1
|
Bhattacharya A, Shepherd C, El-Assaad F, Mather KA. Bacteria in the brain: do they have a role in the pathogenesis of Alzheimer's disease? Curr Opin Psychiatry 2025; 38:252-257. [PMID: 40013501 DOI: 10.1097/yco.0000000000000989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
PURPOSE OF REVIEW Worldwide efforts continue to unravel the complex pathological pathways that lead to Alzheimer's disease. The gut-brain-microbiome axis, a communication pathway between the gut, brain and microorganisms, is emerging as a potential mechanism involved in Alzheimer's disease pathogenesis. While the gut microbiome's role in Alzheimer's disease has gained significant attention, the brain microbiome remains relatively unexplored. This review summarizes the latest research on the brain microbiome in Alzheimer's disease. RECENT FINDINGS In the past 4 years, four out of five studies have found bacteria, such as Streptococcus pneumoniae , in postmortem samples of both control and Alzheimer's disease brains, supporting the idea that the brain is not a sterile environment. Two studies report the overabundance of several bacterial phyla, including Proteobacteria and Actinomycetes, in postmortem Alzheimer's disease brains versus controls. One study reported the presence of Borrelia burgdorferi in a subset of Alzheimer's disease cases compared to controls. SUMMARY Limitations and challenges persist in studying the brain microbiome, including the lack of standardized assays and data analysis methods, small sample sizes, and inconsistent use of controls for environmental microbial contamination during sample processing. Well designed studies that employ reproducible and rigorous methods are required to elucidate whether microbes are involved in the pathogenesis of Alzheimer's disease.
Collapse
Affiliation(s)
- Arnav Bhattacharya
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry and Mental Health, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Kensington
| | - Claire Shepherd
- Sydney Brain Bank, Neuroscience Research Australia (NeuRA), Randwick
- Department of Pathology, School of Biomedical Sciences, University of New South Wales, Kensington
| | - Fatima El-Assaad
- Microbiome Research Centre, Centre, St. George and Sutherland Clinical Campuses, UNSW Sydney, New South Wales, Australia
| | - Karen A Mather
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry and Mental Health, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Kensington
| |
Collapse
|
2
|
Ferreira da Silva A, Gomes A, Gonçalves LMD, Fernandes A, Almeida AJ. Exploring the Link Between Periodontitis and Alzheimer's Disease-Could a Nanoparticulate Vaccine Break It? Pharmaceutics 2025; 17:141. [PMID: 40006510 PMCID: PMC11858903 DOI: 10.3390/pharmaceutics17020141] [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: 12/16/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 02/27/2025] Open
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder, as approximately 55 million people worldwide are affected, with a significant tendency to increase. It reveals three main pathological features: amyloid plaques, neurofibrillary tangles, and neuroinflammation, responsible for the neurodegenerative changes that slowly lead to deterioration of personality and cognitive control. Over a century after the first case report, effective treatments remain elusive, likely due to an incomplete understanding of the precise mechanisms driving its pathogenesis. Recent studies provide growing evidence of an infectious aetiology for AD, a hypothesis reinforced by findings that amyloid beta functions as an antimicrobial peptide. Among the microorganisms already associated with AD, Porphyromonas gingivalis (Pg), the keystone pathogen of periodontitis (PeD), has received particular attention as a possible aetiological agent for AD development. Herein, we review the epidemiological and genetic evidence linking PeD and Pg to AD, highlighting the identification of periodontal bacteria in post mortem analysis of AD patients' brains and identifying putative mechanistic links relevant to the biological plausibility of the association. With the focus on AD research shifting from cure to prevention, the proposed mechanisms linking PeD to AD open the door for unravelling new prophylactic approaches able to reduce the global burden of AD. As hypothesised in this review, these could include a bionanotechnological approach involving the development of an oral nanoparticulate vaccine based on Pg-specific antigens. Such a vaccine could prevent Pg antigens from progressing to the brain and triggering AD pathology, representing a promising step toward innovative and effective AD prevention.
Collapse
Affiliation(s)
| | | | | | | | - António J. Almeida
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003 Lisbon, Portugal; (A.F.d.S.); (A.G.); (L.M.D.G.); (A.F.)
| |
Collapse
|
3
|
Kloske CM, Mahinrad S, Barnum CJ, Batista AF, Bradshaw EM, Butts B, Carrillo MC, Chakrabarty P, Chen X, Craft S, Da Mesquita S, Dabin LC, Devanand D, Duran‐Laforet V, Elyaman W, Evans EE, Fitzgerald‐Bocarsly P, Foley KE, Harms AS, Heneka MT, Hong S, Huang YA, Jackvony S, Lai L, Guen YL, Lemere CA, Liddelow SA, Martin‐Peña A, Orr AG, Quintana FJ, Ramey GD, Rexach JE, Rizzo SJS, Sexton C, Tang AS, Torrellas JG, Tsai AP, van Olst L, Walker KA, Wharton W, Tansey MG, Wilcock DM. Advancements in Immunity and Dementia Research: Highlights from the 2023 AAIC Advancements: Immunity Conference. Alzheimers Dement 2025; 21:e14291. [PMID: 39692624 PMCID: PMC11772715 DOI: 10.1002/alz.14291] [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: 06/02/2024] [Revised: 08/23/2024] [Accepted: 09/07/2024] [Indexed: 12/19/2024]
Abstract
The immune system is a key player in the onset and progression of neurodegenerative disorders. While brain resident immune cell-mediated neuroinflammation and peripheral immune cell (eg, T cell) infiltration into the brain have been shown to significantly contribute to Alzheimer's disease (AD) pathology, the nature and extent of immune responses in the brain in the context of AD and related dementias (ADRD) remain unclear. Furthermore, the roles of the peripheral immune system in driving ADRD pathology remain incompletely elucidated. In March of 2023, the Alzheimer's Association convened the Alzheimer's Association International Conference (AAIC), Advancements: Immunity, to discuss the roles of the immune system in ADRD. A wide range of topics were discussed, such as animal models that replicate human pathology, immune-related biomarkers and clinical trials, and lessons from other fields describing immune responses in neurodegeneration. This manuscript presents highlights from the conference and outlines avenues for future research on the roles of immunity in neurodegenerative disorders. HIGHLIGHTS: The immune system plays a central role in the pathogenesis of Alzheimer's disease. The immune system exerts numerous effects throughout the brain on amyloid-beta, tau, and other pathways. The 2023 AAIC, Advancements: Immunity, encouraged discussions and collaborations on understanding the role of the immune system.
Collapse
|
4
|
Hu R, Li M, Chen S, Wang M, Tao X, Zhu Y, Yan H, Liu Y. Sniffer restricts arboviral brain infections by regulating ROS levels and protecting blood-brain barrier integrity in Drosophila and mosquitoes. PLoS Pathog 2024; 20:e1012797. [PMID: 39680616 PMCID: PMC11684763 DOI: 10.1371/journal.ppat.1012797] [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: 07/15/2024] [Revised: 12/30/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
Arthropod-borne viruses (arboviruses) are transmitted to humans by arthropod vectors and pose a serious threat to global public health. Neurotropic arboviruses including Sindbis virus (SINV) persistently infect the central nervous system (CNS) of vector insects without causing notable pathological changes or affecting their behavior or lifespan. However, the mechanisms by which vector insects evade these viral infections in the brains are poorly understood. In this study, we found that loss of the carbonyl reductase Sniffer (Sni) led to a significant increase in SINV infection in the Drosophila brain. Sni regulates reactive oxygen species (ROS) levels, and its depletion leads to elevated ROS, which in turn disrupts the septate junctions (SJs) between subperineurial glia (SPG) cells, compromising the integrity and barrier function of the blood-brain barrier (BBB). Genetic and pharmacological reduction of ROS restored BBB integrity and reduced viral load in the brains of Sni-depleted flies. Additionally, we identified Sni homologs and revealed that the antiviral function of Sni is highly conserved in mosquitoes, where it regulates ROS and protects BBB integrity. Our results revealed an evolutionarily conserved antiviral mechanism in which Sni acts as an antioxidant that protects BBB integrity and restricts viral infection in the vector insect brain.
Collapse
Affiliation(s)
- Rui Hu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Mengzhu Li
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Shulin Chen
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Man Wang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Xinjun Tao
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Yihan Zhu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Huan Yan
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Yuan Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| |
Collapse
|
5
|
Conceição M, Di Filippo LD, Duarte JL, Beserra FP, Gremião MPD, Chorilli M. Repurposing doxycycline for Alzheimer's treatment: Challenges from a nano-based drug delivery perspective. Brain Behav Immun Health 2024; 42:100894. [PMID: 39525305 PMCID: PMC11550769 DOI: 10.1016/j.bbih.2024.100894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 07/16/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024] Open
Abstract
Drug repurposing, also known as drug repositioning, involves identifying new applications for drugs whose effects in a disease are already established. Doxycycline, a broad-spectrum antibiotic belonging to the tetracycline class, has demonstrated potential activity against neurodegenerative diseases like Alzheimer's and Parkinson's. However, despite its promise, the repurposed use of doxycycline encounters challenges in reaching the brain in adequate concentrations to exert its effects. To address this issue, nanostructured systems offer an innovative approach that can enhance brain targeting and the desired therapeutic outcomes. This review discusses the advances in doxycycline repurposing for Alzheimer's disease, presenting physicochemical and biological aspects that permeate doxycycline's repositioning and its application in nano-based delivery systems.
Collapse
Affiliation(s)
- Mariana Conceição
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Leonardo Delello Di Filippo
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Jonatas Lobato Duarte
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Fernando Pereira Beserra
- Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Maria Palmira Daflon Gremião
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Marlus Chorilli
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| |
Collapse
|
6
|
Kazemi N, Khorasgani MR, Noorbakhshnia M, Razavi SM, Narimani T, Naghsh N. Protective effects of a lactobacilli mixture against Alzheimer's disease-like pathology triggered by Porphyromonas gingivalis. Sci Rep 2024; 14:27283. [PMID: 39516514 PMCID: PMC11549306 DOI: 10.1038/s41598-024-77853-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
Porphyromonas gingivalis (P. gingivalis) is one of the pathogens involved in gingival inflammation, which may trigger neuroinflammatory diseases such as Alzheimer's disease (AD). This study aimed to investigate the protective (preventive and treatment) effects of a lactobacilli mixture combining Lactobacillus reuteri PTCC1655, Lactobacillus brevis CD0817, Lacticaseibacillus rhamnosus PTCC1637, and Lactobacillus plantarum PTCC1058 against P. gingivalis-induced gingival inflammation and AD-like pathology in rats. These probiotic strains exhibited cognitive enhancement effects, but this study proposed to assess their activity in a mixture. To propose a probable mechanism for P. gingivalis cognitive impairments, the TEs balance were analyzed in hippocampus and cortex tissues. Animals were divided into five groups: the control, lactobacilli, P. gingivalis, lactobacilli + P. gingivalis (prevention), and P. gingivalis + lactobacilli group (treatment) groups. The behavioral and histopathological changes were compared among them. Finally, The Trace elements (TEs) levels in the hippocampus and cortex tissues were analyzed. The palatal tissue sections of the P. gingivalis infected rats showed moderate inflammation with dense infiltration of inflammatory cells, a limited area of tissue edema, and vascular congestion. Additionally, passive avoidance learning and spatial memory were impaired. Histopathological tests revealed the presence of Aβ-positive cells in the P. gingivalis group. While the Aβ-positive cells decreased in the treatment group, their formation was inhibited in the preventive group. Administration of a mixture of lactobacilli (orally) effectively mitigated the gingival inflammation, Aβ production, and improved learning and memory functions. Moreover, Zn, Cu, and Mn levels in the hippocampus were dramatically elevated by P. gingivalis infection, whereas lactobacilli mixture mitigated these disruptive effects. The lactobacilli mixture significantly prevented the disruptive effects of P. gingivalis on gingival and brain tissues in rats. Therefore, new formulated combination of lactobacilli may be a good candidate for inhibiting the P. gingivalis infection and its subsequent cognitive effects. The current study aimed to evaluate the effects of a lactobacilli mixture to manage the disruptive effects of P. gingivalis infection on memory.
Collapse
Affiliation(s)
- Niloofar Kazemi
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Mohammad Rabbani Khorasgani
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran.
| | - Maryam Noorbakhshnia
- Department of Plant and Animal Biology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran.
| | - Seyed Mohammad Razavi
- Department of Oral and Maxillofacial Pathology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Tahmineh Narimani
- Department of Bacteriology and Virology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Narges Naghsh
- Department of Periodontology, Torabinejad Dental Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
7
|
Kehs Z, Cross AC, Li YM. From defense to disease: IFITM3 in immunity and Alzheimer's disease. Neurotherapeutics 2024:e00482. [PMID: 39516072 DOI: 10.1016/j.neurot.2024.e00482] [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: 09/30/2024] [Revised: 10/24/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Innate immunity protein interferon induced transmembrane protein 3 (IFITM3) is a transmembrane protein that has a wide array of functions, including in viral infections, Alzheimer's Disease (AD), and cancer. As an interferon stimulated gene (ISG), IFITM3's expression is upregulated by type-I, II, and III interferons. Moreover, the antiviral activity of IFITM3 is modulated by post-translational modifications. IFITM3 functions in innate immunity to disrupt viral fusion and entry to the plasma membrane as well as prevent viral escape from endosomes. As a γ-secretase modulatory protein, IFITM3 distinctly modulates the processing of amyloid precursor protein (APP) to generate amyloid beta peptides (Aβ) and Notch1 cleavages. Increased IFITM3 expression, which can result from aging, cytokine activation, inflammation, and infection, can lead to an upregulation of γ-secretase for Aβ production that causes a risk of AD. Therefore, the prevention of IFITM3 upregulation has potential in the development of novel therapies for the treatment of AD.
Collapse
Affiliation(s)
- Zoe Kehs
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Programs of Pharmacology, Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA
| | - Abigail C Cross
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Programs of Neuroscience, Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA
| | - Yue-Ming Li
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Programs of Pharmacology, Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA.
| |
Collapse
|
8
|
da Silva AF, Gonçalves LMD, Fernandes A, Almeida AJ. Optimization and evaluation of a chitosan-coated PLGA nanocarrier for mucosal delivery of Porphyromonas gingivalis antigens. Eur J Pharm Sci 2024; 202:106896. [PMID: 39250981 DOI: 10.1016/j.ejps.2024.106896] [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: 02/22/2024] [Revised: 07/30/2024] [Accepted: 09/04/2024] [Indexed: 09/11/2024]
Abstract
Recent advances in understanding Alzheimer's disease (AD) suggest the possibility of an infectious etiology, with Porphyromonas gingivalis emerging as a prime suspect in contributing to AD. P. gingivalis may invade systemic circulation via weakened oral/intestinal barriers and then cross the blood-brain barrier (BBB), reaching the brain and precipitating AD pathology. Based on the proposed links between P. gingivalis and AD, a prospective approach is the development of an oral nanovaccine containing P. gingivalis antigens for mucosal delivery. Targeting the gut-associated lymphoid tissue (GALT), the nanovaccine may elicit both mucosal and systemic immunity, thereby hampering P. gingivalis ability to breach the oral/intestinal barriers and the BBB, respectively. The present study describes the optimization, characterization, and in vitro evaluation of a candidate chitosan-coated poly(lactic-co-glycolic acid) (PLGA-CS) nanovaccine containing a P. gingivalis antigen extract. The nanocarrier was prepared using the double emulsion solvent evaporation method and optimized for selected experimental factors, e.g. PLGA amount, surfactant concentration, w1/o phase ratio, applying a d-optimal statistical design to target the desired physicochemical criteria for its intended application. After nanocarrier optimization, the nanovaccine was characterized in terms of particle size, polydispersity index (PdI), ζ-potential, encapsulation efficiency (EE), drug loading (DL), morphology, and in vitro release profile, as well as for mucoadhesivity, stability under simulated gastrointestinal conditions, antigen integrity, in vitro cytotoxicity and uptake using THP-1 macrophages. The candidate PLGA-CS nanovaccine demonstrated appropriate physicochemical, mucoadhesive, and antigen release properties for oral delivery, along with acceptable levels of EE (55.3 ± 3.5 %) and DL (1.84 ± 0.12 %). The integrity of the encapsulated antigens remained uncompromised throughout NPs production and simulated gastrointestinal exposure, as confirmed by SDS-PAGE and Western blotting analyses. Furthermore, the nanovaccine showed effective in vitro uptake, while exhibiting low cytotoxicity. Taken together, these findings underscore the potential of PLGA-CS NPs as carriers for adequate antigen mucosal delivery, paving the way for further investigations into their applicability as vaccine candidates against P. gingivalis.
Collapse
Affiliation(s)
- André Ferreira da Silva
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal.
| | - Lídia M D Gonçalves
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal.
| | - Adelaide Fernandes
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal.
| | - António J Almeida
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
9
|
Min JH, Sarlus H, Harris RA. MAD-microbial (origin of) Alzheimer's disease hypothesis: from infection and the antimicrobial response to disruption of key copper-based systems. Front Neurosci 2024; 18:1467333. [PMID: 39416952 PMCID: PMC11480022 DOI: 10.3389/fnins.2024.1467333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 09/20/2024] [Indexed: 10/19/2024] Open
Abstract
Microbes have been suspected to cause Alzheimer's disease since at least 1908, but this has generally remained unpopular in comparison to the amyloid hypothesis and the dominance of Aβ and Tau. However, evidence has been accumulating to suggest that these earlier theories are but a manifestation of a common cause that can trigger and interact with all the major molecular players recognized in AD. Aβ, Tau and ApoE, in particular appear to be molecules with normal homeostatic functions but also with alternative antimicrobial functions. Their alternative functions confer the non-immune specialized neuron with some innate intracellular defenses that appear to be re-appropriated from their normal functions in times of need. Indeed, signs of infection of the neurons by biofilm-forming microbial colonies, in synergy with herpes viruses, are evident from the clinical and preclinical studies we discuss. Furthermore, we attempt to provide a mechanistic understanding of the AD landscape by discussing the antimicrobial effect of Aβ, Tau and ApoE and Lactoferrin in AD, and a possible mechanistic link with deficiency of vital copper-based systems. In particular, we focus on mitochondrial oxidative respiration via complex 4 and ceruloplasmin for iron homeostasis, and how this is similar and possibly central to neurodegenerative diseases in general. In the case of AD, we provide evidence for the microbial Alzheimer's disease (MAD) theory, namely that AD could in fact be caused by a long-term microbial exposure or even long-term infection of the neurons themselves that results in a costly prolonged antimicrobial response that disrupts copper-based systems that govern neurotransmission, iron homeostasis and respiration. Finally, we discuss potential treatment modalities based on this holistic understanding of AD that incorporates the many separate and seemingly conflicting theories. If the MAD theory is correct, then the reduction of microbial exposure through use of broad antimicrobial and anti-inflammatory treatments could potentially alleviate AD although this requires further clinical investigation.
Collapse
Affiliation(s)
- Jin-Hong Min
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital at Solna, Stockholm, Sweden
| | | | | |
Collapse
|
10
|
Barrera-Ocampo A. Monoclonal antibodies and aptamers: The future therapeutics for Alzheimer's disease. Acta Pharm Sin B 2024; 14:2795-2814. [PMID: 39027235 PMCID: PMC11252463 DOI: 10.1016/j.apsb.2024.03.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/06/2024] [Accepted: 03/14/2024] [Indexed: 07/20/2024] Open
Abstract
Alzheimer's disease (AD) is considered the most common and prevalent form of dementia of adult-onset with characteristic progressive impairment in cognition and memory. The cure for AD has not been found yet and the treatments available until recently were only symptomatic. Regardless of multidisciplinary approaches and efforts made by pharmaceutical companies, it was only in the past two years that new drugs were approved for the treatment of the disease. Amyloid beta (Aβ) immunotherapy is at the core of this therapy, which is one of the most innovative approaches looking to change the course of AD. This technology is based on synthetic peptides or monoclonal antibodies (mAb) to reduce Aβ levels in the brain and slow down the advance of neurodegeneration. Hence, this article reviews the state of the art about AD neuropathogenesis, the traditional pharmacologic treatment, as well as the modern active and passive immunization describing approved drugs, and drug prototypes currently under investigation in different clinical trials. In addition, future perspectives on immunotherapeutic strategies for AD and the rise of the aptamer technology as a non-immunogenic alternative to curb the disease progression are discussed.
Collapse
Affiliation(s)
- Alvaro Barrera-Ocampo
- Facultad de Ingeniería, Diseño y Ciencias Aplicadas, Departamento de Ciencias Farmacéuticas y Químicas, Grupo Natura, Universidad Icesi, Cali 760031, Colombia
| |
Collapse
|
11
|
Barber AJ, Del Genio CL, Swain AB, Pizzi EM, Watson SC, Tapiavala VN, Zanazzi GJ, Gaur AB. Age, sex and Alzheimer's disease: a longitudinal study of 3xTg-AD mice reveals sex-specific disease trajectories and inflammatory responses mirrored in postmortem brains from Alzheimer's patients. Alzheimers Res Ther 2024; 16:134. [PMID: 38909241 PMCID: PMC11193202 DOI: 10.1186/s13195-024-01492-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 06/06/2024] [Indexed: 06/24/2024]
Abstract
BACKGROUND Aging and sex are major risk factors for developing late-onset Alzheimer's disease. Compared to men, women experience worse neuropathological burden and cognitive decline despite living longer with the disease. Similarly, male 3xTg-AD mice, developed to model Alzheimer's disease, no longer consistently exhibit standard Alzheimer's neuropathology yet experience higher rates of mortality - providing a unique opportunity to further elucidate this dichotomy. We hypothesized that sex differences in the biological aging process yield distinct pathological and molecular Alzheimer's disease signatures in males and females, which could be harnessed for therapeutic and biomarker development. METHODS We aged male and female, 3xTg-AD and B6129 control mice across their respective lifespans (n = 3-8 mice per sex, strain, and age group) and longitudinally assessed neuropathological hallmarks of Alzheimer's disease, markers of hepatic inflammation, splenic mass and morphology, as well as plasma cytokine levels. We conducted RNA sequencing analysis on bulk brain tissue and examined differentially expressed genes (DEGs) between 3xTg-AD and B6129 samples and across ages in each sex. We also examined DEGs between clinical Alzheimer's and control parahippocampal gyrus brain tissue samples from the Mount Sinai Brain Bank study in each sex. RESULTS 3xTg-AD females significantly outlived 3xTg-AD males and exhibited progressive Alzheimer's neuropathology, while 3xTg-AD males demonstrated progressive hepatic inflammation, splenomegaly, circulating inflammatory proteins, and minimal Alzheimer's neuropathological hallmarks. Instead, 3xTg-AD males experienced an accelerated upregulation of immune-related gene expression in the brain relative to females. Our clinical investigations revealed that individuals with Alzheimer's disease develop similar sex-specific alterations in neuronal and immune function. In diseased males of both species, we observed greater upregulation of complement-related gene expression, and lipopolysaccharide was predicted as the top upstream regulator of DEGs. CONCLUSIONS Our data demonstrate that chronic inflammation and complement activation are associated with increased mortality, indicating that age-related changes in immune response contribute to sex differences in Alzheimer's disease trajectories. We provide evidence that aging and transgene-driven disease progression trigger a widespread inflammatory response in 3xTg-AD males, which mimics the impact of lipopolysaccharide stimulation despite the absence of infection.
Collapse
Affiliation(s)
- Alicia J Barber
- Department of Neurology, Geisel School of Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| | - Carmen L Del Genio
- Department of Neurology, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| | | | - Elizabeth M Pizzi
- The Jackson Laboratory, Bar Harbor, ME, USA
- Neuroscience Program, Graduate School of Biomedical Sciences, Tufts University, Boston, MA, USA
| | | | | | - George J Zanazzi
- Department of Pathology, Geisel School of Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| | - Arti B Gaur
- Department of Neurology, Geisel School of Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA.
- Integrative Neuroscience at Dartmouth, Dartmouth College, Hanover, NH, USA.
| |
Collapse
|
12
|
Kettunen P, Koistinaho J, Rolova T. Contribution of CNS and extra-CNS infections to neurodegeneration: a narrative review. J Neuroinflammation 2024; 21:152. [PMID: 38845026 PMCID: PMC11157808 DOI: 10.1186/s12974-024-03139-y] [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: 03/17/2024] [Accepted: 05/23/2024] [Indexed: 06/09/2024] Open
Abstract
Central nervous system infections have been suggested as a possible cause for neurodegenerative diseases, particularly sporadic cases. They trigger neuroinflammation which is considered integrally involved in neurodegenerative processes. In this review, we will look at data linking a variety of viral, bacterial, fungal, and protozoan infections to Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, multiple sclerosis and unspecified dementia. This narrative review aims to bring together a broad range of data currently supporting the involvement of central nervous system infections in the development of neurodegenerative diseases. The idea that no single pathogen or pathogen group is responsible for neurodegenerative diseases will be discussed. Instead, we suggest that a wide range of susceptibility factors may make individuals differentially vulnerable to different infectious pathogens and subsequent pathologies.
Collapse
Affiliation(s)
- Pinja Kettunen
- Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Jari Koistinaho
- Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland.
| | - Taisia Rolova
- Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| |
Collapse
|
13
|
Chen Y, Al-Nusaif M, Li S, Tan X, Yang H, Cai H, Le W. Progress on early diagnosing Alzheimer's disease. Front Med 2024; 18:446-464. [PMID: 38769282 PMCID: PMC11391414 DOI: 10.1007/s11684-023-1047-1] [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: 09/23/2023] [Accepted: 11/15/2023] [Indexed: 05/22/2024]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that affects both cognition and non-cognition functions. The disease follows a continuum, starting with preclinical stages, progressing to mild cognitive and behavioral impairment, ultimately leading to dementia. Early detection of AD is crucial for better diagnosis and more effective treatment. However, the current AD diagnostic tests of biomarkers using cerebrospinal fluid and/or brain imaging are invasive or expensive, and mostly are still not able to detect early disease state. Consequently, there is an urgent need to develop new diagnostic techniques with higher sensitivity and specificity during the preclinical stages of AD. Various non-cognitive manifestations, including behavioral abnormalities, sleep disturbances, sensory dysfunctions, and physical changes, have been observed in the preclinical AD stage before occurrence of notable cognitive decline. Recent research advances have identified several biofluid biomarkers as early indicators of AD. This review focuses on these non-cognitive changes and newly discovered biomarkers in AD, specifically addressing the preclinical stages of the disease. Furthermore, it is of importance to explore the potential for developing a predictive system or network to forecast disease onset and progression at the early stage of AD.
Collapse
Affiliation(s)
- Yixin Chen
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, 116021, China
| | - Murad Al-Nusaif
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, 116021, China
| | - Song Li
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, 116021, China
| | - Xiang Tan
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, 116021, China
| | - Huijia Yang
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, 116021, China
| | - Huaibin Cai
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Weidong Le
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, 116021, China.
- Institute of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, 610072, China.
| |
Collapse
|
14
|
Das B, Tk Baidya A, Chakrabarti S, Chouhan D, Thakur B, Darreh-Shori T, Chen G, Tiwari V, Kumar R. Synthesis and biological evaluation of Halogen-Substituted novel α-Ketoamides as potential protein aggregation modulators in Alzheimer's disease. Bioorg Chem 2024; 147:107373. [PMID: 38653149 DOI: 10.1016/j.bioorg.2024.107373] [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: 02/21/2024] [Revised: 04/01/2024] [Accepted: 04/12/2024] [Indexed: 04/25/2024]
Abstract
The escalating prevalence of Alzheimer's disease (AD) has prompted extensive research into potential therapeutic interventions, with a specific focus on molecular targets such as amyloid beta (Aβ) and tau protein aggregation. In this study, a series of α-ketoamide derivatives was synthesized from β,γ-unsaturated α-keto thioesters, achieving high purity and good yield. Thioflavin T based Aβ aggregation assay identified four promising compounds (BD19, BD23, BD24, and BD27) that demonstrated significant inhibitory effects on Aβ aggregation. BD23, selected for its better solubility (0.045 ± 0.0012 mg/ml), was further subjected to in vitro Parallel Artificial Membrane Permeability Assay to determine the Blood-Brain-Barrier permeability and emerged as BBB permeable with permeability rate (Pe) of 10.66 ± 8.11 × 10-6 cm/s. In addition to its Aβ inhibitory properties, BD23 exhibited significant inhibition of heparin-induced tau aggregation and demonstrated non-toxicity in SHSY5Y cell lines. Subsequent in vivo assays were conducted, administering compound BD23 to an Aβ induced mouse model of AD at various doses (1, 2, & 5 mg/kg). The results revealed a noteworthy enhancement in cognitive functions, particularly when BD23 was administered at a dosage of 5 mg/kg, comparable to the effects observed with the standard dose of Donepezil (DNP). In silico investigations, including molecular docking, molecular dynamics simulations, and Density Functional Theory calculations provided insights into BD23's interactions with the targets and electronic properties. These analyses contribute to the understanding of the therapeutic potential of the lead compounds BD23 which further pave the way for further exploration of its therapeutic potential in the context of AD.
Collapse
Affiliation(s)
- Bhanuranjan Das
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (B.H.U.), Varanasi 221005 U.P., India
| | - Anurag Tk Baidya
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (B.H.U.), Varanasi 221005 U.P., India
| | - Sourabh Chakrabarti
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (B.H.U.), Varanasi 221005 U.P., India
| | - Deepak Chouhan
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (B.H.U.), Varanasi 221005 U.P., India
| | - Banita Thakur
- Division of Clinical Geriatric, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, NEO, 7th Floor, 141 52 Stockholm, Sweden
| | - Taher Darreh-Shori
- Division of Clinical Geriatric, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, NEO, 7th Floor, 141 52 Stockholm, Sweden
| | - Gefei Chen
- Department of Biosciences and Nutrition, Karolinska Institutet, 14 183, Huddinge, Sweden
| | - Vinod Tiwari
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (B.H.U.), Varanasi 221005 U.P., India
| | - Rajnish Kumar
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (B.H.U.), Varanasi 221005 U.P., India.
| |
Collapse
|
15
|
Kim AY, Al Jerdi S, MacDonald R, Triggle CR. Alzheimer's disease and its treatment-yesterday, today, and tomorrow. Front Pharmacol 2024; 15:1399121. [PMID: 38868666 PMCID: PMC11167451 DOI: 10.3389/fphar.2024.1399121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 04/25/2024] [Indexed: 06/14/2024] Open
Abstract
Alois Alzheimer described the first patient with Alzheimer's disease (AD) in 1907 and today AD is the most frequently diagnosed of dementias. AD is a multi-factorial neurodegenerative disorder with familial, life style and comorbidity influences impacting a global population of more than 47 million with a projected escalation by 2050 to exceed 130 million. In the USA the AD demographic encompasses approximately six million individuals, expected to increase to surpass 13 million by 2050, and the antecedent phase of AD, recognized as mild cognitive impairment (MCI), involves nearly 12 million individuals. The economic outlay for the management of AD and AD-related cognitive decline is estimated at approximately 355 billion USD. In addition, the intensifying prevalence of AD cases in countries with modest to intermediate income countries further enhances the urgency for more therapeutically and cost-effective treatments and for improving the quality of life for patients and their families. This narrative review evaluates the pathophysiological basis of AD with an initial focus on the therapeutic efficacy and limitations of the existing drugs that provide symptomatic relief: acetylcholinesterase inhibitors (AChEI) donepezil, galantamine, rivastigmine, and the N-methyl-D-aspartate receptor (NMDA) receptor allosteric modulator, memantine. The hypothesis that amyloid-β (Aβ) and tau are appropriate targets for drugs and have the potential to halt the progress of AD is critically analyzed with a particular focus on clinical trial data with anti-Aβ monoclonal antibodies (MABs), namely, aducanumab, lecanemab and donanemab. This review challenges the dogma that targeting Aβ will benefit the majority of subjects with AD that the anti-Aβ MABs are unlikely to be the "magic bullet". A comparison of the benefits and disadvantages of the different classes of drugs forms the basis for determining new directions for research and alternative drug targets that are undergoing pre-clinical and clinical assessments. In addition, we discuss and stress the importance of the treatment of the co-morbidities, including hypertension, diabetes, obesity and depression that are known to increase the risk of developing AD.
Collapse
Affiliation(s)
- A. Y. Kim
- Medical Education, Weill Cornell Medicine—Qatar, Doha, Qatar
| | | | - R. MacDonald
- Health Sciences Library, Weill Cornell Medicine—Qatar, Doha, Qatar
| | - C. R. Triggle
- Department of Pharmacology and Medical Education, Weill Cornell Medicine—Qatar, Doha, Qatar
| |
Collapse
|
16
|
Tao MH, Gordon SC, Wu T, Trudeau S, Rupp LB, Gonzalez HC, Daida YG, Schmidt MA, Lu M. Antiviral Treatment and Response are Associated With Lower Risk of Dementia Among Hepatitis C Virus-Infected Patients. Am J Geriatr Psychiatry 2024; 32:611-621. [PMID: 38199936 DOI: 10.1016/j.jagp.2023.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/15/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024]
Abstract
OBJECTIVE Eradication of hepatitis C virus (HCV) infection has been linked with improvement in neurocognitive function, but few studies have evaluated the effect of antiviral treatment/ response on risk of dementia. Using data from the Chronic Hepatitis Cohort Study (CHeCS), we investigated how antiviral therapy impacts the risk of developing dementia among patients with HCV. METHODS A total of 17,485 HCV patients were followed until incidence of dementia, death, or last follow-up. We used an extended landmark modeling approach, which included time-varying covariates and propensity score justification for treatment selection bias, as well as generalized estimating equations (GEE) with a link function as multinominal distribution for a discrete time-to-event data. Death was considered a competing risk. RESULTS After 15 years of follow-up, 342 patients were diagnosed with incident dementia. Patients who achieved sustained virological response (SVR) had significantly decreased risk of dementia compared to untreated patients, with hazard ratios (HRs) of 0.32 (95% CI 0.22-0.46) among patients who received direct-acting antiviral (DAA) treatment and 0.41 (95% CI 0.26-0.60) for interferon-based (IFN) treatment. Risk reduction remained even when patients failed antiviral treatment (HR 0.38, 95% CI 0.38-0.51). Patients with cirrhosis, Black/African American patients, and those without private insurance were at significantly higher risk of dementia. CONCLUSION Antiviral treatment independently reduced the risk of dementia among HCV patients, regardless of cirrhosis. Our findings support the importance of initiation antiviral therapy in chronic HCV-infected patients.
Collapse
Affiliation(s)
- Meng-Hua Tao
- Department of Public Health Sciences (M-HT, TW, ST, HCG, ML), Henry Ford Health, Detroit MI.
| | - Stuart C Gordon
- Department of Gastroenterology and Hepatology (SCG, HCG), Henry Ford Health, Detroit MI; School of Medicine (SCG, HCG), Wayne State University, Detroit MI
| | - Trueman Wu
- Department of Public Health Sciences (M-HT, TW, ST, HCG, ML), Henry Ford Health, Detroit MI
| | - Sheri Trudeau
- Department of Public Health Sciences (M-HT, TW, ST, HCG, ML), Henry Ford Health, Detroit MI
| | - Loralee B Rupp
- Department of Health Policy and Health Services Research (LBR), Henry Ford Health, Detroit MI
| | - Humberto C Gonzalez
- Department of Public Health Sciences (M-HT, TW, ST, HCG, ML), Henry Ford Health, Detroit MI; Department of Gastroenterology and Hepatology (SCG, HCG), Henry Ford Health, Detroit MI; School of Medicine (SCG, HCG), Wayne State University, Detroit MI
| | - Yihe G Daida
- Center for Integrated Health Care Research (YGD), Kaiser Permanente Hawaii, Honolulu, HI
| | - Mark A Schmidt
- Center for Health Research (MAS), Kaiser Permanente Northwest, Portland, OR
| | - Mei Lu
- Department of Public Health Sciences (M-HT, TW, ST, HCG, ML), Henry Ford Health, Detroit MI
| |
Collapse
|
17
|
Biagio P, Isabella DF, Federica C, Elena S, Ivan G. Alzheimer's disease and herpes viruses: Current events and perspectives. Rev Med Virol 2024; 34:e2550. [PMID: 38801246 DOI: 10.1002/rmv.2550] [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: 01/21/2024] [Revised: 05/19/2024] [Accepted: 05/21/2024] [Indexed: 05/29/2024]
Abstract
Alzheimer's disease (AD) is a real and current scientific and societal challenge. Alzheimer's disease is characterised by a neurodegenerative neuroinflammatory process, but the etiopathogenetic mechanisms are still unclear. The possible infectious aetiology and potential involvement of Herpes viruses as triggers for the formation of extracellular deposits of amyloid beta (Aβ) peptide (amyloid plaques) and intraneuronal aggregates of hyperphosphorylated and misfold could be a possible explanation. In fact, the possible genetic interference of Herpes viruses with the genome of the host neuronal cell or the stimulation of the infection to a continuous immune response with a consequent chronic inflammation could constitute those mechanisms underlying the development of AD, with possible implications in the understanding and management of the disease. Herpes viruses could be significantly involved in the pathogenesis of AD and in particular, their ability to reactivate in particular conditions such as immunocompromise and immunosenescence, could explain the neurological damage characteristic of AD. Our review aims to evaluate the state of the art of knowledge and perspectives regarding the potential relationship between Herpes viruses and AD, in order to be able to identify the possible etiopathogenetic mechanisms and the possible therapeutic implications.
Collapse
Affiliation(s)
- Pinchera Biagio
- Division of Infectious Diseases, Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - Di Filippo Isabella
- Division of Infectious Diseases, Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - Cuccurullo Federica
- Division of Infectious Diseases, Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - Salvatore Elena
- Division of Neurology, Department of Neuroscience Reproductive Sciences and Odontostomatology, University of Naples "Federico II", Naples, Italy
| | - Gentile Ivan
- Division of Infectious Diseases, Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| |
Collapse
|
18
|
Kim HS, Jung H, Park YH, Heo SH, Kim S, Moon M. Skin-brain axis in Alzheimer's disease - Pathologic, diagnostic, and therapeutic implications: A Hypothetical Review. Aging Dis 2024; 16:901-916. [PMID: 38739932 DOI: 10.14336/ad.2024.0406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 04/06/2024] [Indexed: 05/16/2024] Open
Abstract
The dynamic interaction between the brain and the skin is termed the 'skin-brain axis.' Changes in the skin not only reflect conditions in the brain but also exert direct and indirect effects on the brain. Interestingly, the connection between the skin and brain is crucial for understanding aging and neurodegenerative diseases. Several studies have shown an association between Alzheimer's disease (AD) and various skin disorders, such as psoriasis, bullous pemphigoid, and skin cancer. Previous studies have shown a significantly increased risk of new-onset AD in patients with psoriasis. In contrast, skin cancer may reduce the risk of developing AD. Accumulating evidence suggests an interaction between skin disease and AD; however, AD-associated pathological changes mediated by the skin-brain axis are not yet clearly defined. While some studies have reported on the diagnostic implications of the skin-brain axis in AD, few have discussed its potential therapeutic applications. In this review, we address the pathological changes mediated by the skin-brain axis in AD. Furthermore, we summarize (1) the diagnostic implications elucidated through the role of the skin-brain axis in AD and (2) the therapeutic implications for AD based on the skin-brain axis. Our review suggests that a potential therapeutic approach targeting the skin-brain axis will enable significant advances in the treatment of AD.
Collapse
Affiliation(s)
- Hyeon Soo Kim
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea
| | - Haram Jung
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea
| | - Yong Ho Park
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea
| | - Su-Hak Heo
- Department of Medicinal Bioscience, Konkuk University (Glocal Campus), Chungcheongbuk-do 27478, Korea
| | - Sujin Kim
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea
- Research Institute for Dementia Science, Konyang University, Daejeon 35365, Korea
| | - Minho Moon
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea
- Research Institute for Dementia Science, Konyang University, Daejeon 35365, Korea
| |
Collapse
|
19
|
Williams ZAP, Lang L, Nicolas S, Clarke G, Cryan J, Vauzour D, Nolan YM. Do microbes play a role in Alzheimer's disease? Microb Biotechnol 2024; 17:e14462. [PMID: 38593310 PMCID: PMC11003713 DOI: 10.1111/1751-7915.14462] [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: 03/05/2024] [Accepted: 03/19/2024] [Indexed: 04/11/2024] Open
Abstract
Alzheimer's disease is a complex and progressive condition that affects essential neurological functions such as memory and reasoning. In the brain, neuronal loss, synaptic dysfunction, proteinopathy, neurofibrillary tangles, and neuroinflammation are the hallmarks of Alzheimer's disease pathophysiology. In addition, recent evidence has highlighted that microbes, whether commensal or pathogenic, also have the ability to interact with their host and to regulate its immune system, therefore participating in the exchanges that lead to peripheral inflammation and neuropathology. Because of this intimate relationship, bacteria, viruses, fungi, and protozoa have been implicated in the development of Alzheimer's disease. Here, we bring together current and most recent evidence of the role of microbes in Alzheimer's disease, raising burning questions that need to be addressed to guide therapeutic approaches and potential prophylactic strategies.
Collapse
Affiliation(s)
- Zoë A. P. Williams
- Department of Anatomy and NeuroscienceUniversity College CorkCorkIreland
- APC Microbiome IrelandUniversity College CorkCorkIreland
| | - Leonie Lang
- Norwich Medical School, Faculty of Medicine and Health SciencesUniversity of East AngliaNorwichUK
| | - Sarah Nicolas
- Department of Anatomy and NeuroscienceUniversity College CorkCorkIreland
- APC Microbiome IrelandUniversity College CorkCorkIreland
| | - Gerard Clarke
- APC Microbiome IrelandUniversity College CorkCorkIreland
- Department of Psychiatry and Neurobehavioural ScienceUniversity College CorkCorkIreland
| | - John Cryan
- Department of Anatomy and NeuroscienceUniversity College CorkCorkIreland
- APC Microbiome IrelandUniversity College CorkCorkIreland
| | - David Vauzour
- Norwich Medical School, Faculty of Medicine and Health SciencesUniversity of East AngliaNorwichUK
| | - Yvonne M. Nolan
- Department of Anatomy and NeuroscienceUniversity College CorkCorkIreland
- APC Microbiome IrelandUniversity College CorkCorkIreland
| |
Collapse
|
20
|
Aliashrafi M, Nasehi M, Zarrindast MR, Joghataei MT, Zali H, Siadat SD. Intracerebroventricular Cutibacterium acnes Generates Manifestations of Alzheimer's Disease-like Pathology in the Rat Hippocampus. Neuroscience 2024; 540:103-116. [PMID: 38266907 DOI: 10.1016/j.neuroscience.2024.01.013] [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: 04/23/2023] [Revised: 01/13/2024] [Accepted: 01/16/2024] [Indexed: 01/26/2024]
Abstract
The infection hypothesis is a new causative explanation for Alzheimer's disease (AD). In recent decades, various species of bacterial pathogens have been distinguished in the autopsy of Alzheimer's patients; however, the mechanism of bacterial contribution to AD pathology is still unknown. To explore the hypothesis, Cutibacterium acnes (C. acnes) was selected, and effects of its intracerebroventricular (ICV) inoculation in rats was evaluated. The results revealed that C. acnes causes memory impairment, which might be a consequence of upregulated Amyloid β (Aβ) deposits in the hippocampus; Aβ aggregates are co-localized with C. acnes colonies. The key point of our hypothesis is that the activation of the innate immune system by C. acnes through the TLR2/NF-κB/NLRP3 signaling pathway, eventually leads to increased neuroinflammation, which might be resulted from microgliosis and astrogliosis. Neuroinflammation increases oxidative stress and cell apoptosis. Overall, the obtained results of this study support our hypothesis that brain exposure to C. acnes prompted neuroinflammation with similar AD-like pathology.
Collapse
Affiliation(s)
- Morteza Aliashrafi
- Department of Cognitive Neuroscience, Institute for Cognitive Science Studies, Tehran, Iran; Shahid Beheshti University, Tehran, Iran
| | - Mohammad Nasehi
- Cognitive and Neuroscience Research Center, Amir-Almomenin Hospital, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mohammad-Reza Zarrindast
- Department of Cognitive Neuroscience, Institute for Cognitive Science Studies, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad-Taghi Joghataei
- Cellular and Molecular Research Center, Department of Neuroscience, Iran University of Medical Science, Tehran, Iran
| | - Hakimeh Zali
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Seyed Davar Siadat
- Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
21
|
Green RE, Sudre CH, Warren‐Gash C, Butt J, Waterboer T, Hughes AD, Schott JM, Richards M, Chaturvedi N, Williams DM. Common infections and neuroimaging markers of dementia in three UK cohort studies. Alzheimers Dement 2024; 20:2128-2142. [PMID: 38248636 PMCID: PMC10984486 DOI: 10.1002/alz.13613] [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: 07/20/2023] [Revised: 10/13/2023] [Accepted: 11/25/2023] [Indexed: 01/23/2024]
Abstract
INTRODUCTION We aimed to investigate associations between common infections and neuroimaging markers of dementia risk (brain volume, hippocampal volume, white matter lesions) across three population-based studies. METHODS We tested associations between serology measures (pathogen serostatus, cumulative burden, continuous antibody responses) and outcomes using linear regression, including adjustments for total intracranial volume and scanner/clinic information (basic model), age, sex, ethnicity, education, socioeconomic position, alcohol, body mass index, and smoking (fully adjusted model). Interactions between serology measures and apolipoprotein E (APOE) genotype were tested. Findings were meta-analyzed across cohorts (Nmain = 2632; NAPOE-interaction = 1810). RESULTS Seropositivity to John Cunningham virus associated with smaller brain volumes in basic models (β = -3.89 mL [-5.81, -1.97], Padjusted < 0.05); these were largely attenuated in fully adjusted models (β = -1.59 mL [-3.55, 0.36], P = 0.11). No other relationships were robust to multiple testing corrections and sensitivity analyses, but several suggestive associations were observed. DISCUSSION We did not find clear evidence for relationships between common infections and markers of dementia risk. Some suggestive findings warrant testing for replication.
Collapse
Affiliation(s)
- Rebecca E. Green
- MRC Unit for Lifelong Health & Ageing at UCLUniversity College LondonLondonUK
| | - Carole H. Sudre
- MRC Unit for Lifelong Health & Ageing at UCLUniversity College LondonLondonUK
- Dementia Research CentreUCL Queen Square Institute of NeurologyLondonUK
- School of Biomedical Engineering and Imaging SciencesKing's College LondonLondonUK
- Department of Medical Physics and Biomedical EngineeringCentre for Medical Image Computing (CMIC)University College London (UCL)LondonUK
| | - Charlotte Warren‐Gash
- Faculty of Epidemiology and Population HealthLondon School of Hygiene and Tropical MedicineLondonUK
| | - Julia Butt
- Division of Infections and Cancer EpidemiologyGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Tim Waterboer
- Division of Infections and Cancer EpidemiologyGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Alun D. Hughes
- MRC Unit for Lifelong Health & Ageing at UCLUniversity College LondonLondonUK
| | | | - Marcus Richards
- MRC Unit for Lifelong Health & Ageing at UCLUniversity College LondonLondonUK
| | - Nish Chaturvedi
- MRC Unit for Lifelong Health & Ageing at UCLUniversity College LondonLondonUK
| | - Dylan M. Williams
- MRC Unit for Lifelong Health & Ageing at UCLUniversity College LondonLondonUK
| |
Collapse
|
22
|
Seo DO, Holtzman DM. Current understanding of the Alzheimer's disease-associated microbiome and therapeutic strategies. Exp Mol Med 2024; 56:86-94. [PMID: 38172602 PMCID: PMC10834451 DOI: 10.1038/s12276-023-01146-2] [Citation(s) in RCA: 53] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 10/13/2023] [Accepted: 10/16/2023] [Indexed: 01/05/2024] Open
Abstract
Alzheimer's disease (AD) is a fatal progressive neurodegenerative disease. Despite tremendous research efforts to understand this complex disease, the exact pathophysiology of the disease is not completely clear. Recently, anti-Aβ antibodies have been shown to remove amyloid from the brain and slow the clinical progression of mild dementia by ~30%. However, exploring alternative strategies is crucial to understanding and developing more effective therapeutic interventions. In recent years, the microbiota-gut-brain axis has received significant attention in the AD field. Numerous studies have suggested that alterations in the gut microbiota composition are associated with the progression of AD, and several underlying mechanisms have been proposed. However, studies in this area are still in their infancy, and many aspects of this field are just beginning to be explored and understood. Gaining a deeper understanding of the intricate interactions and signaling pathways involved in the microbiota-AD interaction is crucial for optimizing therapeutic strategies targeting gut microbiota to positively impact AD. In this review, we aim to summarize the current understanding of the microbiota-gut-brain axis in AD. We will discuss the existing evidence regarding the role of gut microbiota in AD pathogenesis, suggested underlying mechanisms, biological factors influencing the microbiome-gut-brain axis in AD, and remaining questions in the field. Last, we will discuss potential therapeutic approaches to recondition the community of gut microbiota to alleviate disease progression. An ongoing exploration of the gut-brain axis and the development of microbiota-based therapies hold the potential for advancing AD management in the future.
Collapse
Affiliation(s)
- Dong-Oh Seo
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
| |
Collapse
|
23
|
Limanaqi F, Zecchini S, Saulle I, Strizzi S, Vanetti C, Garziano M, Cappelletti G, Parolin D, Caccia S, Trabattoni D, Fenizia C, Clerici M, Biasin M. Alpha-synuclein dynamics bridge Type-I Interferon response and SARS-CoV-2 replication in peripheral cells. Biol Res 2024; 57:2. [PMID: 38191441 PMCID: PMC10775536 DOI: 10.1186/s40659-023-00482-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 12/15/2023] [Indexed: 01/10/2024] Open
Abstract
BACKGROUND Increasing evidence suggests a double-faceted role of alpha-synuclein (α-syn) following infection by a variety of viruses, including SARS-CoV-2. Although α-syn accumulation is known to contribute to cell toxicity and the development and/or exacerbation of neuropathological manifestations, it is also a key to sustaining anti-viral innate immunity. Consistently with α-syn aggregation as a hallmark of Parkinson's disease, most studies investigating the biological function of α-syn focused on neural cells, while reports on the role of α-syn in periphery are limited, especially in SARS-CoV-2 infection. RESULTS Results herein obtained by real time qPCR, immunofluorescence and western blot indicate that α-syn upregulation in peripheral cells occurs as a Type-I Interferon (IFN)-related response against SARS-CoV-2 infection. Noteworthy, this effect mostly involves α-syn multimers, and the dynamic α-syn multimer:monomer ratio. Administration of excess α-syn monomers promoted SARS-CoV-2 replication along with downregulation of IFN-Stimulated Genes (ISGs) in epithelial lung cells, which was associated with reduced α-syn multimers and α-syn multimer:monomer ratio. These effects were prevented by combined administration of IFN-β, which hindered virus replication and upregulated ISGs, meanwhile increasing both α-syn multimers and α-syn multimer:monomer ratio in the absence of cell toxicity. Finally, in endothelial cells displaying abortive SARS-CoV-2 replication, α-syn multimers, and multimer:monomer ratio were not reduced following exposure to the virus and exogenous α-syn, suggesting that only productive viral infection impairs α-syn multimerization and multimer:monomer equilibrium. CONCLUSIONS Our study provides novel insights into the biology of α-syn, showing that its dynamic conformations are implicated in the innate immune response against SARS-CoV-2 infection in peripheral cells. In particular, our results suggest that promotion of non-toxic α-syn multimers likely occurs as a Type-I IFN-related biological response which partakes in the suppression of viral replication. Further studies are needed to replicate our findings in neuronal cells as well as animal models, and to ascertain the nature of such α-syn conformations.
Collapse
Affiliation(s)
- Fiona Limanaqi
- Department of Biomedical and Clinical Sciences, University of Milan, Via G.B. Grassi, Milan, Italy.
| | - Silvia Zecchini
- Department of Biomedical and Clinical Sciences, University of Milan, Via G.B. Grassi, Milan, Italy
| | - Irma Saulle
- Department of Biomedical and Clinical Sciences, University of Milan, Via G.B. Grassi, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Via Francesco Sforza, Milan, Italy
| | - Sergio Strizzi
- Department of Biomedical and Clinical Sciences, University of Milan, Via G.B. Grassi, Milan, Italy
| | - Claudia Vanetti
- Department of Biomedical and Clinical Sciences, University of Milan, Via G.B. Grassi, Milan, Italy
| | - Micaela Garziano
- Department of Biomedical and Clinical Sciences, University of Milan, Via G.B. Grassi, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Via Francesco Sforza, Milan, Italy
| | - Gioia Cappelletti
- Department of Biomedical and Clinical Sciences, University of Milan, Via G.B. Grassi, Milan, Italy
| | - Debora Parolin
- Department of Biomedical and Clinical Sciences, University of Milan, Via G.B. Grassi, Milan, Italy
| | - Sonia Caccia
- Department of Biomedical and Clinical Sciences, University of Milan, Via G.B. Grassi, Milan, Italy
| | - Daria Trabattoni
- Department of Biomedical and Clinical Sciences, University of Milan, Via G.B. Grassi, Milan, Italy
| | - Claudio Fenizia
- Department of Biomedical and Clinical Sciences, University of Milan, Via G.B. Grassi, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Via Francesco Sforza, Milan, Italy
| | - Mario Clerici
- Department of Pathophysiology and Transplantation, University of Milan, Via Francesco Sforza, Milan, Italy
- IRCCS Fondazione Don Carlo Gnocchi, 20148, Milan, Italy
| | - Mara Biasin
- Department of Biomedical and Clinical Sciences, University of Milan, Via G.B. Grassi, Milan, Italy.
| |
Collapse
|
24
|
James AS, Adil NA, Goltz D, Tangudu D, Chaudhari DS, Shukla R, Kumar V, Kumar A, Masternak MM, Holland P, Labyak C, Golden A, Dangiolo M, Arikawa AY, Kociolek J, Fraser A, Williams C, Agronin M, Aymat M, Jain S, Yadav H. Abnormalities in gut virome signatures linked with cognitive impairment in older adults. Gut Microbes 2024; 16:2431648. [PMID: 39676708 DOI: 10.1080/19490976.2024.2431648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 10/25/2024] [Accepted: 11/11/2024] [Indexed: 12/17/2024] Open
Abstract
Multiple emerging lines of evidence indicate that the microbiome contributes to aging and cognitive health. However, the roles of distinct microbial components, such as viruses (virome) and their interactions with bacteria (bacteriome), as well as their metabolic pathways (metabolome) in relation to aging and cognitive function, remain poorly understood. Here, we present proof-of-concept results from a pilot study using datasets (n = 176) from the Microbiome in Aging Gut and Brain (MiaGB) consortium, demonstrating that the human virome signature significantly differs across the aging continuum (60s vs. 70s vs. 80+ years of age) in older adults. We observed that the predominant virome signature was enriched with bacteriophages, which change considerably with aging continuum. Analyses of interactions between phages and the host bacteriome suggest that lytic or temperate relationships change distinctly across the aging continuum, as well as cognitive impairment. Interestingly, the phage-bacteriome-metabolome interactions develop unique patterns that are distinctly linked to aging and cognitive dysfunction in older adults. The phage-bacteriome interactions affect bacterial metabolic pathways, potentially impacting older adults' health, including the risk of cognitive decline and dementia. Further comprehension of these studies could provide opportunities to target the microbiome by developing phage therapies to improve aging and brain health in older adults.
Collapse
Affiliation(s)
- Adewale S James
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Center for Excellence in Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Noorul A Adil
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Center for Excellence in Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Dayna Goltz
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Center for Excellence in Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Divyani Tangudu
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Center for Excellence in Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Diptaraj S Chaudhari
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Center for Excellence in Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Rohit Shukla
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Center for Excellence in Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Vivek Kumar
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Center for Excellence in Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Ambuj Kumar
- Research Methodology and Biostatistics, Department of Internal Medicine, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Michal M Masternak
- School of Global Health Management and Informatics, University of Central Florida, Orlando, FL, USA
| | - Peter Holland
- Department of Neuroscience, FAU Schmidt College of Medicine/i-Health FAU, Boca Raton, FL, USA
| | - Corinne Labyak
- Department of Nutrition and Dietetics, University of North Florida, Jacksonville, FL, USA
| | - Adam Golden
- School of Global Health Management and Informatics, University of Central Florida, Orlando, FL, USA
| | - Mariana Dangiolo
- School of Global Health Management and Informatics, University of Central Florida, Orlando, FL, USA
| | - Andrea Y Arikawa
- Department of Nutrition and Dietetics, University of North Florida, Jacksonville, FL, USA
| | - Judyta Kociolek
- Department of Neuroscience, FAU Schmidt College of Medicine/i-Health FAU, Boca Raton, FL, USA
- Clinical Research Unit, Division of Research, Florida Atlantic University, Boca Raton, FL, USA
| | - Amoy Fraser
- School of Global Health Management and Informatics, University of Central Florida, Orlando, FL, USA
| | - Cynthia Williams
- School of Global Health Management and Informatics, University of Central Florida, Orlando, FL, USA
| | - Marc Agronin
- Behavioral Health, MIND Institute, Miami Jewish Health, Miami, FL, USA
| | - Mariolga Aymat
- Behavioral Health, MIND Institute, Miami Jewish Health, Miami, FL, USA
| | - Shalini Jain
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Center for Excellence in Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Hariom Yadav
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Center for Excellence in Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| |
Collapse
|
25
|
Barber AJ, del Genio CL, Swain AB, Pizzi EM, Watson SC, Tapiavala VN, Zanazzi GJ, Gaur AB. Age, Sex and Alzheimer's disease: A longitudinal study of 3xTg-AD mice reveals sex-specific disease trajectories and inflammatory responses mirrored in postmortem brains from Alzheimer's patients. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.23.573209. [PMID: 38187539 PMCID: PMC10769453 DOI: 10.1101/2023.12.23.573209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Background Aging and sex are major risk factors for developing late-onset Alzheimer's disease. Compared to men, women are not only nearly twice as likely to develop Alzheimer's, but they also experience worse neuropathological burden and cognitive decline despite living longer with the disease. It remains unclear how and when sex differences in biological aging emerge and contribute to Alzheimer's disease pathogenesis. We hypothesized that these differences lead to distinct pathological and molecular Alzheimer's disease signatures in males and females, which could be harnessed for therapeutic and biomarker development. Methods We aged male and female, 3xTg-AD and B6129 (WT) control mice across their respective lifespans while longitudinally collecting brain, liver, spleen, and plasma samples (n=3-8 mice per sex, strain, and age group). We performed histological analyses on all tissues and assessed neuropathological hallmarks of Alzheimer's disease, markers of hepatic inflammation, as well as splenic mass and morphology. Additionally, we measured concentrations of cytokines, chemokines, and growth factors in the plasma. We conducted RNA sequencing (RNA-Seq) analysis on bulk brain tissue and examined differentially expressed genes (DEGs) between 3xTg-AD and WT samples and across ages in each sex. We also examined DEGs between clinical Alzheimer's and control parahippocampal gyrus brain tissue samples from the Mount Sinai Brain Bank (MSBB) study in each sex. Results 3xTg-AD females significantly outlived 3xTg-AD males and exhibited progressive Alzheimer's neuropathology, while 3xTg-AD males demonstrated progressive hepatic inflammation, splenomegaly, circulating inflammatory proteins, and next to no Alzheimer's neuropathological hallmarks. Instead, 3xTg-AD males experienced an accelerated upregulation of immune-related gene expression in the brain relative to females, further suggesting distinct inflammatory disease trajectories between the sexes. Clinical investigations revealed that 3xTg-AD brain aging phenotypes are not an artifact of the animal model, and individuals with Alzheimer's disease develop similar sex-specific alterations in canonical pathways related to neuronal signaling and immune function. Interestingly, we observed greater upregulation of complement-related gene expression, and lipopolysaccharide (LPS) was predicted as the top upstream regulator of DEGs in diseased males of both species. Conclusions Our data demonstrate that chronic inflammation and complement activation are associated with increased mortality, revealing that age-related changes in immune response act as a primary driver of sex differences in Alzheimer's disease trajectories. We propose a model of disease pathogenesis in 3xTg-AD males in which aging and transgene-driven disease progression trigger an inflammatory response, mimicking the effects of LPS stimulation despite the absence of infection.
Collapse
Affiliation(s)
- Alicia J. Barber
- Department of Neurology, Geisel School of Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
- Integrative Neuroscience at Dartmouth, Dartmouth College, Hanover, NH, USA
| | - Carmen L. del Genio
- Department of Neurology, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| | | | - Elizabeth M. Pizzi
- The Jackson Laboratory, Bar Harbor, ME, USA
- Neuroscience Program, Graduate School of Biomedical Sciences, Tufts University, Boston, MA, USA
| | | | | | - George J. Zanazzi
- Department of Pathology, Geisel School of Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| | - Arti B. Gaur
- Department of Neurology, Geisel School of Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| |
Collapse
|
26
|
Cetin E, Mazzarino M, González-Mateo GT, Kopytina V, Meran S, Fraser D, López-Cabrera M, Labéta MO, Raby AC. Calprotectin blockade inhibits long-term vascular pathology following peritoneal dialysis-associated bacterial infection. Front Cell Infect Microbiol 2023; 13:1285193. [PMID: 38094743 PMCID: PMC10716465 DOI: 10.3389/fcimb.2023.1285193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 11/02/2023] [Indexed: 12/18/2023] Open
Abstract
Bacterial infections and the concurrent inflammation have been associated with increased long-term cardiovascular (CV) risk. In patients receiving peritoneal dialysis (PD), bacterial peritonitis is a common occurrence, and each episode further increases late CV mortality risk. However, the underlying mechanism(s) remains to be elucidated before safe and efficient anti-inflammatory interventions can be developed. Damage-Associated Molecular Patterns (DAMPs) have been shown to contribute to the acute inflammatory response to infections, but a potential role for DAMPs in mediating long-term vascular inflammation and CV risk following infection resolution in PD, has not been investigated. We found that bacterial peritonitis in mice that resolved within 24h led to CV disease-promoting systemic and vascular immune-mediated inflammatory responses that were maintained up to 28 days. These included higher blood proportions of inflammatory leukocytes displaying increased adhesion molecule expression, higher plasma cytokines levels, and increased aortic inflammatory and atherosclerosis-associated gene expression. These effects were also observed in infected nephropathic mice and amplified in mice routinely exposed to PD fluids. A peritonitis episode resulted in elevated plasma levels of the DAMP Calprotectin, both in PD patients and mice, here the increase was maintained up to 28 days. In vitro, the ability of culture supernatants from infected cells to promote key inflammatory and atherosclerosis-associated cellular responses, such as monocyte chemotaxis, and foam cell formation, was Calprotectin-dependent. In vivo, Calprotectin blockade robustly inhibited the short and long-term peripheral and vascular consequences of peritonitis, thereby demonstrating that targeting of the DAMP Calprotectin is a promising therapeutic strategy to reduce the long-lasting vascular inflammatory aftermath of an infection, notably PD-associated peritonitis, ultimately lowering CV risk.
Collapse
Affiliation(s)
- Esra Cetin
- Wales Kidney Research Unit, Division of Infection & Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Morgane Mazzarino
- Wales Kidney Research Unit, Division of Infection & Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Guadalupe T. González-Mateo
- Tissue and Organ Homeostasis Program, Centro de Biología Molecular Severo Ochoa – Consejo Superior de Investigaciones Científicas – Universidad Autónoma de Madrid (CBMSO-CSIC-UAM), Madrid, Spain
- Premium Research, S.L., Guadalajara, Spain
| | - Valeria Kopytina
- Tissue and Organ Homeostasis Program, Centro de Biología Molecular Severo Ochoa – Consejo Superior de Investigaciones Científicas – Universidad Autónoma de Madrid (CBMSO-CSIC-UAM), Madrid, Spain
| | - Soma Meran
- Wales Kidney Research Unit, Division of Infection & Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Donald Fraser
- Wales Kidney Research Unit, Division of Infection & Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Manuel López-Cabrera
- Tissue and Organ Homeostasis Program, Centro de Biología Molecular Severo Ochoa – Consejo Superior de Investigaciones Científicas – Universidad Autónoma de Madrid (CBMSO-CSIC-UAM), Madrid, Spain
| | - Mario O. Labéta
- Wales Kidney Research Unit, Division of Infection & Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Anne-Catherine Raby
- Wales Kidney Research Unit, Division of Infection & Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
27
|
Amadoro G, Latina V, Stigliano E, Micera A. COVID-19 and Alzheimer's Disease Share Common Neurological and Ophthalmological Manifestations: A Bidirectional Risk in the Post-Pandemic Future. Cells 2023; 12:2601. [PMID: 37998336 PMCID: PMC10670749 DOI: 10.3390/cells12222601] [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: 10/04/2023] [Revised: 11/07/2023] [Accepted: 11/08/2023] [Indexed: 11/25/2023] Open
Abstract
A growing body of evidence indicates that a neuropathological cross-talk takes place between the coronavirus disease 2019 (COVID-19) -the pandemic severe pneumonia that has had a tremendous impact on the global economy and health since three years after its outbreak in December 2019- and Alzheimer's Disease (AD), the leading cause of dementia among human beings, reaching 139 million by the year 2050. Even though COVID-19 is a primary respiratory disease, its causative agent, the so-called Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2), is also endowed with high neuro-invasive potential (Neurocovid). The neurological complications of COVID-19, resulting from the direct viral entry into the Central Nervous System (CNS) and/or indirect systemic inflammation and dysregulated activation of immune response, encompass memory decline and anosmia which are typically associated with AD symptomatology. In addition, patients diagnosed with AD are more vulnerable to SARS-CoV-2 infection and are inclined to more severe clinical outcomes. In the present review, we better elucidate the intimate connection between COVID-19 and AD by summarizing the involved risk factors/targets and the underlying biological mechanisms shared by these two disorders with a particular focus on the Angiotensin-Converting Enzyme 2 (ACE2) receptor, APOlipoprotein E (APOE), aging, neuroinflammation and cellular pathways associated with the Amyloid Precursor Protein (APP)/Amyloid beta (Aβ) and tau neuropathologies. Finally, the involvement of ophthalmological manifestations, including vitreo-retinal abnormalities and visual deficits, in both COVID-19 and AD are also discussed. Understanding the common physiopathological aspects linking COVID-19 and AD will pave the way to novel management and diagnostic/therapeutic approaches to cope with them in the post-pandemic future.
Collapse
Affiliation(s)
- Giuseppina Amadoro
- Institute of Translational Pharmacology (IFT), National Research Council (CNR), Via Fosso del Cavaliere 100, 00133 Rome, Italy;
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
| | - Valentina Latina
- Institute of Translational Pharmacology (IFT), National Research Council (CNR), Via Fosso del Cavaliere 100, 00133 Rome, Italy;
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
| | - Egidio Stigliano
- Area of Pathology, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Istituto di Anatomia Patologica, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy;
| | - Alessandra Micera
- Research and Development Laboratory for Biochemical, Molecular and Cellular Applications in Ophthalmological Sciences, IRCCS-Fondazione Bietti, Via Santo Stefano Rotondo, 6, 00184 Rome, Italy
| |
Collapse
|
28
|
Chatanaka MK, Sohaei D, Diamandis EP, Prassas I. Beyond the amyloid hypothesis: how current research implicates autoimmunity in Alzheimer's disease pathogenesis. Crit Rev Clin Lab Sci 2023; 60:398-426. [PMID: 36941789 DOI: 10.1080/10408363.2023.2187342] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 03/01/2023] [Indexed: 03/23/2023]
Abstract
The amyloid hypothesis has so far been at the forefront of explaining the pathogenesis of Alzheimer's Disease (AD), a progressive neurodegenerative disorder that leads to cognitive decline and eventual death. Recent evidence, however, points to additional factors that contribute to the pathogenesis of this disease. These include the neurovascular hypothesis, the mitochondrial cascade hypothesis, the inflammatory hypothesis, the prion hypothesis, the mutational accumulation hypothesis, and the autoimmunity hypothesis. The purpose of this review was to briefly discuss the factors that are associated with autoimmunity in humans, including sex, the gut and lung microbiomes, age, genetics, and environmental factors. Subsequently, it was to examine the rise of autoimmune phenomena in AD, which can be instigated by a blood-brain barrier breakdown, pathogen infections, and dysfunction of the glymphatic system. Lastly, it was to discuss the various ways by which immune system dysregulation leads to AD, immunomodulating therapies, and future directions in the field of autoimmunity and neurodegeneration. A comprehensive account of the recent research done in the field was extracted from PubMed on 31 January 2022, with the keywords "Alzheimer's disease" and "autoantibodies" for the first search input, and "Alzheimer's disease" with "IgG" for the second. From the first search, 19 papers were selected, because they contained recent research on the autoantibodies found in the biofluids of patients with AD. From the second search, four papers were selected. The analysis of the literature has led to support the autoimmune hypothesis in AD. Autoantibodies were found in biofluids (serum/plasma, cerebrospinal fluid) of patients with AD with multiple methods, including ELISA, Mass Spectrometry, and microarray analysis. Through continuous research, the understanding of the synergistic effects of the various components that lead to AD will pave the way for better therapeutic methods and a deeper understanding of the disease.
Collapse
Affiliation(s)
- Miyo K Chatanaka
- Department of Laboratory and Medicine Pathobiology, University of Toronto, Toronto, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada
| | - Dorsa Sohaei
- Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
| | - Eleftherios P Diamandis
- Department of Laboratory and Medicine Pathobiology, University of Toronto, Toronto, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada
- Department of Clinical Biochemistry, University Health Network, Toronto, Canada
| | - Ioannis Prassas
- Laboratory Medicine Program, University Health Network, Toronto, Canada
| |
Collapse
|
29
|
Piotrowski SL, Tucker A, Jacobson S. The elusive role of herpesviruses in Alzheimer's disease: current evidence and future directions. NEUROIMMUNE PHARMACOLOGY AND THERAPEUTICS 2023; 2:253-266. [PMID: 38013835 PMCID: PMC10474380 DOI: 10.1515/nipt-2023-0011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 06/26/2023] [Indexed: 11/29/2023]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia. While pathologic hallmarks, such as extracellular beta-amyloid plaques, are well-characterized in affected individuals, the pathogenesis that causes plaque formation and eventual cognitive decline is not well understood. A recent resurgence of the decades-old "infectious hypothesis" has garnered increased attention on the potential role that microbes may play in AD. In this theory, it is thought that pathogens such as viruses may act as seeds for beta-amyloid aggregation, ultimately leading to plaques. Interest in the infectious hypothesis has also spurred further investigation into additional characteristics of viral infection that may play a role in AD progression, such as neuroinflammation, latency, and viral DNA integration. While a flurry of research in this area has been recently published, with herpesviruses being of particular interest, the role of pathogens in AD remains controversial. In this review, the insights gained thus far into the possible role of herpesviruses in AD are summarized. The challenges and potential future directions of herpesvirus research in AD and dementia are also discussed.
Collapse
Affiliation(s)
- Stacey L. Piotrowski
- Viral Immunology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Comparative Biomedical Scientist Training Program, National Institutes of Health, Bethesda, MD, USA
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
| | - Allison Tucker
- Viral Immunology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Steven Jacobson
- Viral Immunology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
30
|
Chesney AD, Maiti B, Hansmann UHE. SARS-COV-2 spike protein fragment eases amyloidogenesis of α-synuclein. J Chem Phys 2023; 159:015103. [PMID: 37409768 PMCID: PMC10328560 DOI: 10.1063/5.0157331] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 06/19/2023] [Indexed: 07/07/2023] Open
Abstract
Parkinson's disease is accompanied by the presence of amyloids in the brain that are formed of α-synuclein chains. The correlation between COVID-19 and the onset of Parkinson's disease led to the idea that amyloidogenic segments in SARS-COV-2 proteins can induce aggregation of α-synuclein. Using molecular dynamic simulations, we show that the fragment FKNIDGYFKI of the spike protein, which is unique for SARS-COV-2, preferentially shifts the ensemble of α-synuclein monomer toward rod-like fibril seeding conformations and, at the same time, differentially stabilizes this polymorph over the competing twister-like structure. Our results are compared with earlier work relying on a different protein fragment that is not specific for SARS-COV-2.
Collapse
Affiliation(s)
- Andrew D. Chesney
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, USA
| | - Buddhadev Maiti
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, USA
| | - Ulrich H. E. Hansmann
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, USA
| |
Collapse
|
31
|
Olivera E, Sáez A, Carniglia L, Caruso C, Lasaga M, Durand D. Alzheimer's disease risk after COVID-19: a view from the perspective of the infectious hypothesis of neurodegeneration. Neural Regen Res 2023; 18:1404-1410. [PMID: 36571334 PMCID: PMC10075115 DOI: 10.4103/1673-5374.360273] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
In light of the rising evidence of the association between viral and bacterial infections and neurodegeneration, we aimed at revisiting the infectious hypothesis of Alzheimer's disease and analyzing the possible implications of COVID-19 neurological sequelae in long-term neurodegeneration. We wondered how SARS-CoV-2 could be related to the amyloid-β cascade and how it could lead to the pathological hallmarks of the disease. We also predict a paradigm change in clinical medicine, which now has a great opportunity to conduct prospective surveillance of cognitive sequelae and progression to dementia in people who suffered severe infections together with other risk factors for Alzheimer's disease.
Collapse
Affiliation(s)
- Eugenia Olivera
- Instituto de Investigaciones Biomédicas INBIOMED UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | - Albany Sáez
- Instituto de Investigaciones Biomédicas INBIOMED UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | - Lila Carniglia
- Instituto de Investigaciones Biomédicas INBIOMED UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | - Carla Caruso
- Instituto de Investigaciones Biomédicas INBIOMED UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | - Mercedes Lasaga
- Instituto de Investigaciones Biomédicas INBIOMED UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | - Daniela Durand
- Instituto de Investigaciones Biomédicas INBIOMED UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Argentina
| |
Collapse
|
32
|
Madnani RS. Alzheimer's disease: a mini-review for the clinician. Front Neurol 2023; 14:1178588. [PMID: 37426432 PMCID: PMC10325860 DOI: 10.3389/fneur.2023.1178588] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 06/02/2023] [Indexed: 07/11/2023] Open
Abstract
Alzheimer's disease (AD), the most common form of dementia, is a striking example of the connection between neurophysiological abnormalities and higher-order cognitive deficiencies. Since its initial description in 1906, research into the pathophysiology and etiology of AD has led to the illumination of an incredibly complex set of genetic and molecular mechanisms for the disease's progression, characterized by much more than the neuropathological hallmarks of beta-amyloid (Aβ) plaques and neurofibrillary tangles (NFTs). In this review, findings relating the neurodegeneration present in AD to its clinical presentation and treatment are summarized, with an emphasis on the interconnectedness of disease pathophysiology. Further, diagnostic guidelines are provided based on the National Institute on Aging-Alzheimer's Association (NIA-AA) workgroup's clinical recommendations. Through the dissemination of detailed but digestible open access resources such as this one, we can move towards an increase in the equity and accessibility of education for the modern clinician.
Collapse
|
33
|
Wang YR, Liang CR, Heng T, Zhang T, Hu XT, Long Y, Huang L, Dong B, Gao X, Deng J, Xu X, Yao XQ. Circulating antibodies to Helicobacter pylori are associated with biomarkers of neurodegeneration in cognitively intact adults. Asian J Psychiatr 2023; 86:103680. [PMID: 37352754 DOI: 10.1016/j.ajp.2023.103680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/13/2023] [Accepted: 06/17/2023] [Indexed: 06/25/2023]
Abstract
Helicobacter pylori (H. pylori) infection confers risk for Alzheimer's Disease (AD), with the mechanisms unknown. Infections are linked to the etiology of AD partly through modulating the humoral immunity post-infection. This study found increased plasma levels of tTau and pTau181 in H. Pylori infected individuals with intact cognition. Plasma antibodies to H. pylori were positively associated with Aβ40, Aβ42, tTau, and pTau181, adjusting for age, sex, education level, BMI, ApoE ε4 genotype, hypertension, diabetes mellitus, and hypercholesteremia. This study presents novel insights into the relationship between H. pylori infection and AD from an autoimmune perspective.
Collapse
Affiliation(s)
- Ye-Ran Wang
- Center of Health Management, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing 400042, China
| | - Chun-Rong Liang
- Department of Sleep and Psychology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing 400042, China
| | - Tian Heng
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Ting Zhang
- Center of Health Management, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing 400042, China
| | - Xiao-Tong Hu
- Center of Health Management, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing 400042, China
| | - Yan Long
- Center of Health Management, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing 400042, China
| | - Liang Huang
- Center of Health Management, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing 400042, China
| | - Bo Dong
- Center of Health Management, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing 400042, China
| | - Xia Gao
- Center of Health Management, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing 400042, China
| | - Juan Deng
- Center of Health Management, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing 400042, China
| | - Xia Xu
- Center of Health Management, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing 400042, China.
| | - Xiu-Qing Yao
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.
| |
Collapse
|
34
|
Chesney AD, Maiti B, Hansmann UHE. SARS-COV-2 Spike Protein Fragment eases Amyloidogenesis of α-Synuclein. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.06.539715. [PMID: 37214999 PMCID: PMC10197603 DOI: 10.1101/2023.05.06.539715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Parkinson's Disease is accompanied by presence of amyloids in the brain formed of α-synuclein chains. Correlation between COVID-19 and the onset of Parkinson's disease let to the idea that amyloidogenic segments in SARS-COV-2 proteins can induce aggregation of α-synuclein. Using molecular dynamic simulations, we show that the fragment FKNIDGYFKI of the spike protein, which is unique for SARS-COV-2, shifts preferentially the ensemble of α-synuclein monomer towards rod-like fibril seeding conformations, and at the same time stabilizes differentially this polymorph over the competing twister-like structure. Our results are compared with earlier work relying on a different protein fragment that is not specific for SARS-COV-2.
Collapse
|
35
|
Zhang LY, Wang DZ, Wang J, Guo L, Li BH, Wang JH. Associations of Serum Antimicrobial Peptide LL-37 with Longitudinal Cognitive Decline and Neurodegeneration Among Older Adults with Memory Complaints. J Alzheimers Dis 2023; 93:595-603. [PMID: 37066916 DOI: 10.3233/jad-230007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
BACKGROUND A potential role of the antimicrobial peptide LL-37, which is upregulated after infection, in the pathogenesis of Alzheimer's disease (AD) was identified. However, the clinical relevance of LL-37 in AD is not clear yet. OBJECTIVE This study aims to investigate the association of circulating LL-37 with longitudinal cognitive decline and neurodegeneration among older adults with memory complaints. METHODS This cohort study recruited 357 older adults with memory complaints. Participants were followed-up for two years and the cognitive functions were assessed using the Mini-Mental State Examination (MMSE). Serum LL-37, pTau181, and tTau levels were determined at baseline. Associations of baseline LL-37 with longitudinal cognitive decline and change of neurodegenerative biomarkers were analyzed. RESULTS No difference was found in the slope of longitudinal cognitive decline during follow-up between the low and high LL-37 group, adjusting for age, sex, education, body mass index, APOE ɛ4 carrier status, comorbidities, and baseline MMSE scores (difference in slope: 0.226, 95% CI: -0.169 to 0.621). Higher LL-37 levels were associated with longitudinal cognitive decline, as indicated by a decrease of MMSE scores of 3 points or above during follow-up (RR = 2.11, 95% CI: 1.32 to 3.38). The high LL-37 group had larger slopes of the increase in neurofilament light (difference in slope: 3.759, 95% CI: 2.367 to 5.152) and pTau181 (difference in slope: 0.325, 95% CI: 0.151 to 0.499) than the low LL-37 group. CONCLUSION These findings support an association of the antimicrobial peptide LL-37 with AD from a clinical perspective.
Collapse
Affiliation(s)
- Li-Ya Zhang
- Department of Neurology, 363 Hospital, Chengdu, Sichuan Province, China
| | - Duo-Zi Wang
- Department of Neurology, the Affiliated Hospital of University of Electronic Science and Technology, Sichuan Provincial People's Hospital, Chengdu, SichuanProvince, China
| | - Jian Wang
- Department of Neurology, Ya'an People's Hospital, Ya'an, Sichuan Province, China
| | - Lei Guo
- Department of Neurology, the Affiliated Hospital of University of Electronic Science and Technology, Sichuan Provincial People's Hospital, Chengdu, SichuanProvince, China
| | - Bing-Hu Li
- Department of Neurology, the Affiliated Hospital of University of Electronic Science and Technology, Sichuan Provincial People's Hospital, Chengdu, SichuanProvince, China
| | - Jian-Hong Wang
- Department of Neurology, the Affiliated Hospital of University of Electronic Science and Technology, Sichuan Provincial People's Hospital, Chengdu, SichuanProvince, China
| |
Collapse
|
36
|
Xie J, Cools L, Van Imschoot G, Van Wonterghem E, Pauwels MJ, Vlaeminck I, De Witte C, EL Andaloussi S, Wierda K, De Groef L, Haesebrouck F, Van Hoecke L, Vandenbroucke RE. Helicobacter pylori-derived outer membrane vesicles contribute to Alzheimer's disease pathogenesis via C3-C3aR signalling. J Extracell Vesicles 2023; 12:e12306. [PMID: 36792546 PMCID: PMC9931688 DOI: 10.1002/jev2.12306] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 01/06/2023] [Accepted: 01/18/2023] [Indexed: 02/17/2023] Open
Abstract
The gut microbiota represents a diverse and dynamic population of microorganisms that can influence the health of the host. Increasing evidence supports the role of the gut microbiota as a key player in the pathogenesis of neurodegenerative diseases, including Alzheimer's disease (AD). Unfortunately, the mechanisms behind the interplay between gut pathogens and AD are still elusive. It is known that bacteria-derived outer membrane vesicles (OMVs) act as natural carriers of virulence factors that are central players in the pathogenesis of the bacteria. Helicobacter pylori (H. pylori) is a common gastric pathogen and H. pylori infection has been associated with an increased risk to develop AD. Here, we are the first to shed light on the role of OMVs derived from H. pylori on the brain in healthy conditions and on disease pathology in the case of AD. Our results reveal that H. pylori OMVs can cross the biological barriers, eventually reaching the brain. Once in the brain, these OMVs are taken up by astrocytes, which induce activation of glial cells and neuronal dysfunction, ultimately leading to exacerbated amyloid-β pathology and cognitive decline. Mechanistically, we identified a critical role for the complement component 3 (C3)-C3a receptor (C3aR) signalling in mediating the interaction between astrocytes, microglia and neurons upon the presence of gut H. pylori OMVs. Taken together, our study reveals that H. pylori has a detrimental effect on brain functionality and accelerates AD development via OMVs and C3-C3aR signalling.
Collapse
Affiliation(s)
- Junhua Xie
- VIB Center for Inflammation ResearchVIBGhentBelgium
- Department of Biomedical Molecular BiologyGhent UniversityGhentBelgium
- Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary MedicineGhent UniversityMerelbekeBelgium
| | - Lien Cools
- VIB Center for Inflammation ResearchVIBGhentBelgium
- Department of Biomedical Molecular BiologyGhent UniversityGhentBelgium
- Cellular Communication and Neurodegeneration Research Group, Department of Biology, Leuven Brain InstituteKU LeuvenLeuvenBelgium
| | - Griet Van Imschoot
- VIB Center for Inflammation ResearchVIBGhentBelgium
- Department of Biomedical Molecular BiologyGhent UniversityGhentBelgium
| | - Elien Van Wonterghem
- VIB Center for Inflammation ResearchVIBGhentBelgium
- Department of Biomedical Molecular BiologyGhent UniversityGhentBelgium
| | - Marie J. Pauwels
- VIB Center for Inflammation ResearchVIBGhentBelgium
- Department of Biomedical Molecular BiologyGhent UniversityGhentBelgium
| | - Ine Vlaeminck
- VIB Center for Brain & Disease ResearchElectrophysiology Expertise UnitLeuvenBelgium
- KU Leuven ‐ Department of NeurosciencesLeuvenBelgium
| | - Chloë De Witte
- Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary MedicineGhent UniversityMerelbekeBelgium
| | | | - Keimpe Wierda
- VIB Center for Brain & Disease ResearchElectrophysiology Expertise UnitLeuvenBelgium
- KU Leuven ‐ Department of NeurosciencesLeuvenBelgium
| | - Lies De Groef
- Cellular Communication and Neurodegeneration Research Group, Department of Biology, Leuven Brain InstituteKU LeuvenLeuvenBelgium
| | - Freddy Haesebrouck
- Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary MedicineGhent UniversityMerelbekeBelgium
| | - Lien Van Hoecke
- VIB Center for Inflammation ResearchVIBGhentBelgium
- Department of Biomedical Molecular BiologyGhent UniversityGhentBelgium
| | - Roosmarijn E. Vandenbroucke
- VIB Center for Inflammation ResearchVIBGhentBelgium
- Department of Biomedical Molecular BiologyGhent UniversityGhentBelgium
| |
Collapse
|
37
|
Rudnicka-Drożak E, Drożak P, Mizerski G, Zaborowski T, Ślusarska B, Nowicki G, Drożak M. Links between COVID-19 and Alzheimer's Disease-What Do We Already Know? INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:2146. [PMID: 36767513 PMCID: PMC9915236 DOI: 10.3390/ijerph20032146] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/21/2023] [Accepted: 01/22/2023] [Indexed: 06/18/2023]
Abstract
Alzheimer's disease (AD) is a life-changing condition whose etiology is explained by several hypotheses. Recently, a new virus contributed to the evidence of viral involvement in AD: the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which causes the COVID-19 coronavirus disease. AD was found to be one of the most common COVID-19 comorbidities, and it was found to increase mortality from this disease as well. Moreover, AD patients were observed to present with the distinct clinical features of COVID-19, with delirium being prevalent in this group. The SARS-CoV-2 virus enters host cells through the angiotensin-converting enzyme 2 (ACE2) receptor. ACE2 is overexpressed in brains with AD, which thus increases the viral invasion. Furthermore, the inhibition of the ACE2 receptor by the SARS-CoV-2 virus may also decrease the brain-derived neurotrophic factor (BDNF), contributing to neurodegeneration. The ApoE ε4 allele, which increases the risk of AD, was found to facilitate the SARS-CoV-2 entry into cells. Furthermore, the neuroinflammation and oxidative stress existing in AD patients enhance the inflammatory response associated with COVID-19. Moreover, pandemic and associated social distancing measures negatively affected the mental health, cognitive function, and neuro-psychiatric symptoms of AD patients. This review comprehensively covers the links between COVID-19 and Alzheimer's disease, including clinical presentation, molecular mechanisms, and the effects of social distancing.
Collapse
Affiliation(s)
- Ewa Rudnicka-Drożak
- Chair and Department of Family Medicine, Medical University of Lublin, Langiewicza 6a, 20-035 Lublin, Poland
| | - Paulina Drożak
- Student Scientific Society, Chair and Department of Family Medicine, Medical University of Lublin, Langiewicza 6a, 20-035 Lublin, Poland
| | - Grzegorz Mizerski
- Chair and Department of Family Medicine, Medical University of Lublin, Langiewicza 6a, 20-035 Lublin, Poland
| | - Tomasz Zaborowski
- Chair and Department of Family Medicine, Medical University of Lublin, Langiewicza 6a, 20-035 Lublin, Poland
| | - Barbara Ślusarska
- Department of Family and Geriatric Nursing, Faculty of Health Sciences, Medical University of Lublin, 20-081 Lublin, Poland
| | - Grzegorz Nowicki
- Department of Family and Geriatric Nursing, Faculty of Health Sciences, Medical University of Lublin, 20-081 Lublin, Poland
| | - Martyna Drożak
- Student Scientific Society, Chair and Department of Family Medicine, Medical University of Lublin, Langiewicza 6a, 20-035 Lublin, Poland
| |
Collapse
|
38
|
Green R, Mayilsamy K, McGill AR, Martinez TE, Chandran B, Blair LJ, Bickford PC, Mohapatra SS, Mohapatra S. SARS-CoV-2 infection increases the gene expression profile for Alzheimer's disease risk. Mol Ther Methods Clin Dev 2022; 27:217-229. [PMID: 36187720 PMCID: PMC9508696 DOI: 10.1016/j.omtm.2022.09.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 09/21/2022] [Indexed: 02/02/2023]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has caused over 600,000,000 infections globally thus far. Up to 30% of individuals with mild to severe disease develop long COVID, exhibiting diverse neurologic symptoms including dementias. However, there is a paucity of knowledge of molecular brain markers and whether these can precipitate the onset of Alzheimer's disease (AD). Herein, we report the brain gene expression profiles of severe COVID-19 patients showing increased expression of innate immune response genes and genes implicated in AD pathogenesis. The use of a mouse-adapted strain of SARS-CoV-2 (MA10) in an aged mouse model shows evidence of viral neurotropism, prolonged viral infection, increased expression of tau aggregator FKBP51, interferon-inducible gene Ifi204, and complement genes C4 and C5AR1. Brain histopathology shows AD signatures including increased tau-phosphorylation, tau-oligomerization, and α-synuclein expression in aged MA10 infected mice. The results of gene expression profiling of SARS-CoV-2-infected and AD brains and studies in the MA10 aged mouse model taken together, for the first time provide evidence suggesting that SARS-CoV-2 infection alters expression of genes in the brain associated with the development of AD. Future studies of common molecular markers in SARS-CoV-2 infection and AD could be useful for developing novel therapies targeting AD.
Collapse
Affiliation(s)
- Ryan Green
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- James A Haley VA Hospital, Tampa, FL 33612, USA
| | - Karthick Mayilsamy
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- James A Haley VA Hospital, Tampa, FL 33612, USA
| | - Andrew R. McGill
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- James A Haley VA Hospital, Tampa, FL 33612, USA
| | - Taylor E. Martinez
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- James A Haley VA Hospital, Tampa, FL 33612, USA
| | - Bala Chandran
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Laura J. Blair
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- Byrd Alzheimer’s Research Institute, University of South Florida, Tampa, FL 33613, USA
- James A Haley VA Hospital, Tampa, FL 33612, USA
| | - Paula C. Bickford
- Center of Excellence for Aging and Brain Repair, Departments of Neurosurgery and Brain Repair, and Molecular Pharmacology and Physiology, Morsani College of Medicine, Tampa, FL 33613, USA
- James A Haley VA Hospital, Tampa, FL 33612, USA
| | - Shyam S. Mohapatra
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- James A Haley VA Hospital, Tampa, FL 33612, USA
| | - Subhra Mohapatra
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- James A Haley VA Hospital, Tampa, FL 33612, USA
| |
Collapse
|
39
|
Huynh QS, Elangovan S, Holsinger RMD. Non-Pharmacological Therapeutic Options for the Treatment of Alzheimer's Disease. Int J Mol Sci 2022; 23:11037. [PMID: 36232336 PMCID: PMC9570337 DOI: 10.3390/ijms231911037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 09/10/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022] Open
Abstract
Alzheimer's disease is a growing global crisis in need of urgent diagnostic and therapeutic strategies. The current treatment strategy mostly involves immunotherapeutic medications that have had little success in halting disease progress. Hypotheses for pathogenesis and development of AD have been expanded to implicate both organ systems as well as cellular reactions. Non-pharmacologic interventions ranging from minimally to deeply invasive have attempted to address these diverse contributors to AD. In this review, we aim to delineate mechanisms underlying such interventions while attempting to provide explanatory links between the observed differences in disease states and postulated metabolic or structural mechanisms of change. The techniques discussed are not an exhaustive list of non-pharmacological interventions against AD but provide a foundation to facilitate a deeper understanding of the area of study.
Collapse
Affiliation(s)
- Quy-Susan Huynh
- Laboratory of Molecular Neuroscience and Dementia, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
- Neuroscience, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Shalini Elangovan
- Laboratory of Molecular Neuroscience and Dementia, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
| | - R. M. Damian Holsinger
- Laboratory of Molecular Neuroscience and Dementia, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
- Neuroscience, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
40
|
Herpes Simplex Virus Type 1 Preferentially Enhances Neuro-Inflammation and Senescence in Brainstem of Female Mice. J Virol 2022; 96:e0108122. [PMID: 35975996 PMCID: PMC9472638 DOI: 10.1128/jvi.01081-22] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Following acute infection, herpes simplex virus 1 (HSV-1) establishes lifelong latency in neurons. The latency associated transcript (LAT) is the only viral gene abundantly expressed during latency. Wild-type (WT) HSV-1 reactivates more efficiently than LAT mutants because LAT promotes establishment and maintenance of latency. While sensory neurons in trigeminal ganglia (TG) are important sites for latency, brainstem is also a site for latency and reactivation from latency. The principal sensory nucleus of the spinal trigeminal tract (Pr5) likely harbors latent HSV-1 because it receives afferent inputs from TG. The locus coeruleus (LC), an adjacent brainstem region, sends axonal projections to cortical structures and is indirectly linked to Pr5. Senescent cells accumulate in the nervous system during aging and accelerate neurodegenerative processes. Generally senescent cells undergo irreversible cell cycle arrest and produce inflammatory cytokines and chemokines. Based on these observations, we hypothesized HSV-1 influences senescence and inflammation in Pr5 and LC of latently infected mice. This hypothesis was tested using a mouse model of infection. Strikingly, female but not age-matched male mice latently infected with a LAT null mutant (dLAT2903) exhibited significantly higher levels of senescence markers and inflammation in LC, including cell cycle inhibitor p16, NLRP3 (NOD-, LRR- and pyrin domain-containing protein 3), IL-1α, and IL-β. Conversely, Pr5 in female but not male mice latently infected with WT HSV-1 or dLAT2903 exhibited enhanced expression of important inflammatory markers. The predilection of HSV-1 to induce senescence and inflammation in key brainstem regions of female mice infers that enhanced neurodegeneration occurs. IMPORTANCE HSV-1 (herpes simplex virus 1), an important human pathogen, establishes lifelong latency in neurons in trigeminal ganglia and the central nervous system. In contrast to productive infection, the only viral transcript abundantly expressed in latently infected neurons is the latency associated transcript (LAT). The brainstem, including principal sensory nucleus of the spinal trigeminal tract (Pr5) and locus coeruleus (LC), may expedite HSV-1 spread from trigeminal ganglia to the brain. Enhanced senescence and expression of key inflammatory markers were detected in LC of female mice latently infected with a LAT null mutant (dLAT2903) relative to age-matched male or female mice latently infected with wild-type HSV-1. Conversely, wild-type HSV-1 and dLAT2903 induced higher levels of senescence and inflammatory markers in Pr5 of latently infected female mice. In summary, enhanced inflammation and senescence in LC and Pr5 of female mice latently infected with HSV-1 are predicted to accelerate neurodegeneration.
Collapse
|
41
|
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
|
42
|
Li S, Le W. Editorial: Viral infection and brain diseases. Brain Res Bull 2022; 188:108-109. [PMID: 35870738 PMCID: PMC9299986 DOI: 10.1016/j.brainresbull.2022.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Affiliation(s)
- Song Li
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Weidong Le
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, China; Institute of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China; Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, Sichuan Province, China.
| |
Collapse
|
43
|
Nikolaeva NS, Yandulova EY, Aleksandrova YR, Starikov AS, Neganova ME. The Role of a Pathological Interaction between β-amyloid and Mitochondria in the Occurrence and Development of Alzheimer's Disease. Acta Naturae 2022; 14:19-34. [PMID: 36348714 PMCID: PMC9611857 DOI: 10.32607/actanaturae.11723] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 07/05/2022] [Indexed: 11/20/2022] Open
Abstract
Alzheimer's disease (AD) is one of the most common neurodegenerative diseases in existence. It is characterized by an impaired cognitive function that is due to a progressive loss of neurons in the brain. Extracellular β-amyloid (Aβ) plaques are the main pathological features of the disease. In addition to abnormal protein aggregation, increased mitochondrial fragmentation, altered expression of the genes involved in mitochondrial biogenesis, disruptions in the ER-mitochondria interaction, and mitophagy are observed. Reactive oxygen species are known to affect Aβ expression and aggregation. In turn, oligomeric and aggregated Aβ cause mitochondrial disorders. In this review, we summarize available knowledge about the pathological effects of Aβ on mitochondria and the potential molecular targets associated with proteinopathy and mitochondrial dysfunction for the pharmacological treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- N. S. Nikolaeva
- Federal State Budgetary Institution of Science Institute of Physiologically Active Compounds of the Russian Academy of Sciences, Chernogolovka, 142432 Russia
| | - E. Yu. Yandulova
- Federal State Budgetary Institution of Science Institute of Physiologically Active Compounds of the Russian Academy of Sciences, Chernogolovka, 142432 Russia
| | - Yu. R. Aleksandrova
- Federal State Budgetary Institution of Science Institute of Physiologically Active Compounds of the Russian Academy of Sciences, Chernogolovka, 142432 Russia
| | - A. S. Starikov
- Federal State Budgetary Institution of Science Institute of Physiologically Active Compounds of the Russian Academy of Sciences, Chernogolovka, 142432 Russia
| | - M. E. Neganova
- Federal State Budgetary Institution of Science Institute of Physiologically Active Compounds of the Russian Academy of Sciences, Chernogolovka, 142432 Russia
| |
Collapse
|
44
|
Trevisan C, Siviero P, Limongi F, Noale M, Maggi S. Health-Related Predictors of Changes in Cognitive Status in Community-Dwelling Older Individuals. Front Aging Neurosci 2022; 14:876359. [PMID: 35795233 PMCID: PMC9252613 DOI: 10.3389/fnagi.2022.876359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 05/23/2022] [Indexed: 12/02/2022] Open
Abstract
Given the rising numbers of older people living with dementia, this study focuses on identifying modifiable health-related factors associated with changes in cognitive status. The predictors of 1-year conversion from Preserved Cognitive Health (PCH) and Mild Cognitive Impairment (MCI) in older adults were evaluated. Two logistic regression models were performed on data from an Italian multicenter population-based study; both included sociodemographic factors, family history of dementia (FHD), risk behaviors, and depressive symptoms. The first model considered also disease clusters, while the second one included diseases' number and biochemical parameters. The sample involved 459 participants (61.4% women, median age 75 years). Of the 80 PCH individuals at baseline, after 1 year 35 (43.8%) were stable, 44 (55.0%) progressed to MCI, none to dementia, and one to unclassified status. Of the 379 MCI participants at baseline, after 1 year 281 (74.1%) remained stable, 38 (10.0%) reverted to PCH, 15 (4.0%) progressed to dementia, and 45 (11.9%) become unclassifiable. Hypertension/bone and joint diseases cluster was the only predictor of PCH progression to MCI; age and depression were associated with MCI progression to dementia; FHD was associated with MCI reversion to PCH. More diseases and fewer white blood cells were associated with MCI progression to dementia; more diseases and lower platelets were associated with the transition from MCI to unclassifiable; higher Na and lower TSH levels were associated with MCI reversion. The treatment or management of some chronic conditions and electrolyte imbalances may help attenuate cognitive deterioration in older adults with no or MCI.
Collapse
Affiliation(s)
- Caterina Trevisan
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
- Neuroscience Institute, Aging Branch, National Research Council, Padova, Italy
| | - Paola Siviero
- Neuroscience Institute, Aging Branch, National Research Council, Padova, Italy
| | - Federica Limongi
- Neuroscience Institute, Aging Branch, National Research Council, Padova, Italy
| | - Marianna Noale
- Neuroscience Institute, Aging Branch, National Research Council, Padova, Italy
| | - Stefania Maggi
- Neuroscience Institute, Aging Branch, National Research Council, Padova, Italy
| |
Collapse
|
45
|
Xiao J, Savonenko A, Yolken RH. Strain-specific pre-existing immunity: A key to understanding the role of chronic Toxoplasma infection in cognition and Alzheimer's diseases? Neurosci Biobehav Rev 2022; 137:104660. [PMID: 35405182 DOI: 10.1016/j.neubiorev.2022.104660] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/31/2022] [Accepted: 04/06/2022] [Indexed: 12/22/2022]
Abstract
Toxoplasma exposure can elicit cellular and humoral immune responses. In the case of chronic Toxoplasma infection, these immune responses are long-lasting. Some studies suggest that pre-existing immunity from Toxoplasma infection can shape immune responses and resistance to other pathogens and brain insults later in life. Much evidence has been generated suggesting Toxoplasma infection may contribute to cognitive impairment in the elderly. However, there have also been studies that disagree with the conclusion. Toxoplasma has many strain types, with virulence being the most notable difference. There is also considerable variation in the outcomes following Toxoplasma exposure ranging from resolved to persistent infection. Therefore, the brain microenvironment, particularly cellular constituents, differs based on the infecting strain (virulent versus hypovirulent) and infection stage (resolved versus persistent). Such difference might play a critical role in determining the outcome of the host on subsequent challengings to the brain. The ability of Toxoplasma strains to set up distinct stages for neurodegenerative pathology through varying degrees of virulence provides unique experimental tools for characterizing these pathways.
Collapse
Affiliation(s)
- Jianchun Xiao
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA.
| | - Alena Savonenko
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Robert H Yolken
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
46
|
Bruno F, Malvaso A, Canterini S, Bruni AC. Antimicrobial Peptides (AMPs) in the Pathogenesis of Alzheimer's Disease: Implications for Diagnosis and Treatment. Antibiotics (Basel) 2022; 11:726. [PMID: 35740133 PMCID: PMC9220182 DOI: 10.3390/antibiotics11060726] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 05/25/2022] [Accepted: 05/26/2022] [Indexed: 02/04/2023] Open
Abstract
Alzheimer's disease (AD) represents the most frequent type of dementia in elderly people. There are two major forms of the disease: sporadic (SAD)-whose causes are not completely understood-and familial (FAD)-with clear autosomal dominant inheritance. The two main hallmarks of AD are extracellular deposits of amyloid-beta (Aβ) peptide and intracellular deposits of the hyperphosphorylated form of the tau protein (P-tau). An ever-growing body of research supports the infectious hypothesis of sporadic forms of AD. Indeed, it has been documented that some pathogens, such as herpesviruses and certain bacterial species, are commonly present in AD patients, prompting recent clinical research to focus on the characterization of antimicrobial peptides (AMPs) in this pathology. The literature also demonstrates that Aβ can be considered itself as an AMP; thus, representing a type of innate immune defense peptide that protects the host against a variety of pathogens. Beyond Aβ, other proteins with antimicrobial activity, such as lactoferrin, defensins, cystatins, thymosin β4, LL37, histatin 1, and statherin have been shown to be involved in AD. Here, we summarized and discussed these findings and explored the diagnostic and therapeutic potential of AMPs in AD.
Collapse
Affiliation(s)
- Francesco Bruno
- Regional Neurogenetic Centre (CRN), Department of Primary Care, ASP Catanzaro, 88046 Lamezia Terme, Italy
- Association for Neurogenetic Research (ARN), 88046 Lamezia Terme, Italy;
| | - Antonio Malvaso
- Neurology Unit, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy;
| | - Sonia Canterini
- Division of Neuroscience, Department of Psychology, University La Sapienza, 00158 Rome, Italy;
| | | |
Collapse
|
47
|
Park JH, Kang I, Lee HK. γδ T Cells in Brain Homeostasis and Diseases. Front Immunol 2022; 13:886397. [PMID: 35693762 PMCID: PMC9181321 DOI: 10.3389/fimmu.2022.886397] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/25/2022] [Indexed: 11/30/2022] Open
Abstract
γδ T cells are a distinct subset of T cells expressing γδ T cell receptor (TCR) rather than αβTCR. Since their discovery, the critical roles of γδ T cells in multiple physiological systems and diseases have been investigated. γδ T cells are preferentially located at mucosal surfaces, such as the gut, although a small subset of γδ T cells can circulate the blood. Additionally, a subset of γδ T cells reside in the meninges in the central nervous system. Recent findings suggest γδ T cells in the meninges have critical roles in brain function and homeostasis. In addition, several lines of evidence have shown γδ T cells can infiltrate the brain parenchyma and regulate inflammatory responses in multiple diseases, including neurodegenerative diseases. Although the importance of γδ T cells in the brain is well established, their roles are still incompletely understood due to the complexity of their biology. Because γδ T cells rapidly respond to changes in brain status and regulate disease progression, understanding the role of γδ T cells in the brain will provide critical information that is essential for interpreting neuroimmune modulation. In this review, we summarize the complex role of γδ T cells in the brain and discuss future directions for research.
Collapse
|
48
|
Jana AK, Lander CW, Chesney AD, Hansmann UHE. Effect of an Amyloidogenic SARS-COV-2 Protein Fragment on α-Synuclein Monomers and Fibrils. J Phys Chem B 2022; 126:3648-3658. [PMID: 35580331 PMCID: PMC9186263 DOI: 10.1021/acs.jpcb.2c01254] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Aggregates of α-synuclein are thought to be the disease-causing agent in Parkinson's disease. Various case studies have hinted at a correlation between COVID-19 and the onset of Parkinson's disease. For this reason, we use molecular dynamics simulations to study whether amyloidogenic regions in SARS-COV-2 proteins can initiate and modulate aggregation of α-synuclein. As an example, we choose the nine-residue fragment SFYVYSRVK (SK9), located on the C-terminal of the envelope protein of SARS-COV-2. We probe how the presence of SK9 affects the conformational ensemble of α-synuclein monomers and the stability of two resolved fibril polymorphs. We find that the viral protein fragment SK9 may alter α-synuclein amyloid formation by shifting the ensemble toward aggregation-prone and preferentially rod-like fibril seeding conformations. However, SK9 has only a small effect on the stability of pre-existing or newly formed fibrils. A potential mechanism and key residues for potential virus-induced amyloid formation are described.
Collapse
Affiliation(s)
- Asis K Jana
- Department of Chemistry & Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Chance W Lander
- Department of Chemistry & Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Andrew D Chesney
- Department of Chemistry & Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Ulrich H E Hansmann
- Department of Chemistry & Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, United States
| |
Collapse
|
49
|
Silva J, Patricio F, Patricio-Martínez A, Santos-López G, Cedillo L, Tizabi Y, Limón ID. Neuropathological Aspects of SARS-CoV-2 Infection: Significance for Both Alzheimer's and Parkinson's Disease. Front Neurosci 2022; 16:867825. [PMID: 35592266 PMCID: PMC9111171 DOI: 10.3389/fnins.2022.867825] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 04/14/2022] [Indexed: 01/08/2023] Open
Abstract
Evidence suggests that SARS-CoV-2 entry into the central nervous system can result in neurological and/or neurodegenerative diseases. In this review, routes of SARS-Cov-2 entry into the brain via neuroinvasive pathways such as transcribrial, ocular surface or hematogenous system are discussed. It is argued that SARS-Cov-2-induced cytokine storm, neuroinflammation and oxidative stress increase the risk of developing neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease. Further studies on the effects of SARS-CoV-2 and its variants on protein aggregation, glia or microglia activation, and blood-brain barrier are warranted.
Collapse
Affiliation(s)
- Jaime Silva
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Felipe Patricio
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Aleidy Patricio-Martínez
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
- Facultad de Ciencias Biológicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Gerardo Santos-López
- Laboratorio de Biología Molecular y Virología, Centro de Investigación Biomédica de Oriente, Instituto Mexicano del Seguro Social, Atlixco, Mexico
| | - Lilia Cedillo
- Centro de Detección Biomolecular, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, United States
| | - Ilhuicamina Daniel Limón
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| |
Collapse
|
50
|
Nelson AR. Peripheral Pathways to Neurovascular Unit Dysfunction, Cognitive Impairment, and Alzheimer’s Disease. Front Aging Neurosci 2022; 14:858429. [PMID: 35517047 PMCID: PMC9062225 DOI: 10.3389/fnagi.2022.858429] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 03/03/2022] [Indexed: 12/11/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common form of dementia. It was first described more than a century ago, and scientists are acquiring new data and learning novel information about the disease every day. Although there are nuances and details continuously being unraveled, many key players were identified in the early 1900’s by Dr. Oskar Fischer and Dr. Alois Alzheimer, including amyloid-beta (Aβ), tau, vascular abnormalities, gliosis, and a possible role of infections. More recently, there has been growing interest in and appreciation for neurovascular unit dysfunction that occurs early in mild cognitive impairment (MCI) before and independent of Aβ and tau brain accumulation. In the last decade, evidence that Aβ and tau oligomers are antimicrobial peptides generated in response to infection has expanded our knowledge and challenged preconceived notions. The concept that pathogenic germs cause infections generating an innate immune response (e.g., Aβ and tau produced by peripheral organs) that is associated with incident dementia is worthwhile considering in the context of sporadic AD with an unknown root cause. Therefore, the peripheral amyloid hypothesis to cognitive impairment and AD is proposed and remains to be vetted by future research. Meanwhile, humans remain complex variable organisms with individual risk factors that define their immune status, neurovascular function, and neuronal plasticity. In this focused review, the idea that infections and organ dysfunction contribute to Alzheimer’s disease, through the generation of peripheral amyloids and/or neurovascular unit dysfunction will be explored and discussed. Ultimately, many questions remain to be answered and critical areas of future exploration are highlighted.
Collapse
|