1
|
Kordi R, Andrews TJ, Hicar MD. Infections, genetics, and Alzheimer's disease: Exploring the pathogenic factors for innovative therapies. Virology 2025; 607:110523. [PMID: 40174330 DOI: 10.1016/j.virol.2025.110523] [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: 01/10/2025] [Revised: 03/20/2025] [Accepted: 03/26/2025] [Indexed: 04/04/2025]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative condition that creates a significant global health challenge and profoundly affects patients and their families. Recent research has highlighted the critical role of microorganisms, particularly viral infections, in the pathogenesis of AD. The involvement of viral infections in AD pathogenesis is predominantly attributed to their ability to induce neuroinflammation and amyloid beta (Aβ) deposition in the brain. The extant research exploring the relationship between viruses and AD has focused largely on Herpesviridae family. Traces of Herpesviruses, such as Herpes Simplex Virus-1 and Epstein Barr Virus, have been found in the brains of patients with AD. These viruses are thought to contribute to the disease progression by triggering chronic inflammatory responses in the brain. They can remain dormant in the brain, and become reactivated due to stress, a secondary viral infection, or immune-senescence in older adults. This review focuses on the association between Herpesviridae and bacterial infections with AD. We explore the genetic factors that might regulate viral illness and discuss clinical trials investigating antiviral and anti-inflammatory agents as possible therapeutic strategies to mitigate cognitive decline in patients with AD. In summary, understanding the interplay between infections, genetic factors, and AD pathogenesis may pave the way for novel therapeutic approaches, facilitating better management and possibly even prevent this debilitating disease.
Collapse
Affiliation(s)
- Ramesh Kordi
- Department of Pediatrics, Division of Infectious Diseases, State University of New York at Buffalo, Buffalo, NY, 14203, USA
| | - Ted J Andrews
- Department of Pediatrics, Division of Developmental Pediatrics and Rehabilitation, State University of New York at Buffalo, Buffalo, NY, 14203, USA
| | - Mark D Hicar
- Department of Pediatrics, Division of Infectious Diseases, State University of New York at Buffalo, Buffalo, NY, 14203, USA.
| |
Collapse
|
2
|
Kalamvoki M. HSV-1 virions and related particles: biogenesis and implications in the infection. J Virol 2025; 99:e0107624. [PMID: 39898651 PMCID: PMC11915793 DOI: 10.1128/jvi.01076-24] [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] [Indexed: 02/04/2025] Open
Abstract
Virion formation and egress are sophisticated processes that rely on the spatial and temporal organization of host cell membranes and the manipulation of host machineries involved in protein sorting, membrane bending, fusion, and fission. These processes result in the formation of infectious virions, defective particles, and various vesicle-like structures. In herpes simplex virus 1 (HSV-1) infections, virions and capsid-less particles, known as light (L)-particles, are formed. HSV-1 infection also stimulates the release of particles that resemble extracellular vesicles (EVs). In productively infected cells, most EVs are generated through the CD63 tetraspanin biogenesis pathway and lack viral components. A smaller subset of EVs, generated through the endosomal sorting complexes required for transport (ESCRT) pathway, contains both viral and host factors. Viral mechanisms tightly regulate EV biogenesis, including the inhibition of autophagy-a process critical for increased production of CD63+ EVs during HSV-1 infection. Mutant viruses that fail to suppress autophagy instead promote microvesicle production from the plasma membrane. Additionally, the viral protein ICP0 (Infected Cell Protein 0) enhances EV biogenesis during HSV-1 infection. The different types of particles can be separated by density gradients due to their distinct biophysical properties. L-particles and ESCRT+ EVs display a pro-viral role, supporting viral replication, whereas CD63+ EVs exhibit antiviral effects. Overall, these studies highlight that HSV-1 infection yields numerous and diverse particles, with their type and composition shaped by the ability of the virus to evade host responses. These particles likely shape the infectious microenvironment and determine disease outcomes.
Collapse
Affiliation(s)
- Maria Kalamvoki
- Department of Microbiology, Molecular Genetics, and Immunology, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
3
|
Huang Z, Zhao X, Jiang Z, Qiu X, Sun X, Wang D, Zhang H, Chen Q, Tan R, Shen Y. IP6K2 mutations as a novel mechanism of resistance to oncolytic virus therapy. J Transl Med 2025; 23:311. [PMID: 40075351 PMCID: PMC11900485 DOI: 10.1186/s12967-025-06265-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 02/17/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Oncolytic virus therapy (OVT) represents a promising frontier in cancer treatment. Despite its efficacy in clinical trials, variability in patient response, particularly resistance development, highlights the need for tailored therapeutic strategies. METHODS The Inositol Hexakisphosphate Kinase 2 (IP6K2) gene knock out was carried by CRISPR/Cas9 system. The evaluation of biomarkers of apoptosis and relevant pathways was conducted to be assessed. Attachment assay was conducted to verify the binding ability of virus to the host cells. Cell proliferation and apoptosis was assessed. Subcutaneous xenograft model was used to evaluate IP6K2 knock out influence in vivo. cBioPortal and TCGA database were applied to analyze genomic alterations in pan-cancer. RESULTS IP6K2 was essential for effective Herpes Simplex Virus Type1 (HSV-1) replication and subsequent cell apoptosis, acting through the tumor Protein p53 (p53) and Cyclin-Dependent Kinase Inhibitor 1 A (p21) signaling axis. The tumor model demonstrated that tumors lacking IP6K2 exhibited resistance to HSV-1 oncolysis, resulting in diminished therapeutic outcomes. Analysis of cBioPortal and TCGA databases corroborated the potential resistance stemming from IP6K2 mutations across various cancer types, underscoring the necessity for pre-treatment IP6K2 status assessment. CONCLUSIONS This study underscores the role of IP6K2 as potential markers of resistance, which opens avenues for precision medicine approaches in OVT.
Collapse
Affiliation(s)
- Zhijian Huang
- Department of Breast Surgical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, 610041, China
- Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, 02215, USA
| | - Xiangqian Zhao
- The Cancer Center, Union Hospital, Fujian Medical University, Fuzhou, 350001, China
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University Qishan Campus, Fuzhou, 350117, China
| | - Zirong Jiang
- Department of Thyroid and Breast Surgery, Ningde Municipal Hospital of Ningde Normal University, Ningde, 352100, China
| | - Xiaoting Qiu
- Department of Breast Surgical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Xinhao Sun
- College of Science, Northeastern University, Boston, 02115, USA
| | - Dawei Wang
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University Qishan Campus, Fuzhou, 350117, China
| | - Hucheng Zhang
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University Qishan Campus, Fuzhou, 350117, China
| | - Qi Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University Qishan Campus, Fuzhou, 350117, China.
| | - Ruirong Tan
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, 610041, China.
| | - Yangkun Shen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University Qishan Campus, Fuzhou, 350117, China.
| |
Collapse
|
4
|
Wang Z, Zou W, Zeng Q, Song X, Li M, Pang J, Zhu H, La C, Wang X, Wang Y, Zheng K. Novel Hsp90α inhibitor inhibits HSV-1 infection by suppressing the Akt/β-catenin pathway. Int J Antimicrob Agents 2025; 65:107448. [PMID: 39863183 DOI: 10.1016/j.ijantimicag.2025.107448] [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: 07/11/2024] [Revised: 12/06/2024] [Accepted: 01/17/2025] [Indexed: 01/27/2025]
Abstract
OBJECTIVE The prevalence of herpes simplex virus type 1 (HSV-1) infection and the emergence of drug-resistant HSV-1 strains posts a significant global health challenge, necessitating the urgent development of effective anti-HSV-1 drugs. As one of the most prevalent molecular chaperones, heat shock protein 90 α (Hsp90α) has been extensively demonstrated to regulate a range of viral infections, thus representing a promising antiviral target. In this study, we identified JD-13 as a novel Hsp90α inhibitor and explored its capability in inhibiting HSV-1 infection. METHODS The inhibitory effect of JD-13 on Hsp90α activity was confirmed by molecular docking, molecular dynamic stimulations, fluorescence quench titration and cellular thermal shift assay. The antiviral activity of JD-13 was examined by viral plaque assay, RT-qPCR, Western blot, flow cytometry, fluorescence microscopy and time-of-addition assay. The in vivo antiviral efficacy of JD-13 was evaluated in the HSV-1 skin infection guinea pig model by analyzing skin lesions and herpes formation. RESULTS JD-13 significantly inhibited the infection of both normal and acyclovir-resistant HSV-1 strains. In addition, JD-13 alleviated skin damage in guinea pigs caused by cutaneous HSV-1 infection. Further studies revealed that JD-13 impaired HSV-1 early infection and suppressed the Akt/β-catenin signalling pathway by promoting Akt degradation. Consequently, the inhibition of the Akt/β-catenin signalling pathway restricted HSV-1 infection. CONCLUSIONS These results suggest JD-13 as a novel HSP90α inhibitor with the potential to be developed as an antiviral agent for the treatment of HSV-1-related diseases.
Collapse
Affiliation(s)
- Zexu Wang
- Institute of Biomedicine, College of Life Science and Technology, Guangdong Province Key Laboratory of Bioengineering Medicine, Key Laboratory of Innovative Technology Research on Natural Products and Cosmetics Raw Materials, Jinan University, Guangzhou, China
| | - Weixiangmin Zou
- Institute of Biomedicine, College of Life Science and Technology, Guangdong Province Key Laboratory of Bioengineering Medicine, Key Laboratory of Innovative Technology Research on Natural Products and Cosmetics Raw Materials, Jinan University, Guangzhou, China
| | - Qiongzhen Zeng
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China.
| | - Xiaowei Song
- Institute of Biomedicine, College of Life Science and Technology, Guangdong Province Key Laboratory of Bioengineering Medicine, Key Laboratory of Innovative Technology Research on Natural Products and Cosmetics Raw Materials, Jinan University, Guangzhou, China; Center for Mitochondrial Genetics and Health, Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, China
| | - Menghe Li
- Institute of Biomedicine, College of Life Science and Technology, Guangdong Province Key Laboratory of Bioengineering Medicine, Key Laboratory of Innovative Technology Research on Natural Products and Cosmetics Raw Materials, Jinan University, Guangzhou, China
| | - Jiaping Pang
- Institute of Biomedicine, College of Life Science and Technology, Guangdong Province Key Laboratory of Bioengineering Medicine, Key Laboratory of Innovative Technology Research on Natural Products and Cosmetics Raw Materials, Jinan University, Guangzhou, China
| | - Hai Zhu
- Institute of Biomedicine, College of Life Science and Technology, Guangdong Province Key Laboratory of Bioengineering Medicine, Key Laboratory of Innovative Technology Research on Natural Products and Cosmetics Raw Materials, Jinan University, Guangzhou, China
| | - Caiwenjie La
- Institute of Biomedicine, College of Life Science and Technology, Guangdong Province Key Laboratory of Bioengineering Medicine, Key Laboratory of Innovative Technology Research on Natural Products and Cosmetics Raw Materials, Jinan University, Guangzhou, China
| | - Xiao Wang
- Department of Pharmacy, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Yifei Wang
- Institute of Biomedicine, College of Life Science and Technology, Guangdong Province Key Laboratory of Bioengineering Medicine, Key Laboratory of Innovative Technology Research on Natural Products and Cosmetics Raw Materials, Jinan University, Guangzhou, China.
| | - Kai Zheng
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, China.
| |
Collapse
|
5
|
Piacentini R, Grassi C. Interleukin 1β receptor and synaptic dysfunction in recurrent brain infection with Herpes simplex virus type-1. Neural Regen Res 2025; 20:416-423. [PMID: 38819045 PMCID: PMC11317954 DOI: 10.4103/nrr.nrr-d-23-01690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 02/21/2024] [Accepted: 03/21/2024] [Indexed: 06/01/2024] Open
Abstract
Several experimental evidence suggests a link between brain Herpes simplex virus type-1 infection and the occurrence of Alzheimer's disease. However, the molecular mechanisms underlying this association are not completely understood. Among the molecular mediators of synaptic and cognitive dysfunction occurring after Herpes simplex virus type-1 infection and reactivation in the brain neuroinflammatory cytokines seem to occupy a central role. Here, we specifically reviewed literature reports dealing with the impact of neuroinflammation on synaptic dysfunction observed after recurrent Herpes simplex virus type-1 reactivation in the brain, highlighting the role of interleukins and, in particular, interleukin 1β as a possible target against Herpes simplex virus type-1-induced neuronal dysfunctions.
Collapse
Affiliation(s)
- Roberto Piacentini
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | - Claudio Grassi
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| |
Collapse
|
6
|
Giuliani M, Uboldi C, Dellatorre FG, Latour E, Ponce NMA, Stortz CA, Lassalle VL, Ayala-Peña VB. Undaria pinnatifida fucoidan extract inhibits activation of the NF-κB signaling pathway by herpes simplex virus type 1 and prevents amyloid-β peptide synthesis in retinal pigment epithelium cells. Arch Virol 2025; 170:27. [PMID: 39762563 DOI: 10.1007/s00705-024-06212-2] [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: 06/27/2024] [Accepted: 10/31/2024] [Indexed: 02/08/2025]
Abstract
Neurodegenerative pathologies such as age-related macular degeneration currently have no cure or effective treatment. In this type of disease, the presence of amyloid-β peptides, oxidative stress, and inflammation trigger dysregulation of retinal pigment epithelial cells and progression toward the death of these cells, resulting in a loss of vision. The production of amyloid-β peptides, oxidative stress, and inflammation can be triggered in response to viral infections. Fucoidans are sulfated polysaccharides that are present in the cell walls of brown algae. There is a large body of literature reporting a wide range of biological properties of these compounds. In this study, we investigated whether Undaria pinnatifida fucoidan extract can prevent infection with herpes simplex virus type 1 and if the extract has antioxidant activity. We also evaluated whether, under viral infection conditions, the synthesis of amyloid-β peptide and NF-κB activation could be inhibited by the extract. The results showed for the first time that this extract can prevent viral infection in retinal pigment epithelial cells and that they can prevent the formation of amyloid-β peptide and the activation of the NF-κB pathway during viral infection. We also found that Undaria pinnatifida fucoidan extract has antioxidant activity and reduces the levels of reactive oxygen species. These data suggest that Undaria pinnatifida fucoidan extract might be effective for treating diseases triggered by viral infections, such as degenerative retinal diseases.
Collapse
Affiliation(s)
- Macarena Giuliani
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur-Bahía Blanca, Buenos Aires, Argentina
| | - Camila Uboldi
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur-Bahía Blanca, Buenos Aires, Argentina
| | - Fernando Gaspar Dellatorre
- Grupo de Investigación y Desarrollo Tecnológico en Acuicultura y Pesca (GIDTAP), Facultad Regional Chubut, Universidad Tecnológica Nacional, Puerto Madryn, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Ezequiel Latour
- Grupo de Investigación y Desarrollo Tecnológico en Acuicultura y Pesca (GIDTAP), Facultad Regional Chubut, Universidad Tecnológica Nacional, Puerto Madryn, Argentina
| | - Nora Marta Andrea Ponce
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Departamento de Química Orgánica, Facultad de Ciencias Exactas y Naturales and Centro de Investigaciones en Hidratos de Carbono (CIHIDECAR-UBA), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Carlos A Stortz
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Departamento de Química Orgánica, Facultad de Ciencias Exactas y Naturales and Centro de Investigaciones en Hidratos de Carbono (CIHIDECAR-UBA), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Verónica Leticia Lassalle
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Departamento de Química, INQUISUR, Universidad Nacional del Sur, Bahía Blanca, Argentina
| | - Victoria Belen Ayala-Peña
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur-Bahía Blanca, Buenos Aires, Argentina.
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.
| |
Collapse
|
7
|
Kobayashi H, Yasukochi M, Horie M, Orba Y, Sawa H, Fujino K, Taharaguchi S. Neuron-associated retroelement-derived protein Arc/Arg3.1 assists in the early stages of alphaherpesvirus infection in human neuronal cells. PLoS One 2024; 19:e0314980. [PMID: 39666775 PMCID: PMC11637343 DOI: 10.1371/journal.pone.0314980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 11/19/2024] [Indexed: 12/14/2024] Open
Abstract
Alphaherpesviruses, including herpes simplex virus type 1 (HSV-1) and pseudorabies virus (PRV), are neurotropic double-stranded DNA viruses. Alphaherpesviruses control the expression of various host factors to ensure efficient infection and propagation. Recently, HSV-1 was found to upregulate Arc/Arg3.1 (Arc) expression, which is a retroelement-derived domesticated gene. Arc is associated with learning and neuroplasticity in host neuronal cells, and its abnormal expression leads to neurological disorders. However, the detailed mechanisms underlying the upregulation of Arc and its physiological significance in viral infections remain unclear. In this study, we found that PRV infection upregulated Arc expression in vitro and identified ICP0 and EP0, the transcriptional regulatory genes of HSV-1 and PRV, as triggers for enhanced Arc expression. Mass spectrometry and co-immunoprecipitation assays identified VP5, the major capsid protein of PRV and HSV-1, as the viral factor that interacted with Arc. Arc knockdown delayed viral infection during the early stages of the viral life cycle, but did not impact the viral attachment and entry. In conclusion, we provide evidence that alphaherpesvirus ICP0 homologues control Arc expression. Additionally, we demonstrate that Arc interacts with the major capsid protein VP5 and plays an important role in the viral lifecycle after intracellular entry. This study advances our knowledge of herpesvirus and retroelement-derived Arc interactions, providing fundamental insights into the pathogenesis of retroelement-derived domesticated genes and herpesvirus-induced neurological diseases.
Collapse
Affiliation(s)
- Hiroko Kobayashi
- Laboratory of Microbiology, School of Veterinary Medicine, Azabu University, Sagamihara, Kanagawa, Japan
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Mitsuki Yasukochi
- Laboratory of Microbiology, School of Veterinary Medicine, Azabu University, Sagamihara, Kanagawa, Japan
| | - Masayuki Horie
- Laboratory of Veterinary Microbiology, Graduate School of Veterinary Science, Osaka Metropolitan University, Izumisano, Osaka, Japan
- Osaka International Research Center for Infectious Diseases, Osaka Metropolitan University, Osaka, Japan
| | - Yasuko Orba
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido, Japan
- Institute for Vaccine Research and Development, Hokkaido University, Sapporo, Hokkaido, Japan
- One Health Research Center, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Hirofumi Sawa
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido, Japan
- Institute for Vaccine Research and Development, Hokkaido University, Sapporo, Hokkaido, Japan
- One Health Research Center, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Kan Fujino
- Laboratory of Microbiology, School of Veterinary Medicine, Azabu University, Sagamihara, Kanagawa, Japan
| | - Satoshi Taharaguchi
- Laboratory of Microbiology, School of Veterinary Medicine, Azabu University, Sagamihara, Kanagawa, Japan
| |
Collapse
|
8
|
Niemeyer CS, Merle L, Bubak AN, Baxter BD, Gentile Polese A, Colon-Reyes K, Vang S, Hassell JE, Bruce KD, Nagel MA, Restrepo D. Olfactory and trigeminal routes of HSV-1 CNS infection with regional microglial heterogeneity. J Virol 2024; 98:e0096824. [PMID: 39475273 PMCID: PMC11575344 DOI: 10.1128/jvi.00968-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 09/29/2024] [Indexed: 11/06/2024] Open
Abstract
Herpes simplex virus type 1 (HSV-1) primarily targets the oral and nasal epithelia before establishing latency in the trigeminal ganglion (TG) and other peripheral ganglia. HSV-1 can also infect and become latent in the central nervous system (CNS) independent of latency in the TGs. Recent studies suggest entry to the CNS via two distinct routes: the TG-brainstem connection and olfactory nerve; however, to date, there is no characterization of brain regions targeted during HSV-1 primary infection. Furthermore, the immune response by microglia may also contribute to the heterogeneity between different brain regions. However, the response to HSV-1 by microglia has not been characterized in a region-specific manner. This study investigated the time course of HSV-1 spread within the olfactory epithelium (OE) and CNS following intranasal inoculation and the corresponding macrophage/microglial response in a C57BL/6 mouse model. We found an apical to basal spread of HSV-1 within the OE and underlying tissue accompanied by an inflammatory response of macrophages. OE infection was followed by infection of a small subset of brain regions targeted by the TG in the brainstem and other cranial nerve nuclei, including the vagus and hypoglossal nerve. Furthermore, other brain regions were positive for HSV-1 antigens, such as the locus coeruleus (LC), raphe nucleus (RaN), and hypothalamus while sparing the hippocampus and cortex. Within each brain region, microglia activation also varied widely. These findings provide critical insights into the region-specific dissemination of HSV-1 within the CNS, elucidating potential mechanisms linking viral infection to neurological and neurodegenerative diseases.IMPORTANCEThis study shows how herpes simplex virus type 1 (HSV-1) spreads within the brain after infecting the nasal passages. Our data reveal the distinct pattern of HSV-1 through the brain during a non-encephalitic infection. Furthermore, microglial activation was also temporally and spatially specific, with some regions of the brain having sustained microglial activation even in the absence of viral antigens. Previous reports have identified specific brain regions found to be positive for HSV-1 infection; however, to date, there has not been a concise investigation of the anatomical spread of HSV-1 and the brain regions consistently vulnerable to viral entry and spread. Understanding these region-specific differences in infection and immune response is crucial because it links HSV-1 infection to potential triggers for neurological and neurodegenerative diseases.
Collapse
Affiliation(s)
- Christy S. Niemeyer
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Laetitia Merle
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université de Bourgogne, Dijon, France
| | - Andrew N. Bubak
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - B. Dnate’ Baxter
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Arianna Gentile Polese
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Katherine Colon-Reyes
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Sandy Vang
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - James E. Hassell
- Department of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kimberley D. Bruce
- Department of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Maria A. Nagel
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Ophthalmology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Diego Restrepo
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
9
|
Niemeyer CS, Merle L, Bubak AN, Dnate' Baxter B, Polese AG, Colon-Reyes K, Vang S, Hassell JE, Bruce KD, Nagel MA, Restrepo D. Olfactory and Trigeminal Routes of HSV-1 CNS Infection with Regional Microglial Heterogeneity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.22.614340. [PMID: 39386674 PMCID: PMC11463476 DOI: 10.1101/2024.09.22.614340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Herpes simplex virus type 1 (HSV-1) primarily targets the oral and nasal epithelia before establishing latency in the trigeminal and other peripheral ganglia (TG). HSV-1 can also infect and go latent in the central nervous system (CNS) independent of latency in the TGs. Recent studies suggest entry to the CNS via two distinct routes: the TG-brainstem connection and olfactory nerve; however, to date, there is no characterization of brain regions targeted during HSV-1 primary infection. Furthermore, the immune response by microglia may also contribute to the heterogeneity between different brain regions. However, the response to HSV-1 by microglia has not been characterized in a region-specific manner. This study investigated the time course of HSV-1 spread within the olfactory epithelium (OE) and CNS following intranasal inoculation and the corresponding macrophage/microglial response in a C57BL/6 mouse model. We found an apical to basal spread of HSV-1 within the OE and underlying tissue accompanied by an inflammatory response of macrophages. OE Infection was followed by infection of a small subset of brain regions targeted by the TG in the brainstem, as well as other cranial nerve nuclei, including the vagus and hypoglossal nerve. Furthermore, other brain regions were positive for HSV-1 antigens, such as the locus coeruleus (LC), raphe nucleus (RaN), and hypothalamus, while sparing the hippocampus and cortex. Within each brain region, microglia activation also varied widely. These findings provide critical insights into the region-specific dissemination of HSV-1 within the CNS, elucidating potential mechanisms linking viral infection to neurological and neurodegenerative diseases. Importance This study sheds light on how herpes simplex virus type 1 (HSV-1) spreads within the brain after infecting the nasal passages. Our data reveals the distinct pattern of HSV-1 through the brain during a non-encephalitic infection. Furthermore, microglial activation was also temporally and spatially specific, with some regions of the brain having sustained microglial activation even in the absence of viral antigen. Previous reports have identified specific regions of the brain found to be positive for HSV-1 infection; however, to date, there has not been a concise investigation of the anatomical spread of HSV-1 and the regions of the brain consistently vulnerable to viral entry and spread. Understanding these region-specific differences in infection and immune response is crucial because it links HSV-1 infection to potential triggers for neurological and neurodegenerative diseases.
Collapse
|
10
|
Chang JY, Balch C, Oh HS. Toward the Eradication of Herpes Simplex Virus: Vaccination and Beyond. Viruses 2024; 16:1476. [PMID: 39339952 PMCID: PMC11437400 DOI: 10.3390/v16091476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 09/11/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Herpes simplex virus (HSV) has coevolved with Homo sapiens for over 100,000 years, maintaining a tenacious presence by establishing lifelong, incurable infections in over half the human population. As of 2024, an effective prophylactic or therapeutic vaccine for HSV remains elusive. In this review, we independently screened PubMed, EMBASE, Medline, and Google Scholar for clinically relevant articles on HSV vaccines. We identified 12 vaccines from our literature review and found promising candidates across various classes, including subunit vaccines, live vaccines, DNA vaccines, and mRNA vaccines. Notably, several vaccines-SL-V20, HF10, VC2, and mRNA-1608-have shown promising preclinical results, suggesting that an effective HSV vaccine may be within reach. Additionally, several other vaccines such as GEN-003 (a subunit vaccine from Genocea), HerpV (a subunit vaccine from Agenus), 0ΔNLS/RVx201 (a live-attenuated replication-competent vaccine from Rational Vaccines), HSV 529 (a replication-defective vaccine from Sanofi Pasteur), and COR-1 (a DNA-based vaccine from Anteris Technologies) have demonstrated potential in clinical trials. However, GEN-003 and HerpV have not advanced further despite promising results. Continued progress with these candidates brings us closer to a significant breakthrough in preventing and treating HSV infections.
Collapse
Affiliation(s)
- Jane Y Chang
- Ascendant Biotech Inc., Foster City, CA 94404, USA
| | - Curt Balch
- Bioscience Advising, Cincinnati, OH 45208, USA
| | - Hyung Suk Oh
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
11
|
Serrero MC, Paludan SR. Restriction factors regulating human herpesvirus infections. Trends Immunol 2024; 45:662-677. [PMID: 39198098 DOI: 10.1016/j.it.2024.07.010] [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: 07/10/2024] [Revised: 07/29/2024] [Accepted: 07/31/2024] [Indexed: 09/01/2024]
Abstract
Herpesviruses are DNA viruses and the cause of diseases ranging from mild skin conditions to severe brain diseases. Mammalian antiviral host defense comprises an array of mechanisms, including restriction factors (RFs), which block specific steps in viral replication cycles. In recent years, knowledge of RFs that contribute to controlling herpesvirus infections has expanded significantly, along with a new understanding of viral evasion mechanisms and disease pathogenesis. By integrating findings from human genetics, murine models, and cellular studies, this review provides a current view of RF control of herpesvirus infections. We also explore the regulation of RF expression, discuss the roles of RFs in diseases, and point towards their growing potential as candidate therapeutic targets.
Collapse
Affiliation(s)
- Manutea C Serrero
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Center for Immunology of Viral Infections, Aarhus, Denmark
| | - Søren R Paludan
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Center for Immunology of Viral Infections, Aarhus, Denmark.
| |
Collapse
|
12
|
Zhao Y, Xu K, Shu F, Zhang F. Neurotropic virus infection and neurodegenerative diseases: Potential roles of autophagy pathway. CNS Neurosci Ther 2024; 30:e14548. [PMID: 38082503 PMCID: PMC11163195 DOI: 10.1111/cns.14548] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/20/2023] [Accepted: 11/21/2023] [Indexed: 06/11/2024] Open
Abstract
Neurodegenerative diseases (NDs) constitute a group of disorders characterized by the progressive deterioration of nervous system functionality. Currently, the precise etiological factors responsible for NDs remain incompletely elucidated, although it is probable that a combination of aging, genetic predisposition, and environmental stressors participate in this process. Accumulating evidence indicates that viral infections, especially neurotropic viruses, can contribute to the onset and progression of NDs. In this review, emerging evidence supporting the association between viral infection and NDs is summarized, and how the autophagy pathway mediated by viral infection can cause pathological aggregation of cellular proteins associated with various NDs is discussed. Furthermore, autophagy-related genes (ARGs) involved in Herpes simplex virus (HSV-1) infection and NDs are analyzed, and whether these genes could link HSV-1 infection to NDs is discussed. Elucidating the mechanisms underlying NDs is critical for developing targeted therapeutic approaches that prevent the onset and slow the progression of NDs.
Collapse
Affiliation(s)
- Yu‐jia Zhao
- Laboratory Animal CentreZunyi Medical UniversityZunyiGuizhouChina
| | - Kai‐fei Xu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou ProvinceZunyi Medical UniversityZunyiGuizhouChina
| | - Fu‐xing Shu
- Bioresource Institute for Healthy UtilizationZunyi Medical UniversityZunyiGuizhouChina
| | - Feng Zhang
- Laboratory Animal CentreZunyi Medical UniversityZunyiGuizhouChina
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou ProvinceZunyi Medical UniversityZunyiGuizhouChina
| |
Collapse
|
13
|
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 PMCID: PMC11964427 DOI: 10.14336/ad.2024.0406] [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: 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
|
14
|
Sutter J, Brettschneider J, Wigdahl B, Bruggeman PJ, Krebs FC, Miller V. Non-Thermal Plasma Reduces HSV-1 Infection of and Replication in HaCaT Keratinocytes In Vitro. Int J Mol Sci 2024; 25:3839. [PMID: 38612649 PMCID: PMC11011387 DOI: 10.3390/ijms25073839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 03/23/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
Herpes simplex virus type 1 (HSV-1) is a lifelong pathogen characterized by asymptomatic latent infection in the trigeminal ganglia (TG), with periodic outbreaks of cold sores caused by virus reactivation in the TG and subsequent replication in the oral mucosa. While antiviral therapies can provide relief from cold sores, they are unable to eliminate HSV-1. We provide experimental results that highlight non-thermal plasma (NTP) as a new alternative therapy for HSV-1 infection that would resolve cold sores faster and reduce the establishment of latent infection in the TG. Additionally, this study is the first to explore the use of NTP as a therapy that can both treat and prevent human viral infections. The antiviral effect of NTP was investigated using an in vitro model of HSV-1 epithelial infection that involved the application of NTP from two separate devices to cell-free HSV-1, HSV-1-infected cells, and uninfected cells. It was found that NTP reduced the infectivity of cell-free HSV-1, reduced viral replication in HSV-1-infected cells, and diminished the susceptibility of uninfected cells to HSV-1 infection. This triad of antiviral mechanisms of action suggests the potential of NTP as a therapeutic agent effective against HSV-1 infection.
Collapse
Affiliation(s)
- Julia Sutter
- Center for Molecular Virology and Gene Therapy, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA 19102, USA; (J.S.); (J.B.); (B.W.); (F.C.K.)
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA 19102, USA
| | - Jascha Brettschneider
- Center for Molecular Virology and Gene Therapy, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA 19102, USA; (J.S.); (J.B.); (B.W.); (F.C.K.)
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA 19102, USA
| | - Brian Wigdahl
- Center for Molecular Virology and Gene Therapy, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA 19102, USA; (J.S.); (J.B.); (B.W.); (F.C.K.)
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA 19102, USA
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Peter J. Bruggeman
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN 55455, USA;
| | - Fred C. Krebs
- Center for Molecular Virology and Gene Therapy, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA 19102, USA; (J.S.); (J.B.); (B.W.); (F.C.K.)
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA 19102, USA
| | - Vandana Miller
- Center for Molecular Virology and Gene Therapy, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA 19102, USA; (J.S.); (J.B.); (B.W.); (F.C.K.)
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA 19102, USA
| |
Collapse
|
15
|
Liu Y, Xu W, Yang P, Liu X. Revealing Molecular Patterns of Alzheimer's Disease Risk Gene Expression Signatures in COVID-19 Brains. J Alzheimers Dis 2024; 101:31-48. [PMID: 39058446 DOI: 10.3233/jad-240609] [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] [Indexed: 07/28/2024]
Abstract
Background Various virus infections are known to predispose to Alzheimer's disease (AD), and a linkage between COVID-19 and AD has been established. COVID-19 infection modulates the gene expression of the genes implicated in progression of AD. Objective Determination of molecular patterns and codon usage and context analysis for the genes that are modulated during COVID-19 infection and are implicated in AD was the target of the study. Methods Our study employed a comprehensive array of research methods, including relative synonymous codon usage, Codon adaptation index analysis, Neutrality and parity analysis, Rare codon analyses, and codon context analysis. This meticulous approach was crucial in determining the molecular patterns present in genes up or downregulated during COVID-19 infection. Results G/C ending codons were preferred in upregulated genes while not in downregulated genes, and in both gene sets, longer genes have high expressivity. Similarly, T over A nucleotide was preferred, and selection was the major evolutionary force in shaping codon usage in both gene sets. Apart from stops codons, codons CGU - Arg, AUA - Ile, UUA - Leu, UCG - Ser, GUA - Val, and CGA - Arg in upregulated genes, while CUA - Leu, UCG - Ser, and UUA - Leu in downregulated genes were present below the 0.5%. Glutamine-initiated codon pairs have high residual values in upregulated genes. Identical codon pairs GAG-GAG and GUG-GUG were preferred in both gene sets. Conclusions The shared and unique molecular features in the up- and downregulated gene sets provide insights into the complex interplay between COVID-19 infection and AD. Further studies are required to elucidate the relationship of these molecular patterns with AD pathology.
Collapse
Affiliation(s)
- Yan Liu
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, China
| | - Weiyue Xu
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, China
| | - Pan Yang
- TEDA institute of Biological Science and Biotechnology, Nankai University, TEDA, Tianjin, China
| | - Xingshun Liu
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
16
|
Tan S, Kelty E, Page A, Etherton-Beer C, Sanfilippo F, Almeida OP. Cross-Sectional and Longitudinal Associations Between Treatment for Herpes Virus Infection and the Dispensing of Antidementia Medicines: An Analysis of the Australian Pharmaceutical Benefits Scheme Database. J Alzheimers Dis 2024; 100:791-797. [PMID: 38905050 DOI: 10.3233/jad-240391] [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] [Indexed: 06/23/2024]
Abstract
Background Evidence from previous observational studies suggest that infection by herpes simplex virus (HSV) and varicella zoster virus (VZV) increase the risk of dementia. Objective To investigate if older adults exposed to HSV treatment have lower risk of dementia than the rest of the population. Methods We used the 10% Australian Pharmaceutical Benefits Scheme (PBS) database from 2013 to 2022 to ascertain the cross-sectional, time-series and longitudinal association between exposure to HSV treatment and the dispensing of antidementia medicines. Participants were men and women aged 60 years or older. We used Anatomical Therapeutic Chemical (ATC) codes to identify medicines dispensed for the treatment of HSV and dementia. Results During the year 2022 6,868 (1.2%) of 559,561 of participants aged 60 years or over were dispensed antidementia agent. The odds ratio (OR) of being dispensed an antidementia agent among individuals dispensed treatment for HSV was 0.73 (99% CI = 0.56-0.95). Multilevel logistic regression for the 2013-2022 period for those dispensed HSV treatment was 0.87 (99% CI = 0.75-1.00). Split-time span series from 2013 was associated with hazard ratio of 0.98 (99% CI = 0.89-1.07) for individuals dispensed relative to those not dispensed HSV treatment. All analyses were adjusted for age, sex, and the dispensing of medicines for the treatment of diabetes, hyperlipidemia, hypertension, and ischemic heart disease. Conclusions The dispensing of antiviral medicines for the treatment of HSV and VZV is consistently, but not conclusively, associated with decreased dispensing of antidementia medicines. This suggests that treatment of HSV and VZV infections may contribute to reduce the risk of dementia.
Collapse
Affiliation(s)
- Stephanie Tan
- School of Population and Global Health, University of Western Australia, Perth, Australia
- Sir Charles Gairdner Hospital, Perth, Australia
| | - Erin Kelty
- School of Population and Global Health, University of Western Australia, Perth, Australia
| | - Amy Page
- School of Population and Global Health, University of Western Australia, Perth, Australia
- School of Allied Health, University of Western Australia, Perth, Australia
| | | | - Frank Sanfilippo
- School of Population and Global Health, University of Western Australia, Perth, Australia
| | - Osvaldo P Almeida
- Medical School, University of Western Australia, Perth, Australia
- Institute for Health Research, University of Notre Dame, Fremantle, Australia
| |
Collapse
|
17
|
Vestin E, Boström G, Olsson J, Elgh F, Lind L, Kilander L, Lövheim H, Weidung B. Herpes Simplex Viral Infection Doubles the Risk of Dementia in a Contemporary Cohort of Older Adults: A Prospective Study. J Alzheimers Dis 2024; 97:1841-1850. [PMID: 38306033 PMCID: PMC10894565 DOI: 10.3233/jad-230718] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2023] [Indexed: 02/03/2024]
Abstract
Background Evidence indicates that herpes simplex virus (HSV) participates in the pathogenesis of Alzheimer's disease (AD). Objective We investigated AD and dementia risks according to the presence of herpesvirus antibodies in relation to anti-herpesvirus treatment and potential APOE ɛ4 carriership interaction. Methods This study was conducted with 1002 dementia-free 70-year-olds living in Sweden in 2001-2005 who were followed for 15 years. Serum samples were analyzed to detect anti-HSV and anti-HSV-1 immunoglobulin (Ig) G, anti-cytomegalovirus (CMV) IgG, anti-HSV IgM, and anti-HSV and anti-CMV IgG levels. Diagnoses and drug prescriptions were collected from medical records. Cox proportional-hazards regression models were applied. Results Cumulative AD and all-cause dementia incidences were 4% and 7%, respectively. Eighty-two percent of participants were anti-HSV IgG carriers, of whom 6% received anti-herpesvirus treatment. Anti-HSV IgG was associated with a more than doubled dementia risk (fully adjusted hazard ratio = 2.26, p = 0.031). No significant association was found with AD, but the hazard ratio was of the same magnitude as for dementia. Anti-HSV IgM and anti-CMV IgG prevalence, anti-herpesvirus treatment, and anti-HSV and -CMV IgG levels were not associated with AD or dementia, nor were interactions between anti-HSV IgG and APOE ɛ4 or anti-CMV IgG. Similar results were obtained for HSV-1. Conclusions HSV (but not CMV) infection may be indicative of doubled dementia risk. The low AD incidence in this cohort may have impaired the statistical power to detect associations with AD.
Collapse
Affiliation(s)
- Erika Vestin
- Department of Public Health and Caring Sciences, Clinical Geriatrics, Uppsala University, Uppsala, Sweden
| | - Gustaf Boström
- Department of Public Health and Caring Sciences, Clinical Geriatrics, Uppsala University, Uppsala, Sweden
- Centre for Clinical Research, Västmanland and County Hospital, Uppsala University, Västerås, Sweden
| | - Jan Olsson
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden
| | - Fredrik Elgh
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden
| | - Lars Lind
- Department of Medical Sciences, Acute and Internal Medicine, Uppsala University, Uppsala, Sweden
| | - Lena Kilander
- Department of Public Health and Caring Sciences, Clinical Geriatrics, Uppsala University, Uppsala, Sweden
| | - Hugo Lövheim
- Department of Community Medicine and Rehabilitation, Geriatric Medicine, Umeå University, Umeå, Sweden
| | - Bodil Weidung
- Department of Public Health and Caring Sciences, Clinical Geriatrics, Uppsala University, Uppsala, Sweden
| |
Collapse
|
18
|
Espay AJ, Herrup K, Kepp KP, Daly T. The proteinopenia hypothesis: Loss of Aβ 42 and the onset of Alzheimer's Disease. Ageing Res Rev 2023; 92:102112. [PMID: 38270185 DOI: 10.1016/j.arr.2023.102112] [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] [Revised: 10/17/2023] [Accepted: 10/27/2023] [Indexed: 01/26/2024]
Abstract
The dominant protein-lowering strategy in Alzheimer's Disease (AD) has failed to provide a clinically-meaningful treatment for patients. We hypothesize that the loss of functional, soluble Aβ42 during the process of aggregation into amyloid is more detrimental to the brain than the corresponding accrual of insoluble amyloid.
Collapse
Affiliation(s)
- Alberto J Espay
- James J. and Joan A. Gardner Family Center for Parkinson's Disease and Movement Disorders, Department of Neurology, University of Cincinnati, Cincinnati, OH, USA.
| | - Karl Herrup
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kasper P Kepp
- Department of Chemistry, Section of Biophysical and Biomedicinal Chemistry, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Timothy Daly
- Science Norms Democracy, UMR 8011 Sorbonne University, Paris, France; Bioethics Program, FLACSO Argentina, Tucumán 1966, C1050 AAN, Buenos Aires, Argentina.
| |
Collapse
|
19
|
de Souza Carneiro VC, Leon LAA, de Paula VS. miRNAs: Targets to Investigate Herpesvirus Infection Associated with Neurological Disorders. Int J Mol Sci 2023; 24:15876. [PMID: 37958855 PMCID: PMC10650863 DOI: 10.3390/ijms242115876] [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: 10/09/2023] [Revised: 10/30/2023] [Accepted: 10/31/2023] [Indexed: 11/15/2023] Open
Abstract
Herpesvirus is associated with various neurological disorders and a specific diagnosis is associated with a better prognosis. MicroRNAs (miRNAs) are potential diagnostic and prognostic biomarkers of neurological diseases triggered by herpetic infection. In this review, we discuss miRNAs that have been associated with neurological disorders related to the action of herpesviruses. Human miRNAs and herpesvirus-encoded miRNAs were listed and discussed. This review article will be valuable in stimulating the search for new diagnostic and prognosis alternatives and understanding the role of these miRNAs in neurological diseases triggered by herpesviruses.
Collapse
Affiliation(s)
- Vanessa Cristine de Souza Carneiro
- Laboratory of Molecular Virology and Parasitology, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro 21040-360, Brazil; (V.C.d.S.C.); (V.S.d.P.)
- Laboratory of Technological Development in Virology, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro 21040-360, Brazil
| | - Luciane Almeida Amado Leon
- Laboratory of Technological Development in Virology, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro 21040-360, Brazil
| | - Vanessa Salete de Paula
- Laboratory of Molecular Virology and Parasitology, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro 21040-360, Brazil; (V.C.d.S.C.); (V.S.d.P.)
| |
Collapse
|
20
|
Bruno F, Abondio P, Bruno R, Ceraudo L, Paparazzo E, Citrigno L, Luiselli D, Bruni AC, Passarino G, Colao R, Maletta R, Montesanto A. Alzheimer's disease as a viral disease: Revisiting the infectious hypothesis. Ageing Res Rev 2023; 91:102068. [PMID: 37704050 DOI: 10.1016/j.arr.2023.102068] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/05/2023] [Accepted: 09/07/2023] [Indexed: 09/15/2023]
Abstract
Alzheimer's disease (AD) represents the most frequent type of dementia in elderly people. Two major forms of the disease exist: sporadic - the causes of which have not yet been fully understood - and familial - inherited within families from generation to generation, with a clear autosomal dominant transmission of mutations in Presenilin 1 (PSEN1), 2 (PSEN2) or Amyloid Precursors Protein (APP) genes. The main hallmark of AD consists of extracellular deposits of amyloid-beta (Aβ) peptide and intracellular deposits of the hyperphosphorylated form of the tau protein. An ever-growing body of research supports the viral infectious hypothesis of sporadic forms of AD. In particular, it has been shown that several herpes viruses (i.e., HHV-1, HHV-2, HHV-3 or varicella zoster virus, HHV-4 or Epstein Barr virus, HHV-5 or cytomegalovirus, HHV-6A and B, HHV-7), flaviviruses (i.e., Zika virus, Dengue fever virus, Japanese encephalitis virus) as well as Human Immunodeficiency Virus (HIV), hepatitis viruses (HAV, HBV, HCV, HDV, HEV), SARS-CoV2, Ljungan virus (LV), Influenza A virus and Borna disease virus, could increase the risk of AD. Here, we summarized and discussed these results. Based on these findings, significant issues for future studies are also put forward.
Collapse
Affiliation(s)
- Francesco Bruno
- Regional Neurogenetic Centre (CRN), Department of Primary Care, Azienda Sanitaria Provinciale Di Catanzaro, Viale A. Perugini, 88046 Lamezia Terme, CZ, Italy; Association for Neurogenetic Research (ARN), Lamezia Terme, CZ, Italy
| | - Paolo Abondio
- Laboratory of Ancient DNA, Department of Cultural Heritage, University of Bologna, Via degli Ariani 1, 48121 Ravenna, Italy.
| | - Rossella Bruno
- Sudent at the Department of Medical and Surgical Sciences, Magna Graecia University of Catanzaro, 88050 Catanzaro, Italy
| | - Leognano Ceraudo
- Sudent at the Department of Medical and Surgical Sciences, University of Parma, 43121 Parma, Italy
| | - Ersilia Paparazzo
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende 87036, Italy
| | - Luigi Citrigno
- National Research Council (CNR) - Institute for Biomedical Research and Innovation - (IRIB), 87050 Mangone, Cosenza, Italy
| | - Donata Luiselli
- Laboratory of Ancient DNA, Department of Cultural Heritage, University of Bologna, Via degli Ariani 1, 48121 Ravenna, Italy
| | - Amalia C Bruni
- Regional Neurogenetic Centre (CRN), Department of Primary Care, Azienda Sanitaria Provinciale Di Catanzaro, Viale A. Perugini, 88046 Lamezia Terme, CZ, Italy; Association for Neurogenetic Research (ARN), Lamezia Terme, CZ, Italy
| | - Giuseppe Passarino
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende 87036, Italy
| | - Rosanna Colao
- Regional Neurogenetic Centre (CRN), Department of Primary Care, Azienda Sanitaria Provinciale Di Catanzaro, Viale A. Perugini, 88046 Lamezia Terme, CZ, Italy
| | - Raffaele Maletta
- Regional Neurogenetic Centre (CRN), Department of Primary Care, Azienda Sanitaria Provinciale Di Catanzaro, Viale A. Perugini, 88046 Lamezia Terme, CZ, Italy; Association for Neurogenetic Research (ARN), Lamezia Terme, CZ, Italy
| | - Alberto Montesanto
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende 87036, Italy.
| |
Collapse
|
21
|
Zhou X, Kumar P, Bhuyan DJ, Jensen SO, Roberts TL, Münch GW. Neuroinflammation in Alzheimer's Disease: A Potential Role of Nose-Picking in Pathogen Entry via the Olfactory System? Biomolecules 2023; 13:1568. [PMID: 38002250 PMCID: PMC10669446 DOI: 10.3390/biom13111568] [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: 09/13/2023] [Revised: 10/22/2023] [Accepted: 10/23/2023] [Indexed: 11/26/2023] Open
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disorder characterized by progressive cognitive decline and memory impairment. Many possible factors might contribute to the development of AD, including amyloid peptide and tau deposition, but more recent evidence suggests that neuroinflammation may also play an-at least partial-role in its pathogenesis. In recent years, emerging research has explored the possible involvement of external, invading pathogens in starting or accelerating the neuroinflammatory processes in AD. In this narrative review, we advance the hypothesis that neuroinflammation in AD might be partially caused by viral, bacterial, and fungal pathogens entering the brain through the nose and the olfactory system. The olfactory system represents a plausible route for pathogen entry, given its direct anatomical connection to the brain and its involvement in the early stages of AD. We discuss the potential mechanisms through which pathogens may exploit the olfactory pathway to initiate neuroinflammation, one of them being accidental exposure of the olfactory mucosa to hands contaminated with soil and feces when picking one's nose.
Collapse
Affiliation(s)
- Xian Zhou
- NICM Health Research Institute, Western Sydney University, Westmead, NSW 2145, Australia; (X.Z.); (D.J.B.)
| | - Paayal Kumar
- Pharmacology Unit, School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia;
| | - Deep J. Bhuyan
- NICM Health Research Institute, Western Sydney University, Westmead, NSW 2145, Australia; (X.Z.); (D.J.B.)
| | - Slade O. Jensen
- Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia; (S.O.J.); (T.L.R.)
- Microbiology and Infectious Diseases Unit, School of Medicine, Western Sydney University, Liverpool, NSW 2170, Australia
| | - Tara L. Roberts
- Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia; (S.O.J.); (T.L.R.)
- Oncology Unit, School of Medicine, Western Sydney University, Liverpool, NSW 2170, Australia
| | - Gerald W. Münch
- NICM Health Research Institute, Western Sydney University, Westmead, NSW 2145, Australia; (X.Z.); (D.J.B.)
- Pharmacology Unit, School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia;
| |
Collapse
|
22
|
Ghorbani M, Ferreira D, Maioli S. A metagenomic study of gut viral markers in amyloid-positive Alzheimer's disease patients. Alzheimers Res Ther 2023; 15:141. [PMID: 37608325 PMCID: PMC10464408 DOI: 10.1186/s13195-023-01285-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 08/04/2023] [Indexed: 08/24/2023]
Abstract
BACKGROUND Mounting evidence suggests the involvement of viruses in the development and treatment of Alzheimer's disease (AD). However, there remains a significant research gap in metagenomic studies investigating the gut virome of AD patients, leaving gut viral dysbiosis in AD unexplored. This study aimed to fill this gap by conducting a metagenomics analysis of the gut virome in both amyloid-positive AD patients (Aβ + ADs) and healthy controls (HCs), with the objective of identifying viral signatures linked with AD. METHOD Whole-genome sequence (WGS) data from 65 human participants, including 30 Aβ + ADs and 35 HCs, was obtained from the database NCBI SRA (Bio Project: PRJEB47976). The Metaphlan3 pipeline and linear discriminant analysis effect size (LEfSe) analysis were utilized for the bioinformatics process and the detection of viral signatures, respectively. In addition, the Benjamini-Hochberg method was applied with a significance cutoff of 0.05 to evaluate the false discovery rate for all biomarkers identified by LEfSe. The CombiROC model was employed to determine the discriminatory power of the viral signatures identified by LEfSe. RESULTS Compared to HCs, the gut virome profiles of Aβ + ADs showed lower alpha diversity, indicating a lower bacteriophage richness. The Siphoviridae family was decreased in Aβ + ADs. Significant decreases of Lactococcus phages were found in Aβ + ADs, including bIL285, Lactococcus phage bIL286, Lactococcus phage bIL309, and Lactococcus phage BK5 T, Lactococcus phage BM13, Lactococcus phage P335 sensu lato, Lactococcus phage phiLC3, Lactococcus phage r1t, Lactococcus phage Tuc2009, Lactococcus phage ul36, and Lactococcus virus bIL67. The predictive combined model of these viral signatures obtained an area under the curve of 0.958 when discriminating Aβ + ADs from HCs. CONCLUSION This is the first study to identify distinct viral signatures in the intestine that can be used to effectively distinguish individuals with AD from HCs.
Collapse
Affiliation(s)
- Mahin Ghorbani
- Department of Dental Medicine, Karolinska Institute, Stockholm, Sweden.
| | - Daniel Ferreira
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Facultad de Ciencias de la Salud, Universidad Fernando Pessoa Canarias, Las Palmas, España
| | - Silvia Maioli
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
23
|
Yan H, Wang J, Yang J, Xu Z, Li C, Hao C, Wang S, Wang W. Inhibition Effects and Mechanisms of Marine Polysaccharide PSSD against Herpes Simplex Virus Type 2. Mar Drugs 2023; 21:364. [PMID: 37367689 DOI: 10.3390/md21060364] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 06/14/2023] [Accepted: 06/16/2023] [Indexed: 06/28/2023] Open
Abstract
Genital herpes is a common sexually transmitted disease mainly caused by herpes simplex virus type 2 (HSV-2), which can increase the risk of HIV transmission and is a major health problem in the world. Thus, it is of great significance to develop new anti-HSV-2 drugs with high efficiency and low toxicity. In this study, the anti-HSV-2 activities of PSSD, a marine sulfated polysaccharide, was deeply explored both in vitro and in vivo. The results showed that PSSD had marked anti-HSV-2 activities in vitro with low cytotoxicity. PSSD can directly interact with virus particles to inhibit the adsorption of virus to the cell surface. PSSD may also interact with virus surface glycoproteins to block virus-induced membrane fusion. Importantly, PSSD can significantly attenuate the symptoms of genital herpes and weight loss in mice after gel smear treatment, as well as reducing the titer of virus shedding in the reproductive tract of mice, superior to the effect of acyclovir. In summary, the marine polysaccharide PSSD possesses anti-HSV-2 effects both in vitro and in vivo, and has potential to be developed into a novel anti-genital herpes agent in the future.
Collapse
Affiliation(s)
- Han Yan
- Key Laboratory of Marine Drugs, Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Jie Wang
- Key Laboratory of Marine Drugs, Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Jiayi Yang
- Key Laboratory of Marine Drugs, Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Zhongqiu Xu
- Key Laboratory of Marine Drugs, Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
- Medical Research Center, Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Chunxia Li
- Key Laboratory of Marine Drugs, Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
- The Laboratory of Marine Glycodrug Research and Development, Marine Biomedical Research Institute of Qingdao, Qingdao 266100, China
| | - Cui Hao
- Medical Research Center, Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Shixin Wang
- Key Laboratory of Marine Drugs, Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
- The Laboratory of Marine Glycodrug Research and Development, Marine Biomedical Research Institute of Qingdao, Qingdao 266100, China
| | - Wei Wang
- Key Laboratory of Marine Drugs, Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| |
Collapse
|
24
|
Al-Kuraishy HM, Al-Gareeb AI, Kaushik A, Kujawska M, Ahmed EA, Batiha GES. SARS-COV-2 infection and Parkinson's disease: Possible links and perspectives. J Neurosci Res 2023; 101:952-975. [PMID: 36717481 DOI: 10.1002/jnr.25171] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 01/13/2023] [Accepted: 01/15/2023] [Indexed: 02/01/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by the progressive loss of dopaminergic neurons in the substantia nigra. The hallmarks are the presence of Lewy bodies composed mainly of aggregated α-synuclein and immune activation and inflammation in the brain. The neurotropism of SARS-CoV-2 with induction of cytokine storm and neuroinflammation can contribute to the development of PD. Interestingly, overexpression of α-synuclein in PD patients may limit SARS-CoV-2 neuroinvasion and degeneration of dopaminergic neurons; however, on the other hand, this virus can speed up the α-synuclein aggregation. The review aims to discuss the potential link between COVID-19 and the risk of PD, highlighting the need for further studies to authenticate the potential association. We have also overviewed the influence of SARS-CoV-2 infection on the PD course and management. In this context, we presented the prospects for controlling the COVID-19 pandemic and related PD cases that, beyond global vaccination and novel anti-SARS-CoV-2 agents, may include the development of graphene-based nanoscale platforms offering antiviral and anti-amyloid strategies against PD.
Collapse
Affiliation(s)
- Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriyia University, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriyia University, Baghdad, Iraq
| | - Ajeet Kaushik
- NanoBioTech Laboratory, Department of Environmental Engineering, Florida Polytechnic University, Lakeland, Florida, USA
| | - Małgorzata Kujawska
- Department of Toxicology, Poznan University of Medical Sciences, Poznan, Poland
| | - Eman A Ahmed
- Department of Pharmacology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt
| |
Collapse
|
25
|
Salgado B, Sastre I, Bullido MJ, Aldudo J. Herpes Simplex Virus Type 1 Induces AD-like Neurodegeneration Markers in Human Progenitor and Differentiated ReNcell VM Cells. Microorganisms 2023; 11:1205. [PMID: 37317179 DOI: 10.3390/microorganisms11051205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 04/26/2023] [Accepted: 04/27/2023] [Indexed: 06/16/2023] Open
Abstract
An increasing body of evidence strongly suggests that infections or reactivations of herpes simplex virus type 1 (HSV-1) may be closely linked to Alzheimer's disease (AD). Promising results have been obtained using cell and animal models of HSV-1 infection, contributing to the understanding of the molecular mechanisms linking HSV-1 infection and AD neurodegeneration. ReNcell VM is a human neural stem cell line that has been used as a model system to study the impact of various infectious agents on the central nervous system. In this study, we demonstrate the suitability of the ReNcell VM cell line for developing a new in vitro model of HSV-1 infection. By following standard differentiation protocols, we were able to derive various nervous cell types, including neurons, astrocytes, and oligodendrocytes, from neural precursors. Additionally, we demonstrated the susceptibility of ReNcell VM cells, including precursor and differentiated cells, to HSV-1 infection and subsequent viral-induced AD-like neurodegeneration. Our findings support the use of this cell line to generate a new research platform for investigating AD neuropathology and its most significant risk factors, which may lead to important discoveries in the context of this highly impactful disease.
Collapse
Affiliation(s)
- Blanca Salgado
- Centro de Biologia Molecular "Severo Ochoa" (C.S.I.C.-U.A.M.), Universidad Autonoma de Madrid, 28049 Madrid, Spain
- Centro de Investigacion Biomedica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28031 Madrid, Spain
| | - Isabel Sastre
- Centro de Biologia Molecular "Severo Ochoa" (C.S.I.C.-U.A.M.), Universidad Autonoma de Madrid, 28049 Madrid, Spain
- Centro de Investigacion Biomedica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28031 Madrid, Spain
| | - Maria J Bullido
- Centro de Biologia Molecular "Severo Ochoa" (C.S.I.C.-U.A.M.), Universidad Autonoma de Madrid, 28049 Madrid, Spain
- Centro de Investigacion Biomedica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28031 Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Universitario La Paz-IdiPAZ, 28046 Madrid, Spain
| | - Jesus Aldudo
- Centro de Biologia Molecular "Severo Ochoa" (C.S.I.C.-U.A.M.), Universidad Autonoma de Madrid, 28049 Madrid, Spain
- Centro de Investigacion Biomedica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28031 Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Universitario La Paz-IdiPAZ, 28046 Madrid, Spain
| |
Collapse
|
26
|
Bye LJ, Finol-Urdaneta RK, Tae HS, Adams DJ. Nicotinic acetylcholine receptors: Key targets for attenuating neurodegenerative diseases. Int J Biochem Cell Biol 2023; 157:106387. [PMID: 36754161 DOI: 10.1016/j.biocel.2023.106387] [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: 12/18/2022] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 02/08/2023]
Abstract
Nicotinic acetylcholine receptors (nAChRs) are master regulators of immune functions via the cholinergic anti-inflammatory pathway and are expressed in microglia, the brain's resident immune cells. There is an extensive dialogue between the neurons and the glial cells around them from which microglia are tasked with monitoring, nurturing, and defending their microenvironment. Dysregulation of any of these processes can have devastating and long-lasting consequences involving microglia-mediated neuroinflammation associated with neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and Huntington's disease, amongst others. Disease-associated microglia acquire a distinguishing phenotype that emphasizes scavenging and defence functions while nurturing and repairing functions become muted. Attempts to resolve this critical imbalance remain a key focus of research. Furthermore, cholinergic modulation of neuroinflammation represents a promising avenue for treatment.
Collapse
Affiliation(s)
- Lydia J Bye
- Illawarra Health and Medical Research Institute (IHMRI), Faculty of Science, Medicine and Health, University of Wollongong, NSW 2522 Australia
| | - Rocio K Finol-Urdaneta
- Illawarra Health and Medical Research Institute (IHMRI), Faculty of Science, Medicine and Health, University of Wollongong, NSW 2522 Australia
| | - Han-Shen Tae
- Illawarra Health and Medical Research Institute (IHMRI), Faculty of Science, Medicine and Health, University of Wollongong, NSW 2522 Australia
| | - David J Adams
- Illawarra Health and Medical Research Institute (IHMRI), Faculty of Science, Medicine and Health, University of Wollongong, NSW 2522 Australia.
| |
Collapse
|
27
|
Ketogenic diet restrains herpes simplex encephalitis via gut microbes. Microbes Infect 2023; 25:105061. [PMID: 36270600 DOI: 10.1016/j.micinf.2022.105061] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 10/14/2022] [Accepted: 10/14/2022] [Indexed: 11/05/2022]
Abstract
Herpes simplex virus type 1 (HSV-1) infection-associated herpes simplex encephalitis (HSE) is an occasionally but severe neuronal disease that causes behavioral disorder and impairs cognition. Herein, we demonstrate that the consumption of ketogenic diet (KD), a low-carbohydrate high-fat diet, restricts the neurotropic infection of HSV-1 and HSE progression in mice. KD reduced weight loss, neurodegenerative symptoms, virus production and neuroinflammation, resulting in the enhanced survival rate of HSE mice. Notably, depletion of gut microbes by antibiotics attenuated the protective function of KD on HSV-1-related neuroinflammation and HSE development. Therefore, KD represents as an alternative therapeutic strategy to alleviate or prevent HSE via gut microbiota.
Collapse
|
28
|
Harrell TL, Davido DJ, Bertke AS. Herpes Simplex Virus 1 (HSV-1) Infected Cell Protein 0 (ICP0) Targets of Ubiquitination during Productive Infection of Primary Adult Sensory Neurons. Int J Mol Sci 2023; 24:2931. [PMID: 36769256 PMCID: PMC9917815 DOI: 10.3390/ijms24032931] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/18/2023] [Accepted: 01/26/2023] [Indexed: 02/05/2023] Open
Abstract
Herpes simplex virus 1 (HSV-1) enters sensory neurons with the potential for productive or latent infection. For either outcome, HSV-1 must curtail the intrinsic immune response, regulate viral gene expression, and remove host proteins that could restrict viral processes. Infected cell protein 0 (ICP0), a virus-encoded E3 ubiquitin ligase, supports these processes by mediating the transfer of ubiquitin to target proteins to change their location, alter their function, or induce their degradation. To identify ubiquitination targets of ICP0 during productive infection in sensory neurons, we immunoprecipitated ubiquitinated proteins from primary adult sensory neurons infected with HSV-1 KOS (wild-type), HSV-1 n212 (expressing truncated, defective ICP0), and uninfected controls using anti-ubiquitin antibody FK2 (recognizing K29, K48, K63 and monoubiquitinated proteins), followed by LC-MS/MS and comparative analyses. We identified 40 unique proteins ubiquitinated by ICP0 and 17 ubiquitinated by both ICP0 and host mechanisms, of which High Mobility Group Protein I/Y (HMG I/Y) and TAR DNA Binding Protein 43 (TDP43) were selected for further analysis. We show that ICP0 ubiquitinates HMG I/Y and TDP43, altering protein expression at specific time points during productive HSV-1 infection, demonstrating that ICP0 manipulates the sensory neuronal environment in a time-dependent manner to regulate infection outcome in neurons.
Collapse
Affiliation(s)
- Telvin L. Harrell
- Biomedical and Veterinary Science, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA
| | - David J. Davido
- Molecular Biosciences, University of Kansas, Lawrence, KS 66045, USA
| | - Andrea S. Bertke
- Population Health Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA
- Center for Emerging Zoonotic and Arthropod-Borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA
| |
Collapse
|
29
|
Anwar MM. The emerging mechanism behind viral infections and extracellular vesicles hypotheses leading to neuroinflammation and Alzheimer's disease pathology. IBRAIN 2023; 9:63-71. [PMID: 37786515 PMCID: PMC10529198 DOI: 10.1002/ibra.12090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 01/14/2023] [Accepted: 01/14/2023] [Indexed: 10/04/2023]
Abstract
Despite decades of repeated and intense research, the etiology of sudden Alzheimer's disease (AD) symptoms is still unclear. AD progressive pathology mainly involves neuron damage, depositions of amyloid-beta (Aβ), and hyperphosphorylated tau protein. All these defects are manifested by exaggerated cytokine storm and neuroinflammation leading to irreversible brain damage in the long term. Despite the numerous risks and drawbacks associated with AD, it is believed that there is a hidden unknown causative and predisposing factors for AD. Extracellular vesicles (EVs) are small vesicles released by cells as a type of intercellular communication. Several pieces of evidence support the inclusion of viral components within EVs facilitating their penetration into the blood-brain barrier leading to neuroinflammation. In light of the SARS-CoV-19 pandemic and its related neurological complications, it is mandatory to highlight the possibility and viability of viral infections such as varicella-zoster virus (VZV) and herpes simplex virus (HSV) on the onset of AD. Herein, the author is investigating the potential role of VZV and HSV along with highlighting the suggested route of pathogenesis entry resulting in AD manifestations. Additionally, this review aims to summarize the role of EVs in mediating the central nervous system viral infections leading to AD.
Collapse
Affiliation(s)
- Mai M. Anwar
- Department of BiochemistryNational Organization for Drug Control and Research (NODCAR)/Egyptian Drug Authority (EDA)CairoEgypt
| |
Collapse
|
30
|
Osaka R, Kobayashi N, Shimada K, Ishii A, Oka N, Kondo K. VP26, a herpes simplex virus type 1 capsid protein, increases DNA methylation in COASY promoter region. Brain Behav Immun Health 2022; 26:100545. [PMID: 36345321 PMCID: PMC9636445 DOI: 10.1016/j.bbih.2022.100545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 10/20/2022] [Accepted: 10/23/2022] [Indexed: 11/06/2022] Open
Abstract
It has been reported that some specific changes in DNA methylation can be due to aging or infection by tumor-related viruses but the effect of herpes simplex virus type 1 (HSV-1) in this regard is unknown. HSV-1 is a well-known virus that causes cold sores. After the primary infection, the virus switches to latent infection and remains in the body for the whole life. As the location of DNA methylation, we focused on the promoter region of the COASY gene, which codes for coenzyme A synthase, because methylation in this region is reportedly associated with Alzheimer's disease (AD). During HSV-1 lytic infection, compared to non-infected cells, COASY DNA methylation decreased but when HSV-1 replication was inhibited by acyclovir, an anti-herpes agent, COASY DNA methylation increased. In addition, for expression of immediate early protein only, there was no significant change in COASY DNA methylation, while for expression of the capsid protein VP26, a late protein known to bind with DNA methyltransferase DNMT3A, in the nucleus only, COASY DNA methylation significantly increased compared to the control, without changes in DNMT3A mRNA. Our results suggested that DNA methylation occurred not due to transcriptional changes in DNMT3A but through translational regulation. In this research, we showed that host COASY DNA methylation is altered by HSV-1 infection, in particular by HSV-1 VP26. It is a potential cause of various diseases, and this is particularly relevant for AD.
Collapse
Affiliation(s)
- Rui Osaka
- Department of Virology, The Jikei University School of Medicine, Tokyo, Japan
| | - Nobuyuki Kobayashi
- Department of Virology, The Jikei University School of Medicine, Tokyo, Japan
| | - Kazuya Shimada
- Department of Virology, The Jikei University School of Medicine, Tokyo, Japan
| | - Azusa Ishii
- Department of Virology, The Jikei University School of Medicine, Tokyo, Japan
| | - Naomi Oka
- Department of Virology, The Jikei University School of Medicine, Tokyo, Japan
| | - Kazuhiro Kondo
- Department of Virology, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
31
|
Carneiro VCDS, Pereira JG, de Paula VS. Family Herpesviridae and neuroinfections: current status and research in progress. Mem Inst Oswaldo Cruz 2022; 117:e220200. [PMID: 36417627 PMCID: PMC9677594 DOI: 10.1590/0074-02760220200] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 10/10/2022] [Indexed: 11/22/2022] Open
Abstract
This article addresses the relationship between human herpesviruses (HHVs) and neuroinfections. Alphaherpesviruses, betaherpesviruses and gammaherpesviruses are neurotropic viruses that establish latency and exhibit reactivation capacity. Encephalitis and meningitis are common in cases of HHV. The condition promoted by HHV infection is a purported trigger for certain neurodegenerative diseases. Ongoing studies have identified an association between HSV-1 and the occurrence of Alzheimer's disease, multiple sclerosis and infections by HHV-6 and Epstein-Barr virus. In this review, we highlight the importance of research investigating the role of herpesviruses in the pathogenesis of diseases that affect the nervous system and describe other studies in progress.
Collapse
Affiliation(s)
| | | | - Vanessa Salete de Paula
- Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Laboratório de Virologia Molecular, Rio de Janeiro, RJ, Brasil,+ Corresponding author:
| |
Collapse
|
32
|
Trunfio M, Di Girolamo L, Ponzetta L, Russo M, Burdino E, Imperiale D, Atzori C, Di Perri G, Calcagno A. Seropositivity and reactivations of HSV-1, but not of HSV-2 nor VZV, associate with altered blood–brain barrier, beta amyloid, and tau proteins in people living with HIV. J Neurovirol 2022; 29:100-105. [PMID: 36352195 DOI: 10.1007/s13365-022-01105-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/20/2022] [Accepted: 10/27/2022] [Indexed: 11/11/2022]
Abstract
Among 128 adult people living with HIV and no neurological conditions confounding the cerebrospinal fluid results, the presence of HSV-1 chronic infection (detected either by serology or PCR), but not of HSV-2 and VZV, independently associated with higher odds of blood-brain barrier impairment, abnormally increased cerebrospinal fluid levels of tau and phosphorylated-181 tau, and decreased concentrations of fragments 1-42 of beta amyloid compared to the seronegative counterpart. These associations were even stronger for seropositive participants with a positive history of at least one symptomatic reactivation of HSV-1.
Collapse
|
33
|
Janicka M, Ranoszek-Soliwoda K, Chodaczek G, Antos-Bielska M, Brytan M, Tomaszewska E, Celichowski G, Grobelny J, Cymerys J, Krzyżowska M, Chodkowski M. Functionalized Noble Metal Nanoparticles for the Treatment of Herpesvirus Infection. Microorganisms 2022; 10:microorganisms10112161. [PMID: 36363754 PMCID: PMC9695377 DOI: 10.3390/microorganisms10112161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 10/14/2022] [Accepted: 10/26/2022] [Indexed: 11/06/2022] Open
Abstract
Neuroinfections caused by herpesviruses, mainly by HHV-1, represent a significant problem for modern medicine due to the small number of therapeutic substances available in the pharmaceutical sector. Furthermore, HHV-1 infection has been linked to neurodegenerative processes such as Alzheimer’s disease, which justifies the search for new effective therapies. The development of nanotechnology opens up new possibilities for the treatment of neuroinflammation. Gold and silver nanoparticles are gaining popularity, and the number of clinical trials involving metallic nanoparticles is constantly increasing. This paper reviews the research on gold and silver nanoparticles and their potential use in the treatment of herpesvirus neuroinfection.
Collapse
Affiliation(s)
- Martyna Janicka
- Military Institute of Hygiene and Epidemiology, Kozielska 4, 01-163 Warsaw, Poland
- Division of Microbiology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-786 Warsaw, Poland
| | - Katarzyna Ranoszek-Soliwoda
- Department of Materials Technology and Chemistry, Faculty of Chemistry, University of Lodz, Pomorska 163 St., 90-236 Lodz, Poland
| | - Grzegorz Chodaczek
- Bioimaging Laboratory, Łukasiewicz Research Network—PORT Polish Center for Technology Development, 54-066 Wroclaw, Poland
| | | | - Marek Brytan
- Military Institute of Hygiene and Epidemiology, Kozielska 4, 01-163 Warsaw, Poland
| | - Emilia Tomaszewska
- Department of Materials Technology and Chemistry, Faculty of Chemistry, University of Lodz, Pomorska 163 St., 90-236 Lodz, Poland
| | - Grzegorz Celichowski
- Department of Materials Technology and Chemistry, Faculty of Chemistry, University of Lodz, Pomorska 163 St., 90-236 Lodz, Poland
| | - Jarosław Grobelny
- Department of Materials Technology and Chemistry, Faculty of Chemistry, University of Lodz, Pomorska 163 St., 90-236 Lodz, Poland
| | - Joanna Cymerys
- Division of Microbiology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-786 Warsaw, Poland
| | - Małgorzata Krzyżowska
- Military Institute of Hygiene and Epidemiology, Kozielska 4, 01-163 Warsaw, Poland
- Correspondence: (M.K.); (M.C.)
| | - Marcin Chodkowski
- Military Institute of Hygiene and Epidemiology, Kozielska 4, 01-163 Warsaw, Poland
- Correspondence: (M.K.); (M.C.)
| |
Collapse
|
34
|
Gompels UA, Bravo FJ, Briggs S, Ameri S, Cardin RD, Bernstein DI. Immunisation Using Novel DNA Vaccine Encoding Virus Membrane Fusion Complex and Chemokine Genes Shows High Protection from HSV-2. Viruses 2022; 14:v14112317. [PMID: 36366414 PMCID: PMC9698128 DOI: 10.3390/v14112317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/18/2022] [Accepted: 10/20/2022] [Indexed: 11/07/2022] Open
Abstract
Herpes simplex virus 1 and 2 infections cause high unmet disease burdens worldwide. Mainly HSV-2 causes persistent sexually transmitted disease, fatal neonatal disease and increased transmission of HIV/AIDS. Thus, there is an urgent requirement to develop effective vaccines. We developed nucleic acid vaccines encoding a novel virus entry complex stabilising cell membrane fusion, 'virus-like membranes', VLM. Two dose intramuscular immunisations using DNA expression plasmids in a guinea pig model gave 100% protection against acute disease and significantly reduced virus replication after virus intravaginal challenge. There was also reduced establishment of latency within the dorsal root ganglia and spinal cord, but recurrent disease and recurrent virus shedding remained. To increase cellular immunity and protect against recurrent disease, cDNA encoding an inhibitor of chemokine receptors on T regulatory cells was added and compared to chemokine CCL5 effects. Immunisation including this novel human chemokine gene, newly defined splice variant from an endogenous virus genome, 'virokine immune therapeutic', VIT, protected most guinea pigs from recurrent disease and reduced recurrent virus shedding distinct from a gD protein vaccine similar to that previously evaluated in clinical trials. All DNA vaccines induced significant neutralising antibodies and warrant evaluation for new therapeutic treatments.
Collapse
Affiliation(s)
- Ursula A. Gompels
- Virothera, Milner Therapeutics Institute, Cambridge Biomedical Campus, Cambridge CB4 0WS, UK
- Correspondence:
| | - Fernando J. Bravo
- Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Sean Briggs
- Virothera, Milner Therapeutics Institute, Cambridge Biomedical Campus, Cambridge CB4 0WS, UK
| | - Shima Ameri
- Virothera, Milner Therapeutics Institute, Cambridge Biomedical Campus, Cambridge CB4 0WS, UK
| | - Rhonda D. Cardin
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - David I. Bernstein
- Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45221, USA
| |
Collapse
|
35
|
Implications of Microorganisms in Alzheimer's Disease. Curr Issues Mol Biol 2022; 44:4584-4615. [PMID: 36286029 PMCID: PMC9600878 DOI: 10.3390/cimb44100314] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 09/23/2022] [Accepted: 09/28/2022] [Indexed: 11/20/2022] Open
Abstract
Alzheimer’s disease (AD) is a deadly brain degenerative disorder that leads to brain shrinkage and dementia. AD is manifested with hyperphosphorylated tau protein levels and amyloid beta (Aβ) peptide buildup in the hippocampus and cortex regions of the brain. The nervous tissue of AD patients also contains fungal proteins and DNA which are linked to bacterial infections, suggesting that polymicrobial infections also occur in the brains of those with AD. Both immunohistochemistry and next-generation sequencing (NGS) techniques were employed to assess fungal and bacterial infections in the brain tissue of AD patients and non-AD controls, with the most prevalent fungus genera detected in AD patients being Alternaria, Botrytis, Candida, and Malassezia. Interestingly, Fusarium was the most common genus detected in the control group. Both AD patients and controls were also detectable for Proteobacteria, followed by Firmicutes, Actinobacteria, and Bacteroides for bacterial infection. At the family level, Burkholderiaceae and Staphylococcaceae exhibited higher levels in the brains of those with AD than the brains of the control group. Accordingly, there is thought to be a viscous cycle of uncontrolled neuroinflammation and neurodegeneration in the brain, caused by agents such as the herpes simplex virus type 1 (HSV1), Chlamydophilapneumonia, and Spirochetes, and the presence of apolipoprotein E4 (APOE4), which is associated with an increased proinflammatory response in the immune system. Systemic proinflammatory cytokines are produced by microorganisms such as Cytomegalovirus, Helicobacter pylori, and those related to periodontal infections. These can then cross the blood–brain barrier (BBB) and lead to the onset of dementia. Here, we reviewed the relationship between the etiology of AD and microorganisms (such as bacterial pathogens, Herpesviridae viruses, and periodontal pathogens) according to the evidence available to understand the pathogenesis of AD. These findings might guide a targeted anti-inflammatory therapeutic approach to AD.
Collapse
|
36
|
Lukiw WJ, Jaber VR, Pogue AI, Zhao Y. SARS-CoV-2 Invasion and Pathological Links to Prion Disease. Biomolecules 2022; 12:1253. [PMID: 36139092 PMCID: PMC9496025 DOI: 10.3390/biom12091253] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 08/26/2022] [Accepted: 08/30/2022] [Indexed: 12/19/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of the COVID-19 disease, is a highly infectious and transmissible viral pathogen that continues to impact human health globally. Nearly ~600 million people have been infected with SARS-CoV-2, and about half exhibit some degree of continuing health complication, generically referred to as long COVID. Lingering and often serious neurological problems for patients in the post-COVID-19 recovery period include brain fog, behavioral changes, confusion, delirium, deficits in intellect, cognition and memory issues, loss of balance and coordination, problems with vision, visual processing and hallucinations, encephalopathy, encephalitis, neurovascular or cerebrovascular insufficiency, and/or impaired consciousness. Depending upon the patient’s age at the onset of COVID-19 and other factors, up to ~35% of all elderly COVID-19 patients develop a mild-to-severe encephalopathy due to complications arising from a SARS-CoV-2-induced cytokine storm and a surge in cytokine-mediated pro-inflammatory and immune signaling. In fact, this cytokine storm syndrome: (i) appears to predispose aged COVID-19 patients to the development of other neurological complications, especially those who have experienced a more serious grade of COVID-19 infection; (ii) lies along highly interactive and pathological pathways involving SARS-CoV-2 infection that promotes the parallel development and/or intensification of progressive and often lethal neurological conditions, and (iii) is strongly associated with the symptomology, onset, and development of human prion disease (PrD) and other insidious and incurable neurological syndromes. This commentary paper will evaluate some recent peer-reviewed studies in this intriguing area of human SARS-CoV-2-associated neuropathology and will assess how chronic, viral-mediated changes to the brain and CNS contribute to cognitive decline in PrD and other progressive, age-related neurodegenerative disorders.
Collapse
Affiliation(s)
- Walter J. Lukiw
- LSU Neuroscience Center, Louisiana State University Health Science Center, New Orleans, LA 70112, USA
- Alchem Biotek Research, Toronto, ON M5S 1A8, Canada
- Department of Ophthalmology, LSU Health Science Center, New Orleans, LA 70112, USA
- Department Neurology, LSU Health Science Center, New Orleans, LA 70112, USA
| | - Vivian R. Jaber
- LSU Neuroscience Center, Louisiana State University Health Science Center, New Orleans, LA 70112, USA
| | | | - Yuhai Zhao
- LSU Neuroscience Center, Louisiana State University Health Science Center, New Orleans, LA 70112, USA
- Department of Cell Biology & Anatomy, LSU Health Science Center, New Orleans, LA 70112, USA
| |
Collapse
|
37
|
Thiele M, Donnelly SC, Mitchell RA. OxMIF: a druggable isoform of macrophage migration inhibitory factor in cancer and inflammatory diseases. J Immunother Cancer 2022; 10:e005475. [PMID: 36180072 PMCID: PMC9528626 DOI: 10.1136/jitc-2022-005475] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/12/2022] [Indexed: 11/04/2022] Open
Abstract
Macrophage migration inhibitory factor (MIF) is a proinflammatory cytokine with a pleiotropic spectrum of biological functions implicated in the pathogenesis of cancer and inflammatory diseases. MIF is constitutively present in several cell types and non-lymphoid tissues and is secreted after acute stress or inflammation. MIF triggers the release of proinflammatory cytokines, overrides the anti-inflammatory effects of glucocorticoids, and exerts chemokine function, resulting in increased migration and recruitment of leukocytes into inflamed tissue. Despite this, MIF is a challenging target for therapeutic intervention because of its ubiquitous nature and presence in the circulation and tissue of healthy individuals. Oxidized MIF (oxMIF) is an immunologically distinct disease-related structural isoform found in the plasma and tissues of patients with inflammatory diseases and in solid tumor tissues. MIF converts to oxMIF in an oxidizing, inflammatory environment. This review discusses the biology and activity of MIF and the potential for autoimmune disease and cancer modification by targeting oxMIF. Anti-oxMIF antibodies reduce cancer cell invasion/migration, angiogenesis, proinflammatory cytokine production, and ERK and AKT activation. Anti-oxMIF antibodies also elicit apoptosis and alter immune cell function and/or migration. When co-administered with a glucocorticoid, anti-oxMIF antibodies produced a synergistic response in inflammatory models. Anti-oxMIF antibodies therefore counterregulate biological activities attributed to MIF. oxMIF expression has been observed in inflammatory diseases (eg, sepsis, psoriasis, asthma, inflammatory bowel disease, and systemic lupus erythematosus) and oxMIF has been detected in ovarian, colorectal, lung, and pancreatic cancers. In contrast to MIF, oxMIF is specifically detected in plasma and/or tissues of diseased patients, but not in healthy individuals. Therefore, as a druggable isoform of MIF, oxMIF represents a potential new therapeutic target in inflammatory diseases and cancer. Fully human, monoclonal anti-oxMIF antibodies have been shown to selectively bind oxMIF in preclinical and phase I studies; however, additional clinical assessments are necessary to validate their use as either a monotherapy or in combination with standard-of-care regimens (ie, immunomodulatory agents/checkpoint inhibitors, anti-angiogenic drugs, chemotherapeutics, and glucocorticoids).
Collapse
Affiliation(s)
- Michael Thiele
- Biology Research, OncoOne Research & Development GmbH, Vienna, Austria
| | - Seamas C Donnelly
- Department of Medicine, Tallaght University Hospital & Trinity College Dublin, Dublin, Ireland
| | - Robert A Mitchell
- Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, Kentucky, USA
- Department of Surgery, J.G. Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA
- Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky, USA
- Division of Immunotherapy, Department of Surgery, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
38
|
Cheng J, Wang Q, Hu Y, Mou T, Wang J, Wang L, Zhang Y, Wang T, Li Q. Understanding global changes of the mouse brain proteome after vaginal infection with HSV-2 using a label-free shotgun approach. Front Cell Infect Microbiol 2022; 12:942334. [PMID: 36061859 PMCID: PMC9433710 DOI: 10.3389/fcimb.2022.942334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
Herpes simplex virus type 2 (HSV-2) is a common human pathogen that establishes lifelong latency in neurons of the nervous system. The number of severe central nervous system infections caused by the virus has increased recently. However, the pathogenesis of HSV-2 infection in the nervous system is not fully understood. Here, we demonstrated global proteomic changes in the brain tissue in BALB/c mice vaginally infected with HSV-2. Data are available via ProteomeXchange with identifier PXD034186. A total of 249 differentially expressed proteins were identified in infected brain tissue. The GO and KEGG enrichment analysis of these proteins indicated that they were mainly involved in the regulation of synapse formation and synaptic excitability. In addition, genes affecting autophagy, the development of other neurodegenerative diseases, and signaling pathways relevant to other neurologic diseases were identified. Additional experiments, comparing the brain tissue of asymptomatic and symptomatic mice showed a differential expression of proteins involved in synapse formation and synaptic transmission. Others were involved in autophagy, addiction, and signaling pathways of other neurologic diseases. These results suggest that changes in synaptic structure and function, as well as autophagy, may be related to the development of neurologic abnormalities that follow HSV-2 infection. We also identified a protein GluN2A encoded by Grin2a was continuously expressed at high levels after infection. We propose that GluN2A may be a key molecule in the pathogenesis of HSV-2-induced neurologic diseases.
Collapse
Affiliation(s)
- Jishuai Cheng
- Animal Zoology Department, Institute of Neuroscience, Kunming Medical University, Kunming, China
- Yunnan Key Laboratory of Vaccine Research and Development for Severe Infectious Diseases, Institute of Medical Biology, Chinese Academy of Medicine Sciences and Peking Union Medical College, Kunming, China
| | - Qingzhen Wang
- Animal Zoology Department, Institute of Neuroscience, Kunming Medical University, Kunming, China
| | - Yiwen Hu
- Animal Zoology Department, Institute of Neuroscience, Kunming Medical University, Kunming, China
| | - Tangwei Mou
- Yunnan Key Laboratory of Vaccine Research and Development for Severe Infectious Diseases, Institute of Medical Biology, Chinese Academy of Medicine Sciences and Peking Union Medical College, Kunming, China
| | - Jianbin Wang
- Yunnan Key Laboratory of Vaccine Research and Development for Severe Infectious Diseases, Institute of Medical Biology, Chinese Academy of Medicine Sciences and Peking Union Medical College, Kunming, China
| | - Lichun Wang
- Yunnan Key Laboratory of Vaccine Research and Development for Severe Infectious Diseases, Institute of Medical Biology, Chinese Academy of Medicine Sciences and Peking Union Medical College, Kunming, China
| | - Ying Zhang
- Yunnan Key Laboratory of Vaccine Research and Development for Severe Infectious Diseases, Institute of Medical Biology, Chinese Academy of Medicine Sciences and Peking Union Medical College, Kunming, China
| | - Tinghua Wang
- Animal Zoology Department, Institute of Neuroscience, Kunming Medical University, Kunming, China
| | - Qihan Li
- Yunnan Key Laboratory of Vaccine Research and Development for Severe Infectious Diseases, Institute of Medical Biology, Chinese Academy of Medicine Sciences and Peking Union Medical College, Kunming, China
| |
Collapse
|
39
|
New Insights into the Molecular Interplay between Human Herpesviruses and Alzheimer’s Disease—A Narrative Review. Brain Sci 2022; 12:brainsci12081010. [PMID: 36009073 PMCID: PMC9406069 DOI: 10.3390/brainsci12081010] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 07/24/2022] [Accepted: 07/28/2022] [Indexed: 12/15/2022] Open
Abstract
Human herpesviruses (HHVs) have been implicated as possible risk factors in Alzheimer’s disease (AD) pathogenesis. Persistent lifelong HHVs infections may directly or indirectly contribute to the generation of AD hallmarks: amyloid beta (Aβ) plaques, neurofibrillary tangles composed of hyperphosphorylated tau proteins, and synaptic loss. The present review focuses on summarizing current knowledge on the molecular mechanistic links between HHVs and AD that include processes involved in Aβ accumulation, tau protein hyperphosphorylation, autophagy, oxidative stress, and neuroinflammation. A PubMed search was performed to collect all the available research data regarding the above mentioned mechanistic links between HHVs and AD pathology. The vast majority of research articles referred to the different pathways exploited by Herpes Simplex Virus 1 that could lead to AD pathology, while a few studies highlighted the emerging role of HHV 6, cytomegalovirus, and Epstein–Barr Virus. The elucidation of such potential links may guide the development of novel diagnostics and therapeutics to counter this devastating neurological disorder that until now remains incurable.
Collapse
|
40
|
Zhao Y, Lukiw WJ. SARS-CoV-2 Neuroinvasion, Inflammatory Neurodegeneration and Alzheimer's Disease. Front Cell Neurosci 2022; 16:937961. [PMID: 35783095 PMCID: PMC9247146 DOI: 10.3389/fncel.2022.937961] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 05/31/2022] [Indexed: 12/24/2022] Open
Affiliation(s)
- Yuhai Zhao
- LSU Neuroscience Center, Louisiana State University Health Science Center, New Orleans, LA, United States
- Department of Cell Biology and Anatomy, LSU Health Science Center, New Orleans, LA, United States
| | - Walter J. Lukiw
- LSU Neuroscience Center, Louisiana State University Health Science Center, New Orleans, LA, United States
- Department of Ophthalmology, LSU Health Science Center, New Orleans, LA, United States
- Department of Neurology, Louisiana State University Health Science Center, New Orleans, LA, United States
| |
Collapse
|
41
|
Qiao H, Zhao W, Guo M, Zhu L, Chen T, Wang J, Xu X, Zhang Z, Wu Y, Chen P. Cerebral Organoids for Modeling of HSV-1-Induced-Amyloid β Associated Neuropathology and Phenotypic Rescue. Int J Mol Sci 2022; 23:ijms23115981. [PMID: 35682661 PMCID: PMC9181143 DOI: 10.3390/ijms23115981] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/20/2022] [Accepted: 05/23/2022] [Indexed: 11/16/2022] Open
Abstract
Herpes simplex virus type I (HSV-1) infection is a potential risk factor involved in the Amyloid β (Aβ) associated neuropathology. However, further understanding of the neuropathological effects of the HSV-1 infection is hampered by the limitations of existing infection models due to the distinct differences between human brains and other mammalians’ brains. Here we generated cerebral organoid models derived from pluripotent stem cells to investigate the HSV-induced Aβ associated neuropathology and the role of antiviral drugs in the phenotypic rescue. Our results identified that the HSV-1-infected cerebral organoids recapitulated Aβ associated neuropathology including the multicellular Aβ deposition, dysregulated endogenous AD mediators, reactive gliosis, neuroinflammation, and neural loss, indicating that cerebral organoids offer an opportunity for modeling the interaction of HSV-1 with the complex phenotypes across the genetic, cellular, and tissue levels of the human Alzheimer’s disease (AD). Furthermore, we identified that two antiviral drugs, namely Ribavirin (RBV) and Valacyclovir (VCV), inhibited HSV-1 replication and rescued the neuropathological phenotypes associated with AD in the HSV-1-infected cerebral organoids, implying their therapeutic potential to slow down the progression of AD. Our study provides a high-fidelity human-relevant in-vitro HSV-1 infection model to reconstitute the multiscale neuropathological features associated with AD and discover therapeutic drug candidates relevant to the AD viral hypothesis.
Collapse
Affiliation(s)
- Haowen Qiao
- Tissue Engineering and Organ Manufacturing (TEOM) Lab, Department of Biomedical Engineering, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan 430071, China; (H.Q.); (W.Z.); (L.Z.); (T.C.); (J.W.); (X.X.)
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan 430071, China;
- State Key Laboratory of Virology, Wuhan University, Wuhan 430071, China
| | - Wen Zhao
- Tissue Engineering and Organ Manufacturing (TEOM) Lab, Department of Biomedical Engineering, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan 430071, China; (H.Q.); (W.Z.); (L.Z.); (T.C.); (J.W.); (X.X.)
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan 430071, China;
| | - Moujian Guo
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan 430071, China;
- State Key Laboratory of Virology, Wuhan University, Wuhan 430071, China
- Institute of Medical Virology, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan 430071, China
| | - Lili Zhu
- Tissue Engineering and Organ Manufacturing (TEOM) Lab, Department of Biomedical Engineering, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan 430071, China; (H.Q.); (W.Z.); (L.Z.); (T.C.); (J.W.); (X.X.)
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan 430071, China;
| | - Tao Chen
- Tissue Engineering and Organ Manufacturing (TEOM) Lab, Department of Biomedical Engineering, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan 430071, China; (H.Q.); (W.Z.); (L.Z.); (T.C.); (J.W.); (X.X.)
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan 430071, China;
| | - Jibo Wang
- Tissue Engineering and Organ Manufacturing (TEOM) Lab, Department of Biomedical Engineering, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan 430071, China; (H.Q.); (W.Z.); (L.Z.); (T.C.); (J.W.); (X.X.)
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan 430071, China;
| | - Xiaodong Xu
- Tissue Engineering and Organ Manufacturing (TEOM) Lab, Department of Biomedical Engineering, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan 430071, China; (H.Q.); (W.Z.); (L.Z.); (T.C.); (J.W.); (X.X.)
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan 430071, China;
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430050, China;
| | - Ying Wu
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan 430071, China;
- State Key Laboratory of Virology, Wuhan University, Wuhan 430071, China
- Institute of Medical Virology, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan 430071, China
- Correspondence: (Y.W.); (P.C.)
| | - Pu Chen
- Tissue Engineering and Organ Manufacturing (TEOM) Lab, Department of Biomedical Engineering, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan 430071, China; (H.Q.); (W.Z.); (L.Z.); (T.C.); (J.W.); (X.X.)
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan 430071, China;
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430050, China;
- Correspondence: (Y.W.); (P.C.)
| |
Collapse
|
42
|
Tettevi EJ, Maina M, Simpong DL, Osei-Atweneboana MY, Ocloo A. A Review of African Medicinal Plants and Functional Foods for the Management of Alzheimer's Disease-related Phenotypes, Treatment of HSV-1 Infection and/or Improvement of Gut Microbiota. J Evid Based Integr Med 2022; 27:2515690X221114657. [PMID: 35866220 PMCID: PMC9310297 DOI: 10.1177/2515690x221114657] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Alzheimer's disease (AD), which is a progressive neurodegenerative disorder is the most common form of dementia globally. Several studies have suggested alteration in the gut microbiota and HSV-1 infection as contributing factors to the development of the disease. As at now, there are no AD attenuating agents and AD pharmacotherapy is focused on managing symptoms while plants used in ethnomedicine remain potential sources of drugs for the treatment of the condition. Here, we reviewed published databases for African ethnomedicinal plants and functional foods of African origin that are used in the management of AD-related phenotypes, treatment of herpes simplex virus −1 (HSV-1) and/or improvement of gut microbiota. A total of 101 unique plant species and 24 different types of traditionally prepared African functional foodstuff were identified. Of the 101 identified plant species, 50 species serve as functional foodstuffs. Twenty-three (23) of the ethnomedicinal plant families were successfully identified for the treatment and management of AD-related phenotypes and age-related dementia. Eighteen (18) African plant species from 15 families were also identified as potent remedies for HSV-1; while many African wild fruits (3 species), roots and tubers (7 species), leafy vegetables (14 species), and seaweeds (26 species) were functional foods for modifying AD-related phenotypes. It was concluded that African medicinal plants are potential sources of both AD attenuating agents and phytocompounds that may be used against HSV-1 infection and alteration of gut microbiota. Additionally, a number of African functional foods are important sources of prebiotics and probiotics.
Collapse
Affiliation(s)
- Edward Jenner Tettevi
- Department of Biochemistry, Cell and Molecular Biology, School of Biological Science, University of Ghana, Legon, Ghana
- West African Centre for Cell Biology of Infectious Pathogens, School of Biological Science, University of Ghana, Legon, Ghana
- Biomedical and Public Health Research Unit, Council for Scientific and Industrial Research—Water Research Institute, Accra, Ghana
| | - Mahmoud Maina
- Serpell Laboratory, Sussex Neuroscience, School of Life Sciences, University of Sussex, Sussex, UK
- Biomedical Science Research and Training Centre, College of Medical Sciences, Yobe State University, Damaturu, Nigeria
| | - David Larbi Simpong
- Department of Medical Laboratory Sciences, College of Health and Allied Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Mike Y. Osei-Atweneboana
- Biomedical and Public Health Research Unit, Council for Scientific and Industrial Research—Water Research Institute, Accra, Ghana
- CSIR-College of Science and Technology, 2nd CSIR Close, Airport Residential Area, Behind Golden Tulip Hotel, Accra, Ghana
| | - Augustine Ocloo
- Department of Biochemistry, Cell and Molecular Biology, School of Biological Science, University of Ghana, Legon, Ghana
- Augustine Ocloo, Department of Biochemistry, Cell and Molecular Biology, School of Biological Science, University of Ghana, Volta Road, Legon LG54, Ghana.
| |
Collapse
|