08/26/2015] [Indexed: 02/07/2023] Open
Abstract
Alphaherpesviruses like herpes simplex virus are large DNA viruses characterized by their ability to establish lifelong latent infection in neurons. As for all herpesviruses, alphaherpesvirus virions contain a protein-rich layer called "tegument" that links the DNA-containing capsid to the glycoprotein-studded membrane envelope. Tegument proteins mediate a diverse range of functions during the virus lifecycle, including modulation of the host-cell environment immediately after entry, transport of virus capsids to the nucleus during infection, and wrapping of cytoplasmic capsids with membranes (secondary envelopment) during virion assembly. Eleven tegument proteins that are conserved across alphaherpesviruses have been implicated in the formation of the tegument layer or in secondary envelopment. Tegument is assembled via a dense network of interactions between tegument proteins, with the redundancy of these interactions making it challenging to determine the precise function of any specific tegument protein. However, recent studies have made great headway in defining the interactions between tegument proteins, conserved across alphaherpesviruses, which facilitate tegument assembly and secondary envelopment. We summarize these recent advances and review what remains to be learned about the molecular interactions required to assemble mature alphaherpesvirus virions following the release of capsids from infected cell nuclei.
Collapse
Affiliation(s)
- Danielle J Owen
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK
| | - Colin M Crump
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK.
| | - Stephen C Graham
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK.
| |
Collapse
|
36
|
Cellular Protein WDR11 Interacts with Specific Herpes Simplex Virus Proteins at the trans-Golgi Network To Promote Virus Replication. J Virol 2015; 89:9841-52. [PMID: 26178983 DOI: 10.1128/jvi.01705-15] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 07/12/2015] [Indexed: 12/23/2022] Open
Abstract
UNLABELLED It has recently been proposed that the herpes simplex virus (HSV) protein ICP0 has cytoplasmic roles in blocking antiviral signaling and in promoting viral replication in addition to its well-known proteasome-dependent functions in the nucleus. However, the mechanisms through which it produces these effects remain unclear. While investigating this further, we identified a novel cytoplasmic interaction between ICP0 and the poorly characterized cellular protein WDR11. During an HSV infection, WDR11 undergoes a dramatic change in localization at late times in the viral replication cycle, moving from defined perinuclear structures to a dispersed cytoplasmic distribution. While this relocation was not observed during infection with viruses other than HSV-1 and correlated with efficient HSV-1 replication, the redistribution was found to occur independently of ICP0 expression, instead requiring viral late gene expression. We demonstrate for the first time that WDR11 is localized to the trans-Golgi network (TGN), where it interacts specifically with some, but not all, HSV virion components, in addition to ICP0. Knockdown of WDR11 in cultured human cells resulted in a modest but consistent decrease in yields of both wild-type and ICP0-null viruses, in the supernatant and cell-associated fractions, without affecting viral gene expression. Although further study is required, we propose that WDR11 participates in viral assembly and/or secondary envelopment. IMPORTANCE While the TGN has been proposed to be the major site of HSV-1 secondary envelopment, this process is incompletely understood, and in particular, the role of cellular TGN components in this pathway is unknown. Additionally, little is known about the cellular functions of WDR11, although the disruption of this protein has been implicated in multiple human diseases. Therefore, our finding that WDR11 is a TGN-resident protein that interacts with specific viral proteins to enhance viral yields improves both our understanding of basic cellular biology as well as how this protein is co-opted by HSV.
Collapse
|
37
|
Itzhaki RF. Herpes simplex virus type 1 and Alzheimer's disease: increasing evidence for a major role of the virus. Front Aging Neurosci 2014; 6:202. [PMID: 25157230 PMCID: PMC4128394 DOI: 10.3389/fnagi.2014.00202] [Citation(s) in RCA: 146] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 07/23/2014] [Indexed: 12/15/2022] Open
Abstract
Herpes simplex virus type 1 (HSV1), when present in brain of carriers of the type 4 allele of the apolipoprotein E gene (APOE), has been implicated as a major factor in Alzheimer's disease (AD). It is proposed that virus is normally latent in many elderly brains but reactivates periodically (as in the peripheral nervous system) under certain conditions, for example stress, immunosuppression, and peripheral infection, causing cumulative damage and eventually development of AD. Diverse approaches have provided data that explicitly support, directly or indirectly, these concepts. Several have confirmed HSV1 DNA presence in human brains, and the HSV1-APOE-ε4 association in AD. Further, studies on HSV1-infected APOE-transgenic mice have shown that APOE-e4 animals display a greater potential for viral damage. Reactivated HSV1 can cause direct and inflammatory damage, probably involving increased formation of beta amyloid (Aβ) and of AD-like tau (P-tau)-changes found to occur in HSV1-infected cell cultures. Implicating HSV1 further in AD is the discovery that HSV1 DNA is specifically localized in amyloid plaques in AD. Other relevant, harmful effects of infection include the following: dynamic interactions between HSV1 and amyloid precursor protein (APP), which would affect both viral and APP transport; induction of toll-like receptors (TLRs) in HSV1-infected astrocyte cultures, which has been linked to the likely effects of reactivation of the virus in brain. Several epidemiological studies have shown, using serological data, an association between systemic infections and cognitive decline, with HSV1 particularly implicated. Genetic studies too have linked various pathways in AD with those occurring on HSV1 infection. In relation to the potential usage of antivirals to treat AD patients, acyclovir (ACV) is effective in reducing HSV1-induced AD-like changes in cell cultures, and valacyclovir, the bioactive form of ACV, might be most effective if combined with an antiviral that acts by a different mechanism, such as intravenous immunoglobulin (IVIG).
Collapse
Affiliation(s)
- Ruth F. Itzhaki
- Faculty of Life Sciences, University of ManchesterManchester, Lancs, UK
| |
Collapse
|
38
|
Piacentini R, De Chiara G, Li Puma DD, Ripoli C, Marcocci ME, Garaci E, Palamara AT, Grassi C. HSV-1 and Alzheimer's disease: more than a hypothesis. Front Pharmacol 2014; 5:97. [PMID: 24847267 PMCID: PMC4019841 DOI: 10.3389/fphar.2014.00097] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 04/16/2014] [Indexed: 12/22/2022] Open
Abstract
Among the multiple factors concurring to Alzheimer’s disease (AD) pathogenesis, greater attention should be devoted to the role played by infectious agents. Growing epidemiological and experimental evidence suggests that recurrent herpes simplex virus type-1 (HSV-1) infection is a risk factor for AD although the underlying molecular and functional mechanisms have not been fully elucidated yet. Here, we review literature suggesting the involvement of HSV-1 infection in AD also briefly mentioning possible pharmacological implications of these findings.
Collapse
Affiliation(s)
- Roberto Piacentini
- Institute of Human Physiology, Medical School, Università Cattolica del Sacro Cuore Rome, Italy
| | - Giovanna De Chiara
- Institute of Translational Pharmacology, National Research Council Rome, Italy
| | - Domenica D Li Puma
- Institute of Human Physiology, Medical School, Università Cattolica del Sacro Cuore Rome, Italy
| | - Cristian Ripoli
- Institute of Human Physiology, Medical School, Università Cattolica del Sacro Cuore Rome, Italy
| | - Maria E Marcocci
- Department of Public Health and Infectious Diseases, Sapienza University of Rome Rome, Italy
| | - Enrico Garaci
- San Raffaele Pisana Scientific Institute for Research, Hospitalization and Health Care, Telematic University Rome, Italy
| | - Anna T Palamara
- Department of Public Health and Infectious Diseases, Institute Pasteur Cenci Bolognetti Foundation, Sapienza University of Rome Rome, Italy ; San Raffaele Pisana Scientific Institute for Research, Hospitalization and Health Care Rome, Italy
| | - Claudio Grassi
- Institute of Human Physiology, Medical School, Università Cattolica del Sacro Cuore Rome, Italy
| |
Collapse
|
39
|
Affiliation(s)
- Lisa M. Gargano
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia 30322; ,
| | - James M. Hughes
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia 30322; ,
| |
Collapse
|
40
|
The herpes virus Fc receptor gE-gI mediates antibody bipolar bridging to clear viral antigens from the cell surface. PLoS Pathog 2014; 10:e1003961. [PMID: 24604090 PMCID: PMC3946383 DOI: 10.1371/journal.ppat.1003961] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 01/16/2014] [Indexed: 11/19/2022] Open
Abstract
The Herpes Simplex Virus 1 (HSV-1) glycoprotein gE-gI is a transmembrane Fc receptor found on the surface of infected cells and virions that binds human immunoglobulin G (hIgG). gE-gI can also participate in antibody bipolar bridging (ABB), a process by which the antigen-binding fragments (Fabs) of the IgG bind a viral antigen while the Fc binds to gE-gI. IgG Fc binds gE-gI at basic, but not acidic, pH, suggesting that IgG bound at extracellular pH by cell surface gE-gI would dissociate and be degraded in acidic endosomes/lysosomes if endocytosed. The fate of viral antigens associated with gE-gI-bound IgG had been unknown: they could remain at the cell surface or be endocytosed with IgG. Here, we developed an in vitro model system for ABB and investigated the trafficking of ABB complexes using 4-D confocal fluorescence imaging of ABB complexes with transferrin or epidermal growth factor, well-characterized intracellular trafficking markers. Our data showed that cells expressing gE-gI and the viral antigen HSV-1 gD endocytosed anti-gD IgG and gD in a gE-gI-dependent process, resulting in lysosomal localization. These results suggest that gE-gI can mediate clearance of infected cell surfaces of anti-viral host IgG and viral antigens to evade IgG-mediated responses, representing a general mechanism for viral Fc receptors in immune evasion and viral pathogenesis.
Collapse
|
41
|
Agostini S, Clerici M, Mancuso R. How plausible is a link between HSV-1 infection and Alzheimer's disease? Expert Rev Anti Infect Ther 2014; 12:275-8. [PMID: 24502805 DOI: 10.1586/14787210.2014.887442] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Alzheimer's disease is a leading cause of dementia with a multifactorial and still barely understood etiology. A growing body of epidemiologic and experimental data support a role for infectious agents in this process; herpes simplex virus 1 (HSV-1), in particular, is a strong suspect. We briefly summarize the data pointing at a pathogenic role for HSV-1 in the development of Alzheimer's disease and review results indicating that antiviral might be beneficial in the therapy of this condition.
Collapse
|
42
|
Zhou L, Miranda-Saksena M, Saksena NK. Viruses and neurodegeneration. Virol J 2013; 10:172. [PMID: 23724961 PMCID: PMC3679988 DOI: 10.1186/1743-422x-10-172] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Accepted: 05/20/2013] [Indexed: 11/10/2022] Open
Abstract
Neurodegenerative diseases (NDs) are chronic degenerative diseases of the central nervous system (CNS), which affect 37 million people worldwide. As the lifespan increases, the NDs are the fourth leading cause of death in the developed countries and becoming increasingly prevalent in developing countries. Despite considerable research, the underlying mechanisms remain poorly understood. Although the large majority of studies do not show support for the involvement of pathogenic aetiology in classical NDs, a number of emerging studies show support for possible association of viruses with classical neurodegenerative diseases in humans. Space does not permit for extensive details to be discussed here on non-viral-induced neurodegenerative diseases in humans, as they are well described in literature.Viruses induce alterations and degenerations of neurons both directly and indirectly. Their ability to attack the host immune system, regions of nervous tissue implies that they can interfere with the same pathways involved in classical NDs in humans. Supporting this, many similarities between classical NDs and virus-mediated neurodegeneration (non-classical) have been shown at the anatomic, sub-cellular, genomic and proteomic levels suggesting that viruses can explain neurodegenerative disorders mechanistically. The main objective of this review is to provide readers a detailed snapshot of similarities viral and non-viral neurodegenerative diseases share, so that mechanistic pathways of neurodegeneration in human NDs can be clearly understood. Viruses can guide us to unveil these pathways in human NDs. This will further stimulate the birth of new concepts in the biological research, which is needed for gaining deeper insights into the treatment of human NDs and delineate mechanisms underlying neurodegeneration.
Collapse
Affiliation(s)
- Li Zhou
- Retroviral Genetics Division, Center for Virus Research, Westmead Millennium Institute, Westmead Hospital, The University of Sydney, Westmead NSW 2145, Sydney Australia
| | | | | |
Collapse
|
43
|
Abstract
Late-onset Alzheimer's disease (AD) is the most prevalent cause of dementia among older adults, yet more than a century of research has not determined why this disease develops. One prevailing hypothesis is that late-onset AD is caused by infectious pathogens, an idea widely studied in both humans and experimental animal models. This review examines the infectious AD etiology hypothesis and summarizes existing evidence associating infectious agents with AD in humans. The various mechanisms through which different clinical and subclinical infections could cause or promote the progression of AD are considered, as is the concordance between putative infectious agents and the epidemiology of AD. We searched the PubMed, Web of Science, and EBSCO databases for research articles pertaining to infections and AD and systematically reviewed the evidence linking specific infectious pathogens to AD. The evidence compiled from the literature linking AD to an infectious cause is inconclusive, but the amount of evidence suggestive of an association is too substantial to ignore. Epidemiologic, clinical, and basic science studies that could improve on current understanding of the associations between AD and infections and possibly uncover ways to control this highly prevalent and debilitating disease are suggested.
Collapse
Affiliation(s)
| | - Robert Wallace
- Correspondence to Dr. Robert Wallace, Department of Epidemiology, College of Public Health, The University of Iowa, 105 River St. Iowa City, IA 52242 (e-mail: )
| |
Collapse
|
44
|
Kobayashi N, Nagata T, Shinagawa S, Oka N, Shimada K, Shimizu A, Tatebayashi Y, Yamada H, Nakayama K, Kondo K. Increase in the IgG avidity index due to herpes simplex virus type 1 reactivation and its relationship with cognitive function in amnestic mild cognitive impairment and Alzheimer’s disease. Biochem Biophys Res Commun 2013; 430:907-11. [DOI: 10.1016/j.bbrc.2012.12.054] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Accepted: 12/11/2012] [Indexed: 11/17/2022]
|
45
|
Seamster PE, Loewenberg M, Pascal J, Chauviere A, Gonzales A, Cristini V, Bearer EL. Quantitative measurements and modeling of cargo-motor interactions during fast transport in the living axon. Phys Biol 2012; 9:055005. [PMID: 23011729 DOI: 10.1088/1478-3975/9/5/055005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The kinesins have long been known to drive microtubule-based transport of sub-cellular components, yet the mechanisms of their attachment to cargo remain a mystery. Several different cargo-receptors have been proposed based on their in vitro binding affinities to kinesin-1. Only two of these-phosphatidyl inositol, a negatively charged lipid, and the carboxyl terminus of the amyloid precursor protein (APP-C), a trans-membrane protein-have been reported to mediate motility in living systems. A major question is how these many different cargo, receptors and motors interact to produce the complex choreography of vesicular transport within living cells. Here we describe an experimental assay that identifies cargo-motor receptors by their ability to recruit active motors and drive transport of exogenous cargo towards the synapse in living axons. Cargo is engineered by derivatizing the surface of polystyrene fluorescent nanospheres (100 nm diameter) with charged residues or with synthetic peptides derived from candidate motor receptor proteins, all designed to display a terminal COOH group. After injection into the squid giant axon, particle movements are imaged by laser-scanning confocal time-lapse microscopy. In this report we compare the motility of negatively charged beads with APP-C beads in the presence of glycine-conjugated non-motile beads using new strategies to measure bead movements. The ensuing quantitative analysis of time-lapse digital sequences reveals detailed information about bead movements: instantaneous and maximum velocities, run lengths, pause frequencies and pause durations. These measurements provide parameters for a mathematical model that predicts the spatiotemporal evolution of distribution of the two different types of bead cargo in the axon. The results reveal that negatively charged beads differ from APP-C beads in velocity and dispersion, and predict that at long time points APP-C will achieve greater progress towards the presynaptic terminal. The significance of this data and accompanying model pertains to the role transport plays in neuronal function, connectivity, and survival, and has implications in the pathogenesis of neurological disorders, such as Alzheimer's, Huntington and Parkinson's diseases.
Collapse
Affiliation(s)
- Pamela E Seamster
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
Bearer EL. HSV, axonal transport and Alzheimer's disease: in vitro and in vivo evidence for causal relationships. Future Virol 2012; 7:885-899. [PMID: 23335944 DOI: 10.2217/fvl.12.81] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
HSV, a neurotropic virus, travels within neuronal processes by fast axonal transport. During neuronal infection HSV travels retrograde from the sensory nerve terminus to the neuronal cell body, where it replicates or enters latency. During replication HSV travels anterograde from the cell body to the nerve terminus. Postmortem studies find a high frequency of HSV DNA in the trigeminal ganglia as well as the brain. Studies correlating HSV with Alzheimer's disease (AD) have been controversial. Here we review clinical evidence supporting such a link. Furthermore, the author describes experimental data showing physical interactions between nascent HSV particles and host transport machinery implicated in AD. The author concludes that the complexity of this relationship has been insufficiently explored, although the relative ease and nontoxicity of a potential anti-HSV treatment for AD demands further study.
Collapse
Affiliation(s)
- Elaine L Bearer
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM 81131, USA
| |
Collapse
|
47
|
Nagel CH, Döhner K, Binz A, Bauerfeind R, Sodeik B. Improper tagging of the non-essential small capsid protein VP26 impairs nuclear capsid egress of herpes simplex virus. PLoS One 2012; 7:e44177. [PMID: 22952920 PMCID: PMC3432071 DOI: 10.1371/journal.pone.0044177] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Accepted: 07/30/2012] [Indexed: 01/10/2023] Open
Abstract
To analyze the subcellular trafficking of herpesvirus capsids, the small capsid protein has been labeled with different fluorescent proteins. Here, we analyzed the infectivity of several HSV1(17(+)) strains in which the N-terminal region of the non-essential small capsid protein VP26 had been tagged at different positions. While some variants replicated with similar kinetics as their parental wild type strain, others were not infectious at all. Improper tagging resulted in the aggregation of VP26 in the nucleus, prevented efficient nuclear egress of viral capsids, and thus virion formation. Correlative fluorescence and electron microscopy showed that these aggregates had sequestered several other viral proteins, but often did not contain viral capsids. The propensity for aggregate formation was influenced by the type of the fluorescent protein domain, the position of the inserted tag, the cell type, and the progression of infection. Among the tags that we have tested, mRFPVP26 had the lowest tendency to induce nuclear aggregates, and showed the least reduction in replication when compared to wild type. Our data suggest that bona fide monomeric fluorescent protein tags have less impact on proper assembly of HSV1 capsids and nuclear capsid egress than tags that tend to dimerize. Small chemical compounds capable of inducing aggregate formation of VP26 may lead to new antiviral drugs against HSV infections.
Collapse
Affiliation(s)
| | - Katinka Döhner
- Institute of Virology, Hanover Medical School, Hanover, Germany
| | - Anne Binz
- Institute of Virology, Hanover Medical School, Hanover, Germany
| | | | - Beate Sodeik
- Institute of Virology, Hanover Medical School, Hanover, Germany
| |
Collapse
|
48
|
De Chiara G, Marcocci ME, Sgarbanti R, Civitelli L, Ripoli C, Piacentini R, Garaci E, Grassi C, Palamara AT. Infectious agents and neurodegeneration. Mol Neurobiol 2012; 46:614-38. [PMID: 22899188 PMCID: PMC3496540 DOI: 10.1007/s12035-012-8320-7] [Citation(s) in RCA: 161] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Accepted: 07/31/2012] [Indexed: 12/19/2022]
Abstract
A growing body of epidemiologic and experimental data point to chronic bacterial and viral infections as possible risk factors for neurodegenerative diseases, including Alzheimer’s disease, Parkinson’s disease and amyotrophic lateral sclerosis. Infections of the central nervous system, especially those characterized by a chronic progressive course, may produce multiple damage in infected and neighbouring cells. The activation of inflammatory processes and host immune responses cause chronic damage resulting in alterations of neuronal function and viability, but different pathogens can also directly trigger neurotoxic pathways. Indeed, viral and microbial agents have been reported to produce molecular hallmarks of neurodegeneration, such as the production and deposit of misfolded protein aggregates, oxidative stress, deficient autophagic processes, synaptopathies and neuronal death. These effects may act in synergy with other recognized risk factors, such as aging, concomitant metabolic diseases and the host’s specific genetic signature. This review will focus on the contribution given to neurodegeneration by herpes simplex type-1, human immunodeficiency and influenza viruses, and by Chlamydia pneumoniae.
Collapse
Affiliation(s)
- Giovanna De Chiara
- Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, Rome, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Henaff D, Radtke K, Lippé R. Herpesviruses exploit several host compartments for envelopment. Traffic 2012; 13:1443-9. [PMID: 22805610 DOI: 10.1111/j.1600-0854.2012.01399.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Revised: 07/13/2012] [Accepted: 07/17/2012] [Indexed: 01/16/2023]
Abstract
Enveloped viruses acquire their host-derived membrane at a variety of intracellular locations. Herpesviruses are complex entities that undergo several budding and fusion events during an infection. All members of this large family are believed to share a similar life cycle. However, they seemingly differ in terms of acquisition of their mature envelope. Herpes simplex virus is often believed to bud into an existing intracellular compartment, while the related cytomegalovirus may acquire its final envelope from a novel virus-induced assembly compartment. This review focuses on recent advances in the characterization of cellular compartment(s) potentially contributing to herpes virion final envelopment. It also examines the common points between seemingly distinct envelopment pathways and highlights the dynamic nature of intracellular compartments in the context of herpesvirus infections.
Collapse
Affiliation(s)
- Daniel Henaff
- Department of Pathology and Cell Biology, University of Montreal, Montreal, QC, Canada
| | | | | |
Collapse
|
50
|
Itzhaki RF, Wozniak MA. Could antivirals be used to treat Alzheimer’s disease? Future Microbiol 2012; 7:307-9. [DOI: 10.2217/fmb.12.10] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Ruth F Itzhaki
- Faculty of Life Sciences, The University of Manchester, Stopford Building, Oxford Road, Manchester, M13 9PT, UK
| | - Matthew A Wozniak
- Faculty of Life Sciences, The University of Manchester, Stopford Building, Oxford Road, Manchester, M13 9PT, UK
| |
Collapse
|