1
|
Chauhan P, Begum MY, Narapureddy BR, Gupta S, Wadhwa K, Singh G, Kumawat R, Sharma N, Ballal S, Jha SK, Abomughaid MM, B D, Ojha S, Jha NK. Unveiling the Involvement of Herpes Simplex Virus-1 in Alzheimer's Disease: Possible Mechanisms and Therapeutic Implications. Mol Neurobiol 2025; 62:5850-5874. [PMID: 39648189 DOI: 10.1007/s12035-024-04535-4] [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: 06/27/2023] [Accepted: 10/01/2024] [Indexed: 12/10/2024]
Abstract
Viruses pose a significant challenge and threat to human health, as demonstrated by the current COVID-19 pandemic. Neurodegeneration, particularly in the case of Alzheimer's disease (AD), is significantly influenced by viral infections. AD is a neurodegenerative disease that affects people of all ages and poses a significant threat to millions of individuals worldwide. The precise mechanism behind its development is not yet fully understood; however, the emergence and advancement of AD can be hastened by various environmental factors, such as bacterial and viral infections. There has been a longstanding suspicion that the herpes simplex virus-1 (HSV-1) may have a role to play in the development or advancement of AD. Reactivation of HSV-1 could potentially lead to damage to neurons, either by direct means or indirectly by triggering inflammation. This article provides an overview of the connection between HSV-1 infections and immune cells (astrocytes, microglia, and oligodendrocytes) in the progression of AD. It summarizes recent scientific research on how HSV-1 affects neurons, which could potentially shed light on the clinical features and treatment options for AD. In addition, the paper has explored the impact of HSV-1 on neurons and its role in various aspects of AD, such as Aβ secretion, tau hyperphosphorylation, metabolic dysregulation, oxidative damage, apoptosis, and autophagy. It is believed that the immune response triggered by HSV-1 reactivation plays a role in the development of neurodegeneration in AD. Despite the lack of a cure for AD, researchers have made significant efforts to study the clinical and pathological aspects of the disease, identify biomarkers, and gain insight into its underlying causes. The goal is to achieve early diagnosis and develop treatments that can modify the progression of the disease. The current article discusses the most promising therapy for combating the viral impacts, which provides additional evidence for the frequent reactivations of latent HSV-1 in the AD brain. However, further research is still required to establish the molecular and cellular mechanisms underlying the development of AD through the reactivation of HSV-1. This could potentially lead to new insights in drug development aimed at preventing HSV-1 reactivation and the subsequent development and progression of AD.
Collapse
Affiliation(s)
- Payal Chauhan
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, 124001, India
| | - M Yasmin Begum
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Bayapa Reddy Narapureddy
- Department of Public Health, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Saurabh Gupta
- Department of Biotechnology, GLA University, Mathura, Uttar Pradesh, India
| | - Karan Wadhwa
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, 124001, India
| | - Govind Singh
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, 124001, India.
| | - Rohit Kumawat
- Department of Neurology, National Institute of Medical Sciences, NIMS University Rajsthan, Jaipur, India
| | - Naveen Sharma
- Chandigarh Pharmacy College, Chandigarh Group of Colleges Jhanjeri, Mohali, 140307, Punjab, India
| | - Suhas Ballal
- Departmant of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Saurabh Kumar Jha
- Department of Zoology, Kalindi College, University of Delhi, Delhi, 110008, India
| | - Mosleh Mohammad Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, 61922, Bisha, Saudi Arabia
| | - Dheepak B
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 15551, Al Ain, United Arab Emirates
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Biosciences & Technology, Galgotias University, Greater Noida, India.
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India.
- School of Bioengineering & Biosciences, Lovely Professional University, Phagwara, 144411, India.
| |
Collapse
|
2
|
Linard M, Garrigue I, Vellas B, Coley N, Zetterberg H, Blennow K, Ashton NJ, Payoux P, Salabert AS, Dartigues JF, Mazere J, Andrieu S, Helmer C. Association between herpes simplex virus infection and Alzheimer's disease biomarkers: analysis within the MAPT trial. Sci Rep 2025; 15:2362. [PMID: 39825066 PMCID: PMC11748617 DOI: 10.1038/s41598-024-84583-x] [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: 04/19/2024] [Accepted: 12/24/2024] [Indexed: 01/20/2025] Open
Abstract
In vitro and animal studies have suggested that inoculation with herpes simplex virus 1 (HSV-1) can lead to amyloid deposits, hyperphosphorylation of tau, and/or neuronal loss. Here, we studied the association between HSV-1 and Alzheimer's disease biomarkers in humans. Our sample included 182 participants at risk of cognitive decline from the Multidomain Alzheimer Preventive Trial who had HSV-1 plasma serology and an amyloid PET scan. Plasma Aβ42/40 ratio, neurofilament light chain and p-tau181 were also available for a sub-sample of participants. Multivariate linear regressions were performed and stratified by APOE4 genotype. The median age was 74.0 years, 85.2% were infected with HSV-1. Infected participants tended to have a lower cortical amyloid load than uninfected participants (β = -0.08, p = 0.06), especially those suspected of reactivating HSV-1 most frequently (i.e. with a high anti-HSV-1 IgG level; n = 58, β = -0.09 p = 0.04). After stratification, the association was only significant in APOE4 carriers (n = 43, β = -0.21 p = 0.01). No association was found with the plasma biomarkers. The trend toward lower cortical amyloid load in HSV-1-infected participants was unexpected given the pre-existing literature and may be explained either by a modified immune response in HSV-1 infected subjects which could favour the clearance of amyloid deposits or by a selection bias.
Collapse
Affiliation(s)
- Morgane Linard
- INSERM U1219 Bordeaux Population Health Research Center, University of Bordeaux, 146, rue Léo Saignat, 33076, Bordeaux Cedex, France.
| | - Isabelle Garrigue
- CNRS, MFP, UMR 5234, University of Bordeaux, Bordeaux, France
- Virology Department, University Hospital of Bordeaux, Bordeaux, France
| | - Bruno Vellas
- Gérontopôle de Toulouse, Institut du Vieillissement, University Hospital of Toulouse, Toulouse, France
- INSERM, CERPOP, U1295, University of Toulouse, Toulouse, France
| | - Nicola Coley
- INSERM, CERPOP, U1295, University of Toulouse, Toulouse, France
- Department of Clinical Epidemiology and Public Health, University Hospital of Toulouse, Toulouse, France
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Paris Brain Institute, ICM, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
- Division of Life Sciences and Medicine, and Department of Neurology, Institute on Aging and Brain Disorders, Neurodegenerative Disorder Research Center, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, People's Republic of China
| | - Nicholas James Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, Maurice Wohl Institute Clinical Neuroscience Institute, London, UK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
| | - Pierre Payoux
- Nuclear Medicine Department, University Hospital of Toulouse, Toulouse, France
- INSERM ToNIC, U1214, University of Toulouse, Toulouse, France
| | - Anne-Sophie Salabert
- INSERM ToNIC, U1214, University of Toulouse, Toulouse, France
- Radiopharmacy Department, University Hospital of Toulouse, Toulouse, France
| | - Jean-François Dartigues
- INSERM U1219 Bordeaux Population Health Research Center, University of Bordeaux, 146, rue Léo Saignat, 33076, Bordeaux Cedex, France
- Memory Consultation, CMRR, University Hospital of Bordeaux, Bordeaux, France
| | - Joachim Mazere
- Nuclear Medicine Department, University Hospital of Bordeaux, Bordeaux, France
- CNRS, INCIA, UMR 5287, University of Bordeaux, Bordeaux, France
| | - Sandrine Andrieu
- INSERM, CERPOP, U1295, University of Toulouse, Toulouse, France
- Department of Clinical Epidemiology and Public Health, University Hospital of Toulouse, Toulouse, France
| | - Catherine Helmer
- INSERM, Bergonié Institute, BPH, U1219, CIC-P 1401, University of Bordeaux, Bordeaux, France
| |
Collapse
|
3
|
Tacke C, Landgraf P, Dieterich DC, Kröger A. The fate of neuronal synapse homeostasis in aging, infection, and inflammation. Am J Physiol Cell Physiol 2024; 327:C1546-C1563. [PMID: 39495249 DOI: 10.1152/ajpcell.00466.2024] [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: 07/12/2024] [Revised: 10/18/2024] [Accepted: 10/20/2024] [Indexed: 11/05/2024]
Abstract
Neuroplasticity is the brain's ability to reorganize and modify its neuronal connections in response to environmental stimuli, experiences, learning, and disease processes. This encompasses a variety of mechanisms, including changes in synaptic strength and connectivity, the formation of new synapses, alterations in neuronal structure and function, and the generation of new neurons. Proper functioning of synapses, which facilitate neuron-to-neuron communication, is crucial for brain activity. Neuronal synapse homeostasis, which involves regulating and maintaining synaptic strength and function in the central nervous system (CNS), is vital for this process. Disruptions in synaptic balance, due to factors like inflammation, aging, or infection, can lead to impaired brain function. This review highlights the main aspects and mechanisms underlying synaptic homeostasis, particularly in the context of aging, infection, and inflammation.
Collapse
Affiliation(s)
- Charlotte Tacke
- Institute of Medical Microbiology and Hospital Hygiene, Molecular Microbiology Group, Otto-von-Guericke University, Magdeburg, Germany
| | - Peter Landgraf
- Institute of Pharmacology and Toxicology, Otto-von-Guericke University, Magdeburg, Germany
| | - Daniela C Dieterich
- Institute of Pharmacology and Toxicology, Otto-von-Guericke University, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| | - Andrea Kröger
- Institute of Medical Microbiology and Hospital Hygiene, Molecular Microbiology Group, Otto-von-Guericke University, Magdeburg, Germany
- Helmholtz Center for Infection Research, Innate Immunity and Infection Group, Braunschweig, Germany
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
4
|
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
|
5
|
Kim HS, Jung H, Park YH, Heo SH, Kim S, Moon M. Skin-brain axis in Alzheimer's disease - Pathologic, diagnostic, and therapeutic implications: A Hypothetical Review. Aging Dis 2024; 16:901-916. [PMID: 38739932 DOI: 10.14336/ad.2024.0406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 04/06/2024] [Indexed: 05/16/2024] Open
Abstract
The dynamic interaction between the brain and the skin is termed the 'skin-brain axis.' Changes in the skin not only reflect conditions in the brain but also exert direct and indirect effects on the brain. Interestingly, the connection between the skin and brain is crucial for understanding aging and neurodegenerative diseases. Several studies have shown an association between Alzheimer's disease (AD) and various skin disorders, such as psoriasis, bullous pemphigoid, and skin cancer. Previous studies have shown a significantly increased risk of new-onset AD in patients with psoriasis. In contrast, skin cancer may reduce the risk of developing AD. Accumulating evidence suggests an interaction between skin disease and AD; however, AD-associated pathological changes mediated by the skin-brain axis are not yet clearly defined. While some studies have reported on the diagnostic implications of the skin-brain axis in AD, few have discussed its potential therapeutic applications. In this review, we address the pathological changes mediated by the skin-brain axis in AD. Furthermore, we summarize (1) the diagnostic implications elucidated through the role of the skin-brain axis in AD and (2) the therapeutic implications for AD based on the skin-brain axis. Our review suggests that a potential therapeutic approach targeting the skin-brain axis will enable significant advances in the treatment of AD.
Collapse
Affiliation(s)
- Hyeon Soo Kim
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea
| | - Haram Jung
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea
| | - Yong Ho Park
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea
| | - Su-Hak Heo
- Department of Medicinal Bioscience, Konkuk University (Glocal Campus), Chungcheongbuk-do 27478, Korea
| | - Sujin Kim
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea
- Research Institute for Dementia Science, Konyang University, Daejeon 35365, Korea
| | - Minho Moon
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea
- Research Institute for Dementia Science, Konyang University, Daejeon 35365, Korea
| |
Collapse
|
6
|
Cantero JL, Atienza M, Sastre I, Bullido MJ. Human in vivo evidence of associations between herpes simplex virus and cerebral amyloid-beta load in normal aging. Alzheimers Res Ther 2024; 16:68. [PMID: 38570885 PMCID: PMC10988886 DOI: 10.1186/s13195-024-01437-4] [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: 02/17/2024] [Accepted: 03/25/2024] [Indexed: 04/05/2024]
Abstract
BACKGROUND Mounting data suggests that herpes simplex virus type 1 (HSV-1) is involved in the pathogenesis of AD, possibly instigating amyloid-beta (Aβ) accumulation decades before the onset of clinical symptoms. However, human in vivo evidence linking HSV-1 infection to AD pathology is lacking in normal aging, which may contribute to the elucidation of the role of HSV-1 infection as a potential AD risk factor. METHODS To shed light into this question, serum anti-HSV IgG levels were correlated with 18F-Florbetaben-PET binding to Aβ deposits and blood markers of neurodegeneration (pTau181 and neurofilament light chain) in cognitively normal older adults. Additionally, we investigated whether associations between anti-HSV IgG and AD markers were more evident in APOE4 carriers. RESULTS We showed that increased anti-HSV IgG levels are associated with higher Aβ load in fronto-temporal regions of cognitively normal older adults. Remarkably, these cortical regions exhibited abnormal patterns of resting state-functional connectivity (rs-FC) only in those individuals showing the highest levels of anti-HSV IgG. We further found that positive relationships between anti-HSV IgG levels and Aβ load, particularly in the anterior cingulate cortex, are moderated by the APOE4 genotype, the strongest genetic risk factor for AD. Importantly, anti-HSV IgG levels were unrelated to either subclinical cognitive deficits or to blood markers of neurodegeneration. CONCLUSIONS All together, these results suggest that HSV infection is selectively related to cortical Aβ deposition in normal aging, supporting the inclusion of cognitively normal older adults in prospective trials of antimicrobial therapy aimed at decreasing the AD risk in the aging population.
Collapse
Affiliation(s)
- Jose L Cantero
- Laboratory of Functional Neuroscience, Pablo de Olavide University, Ctra. de Utrera Km 1, Seville, 41013, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.
| | - Mercedes Atienza
- Laboratory of Functional Neuroscience, Pablo de Olavide University, Ctra. de Utrera Km 1, Seville, 41013, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Isabel Sastre
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Universitario La Paz, IdiPAZ (Hospital Universitario La Paz - Universidad Autónoma de Madrid), Madrid, Spain
- Centro de Biología Molecular "Severo Ochoa" (C.S.I.C.-U.A.M.), Universidad Autónoma de Madrid, Madrid, Spain
| | - María Jesús Bullido
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Universitario La Paz, IdiPAZ (Hospital Universitario La Paz - Universidad Autónoma de Madrid), Madrid, Spain
- Centro de Biología Molecular "Severo Ochoa" (C.S.I.C.-U.A.M.), Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
7
|
Chatanaka MK, Sohaei D, Diamandis EP, Prassas I. Beyond the amyloid hypothesis: how current research implicates autoimmunity in Alzheimer's disease pathogenesis. Crit Rev Clin Lab Sci 2023; 60:398-426. [PMID: 36941789 DOI: 10.1080/10408363.2023.2187342] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 03/01/2023] [Indexed: 03/23/2023]
Abstract
The amyloid hypothesis has so far been at the forefront of explaining the pathogenesis of Alzheimer's Disease (AD), a progressive neurodegenerative disorder that leads to cognitive decline and eventual death. Recent evidence, however, points to additional factors that contribute to the pathogenesis of this disease. These include the neurovascular hypothesis, the mitochondrial cascade hypothesis, the inflammatory hypothesis, the prion hypothesis, the mutational accumulation hypothesis, and the autoimmunity hypothesis. The purpose of this review was to briefly discuss the factors that are associated with autoimmunity in humans, including sex, the gut and lung microbiomes, age, genetics, and environmental factors. Subsequently, it was to examine the rise of autoimmune phenomena in AD, which can be instigated by a blood-brain barrier breakdown, pathogen infections, and dysfunction of the glymphatic system. Lastly, it was to discuss the various ways by which immune system dysregulation leads to AD, immunomodulating therapies, and future directions in the field of autoimmunity and neurodegeneration. A comprehensive account of the recent research done in the field was extracted from PubMed on 31 January 2022, with the keywords "Alzheimer's disease" and "autoantibodies" for the first search input, and "Alzheimer's disease" with "IgG" for the second. From the first search, 19 papers were selected, because they contained recent research on the autoantibodies found in the biofluids of patients with AD. From the second search, four papers were selected. The analysis of the literature has led to support the autoimmune hypothesis in AD. Autoantibodies were found in biofluids (serum/plasma, cerebrospinal fluid) of patients with AD with multiple methods, including ELISA, Mass Spectrometry, and microarray analysis. Through continuous research, the understanding of the synergistic effects of the various components that lead to AD will pave the way for better therapeutic methods and a deeper understanding of the disease.
Collapse
Affiliation(s)
- Miyo K Chatanaka
- Department of Laboratory and Medicine Pathobiology, University of Toronto, Toronto, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada
| | - Dorsa Sohaei
- Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
| | - Eleftherios P Diamandis
- Department of Laboratory and Medicine Pathobiology, University of Toronto, Toronto, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada
- Department of Clinical Biochemistry, University Health Network, Toronto, Canada
| | - Ioannis Prassas
- Laboratory Medicine Program, University Health Network, Toronto, Canada
| |
Collapse
|
8
|
Mody PH, Marvin KN, Hynds DL, Hanson LK. Cytomegalovirus infection induces Alzheimer's disease-associated alterations in tau. J Neurovirol 2023; 29:400-415. [PMID: 37436577 DOI: 10.1007/s13365-022-01109-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 11/21/2022] [Accepted: 12/01/2022] [Indexed: 07/13/2023]
Abstract
Alzheimer's disease (AD) manifests with loss of neurons correlated with intercellular deposition of amyloid (amyloid plaques) and intracellular neurofibrillary tangles of hyperphosphorylated tau. However, targeting AD hallmarks has not as yet led to development of an effective treatment despite numerous clinical trials. A better understanding of the early stages of neurodegeneration may lead to development of more effective treatments. One underexplored area is the clinical correlation between infection with herpesviruses and increased risk of AD. We hypothesized that similar to work performed with herpes simplex virus 1 (HSV1), infection with the cytomegalovirus (CMV) herpesvirus increases levels and phosphorylation of tau, similar to AD tauopathy. We used murine CMV (MCMV) to infect mouse fibroblasts and rat neuronal cells to test our hypothesis. MCMV infection increased steady-state levels of primarily high molecular weight forms of tau and altered the patterns of tau phosphorylation. Both changes required viral late gene products. Glycogen synthase kinase 3 beta (GSK3β) was upregulated in the HSVI model, but inhibition with lithium chloride suggested that this enzyme is unlikely to be involved in MCMV infection mediated tau phosphorylation. Thus, we confirm that MCMV, a beta herpes virus, like alpha herpes viruses (e.g., HSV1), can promote tau pathology. This suggests that CMV infection can be useful as another model system to study mechanisms leading to neurodegeneration. Since MCMV infects both mice and rats as permissive hosts, our findings from tissue culture can likely be applied to a variety of AD models to study development of abnormal tau pathology.
Collapse
Affiliation(s)
- Prapti H Mody
- Division of Biology, Texas Woman's University, 304 Administration Drive, Denton, TX, 76204, USA
- Current affiliation: University of Texas Southwestern Medical Center, Dallas, USA
| | - Kelsey N Marvin
- Division of Biology, Texas Woman's University, 304 Administration Drive, Denton, TX, 76204, USA
| | - DiAnna L Hynds
- Division of Biology, Texas Woman's University, 304 Administration Drive, Denton, TX, 76204, USA
| | - Laura K Hanson
- Division of Biology, Texas Woman's University, 304 Administration Drive, Denton, TX, 76204, USA.
| |
Collapse
|
9
|
HSV-1 cellular model reveals links between aggresome formation and early step of Alzheimer's disease. Transl Psychiatry 2023; 13:86. [PMID: 36898995 PMCID: PMC10006237 DOI: 10.1038/s41398-023-02376-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/09/2023] [Accepted: 02/20/2023] [Indexed: 03/12/2023] Open
Abstract
Many studies highlight the potential link between the chronic degenerative Alzheimer's disease and the infection by the herpes simplex virus type-1 (HSV-1). However, the molecular mechanisms making possible this HSV-1-dependent process remain to be understood. Using neuronal cells expressing the wild type form of amyloid precursor protein (APP) infected by HSV-1, we characterized a representative cellular model of the early stage of the sporadic form of the disease and unraveled a molecular mechanism sustaining this HSV-1- Alzheimer's disease interplay. Here, we show that HSV-1 induces caspase-dependent production of the 42 amino-acid long amyloid peptide (Aβ42) oligomers followed by their accumulation in neuronal cells. Aβ42 oligomers and activated caspase 3 (casp3A) concentrate into intracytoplasmic structures observed in Alzheimer's disease neuronal cells called aggresomes. This casp3A accumulation in aggresomes during HSV-1 infection limits the execution of apoptosis until its term, similarly to an abortosis-like event occurring in Alzheimer's disease neuronal cells patients. Indeed, this particular HSV-1 driven cellular context, representative of early stages of the disease, sustains a failed apoptosis mechanism that could explain the chronic amplification of Aβ42 production characteristic of Alzheimer's disease patients. Finally, we show that combination of flurbiprofen, a non-steroidal anti-inflammatory drug (NSAID), with caspase inhibitor reduced drastically HSV-1-induced Aβ42 oligomers production. This provided mechanistic insights supporting the conclusion of clinical trials showing that NSAIDs reduced Alzheimer's disease incidence in early stage of the disease. Therefore, from our study we propose that caspase-dependent production of Aβ42 oligomers together with the abortosis-like event represents a vicious circle in early Alzheimer's disease stages leading to a chronic amplification of Aβ42 oligomers that contributes to the establishment of degenerative disorder like Alzheimer's disease in patients infected by HSV-1. Interestingly this process could be targeted by an association of NSAID with caspase inhibitors.
Collapse
|
10
|
Goldhardt O, Freiberger R, Dreyer T, Wilner L, Yakushev I, Ortner M, Förstl H, Diehl‐Schmid J, Milz E, Priller J, Ramirez A, Magdolen V, Thaler M, Grimmer T. Herpes simplex virus alters Alzheimer's disease biomarkers ‐ A hypothesis paper. Alzheimers Dement 2022; 19:2117-2134. [PMID: 36396609 DOI: 10.1002/alz.12834] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 09/19/2022] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Human herpes simplex virus 1 (HSV1) is discussed to induce amyloid-β (Aβ) accumulation and neurofibrillary tangles of hyperphosphorylated tau (pTau) in Alzheimer's disease (AD) in cell culture and animal models. Aβ appears to be virostatic. We investigated the association between intrathecal antibodies against HSV or cytomegalovirus (CMV) and cerebrospinal fluid (CSF) AD biomarkers. METHODS Aβ42 /Aβ40 ratio, pTau, and tTau were measured in CSF of 117 patients with early AD positive for amyloid pathology (A+) and 30 healthy controls (A-). CSF-to-serum anti-HSV1/2-IgG antibody indices (AI-IgGHSV1/2 ) and CMV (AI-IgGCMV ) were determined by enzyme-linked immunosorbent assay (ELISA). RESULTS Exclusively in HSV1-seropositive AD, pTau was positively and significantly predicted by AI-IgGHSV1/2 and negatively by the Aβ42 /Aβ40 ratio in both univariate and multivariate regression analyses. Furthermore, a significant and negative interaction between the AI-IgGHSV1/2 and Aβ42 /Aβ40 ratio on pTau was found. DISCUSSION The results support the hypothesis that HSV infection contributes to AD. HIGHLIGHTS HSV antibody index is positively associated with tau pathology in patients with AD. HSV antibody index is negatively associated with cerebral FDG metabolism. Amyloid modulates the association of HSV antibody index with CSF-pTau. HSV in AD offers a pathophysiological model connecting tau and amyloid.
Collapse
Affiliation(s)
- Oliver Goldhardt
- Department of Psychiatry and Psychotherapy School of Medicine Klinikum rechts der Isar Technical University of Munich Munich Germany
| | - Robert Freiberger
- Department of Psychiatry and Psychotherapy School of Medicine Klinikum rechts der Isar Technical University of Munich Munich Germany
| | - Tobias Dreyer
- Department of Obstetrics and Gynecology School of Medicine, Klinikum rechts der Isar, Technical University of Munich Munich Germany
| | - Luisa Wilner
- Department of Psychiatry and Psychotherapy School of Medicine Klinikum rechts der Isar Technical University of Munich Munich Germany
- Department of Nuclear Medicine, School of Medicine Klinikum rechts der Isar, Technical University of Munich Munich Germany
| | - Igor Yakushev
- Department of Nuclear Medicine, School of Medicine Klinikum rechts der Isar, Technical University of Munich Munich Germany
| | - Marion Ortner
- Department of Psychiatry and Psychotherapy School of Medicine Klinikum rechts der Isar Technical University of Munich Munich Germany
| | - Hans Förstl
- Department of Psychiatry and Psychotherapy School of Medicine Klinikum rechts der Isar Technical University of Munich Munich Germany
| | - Janine Diehl‐Schmid
- Department of Psychiatry and Psychotherapy School of Medicine Klinikum rechts der Isar Technical University of Munich Munich Germany
| | - Esther Milz
- Division of Neurogenetics and Molecular Psychiatry Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne Cologne Germany
| | - Josef Priller
- Department of Psychiatry and Psychotherapy School of Medicine Klinikum rechts der Isar Technical University of Munich Munich Germany
| | - Alfredo Ramirez
- Division of Neurogenetics and Molecular Psychiatry Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne Cologne Germany
- Department of Neurodegenerative Diseases and Geriatric Psychiatry Medical Faculty University Hospital Bonn Bonn Germany
- German Center for Neurodegenerative Diseases (DZNE) Bonn Germany
- Department of Psychiatry and Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases San Antonio Texas USA
- Cluster of Excellence Cellular Stress Responses in Aging‐associated Diseases (CECAD) University of Cologne Cologne Germany
| | - Viktor Magdolen
- Department of Obstetrics and Gynecology School of Medicine, Klinikum rechts der Isar, Technical University of Munich Munich Germany
| | - Markus Thaler
- Institute for Clinical Chemistry and Pathobiochemistry School of Medicine, Klinikum rechts der Isar, Technical University of Munich Munich Germany
| | - Timo Grimmer
- Department of Psychiatry and Psychotherapy School of Medicine Klinikum rechts der Isar Technical University of Munich Munich Germany
| |
Collapse
|
11
|
Bocharova OV, Fisher A, Pandit NP, Molesworth K, Mychko O, Scott AJ, Makarava N, Ritzel R, Baskakov IV. Aβ plaques do not protect against HSV-1 infection in a mouse model of familial Alzheimer's disease, and HSV-1 does not induce Aβ pathology in a model of late onset Alzheimer's disease. Brain Pathol 2022; 33:e13116. [PMID: 36064300 PMCID: PMC9836376 DOI: 10.1111/bpa.13116] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 07/11/2022] [Indexed: 01/21/2023] Open
Abstract
The possibility that the etiology of late onset Alzheimer's disease is linked to viral infections of the CNS has been actively debated in recent years. According to the antiviral protection hypothesis, viral pathogens trigger aggregation of Aβ peptides that are produced as a defense mechanism in response to infection to entrap and neutralize pathogens. To test the causative relationship between viral infection and Aβ aggregation, the current study examined whether Aβ plaques protect the mouse brain against Herpes Simplex Virus 1 (HSV-1) infection introduced via a physiological route and whether HSV-1 infection triggers formation of Aβ plaques in a mouse model of late-onset AD that does not develop Aβ pathology spontaneously. In aged 5XFAD mice infected via eye scarification, high density of Aβ aggregates did not improve survival time or rate when compared with wild type controls. In 5XFADs, viral replication sites were found in brain areas with a high density of extracellular Aβ deposits, however, no association between HSV-1 and Aβ aggregates could be found. To test whether HSV-1 triggers Aβ aggregation in a mouse model that lacks spontaneous Aβ pathology, 13-month-old hAβ/APOE4/Trem2*R47H mice were infected with HSV-1 via eye scarification with the McKrae HSV-1 strain, intracranial inoculation with McKrae, intracranial inoculation after priming with LPS for 6 weeks, or intracranial inoculation with high doses of McKrae or 17syn + strains that represent different degrees of neurovirulence. No signs of Aβ aggregation were found in any of the experimental groups. Instead, extensive infiltration of peripheral leukocytes was observed during the acute stage of HSV-1 infection, and phagocytic activity of myeloid cells was identified as the primary defense mechanism against HSV-1. The current results argue against a direct causative relationship between HSV-1 infection and Aβ pathology.
Collapse
Affiliation(s)
- Olga V. Bocharova
- Center for Biomedical Engineering and TechnologyUniversity of Maryland School of MedicineBaltimoreMarylandUSA,Department of Anatomy and NeurobiologyUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Aidan Fisher
- Center for Biomedical Engineering and TechnologyUniversity of Maryland School of MedicineBaltimoreMarylandUSA,Department of Anatomy and NeurobiologyUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Narayan P. Pandit
- Center for Biomedical Engineering and TechnologyUniversity of Maryland School of MedicineBaltimoreMarylandUSA,Department of Anatomy and NeurobiologyUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Kara Molesworth
- Center for Biomedical Engineering and TechnologyUniversity of Maryland School of MedicineBaltimoreMarylandUSA,Department of Anatomy and NeurobiologyUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Olga Mychko
- Center for Biomedical Engineering and TechnologyUniversity of Maryland School of MedicineBaltimoreMarylandUSA,Department of Anatomy and NeurobiologyUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Alison J. Scott
- Department of Microbial PathogenesisUniversity of Maryland School of DentistryBaltimoreMarylandUSA
| | - Natallia Makarava
- Center for Biomedical Engineering and TechnologyUniversity of Maryland School of MedicineBaltimoreMarylandUSA,Department of Anatomy and NeurobiologyUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Rodney Ritzel
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR)University of Maryland School of MedicineBaltimoreMarylandUSA
| | - Ilia V. Baskakov
- Center for Biomedical Engineering and TechnologyUniversity of Maryland School of MedicineBaltimoreMarylandUSA,Department of Anatomy and NeurobiologyUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
12
|
Gallego Villarejo L, Bachmann L, Marks D, Brachthäuser M, Geidies A, Müller T. Role of Intracellular Amyloid β as Pathway Modulator, Biomarker, and Therapy Target. Int J Mol Sci 2022; 23:ijms23094656. [PMID: 35563046 PMCID: PMC9103247 DOI: 10.3390/ijms23094656] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/15/2022] [Accepted: 04/19/2022] [Indexed: 02/04/2023] Open
Abstract
The β- and γ-secretase-driven cleavage of the amyloid precursor protein (APP) gives rise to the amyloid β peptide, which is believed to be the main driver of neurodegeneration in Alzheimer’s disease (AD). As it is prominently detectable in extracellular plaques in post-mortem AD brain samples, research in recent decades focused on the pathological role of extracellular amyloid β aggregation, widely neglecting the potential meaning of very early generation of amyloid β inside the cell. In the last few years, the importance of intracellular amyloid β (iAβ) as a strong player in neurodegeneration has been indicated by a rising number of studies. In this review, iAβ is highlighted as a crucial APP cleavage fragment, able to manipulate intracellular pathways and foster neurodegeneration. We demonstrate its relevance as a pathological marker and shed light on initial studies aiming to modulate iAβ through pharmacological treatment, which has been shown to have beneficial effects on cognitive properties in animal models. Finally, we display the relevance of viral infections on iAβ generation and point out future directions urgently needed to manifest the potential relevance of iAβ in Alzheimer’s disease.
Collapse
Affiliation(s)
- Lucia Gallego Villarejo
- Department of Molecular Biochemistry, Cell Signalling, Ruhr University Bochum, 44801 Bochum, Germany; (L.G.V.); (L.B.); (D.M.); (M.B.); (A.G.)
| | - Lisa Bachmann
- Department of Molecular Biochemistry, Cell Signalling, Ruhr University Bochum, 44801 Bochum, Germany; (L.G.V.); (L.B.); (D.M.); (M.B.); (A.G.)
| | - David Marks
- Department of Molecular Biochemistry, Cell Signalling, Ruhr University Bochum, 44801 Bochum, Germany; (L.G.V.); (L.B.); (D.M.); (M.B.); (A.G.)
| | - Maite Brachthäuser
- Department of Molecular Biochemistry, Cell Signalling, Ruhr University Bochum, 44801 Bochum, Germany; (L.G.V.); (L.B.); (D.M.); (M.B.); (A.G.)
| | - Alexander Geidies
- Department of Molecular Biochemistry, Cell Signalling, Ruhr University Bochum, 44801 Bochum, Germany; (L.G.V.); (L.B.); (D.M.); (M.B.); (A.G.)
| | - Thorsten Müller
- Department of Molecular Biochemistry, Cell Signalling, Ruhr University Bochum, 44801 Bochum, Germany; (L.G.V.); (L.B.); (D.M.); (M.B.); (A.G.)
- Institute of Psychiatric Phenomics and Genomics (IPPG), LMU University Hospital, LMU Munich, 80336 Munich, Germany
- Correspondence:
| |
Collapse
|
13
|
Role of HSV-1 in Alzheimer's disease pathogenesis: A challenge for novel preventive/therapeutic strategies. Curr Opin Pharmacol 2022; 63:102200. [PMID: 35276497 DOI: 10.1016/j.coph.2022.102200] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/26/2022] [Accepted: 01/28/2022] [Indexed: 01/03/2023]
Abstract
Herpes simplex virus-1 (HSV-1) is a ubiquitous DNA virus able to establish a life-long latent infection in host sensory ganglia. Following periodic reactivations, the neovirions usually target the site of primary infection causing recurrent diseases in susceptible individuals. However, reactivated HSV-1 may also reach the brain resulting in severe herpetic encephalitis or in asymptomatic infections. These have been reportedly linked to neurodegenerative disorders, such as Alzheimer's disease (AD), suggesting antiviral preventive or/therapeutic treatments as possible strategies to counteract AD onset and progression. Here, we provide an overview of the AD-like mechanisms driven by HSV-1-infection in neurons and discuss the ongoing trials repurposing anti-herpetic drugs to treat AD as well as preventive strategies aimed at blocking virus infection.
Collapse
|
14
|
Xie J, Tian S, Liu J, Cao R, Yue P, Cai X, Shang Q, Yang M, Han L, Zhang DK. Dual role of the nasal microbiota in neurological diseases—An unignorable risk factor or a potential therapy carrier. Pharmacol Res 2022; 179:106189. [DOI: 10.1016/j.phrs.2022.106189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/06/2022] [Accepted: 03/17/2022] [Indexed: 12/11/2022]
|
15
|
Tran DN, Bakx ATCM, van Dis V, Aronica E, Verdijk RM, Ouwendijk WJD. No evidence of aberrant amyloid β and phosphorylated tau expression in herpes simplex virus-infected neurons of the trigeminal ganglia and brain. Brain Pathol 2021; 32:e13044. [PMID: 34913212 PMCID: PMC9245940 DOI: 10.1111/bpa.13044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/26/2021] [Accepted: 12/03/2021] [Indexed: 11/29/2022] Open
Abstract
Increasing evidence supports the role of neurotropic herpes simplex virus 1 (HSV‐1) in the pathogenesis of Alzheimer's disease (AD). However, it is unclear whether previously reported findings in HSV‐1 cell culture and animal models can be translated to humans. Here, we analyzed clinical specimens from latently HSV‐1 infected individuals and individuals with lytic HSV infection of the brain (herpes simplex encephalitis; HSE). Latent HSV‐1 DNA load and latency‐associated transcript (LAT) expression were identical between trigeminal ganglia (TG) of AD patients and controls. Amyloid β (Aβ) and hyperphosphorylated tau (pTau) were not detected in latently HSV‐infected TG neurons. Aging‐related intraneuronal Aβ accumulations, neurofibrillary tangles (NFT), and/or extracellular Aβ plaques were observed in the brain of some HSE patients, but these were neither restricted to HSV‐infected neurons nor brain regions containing virus‐infected cells. Analysis of unique brain material from an AD patient with concurrent HSE showed that HSV‐infected cells frequently localized close to Aβ plaques and NFT, but were not associated with exacerbated AD‐related pathology. HSE‐associated neuroinflammation was not associated with specific Aβ or pTau phenotypes. Collectively, we observed that neither latent nor lytic HSV infection of human neurons is directly associated with aberrant Aβ or pTau protein expression in ganglia and brain.
Collapse
Affiliation(s)
- Diana N Tran
- Department of Viroscience, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Amy T C M Bakx
- Department of Viroscience, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Vera van Dis
- Department of Pathology, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Eleonora Aronica
- Department of (Neuro)Pathology, Amsterdam Neuroscience, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Robert M Verdijk
- Department of Pathology, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Werner J D Ouwendijk
- Department of Viroscience, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| |
Collapse
|
16
|
Bohmwald K, Andrade CA, Gálvez NMS, Mora VP, Muñoz JT, Kalergis AM. The Causes and Long-Term Consequences of Viral Encephalitis. Front Cell Neurosci 2021; 15:755875. [PMID: 34916908 PMCID: PMC8668867 DOI: 10.3389/fncel.2021.755875] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 11/01/2021] [Indexed: 12/15/2022] Open
Abstract
Reports regarding brain inflammation, known as encephalitis, have shown an increasing frequency during the past years. Encephalitis is a relevant concern to public health due to its high morbidity and mortality. Infectious or autoimmune diseases are the most common cause of encephalitis. The clinical symptoms of this pathology can vary depending on the brain zone affected, with mild ones such as fever, headache, confusion, and stiff neck, or severe ones, such as seizures, weakness, hallucinations, and coma, among others. Encephalitis can affect individuals of all ages, but it is frequently observed in pediatric and elderly populations, and the most common causes are viral infections. Several viral agents have been described to induce encephalitis, such as arboviruses, rhabdoviruses, enteroviruses, herpesviruses, retroviruses, orthomyxoviruses, orthopneumovirus, and coronaviruses, among others. Once a neurotropic virus reaches the brain parenchyma, the resident cells such as neurons, astrocytes, and microglia, can be infected, promoting the secretion of pro-inflammatory molecules and the subsequent immune cell infiltration that leads to brain damage. After resolving the viral infection, the local immune response can remain active, contributing to long-term neuropsychiatric disorders, neurocognitive impairment, and degenerative diseases. In this article, we will discuss how viruses can reach the brain, the impact of viral encephalitis on brain function, and we will focus especially on the neurocognitive sequelae reported even after viral clearance.
Collapse
Affiliation(s)
- Karen Bohmwald
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Catalina A Andrade
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nicolás M S Gálvez
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Valentina P Mora
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - José T Muñoz
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
17
|
Vasefi M, Ghaboolian-Zare E, Abedelwahab H, Osu A. Environmental toxins and Alzheimer's disease progression. Neurochem Int 2020; 141:104852. [PMID: 33010393 DOI: 10.1016/j.neuint.2020.104852] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/05/2020] [Accepted: 09/18/2020] [Indexed: 12/29/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia, which causes progressive memory loss and cognitive decline. Effective strategies to treat or prevent remains one of the most challenging undertakings in the medical field. AD is a complex and multifactorial disease that involves several risk factors. Aging and genetic factors both play important roles in the onset of the AD, however; certain environmental factors have been reported to increase the risk of AD. Chronic exposure to toxins has been seen as an environmental factor that may increase the risk of developing a neurodegenerative disease such as AD. Exposure to metals and biotoxins produced by bacteria, molds, and viruses may contribute to the cognitive decline and pathophysiology associated with AD. Toxins may contribute to the pathology of the disease through various mechanisms such as deposition of amyloid-beta (Aβ) plaques and tangles in the brain, induction of apoptosis, inflammation, or oxidative damage. Here, we will review how toxins affect brain physiology with a focus on mechanisms by which toxins may contribute to the development and progression of AD. A better understanding of these mechanisms may help contribute towards the development of an effective strategy to slow the progression of AD.
Collapse
Affiliation(s)
- Maryam Vasefi
- Department Biology, Lamar University, Beaumont, TX, United States.
| | | | | | - Anthony Osu
- Department Biology, Lamar University, Beaumont, TX, United States
| |
Collapse
|
18
|
Zhang LN, Li MJ, Shang YH, Zhao FF, Huang HC, Lao FX. Independent and Correlated Role of Apolipoprotein E ɛ4 Genotype and Herpes Simplex Virus Type 1 in Alzheimer's Disease. J Alzheimers Dis 2020; 77:15-31. [PMID: 32804091 DOI: 10.3233/jad-200607] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The ɛ4 allele of the Apolipoprotein E (APOE) gene in individuals infected by Herpes simplex virus type 1 (HSV-1) has been demonstrated to be a risk factor in Alzheimer's disease (AD). APOE-ɛ4 reduces the levels of neuronal cholesterol, interferes with the transportation of cholesterol, impairs repair of synapses, decreases the clearance of neurotoxic peptide amyloid-β (Aβ), and promotes the deposition of amyloid plaque, and eventually may cause development of AD. HSV-1 enters host cells and can infect the olfactory system, trigeminal ganglia, entorhinal cortex, and hippocampus, and may cause AD-like pathological changes. The lifecycle of HSV-1 goes through a long latent phase. HSV-1 induces neurotropic cytokine expression with pro-inflammatory action and inhibits antiviral cytokine production in AD. It should be noted that interferons display antiviral activity in HSV-1-infected AD patients. Reactivated HSV-1 is associated with infectious burden in cognitive decline and AD. Finally, HSV-1 DNA has been confirmed as present in human brains and is associated with APOEɛ4 in AD. HSV-1 and APOEɛ4 increase the risk of AD and relate to abnormal autophagy, higher concentrations of HSV-1 DNA in AD, and formation of Aβ plaques and neurofibrillary tangles.
Collapse
Affiliation(s)
- Li-Na Zhang
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, P.R. China.,Institute of Functional Factors and Brain Science, Beijing Union University, Beijing, P.R. China.,College of Biochemical Engineering, Beijing Union University, Beijing, P.R. China
| | - Meng-Jie Li
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, P.R. China.,Institute of Functional Factors and Brain Science, Beijing Union University, Beijing, P.R. China.,College of Biochemical Engineering, Beijing Union University, Beijing, P.R. China
| | - Ying-Hui Shang
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, P.R. China.,Institute of Functional Factors and Brain Science, Beijing Union University, Beijing, P.R. China.,College of Biochemical Engineering, Beijing Union University, Beijing, P.R. China
| | - Fan-Fan Zhao
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, P.R. China.,Institute of Functional Factors and Brain Science, Beijing Union University, Beijing, P.R. China.,College of Biochemical Engineering, Beijing Union University, Beijing, P.R. China
| | - Han-Chang Huang
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, P.R. China.,Institute of Functional Factors and Brain Science, Beijing Union University, Beijing, P.R. China.,College of Biochemical Engineering, Beijing Union University, Beijing, P.R. China
| | - Feng-Xue Lao
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, P.R. China.,Institute of Functional Factors and Brain Science, Beijing Union University, Beijing, P.R. China.,College of Biochemical Engineering, Beijing Union University, Beijing, P.R. China
| |
Collapse
|
19
|
Multiple Herpes Simplex Virus-1 (HSV-1) Reactivations Induce Protein Oxidative Damage in Mouse Brain: Novel Mechanisms for Alzheimer's Disease Progression. Microorganisms 2020; 8:microorganisms8070972. [PMID: 32610629 PMCID: PMC7409037 DOI: 10.3390/microorganisms8070972] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/26/2020] [Accepted: 06/27/2020] [Indexed: 12/24/2022] Open
Abstract
Compelling evidence supports the role of oxidative stress in Alzheimer's disease (AD) pathophysiology. Interestingly, Herpes simplex virus-1 (HSV-1), a neurotropic virus that establishes a lifelong latent infection in the trigeminal ganglion followed by periodic reactivations, has been reportedly linked both to AD and to oxidative stress conditions. Herein, we analyzed, through biochemical and redox proteomic approaches, the mouse model of recurrent HSV-1 infection we previously set up, to investigate whether multiple virus reactivations induced oxidative stress in the mouse brain and affected protein function and related intracellular pathways. Following multiple HSV-1 reactivations, we found in mouse brains increased levels of oxidative stress hallmarks, including 4-hydroxynonenal (HNE), and 13 HNE-modified proteins whose levels were found significantly altered in the cortex of HSV-1-infected mice compared to controls. We focused on two proteins previously linked to AD pathogenesis, i.e., glucose-regulated protein 78 (GRP78) and collapsin response-mediated protein 2 (CRMP2), which are involved in the unfolded protein response (UPR) and in microtubule stabilization, respectively. We found that recurrent HSV-1 infection disables GRP78 function and activates the UPR, whereas it prevents CRMP2 function in mouse brains. Overall, these data suggest that repeated HSV-1 reactivation into the brain may contribute to neurodegeneration also through oxidative damage.
Collapse
|
20
|
Marcocci ME, Napoletani G, Protto V, Kolesova O, Piacentini R, Li Puma DD, Lomonte P, Grassi C, Palamara AT, De Chiara G. Herpes Simplex Virus-1 in the Brain: The Dark Side of a Sneaky Infection. Trends Microbiol 2020; 28:808-820. [PMID: 32386801 DOI: 10.1016/j.tim.2020.03.003] [Citation(s) in RCA: 167] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 02/27/2020] [Accepted: 03/25/2020] [Indexed: 12/22/2022]
Abstract
Herpes simplex virus-1 (HSV-1) establishes latency preferentially in sensory neurons of peripheral ganglia. A variety of stresses can induce recurrent reactivations of the virus, which spreads and then actively replicates to the site of primary infection (usually the lips or eyes). Viral particles produced following reactivation can also reach the brain, causing a rare but severe form of diffuse acute infection, namely herpes simplex encephalitis. Most of the time, this infection is clinically asymptomatic. However, it was recently correlated with the production and accumulation of neuropathological biomarkers of Alzheimer's disease. In this review we discuss the different cellular and molecular mechanisms underlying the acute and long-term damage caused by HSV-1 infection in the brain.
Collapse
Affiliation(s)
- Maria Elena Marcocci
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy
| | - Giorgia Napoletani
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy
| | - Virginia Protto
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy
| | - Olga Kolesova
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy
| | - Roberto Piacentini
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Domenica Donatella Li Puma
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Patrick Lomonte
- Université de Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U 1217, Institut NeuroMyoGène (INMG), Lyon, France
| | - Claudio Grassi
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Anna Teresa Palamara
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy; San Raffaele Pisana, IRCCS, Telematic University, Rome, Italy.
| | - Giovanna De Chiara
- Institute of Translational Pharmacology, National Research Council, Rome, Italy
| |
Collapse
|
21
|
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: 50] [Impact Index Per Article: 8.3] [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
|
22
|
Agostini S, Mancuso R, Hernis A, Costa AS, Nemni R, Clerici M. HSV-1-Specific IgG Subclasses Distribution and Serum Neutralizing Activity in Alzheimer's Disease and in Mild Cognitive Impairment. J Alzheimers Dis 2019; 63:131-138. [PMID: 29578484 DOI: 10.3233/jad-170966] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Human Herpes Simplex Virus type 1 (HSV-1) infection is suggested to play a role in the development of Alzheimer's disease (AD). Immunoglobulin G (IgG) neutralize HSV-1 activity, but the virus can evade IgG-mediated immune responses by expressing receptor that efficiently binds the Fc portion of all IgG subclasses with the exception of IgG3. We analyzed HSV-1-specific IgG subclasses and IgG-mediated serum neutralization activity against HSV-1 in individuals with a diagnosis of either AD or mild cognitive impairment (MCI), comparing the results with those obtained in age-matched healthy controls (HC). 186 individuals were enrolled in the study: 67 AD, 58 MCI, and 61 HC. HSV-1 IgG titers and subclasses, neutralizing antibody (NAb) titers, and complement C3 concentration-critical component of antibody-mediated effector activity-were measured in sera by ELISA; IgG neutralizing activity was performed on HSV-1 infected Vero cells. Results showed that, whereas HSV-1-specific IgG1, IgG2, and IgG4 titers as well as complement C3 serum concentration were comparable in all groups of individuals, IgG3 were more frequently detected in MCI (89%) compared to AD (75%; p < 0.05) and HC (68%; p = 0.003), whereas the titer is similar among the three groups (AD: 0.66±0.21 OD; MCI: 0.68±0.24 OD; HC: 0.72±0.28 OD). Notably, HSV-1 specific neutralizing ability of AD sera was reduced even in the presence of high quantity of IgG3. As IgG3 plays a key role in counteracting the ability of HSV-1 to evade immune responses, these data reinforce the hypothesis of a pathogenetic role of HSV-1 in AD.
Collapse
Affiliation(s)
| | | | - Ambra Hernis
- Don Carlo Gnocchi Foundation IRCCS - ONLUS, Milan, Italy
| | | | - Raffaello Nemni
- Don Carlo Gnocchi Foundation IRCCS - ONLUS, Milan, Italy.,Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Mario Clerici
- Don Carlo Gnocchi Foundation IRCCS - ONLUS, Milan, Italy.,Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| |
Collapse
|
23
|
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: 2.5] [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
|
24
|
Gosztyla ML, Brothers HM, Robinson SR. Alzheimer's Amyloid-β is an Antimicrobial Peptide: A Review of the Evidence. J Alzheimers Dis 2019; 62:1495-1506. [PMID: 29504537 DOI: 10.3233/jad-171133] [Citation(s) in RCA: 164] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The amyloid-β (Aβ) peptide has long been considered to be the driving force behind Alzheimer's disease (AD). However, clinical trials that have successfully reduced Aβ burden in the brain have not slowed the cognitive decline, and in some instances, have resulted in adverse outcomes. While these results can be interpreted in different ways, a more nuanced picture of Aβ is emerging that takes into account the facts that the peptide is evolutionarily conserved and is present throughout life in cognitively normal individuals. Recent evidence indicates a role for Aβ as an antimicrobial peptide (AMP), a class of innate immune defense molecule that utilizes fibrillation to protect the host from a wide range of infectious agents. In humans and in animal models, infection of the brain frequently leads to increased amyloidogenic processing of the amyloid-β protein precursor (AβPP) and resultant fibrillary aggregates of Aβ. Evidence from in vitro and in vivo studies demonstrates that Aβ oligomers have potent, broad-spectrum antimicrobial properties by forming fibrils that entrap pathogens and disrupt cell membranes. Importantly, overexpression of Aβ confers increased resistance to infection from both bacteria and viruses. The antimicrobial role of Aβ may explain why increased rates of infection have been observed in some of the AD clinical trials that depleted Aβ. Perhaps progress toward a cure for AD will accelerate once treatment strategies begin to take into account the likely physiological functions of this enigmatic peptide.
Collapse
Affiliation(s)
- Maya L Gosztyla
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| | - Holly M Brothers
- Department of Psychology, The Ohio State University, Columbus, OH, USA
| | - Stephen R Robinson
- Discipline of Psychology, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| |
Collapse
|
25
|
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
|
26
|
Harris SA, Harris EA. Molecular Mechanisms for Herpes Simplex Virus Type 1 Pathogenesis in Alzheimer's Disease. Front Aging Neurosci 2018; 10:48. [PMID: 29559905 PMCID: PMC5845560 DOI: 10.3389/fnagi.2018.00048] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Accepted: 02/12/2018] [Indexed: 12/12/2022] Open
Abstract
This review focuses on research in the areas of epidemiology, neuropathology, molecular biology and genetics that implicates herpes simplex virus type 1 (HSV-1) as a causative agent in the pathogenesis of sporadic Alzheimer’s disease (AD). Molecular mechanisms whereby HSV-1 induces AD-related pathophysiology and pathology, including neuronal production and accumulation of amyloid beta (Aβ), hyperphosphorylation of tau proteins, dysregulation of calcium homeostasis, and impaired autophagy, are discussed. HSV-1 causes additional AD pathologies through mechanisms that promote neuroinflammation, oxidative stress, mitochondrial damage, synaptic dysfunction, and neuronal apoptosis. The AD susceptibility genes apolipoprotein E (APOE), phosphatidylinositol binding clathrin assembly protein (PICALM), complement receptor 1 (CR1) and clusterin (CLU) are involved in the HSV lifecycle. Polymorphisms in these genes may affect brain susceptibility to HSV-1 infection. APOE, for example, influences susceptibility to certain viral infections, HSV-1 viral load in the brain, and the innate immune response. The AD susceptibility gene cholesterol 25-hydroxylase (CH25H) is upregulated in the AD brain and is involved in the antiviral immune response. HSV-1 interacts with additional genes to affect cognition-related pathways and key enzymes involved in Aβ production, Aβ clearance, and hyperphosphorylation of tau proteins. Aβ itself functions as an antimicrobial peptide (AMP) against various pathogens including HSV-1. Evidence is presented supporting the hypothesis that Aβ is produced as an AMP in response to HSV-1 and other brain infections, leading to Aβ deposition and plaque formation in AD. Epidemiologic studies associating HSV-1 infection with AD and cognitive impairment are discussed. Studies are reviewed supporting subclinical chronic reactivation of latent HSV-1 in the brain as significant in the pathogenesis of AD. Finally, the rationale for and importance of clinical trials treating HSV-1-infected MCI and AD patients with antiviral medication is discussed.
Collapse
Affiliation(s)
- Steven A Harris
- St. Vincent Medical Group, Northside Internal Medicine, Indianapolis, IN, United States
| | - Elizabeth A Harris
- Department of Neurology, University of Chicago Medical Center, Chicago, IL, United States
| |
Collapse
|
27
|
Wong SW, Sil P, Martinez J. Rubicon: LC3-associated phagocytosis and beyond. FEBS J 2017; 285:1379-1388. [PMID: 29215797 DOI: 10.1111/febs.14354] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 11/06/2017] [Accepted: 12/04/2017] [Indexed: 02/07/2023]
Abstract
Rubicon (Rubcn) was initially identified as a component of the Class III PI3K complex and a negative regulator of canonical autophagy and endosomal trafficking. However, Rubicon has attracted the most notoriety because of its critical role in LC3-associated phagocytosis (LAP), a form of noncanonical autophagy that utilizes some components of the autophagy machinery to process extracellular cargo. Additionally, Rubicon has been identified as a key modulator of the inflammatory response and viral replication. In this review, we discuss the known functions of Rubicon in LAP and other signaling pathways and examine the disease pathologies associated with Rubicon dysfunction in animal models and humans.
Collapse
Affiliation(s)
- Sing-Wai Wong
- Immunity, Inflammation, and Disease Laboratory, NIEHS, National Institutes of Health, Research Triangle Park, NC, USA.,Oral and Craniofacial Biomedicine Curriculum, School of Dentistry, University of North Carolina at Chapel Hill, NC, USA
| | - Payel Sil
- Immunity, Inflammation, and Disease Laboratory, NIEHS, National Institutes of Health, Research Triangle Park, NC, USA
| | - Jennifer Martinez
- Immunity, Inflammation, and Disease Laboratory, NIEHS, National Institutes of Health, Research Triangle Park, NC, USA
| |
Collapse
|
28
|
Olsson J, Lövheim H, Honkala E, Karhunen PJ, Elgh F, Kok EH. HSV presence in brains of individuals without dementia: the TASTY brain series. Dis Model Mech 2016; 9:1349-1355. [PMID: 27664135 PMCID: PMC5117234 DOI: 10.1242/dmm.026674] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 09/20/2016] [Indexed: 12/28/2022] Open
Abstract
Herpes simplex virus (HSV) type 1 affects a majority of the population and recent evidence suggests involvement in Alzheimer's disease aetiology. We investigated the prevalence of HSV type 1 and 2 in the Tampere Autopsy Study (TASTY) brain samples using PCR and sero-positivity in plasma, and associations with Alzheimer's disease neuropathology. HSV was shown to be present in human brain tissue in 11/584 (1.9%) of samples in the TASTY cohort, of which six had Alzheimer's disease neuropathological amyloid beta (Aβ) aggregations. Additionally, serological data revealed 86% of serum samples tested were IgG-positive for HSV. In conclusion, we report epidemiological evidence of the presence of HSV in brain tissue free from encephalitis symptoms in a cohort most closely representing the general population (a minimum prevalence of 1.9%). Whereas 6/11 samples with HSV DNA in the brain tissue had Aβ aggregations, most of those with Aβ aggregations did not have HSV present in the brain tissue. Summary: We assessed the presence of HSV types 1 and 2 in the brain tissue of a large non-institutionalised autopsy cohort, providing evidence of asymptomatic access of HSV to the brain.
Collapse
Affiliation(s)
- Jan Olsson
- Department of Clinical Microbiology, Virology, Umeå University, Umeå 90185, Sweden
| | - Hugo Lövheim
- Department of Community Medicine and Rehabilitation, Geriatric Medicine, Umeå University, Umeå 90185, Sweden
| | - Emma Honkala
- Department of Clinical Microbiology, Virology, Umeå University, Umeå 90185, Sweden
| | - Pekka J Karhunen
- Department of Forensic Medicine, University of Tampere, Tampere 33520, Finland
| | - Fredrik Elgh
- Department of Clinical Microbiology, Virology, Umeå University, Umeå 90185, Sweden
| | - Eloise H Kok
- Department of Forensic Medicine, University of Tampere, Tampere 33520, Finland
| |
Collapse
|
29
|
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: 175] [Impact Index Per Article: 19.4] [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
|
30
|
Redox Imbalance and Viral Infections in Neurodegenerative Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:6547248. [PMID: 27110325 PMCID: PMC4826696 DOI: 10.1155/2016/6547248] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 03/07/2016] [Accepted: 03/10/2016] [Indexed: 12/12/2022]
Abstract
Reactive oxygen species (ROS) are essential molecules for many physiological functions and act as second messengers in a large variety of tissues. An imbalance in the production and elimination of ROS is associated with human diseases including neurodegenerative disorders. In the last years the notion that neurodegenerative diseases are accompanied by chronic viral infections, which may result in an increase of neurodegenerative diseases progression, emerged. It is known in literature that enhanced viral infection risk, observed during neurodegeneration, is partly due to the increase of ROS accumulation in brain cells. However, the molecular mechanisms of viral infection, occurring during the progression of neurodegeneration, remain unclear. In this review, we discuss the recent knowledge regarding the role of influenza, herpes simplex virus type-1, and retroviruses infection in ROS/RNS-mediated Parkinson's disease (PD), Alzheimer's disease (AD), and amyotrophic lateral sclerosis (ALS).
Collapse
|
31
|
Gurav AN. Alzheimer's disease and periodontitis--an elusive link. Rev Assoc Med Bras (1992) 2015; 60:173-80. [PMID: 24919005 DOI: 10.1590/1806-9282.60.02.015] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Accepted: 03/27/2013] [Indexed: 12/21/2022] Open
Abstract
Alzheimer's disease is the preeminent cause and commonest form of dementia. It is clinically characterized by a progressive descent in the cognitive function, which commences with deterioration in memory. The exact etiology and pathophysiologic mechanism of Alzheimer's disease is still not fully understood. However it is hypothesized that, neuroinflammation plays a critical role in the pathogenesis of Alzheimer's disease. Alzheimer's disease is marked by salient inflammatory features, characterized by microglial activation and escalation in the levels of pro-inflammatory cytokines in the affected regions. Studies have suggested a probable role of systemic infection conducing to inflammatory status of the central nervous system. Periodontitis is common oral infection affiliated with gram negative, anaerobic bacteria, capable of orchestrating localized and systemic infections in the subject. Periodontitis is known to elicit a "low grade systemic inflammation" by release of pro-inflammatory cytokines into systemic circulation. This review elucidates the possible role of periodontitis in exacerbating Alzheimer's disease. Periodontitis may bear the potential to affect the onset and progression of Alzheimer's disease. Periodontitis shares the two important features of Alzheimer's disease namely oxidative damage and inflammation, which are exhibited in the brain pathology of Alzheimer's disease. Periodontitis can be treated and hence it is a modifiable risk factor for Alzheimer's disease.
Collapse
Affiliation(s)
- Abhijit N Gurav
- Department of Periodontics, Tatyasaheb Kore Dental College & Research Centre
| |
Collapse
|
32
|
Herpes simplex infection and the risk of Alzheimer's disease: A nested case-control study. Alzheimers Dement 2015; 11:587-92. [DOI: 10.1016/j.jalz.2014.07.157] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Revised: 05/07/2014] [Accepted: 07/29/2014] [Indexed: 11/24/2022]
|
33
|
Civitelli L, Marcocci ME, Celestino I, Piacentini R, Garaci E, Grassi C, De Chiara G, Palamara AT. Herpes simplex virus type 1 infection in neurons leads to production and nuclear localization of APP intracellular domain (AICD): implications for Alzheimer's disease pathogenesis. J Neurovirol 2015; 21:480-90. [PMID: 25925093 DOI: 10.1007/s13365-015-0344-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 03/27/2015] [Accepted: 04/09/2015] [Indexed: 11/28/2022]
Abstract
Several data indicate that neuronal infection with herpes simplex virus type 1 (HSV-1) causes biochemical alterations reminiscent of Alzheimer's disease (AD) phenotype. They include accumulation of amyloid-β (Aβ), which originates from the cleavage of amyloid precursor protein (APP), and hyperphosphorylation of tau protein, which leads to neurofibrillary tangle deposition. HSV-1 infection triggers APP processing and drives the production of several fragments including APP intracellular domain (AICD) that exerts transactivating properties. Herein, we analyzed the production and intracellular localization of AICD following HSV-1 infection in neurons. We also checked whether AICD induced the transcription of two target genes, neprilysin (nep) and glycogen synthase kinase 3β (gsk3β), whose products play a role in Aβ clearance and tau phosphorylation, respectively. Our data indicate that HSV-1 led to the accumulation and nuclear translocation of AICD in neurons. Moreover, results from chromatin immunoprecipitation assay showed that AICD binds the promoter region of both nep and gsk3β. Time course analysis of NEP and GSK3β expression at both mRNA and protein levels demonstrated that they are differently modulated during infection. NEP expression and enzymatic activity were initially stimulated but, with the progression of infection, they were down-regulated. In contrast, GSK3β expression remained nearly unchanged, but the analysis of its phosphorylation suggests that it was inactivated only at later stages of HSV-1 infection. Thus, our data demonstrate that HSV-1 infection induces early upstream events in the cell that may eventually lead to Aβ deposition and tau hyperphosphorylation and further suggest HSV-1 as a possible risk factor for AD.
Collapse
Affiliation(s)
- Livia Civitelli
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy. .,Experimental Pathology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden.
| | - Maria Elena Marcocci
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Ignacio Celestino
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Roberto Piacentini
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Enrico Garaci
- IRCCS San Raffaele Pisana, Telematic University, Rome, Italy
| | - Claudio Grassi
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome, Italy
| | | | - Anna Teresa Palamara
- Department of Public Health and Infectious Diseases, Institute Pasteur Cenci Bolognetti Foundation, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
34
|
Lövheim H, Gilthorpe J, Adolfsson R, Nilsson LG, Elgh F. Reactivated herpes simplex infection increases the risk of Alzheimer's disease. Alzheimers Dement 2014; 11:593-9. [PMID: 25043910 DOI: 10.1016/j.jalz.2014.04.522] [Citation(s) in RCA: 145] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 03/06/2014] [Accepted: 04/14/2014] [Indexed: 12/14/2022]
Abstract
BACKGROUND Previous studies have suggested a link between herpes simplex virus (HSV) type 1 and the development of Alzheimer's disease (AD). METHODS The present analysis included 3432 persons (53.9% women, mean age at inclusion 62.7 ± 14.4 years) with a mean follow-up time of 11.3 years. The number of incident AD cases was 245. Serum samples were analyzed for anti-HSV antibodies (immunoglobulin (Ig)G and IgM) by enzyme-linked immunosorbent assays. RESULTS The presence of anti-HSV IgG antibodies was not associated with an increased risk for AD, controlled for age and sex (hazard ratio, HR, 0.993, P = .979). However, the presence of anti-HSV IgM at baseline was associated with an increased risk of developing AD (HR 1.959, P = .012). CONCLUSION Positivity for anti-HSV IgM, a sign of reactivated infection, was found to almost double the risk for AD, whereas the presence of anti-HSV IgG antibodies did not affect the risk.
Collapse
Affiliation(s)
- Hugo Lövheim
- Department of Community Medicine and Rehabilitation, Geriatric Medicine, Umeå University, Umeå, Sweden.
| | - Jonathan Gilthorpe
- Department of Pharmacology and Clinical Neuroscience, Umeå University, Umeå, Sweden
| | - Rolf Adolfsson
- Department of Clinical Sciences, Psychiatry, Umeå University, Umeå, Sweden
| | - Lars-Göran Nilsson
- Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå, Sweden; Aging Research Center, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | - Fredrik Elgh
- Department of Clinical Microbiology, Virology, Umeå University, Umeå, Sweden
| |
Collapse
|
35
|
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: 83] [Impact Index Per Article: 7.5] [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
|
36
|
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
|
37
|
Cerebrospinal fluid Alzheimer's biomarker profiles in CNS infections. J Neurol 2012; 260:620-6. [PMID: 23052602 DOI: 10.1007/s00415-012-6688-y] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Revised: 09/14/2012] [Accepted: 09/25/2012] [Indexed: 10/27/2022]
Abstract
The cerebrospinal fluid (CSF) biomarker profile in Alzheimer's disease (AD) is characterized by decreased beta amyloid (Aβ(1-42)), increased total and hyperphosphorylated tau (t-tau and p-tau, respectively), which is a useful diagnostic tool and gives insight in the pathogenesis of AD. It is of importance to study how these biomarkers react in other CNS diseases; therefore, we decided to analyse amyloid and tau biomarkers in different CNS infections. We also included analysis of soluble amyloid precursor proteins (sAPPα and -β). CSF Aβ(1-42), sAPPα and -β, t-tau and p-tau were analysed in bacterial meningitis (n = 12), Lyme neuroborreliosis (n = 13), herpes simplex virus type 1 (HSV-1) encephalitis (n = 10), HIV-associated dementia (HAD) (n = 21), AD (n = 21) and healthy controls (n = 42). Concurrent with AD, Aβ(1-42) was decreased in all groups except neuroborreliosis compared to controls. HSV-1 encephalitis, bacterial meningitis and HAD showed lower concentrations of sAPPα and -β compared to AD. T-tau was increased in AD and HSV-1 encephalitis compared to all other groups. P-tau was higher in AD and HSV-1 encephalitis compared to bacterial meningitis, HAD and control. Decreased CSF Aβ(1-42), sAPPα and -β in various CNS infections imply an effect of neuroinflammation on amyloid metabolism which is similar in regard to AD concerning Aβ(1-42), but differs concerning sAPPα and -β. These results clearly indicate different pathologic pathways in AD and infectious CNS disease and may provide help in the differential biomarker diagnostics. Increased p-tau in HSV-1 encephalitis probably reflect acute neuronal damage and necrosis.
Collapse
|
38
|
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: 169] [Impact Index Per Article: 13.0] [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
|
39
|
Association between IgM anti-herpes simplex virus and plasma amyloid-beta levels. PLoS One 2011; 6:e29480. [PMID: 22216291 PMCID: PMC3247269 DOI: 10.1371/journal.pone.0029480] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2011] [Accepted: 11/29/2011] [Indexed: 11/20/2022] Open
Abstract
Objective Herpes simplex virus (HSV) reactivation has been identified as a possible risk factor for Alzheimer's disease (AD) and plasma amyloid-beta (Aβ) levels might be considered as possible biomarkers of the risk of AD. The aim of our study was to investigate the association between anti-HSV antibodies and plasma Aβ levels. Methods The study sample consisted of 1222 subjects (73.9 y in mean) from the Three-City cohort. IgM and IgG anti-HSV antibodies were quantified using an ELISA kit, and plasma levels of Aβ1–40 and Aβ1–42 were measured using an xMAP-based assay technology. Cross-sectional analyses of the associations between anti-HSV antibodies and plasma Aβ levels were performed by multi-linear regression. Results After adjustment for study center, age, sex, education, and apolipoprotein E-e4 polymorphism, plasma Aβ1–42 and Aβ1–40 levels were specifically inversely associated with anti-HSV IgM levels (β = −20.7, P = 0.001 and β = −92.4, P = 0.007, respectively). In a sub-sample with information on CLU- and CR1-linked SNPs genotyping (n = 754), additional adjustment for CR1 or CLU markers did not modify these associations (adjustment for CR1 rs6656401, β = −25.6, P = 0.002 for Aβ1–42 and β = −132.7, P = 0.002 for Aβ1–40; adjustment for CLU rs2279590, β = −25.6, P = 0.002 for Aβ1–42 and β = −134.8, P = 0.002 for Aβ1–40). No association between the plasma Aβ1–42-to-Aβ1–40 ratio and anti-HSV IgM or IgG were evidenced. Conclusion High anti-HSV IgM levels, markers of HSV reactivation, are associated with lower plasma Aβ1–40 and Aβ1–42 levels, which suggest a possible involvement of the virus in the alterations of the APP processing and potentially in the pathogenesis of AD in human.
Collapse
|
40
|
Abstract
AbstractAlzheimer’s disease (AD) is the most frequent cause of dementia in the elderly, characterized by the presence of cerebral amyloid plaques and neurofibrillary tangles. The causes of the disease are not well understood, especially considering that more than 95% of AD patients are non-familial. Due to the similarity of brain regions affected in herpes simplex encephalitis to those mainly affected in AD, and owing to the very high prevalence of latent herpes simplex virus type 1 (HSV1) infection, reactivation of HSV1 was proposed as one of the possible causes of AD. The trigeminal ganglion, located only a few millimeters from the entorhinal cortex, is the primary site of HSV1 latency, although other sites including the sensory neurons, the nodose ganglion of the vagus nerve and other regions of the brain may be involved, possibly in relation to very early neurofibrillary AD changes in the dorsal raphe, locus coeruleus and other brainstem nuclei. Novel data obtained upon infection of cultured neuronal cells and mouse brain with HSV1 further show that HSV1 infection causes intracellular amyloid-beta protein accumulation, as well as abnormal phosphorylation of tau protein, the major component of tangles. Another interesting fact is the existence of a significant degree of homology between HSV1 components and AD susceptibility genes. In this review we summarize findings that reveal connections between the two conditions, as well as different suggestions for the mechanisms of HSV1-induced AD. As most of the available results support a connection of AD and HSV1 infection, antiviral therapy should be taken into consideration for AD treatment following early diagnosis.
Collapse
|
41
|
APP processing induced by herpes simplex virus type 1 (HSV-1) yields several APP fragments in human and rat neuronal cells. PLoS One 2010; 5:e13989. [PMID: 21085580 PMCID: PMC2981559 DOI: 10.1371/journal.pone.0013989] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2010] [Accepted: 10/25/2010] [Indexed: 11/30/2022] Open
Abstract
Lifelong latent infections of the trigeminal ganglion by the neurotropic herpes simplex virus type 1 (HSV-1) are characterized by periodic reactivation. During these episodes, newly produced virions may also reach the central nervous system (CNS), causing productive but generally asymptomatic infections. Epidemiological and experimental findings suggest that HSV-1 might contribute to the pathogenesis of Alzheimer's disease (AD). This multifactorial neurodegenerative disorder is related to an overproduction of amyloid beta (Aβ) and other neurotoxic peptides, which occurs during amyloidogenic endoproteolytic processing of the transmembrane amyloid precursor protein (APP). The aim of our study was to identify the effects of productive HSV-1 infection on APP processing in neuronal cells. We found that infection of SH-SY5Y human neuroblastoma cells and rat cortical neurons is followed by multiple cleavages of APP, which result in the intra- and/or extra-cellular accumulation of various neurotoxic species. These include: i) APP fragments (APP-Fs) of 35 and 45 kDa (APP-F35 and APP-F45) that comprise portions of Aβ; ii) N-terminal APP-Fs that are secreted; iii) intracellular C-terminal APP-Fs; and iv) Aβ1-40 and Aβ1-42. Western blot analysis of infected-cell lysates treated with formic acid suggests that APP-F35 may be an Aβ oligomer. The multiple cleavages of APP that occur in infected cells are produced in part by known components of the amyloidogenic APP processing pathway, i.e., host-cell β-secretase, γ-secretase, and caspase-3-like enzymes. These findings demonstrate that HSV-1 infection of neuronal cells can generate multiple APP fragments with well-documented neurotoxic potentials. It is tempting to speculate that intra- and extracellular accumulation of these species in the CNS resulting from repeated HSV-1 reactivation could, in the presence of other risk factors, play a co-factorial role in the development of AD.
Collapse
|
42
|
Holmes C, Cotterell D. Role of infection in the pathogenesis of Alzheimer's disease: implications for treatment. CNS Drugs 2009; 23:993-1002. [PMID: 19958038 DOI: 10.2165/11310910-000000000-00000] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
While our understanding of the neuropathology of Alzheimer's disease continues to grow, its pathogenesis remains a subject of intense debate. Genetic mutations contribute to a minority of early-onset autosomal dominant cases, but most cases are of either late-onset familial or sporadic form. CNS infections, most notably herpes simplex virus type 1, Chlamydophila pneumoniae and several types of spirochetes, have been previously suggested as possible aetiological agents in the development of sporadic Alzheimer's disease but with little consistent evidence. However, peripheral infections may have a role to play in accelerating neurodegeneration in Alzheimer's disease by activating already primed microglial cells within the CNS. Potential pharmacological interventions could aim at modification of this peripheral inflammatory response through targeting various agents involved in this inflammatory pathway. However, benefit could also be gained clinically through the meticulous detection, treatment and prevention of infections in individuals either alone or in combination with anti-inflammatory therapy.
Collapse
Affiliation(s)
- Clive Holmes
- University of Southampton, Memory Assessment and Research Centre, Moorgreen Hospital, Southampton, UK.
| | | |
Collapse
|
43
|
Donev R, Kolev M, Millet B, Thome J. Neuronal death in Alzheimer's disease and therapeutic opportunities. J Cell Mol Med 2009; 13:4329-48. [PMID: 19725918 PMCID: PMC4515050 DOI: 10.1111/j.1582-4934.2009.00889.x] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Alzheimer’s disease (AD) is an age-related neurodegenerative disease that affects approximately 24 million people worldwide. A number of different risk factors have been implicated in AD; however, neuritic (amyloid) plaques are considered as one of the defining risk factors and pathological hallmarks of the disease. In the past decade, enormous efforts have been devoted to understand the genetics and molecular pathogenesis leading to neuronal death in AD, which has been transferred into extensive experimental approaches aimed at reversing disease progression. Modern medicine is facing an increasing number of treatments available for vascular and neurodegenerative brain diseases, but no causal or neuroprotective treatment has yet been established. Almost all neurological conditions are characterized by progressive neuronal dysfunction, which, regardless of the pathogenetic mechanism, finally leads to neuronal death. The particular emphasis of this review is on risk factors and mechanisms resulting in neuronal loss in AD and current and prospective opportunities for therapeutic interventions. This review discusses these issues with a view to inspiring the development of new agents that could be useful for the treatment of AD.
Collapse
Affiliation(s)
- Rossen Donev
- Department of Medical Biochemistry and Immunology, School of Medicine, Cardiff University, Heath Park, Cardiff, UK
| | | | | | | |
Collapse
|
44
|
Watts A, Crimmins EM, Gatz M. Inflammation as a potential mediator for the association between periodontal disease and Alzheimer's disease. Neuropsychiatr Dis Treat 2008; 4:865-76. [PMID: 19183779 PMCID: PMC2626915 DOI: 10.2147/ndt.s3610] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Periodontal disease (PDD) is associated with increased risk of cardiovascular disease, cerebrovascular disease, and mortality in many studies, while other studies have begun to suggest an association of PDD with Alzheimer's disease (AD). This paper discusses how infectious pathogens and systemic infection may play a role in AD. The roles of infection and inflammation are addressed specifically with regard to known AD pathologic lesions including senile plaques, neuron death, neurofibrillary tangles, and cerebrovascular changes. A testable model of proposed pathways between periodontal infection and AD is presented including three possible mechanisms: a) direct effects of infectious pathogens, b) inflammatory response to pathogens, and c) the effects on vascular integrity. The role of gene polymorphisms is discussed, including apolipoprotein (APOE) varepsilon4 as a pro-inflammatory and pro-infection genotype.
Collapse
Affiliation(s)
- Amber Watts
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA.
| | | | | |
Collapse
|
45
|
Wozniak MA, Mee AP, Itzhaki RF. Herpes simplex virus type 1 DNA is located within Alzheimer's disease amyloid plaques. J Pathol 2008; 217:131-8. [DOI: 10.1002/path.2449] [Citation(s) in RCA: 243] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
46
|
Abstract
As terminally differentiated vital cells, neurons may be specialized to fight viral infections without undergoing cellular self-destruction. The cellular lysosomal degradation pathway, autophagy, is emerging as one such mechanism of neuronal antiviral defence. Autophagy has diverse physiological functions, such as cellular adaptation to stress, routine organelle and protein turnover, and innate immunity against intracellular pathogens, including viruses. Most of the in vivo evidence for an antiviral role of autophagy is related to viruses that specifically target neurons, including the prototype alphavirus, Sindbis virus, and the alpha-herpesvirus, herpes simplex virus type 1 (HSV-1). In the case of HSV-1, viral evasion of autophagy is essential for lethal encephalitis. As basal autophagy is important in preventing neurodegeneration, and induced autophagy is important in promoting cellular survival during stress, viral antagonism of autophagy in neurons may lead to neuronal dysfunction and/or neuronal cell death. This review provides background information on the roles of autophagy in immunity and neuroprotection, and then discusses the relationships between autophagy and viral neurovirulence.
Collapse
Affiliation(s)
- Anthony Orvedahl
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | |
Collapse
|
47
|
Herpes simplex virus infection causes cellular β-amyloid accumulation and secretase upregulation. Neurosci Lett 2007; 429:95-100. [DOI: 10.1016/j.neulet.2007.09.077] [Citation(s) in RCA: 215] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2007] [Revised: 09/18/2007] [Accepted: 09/28/2007] [Indexed: 11/20/2022]
|
48
|
Interactions between the products of the Herpes simplex genome and Alzheimer's disease susceptibility genes: relevance to pathological-signalling cascades. Neurochem Int 2007; 52:920-34. [PMID: 18164103 DOI: 10.1016/j.neuint.2007.11.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2007] [Revised: 11/08/2007] [Accepted: 11/19/2007] [Indexed: 01/07/2023]
Abstract
The products of the Herpes simplex (HSV-1) genome interact with many Alzheimer's disease susceptibility genes or proteins. These in turn affect those of the virus. For example, HSV-1 binds to heparan sulphate proteoglycans (HSPG2), or alpha-2-macroglobulin (A2M), and enters cells via nectin receptors, which are cleaved by gamma-secretase (APH1B, PSEN1, PSEN2, PEN2, NCSTN). The virus also binds to blood-borne lipoproteins and apolipoprotein E (APOE) is able to modify its infectivity. Viral uptake is cholesterol- and lipid raft-dependent (DHCR24, HMGCR, FDPS, RAFTLIN, SREBF1). The virus is transported to the nucleus via the dynein and kinesin (KNS2) motors associated with the microtubule network (MAPT). Amyloid precursor protein (APP) plays a role in this transport. Nuclear export is mediated via disruption of the nuclear lamina and binding to LMNA. Herpes simplex activates kinases (CDC2 and casein kinase 2) whose substrates include APOE, APP, MAPT, PSEN2, and SREBF1. A viral protein is also able to delete mitochondrial DNA, a situation prevalent in Alzheimer's disease. The virus binds to the host transcription factors transcription factor CP2 (TFCP2) and POU2F1 that control many other genes associated with Alzheimer's disease. Viral latency is controlled by IL6 and IL1B and at different stages of its life cycle the virus can either promote or attenuate apoptosis via Fas and tumor necrosis factor pathways (FAS, TNF, DAPK1, PARP1). Viral evasion strategies include inhibition of the antigen processor TAP2, the production of an Fc immunoglobulin receptor mimic (FCER1G) and inhibition of the viral-activated kinase EIF2AK2. These and other host/viral interactions, targeted to certain Alzheimer's disease susceptibility genes, support the idea that some form of synergy between the pathogen and genetic factors may play a role in the pathology of late-onset Alzheimer's disease.
Collapse
|