1
|
Liu YL, Xu S, Xu X, Tang Y, Shao J, Chen J, Li YG. Integrating network pharmacology and multi-omics to explore the mechanism of Callicarpa kwangtungensis Chun in ameliorating Alzheimer's disease pathology in APP/PS1 mice. JOURNAL OF ETHNOPHARMACOLOGY 2025; 339:119148. [PMID: 39586557 DOI: 10.1016/j.jep.2024.119148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 11/12/2024] [Accepted: 11/21/2024] [Indexed: 11/27/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Callicarpa kwangtungensis Chun (CK) is a traditional herb for the treatment of blood stasis, hemostasis, anti-inflammation, and antidepressant. Previous studies have showen that CK extract has significant anti-neuroinflammatory activity. However, the mechanism by which it treats AD is still unclear. AIM OF STUDY This study aimed to investigate the effects and mechanisms of CK in ameliorating AD pathology using in vivo and in vitro models, supported by a multi-omics analysis approach. MATERIALS AND METHODS The chemical composition of CK was characterized using UPLC-QE Plus-MS/MS. The effects and mechanisms of CK on AD pathology were then investigated using APP/PS1 mice and BV2 and HT22 cell models, with comprehensive insights provided by network pharmacology, transcriptomics, and metabolomics analyses. RESULTS This study is the first to report the identification of 146 compounds from CK. CK administration led to significant improvements in cognitive function, reduced amyloid-beta and neurofibrillary tangle formation, and inhibited the activation of microglia and astrocytes in APP/PS1 mice. Comprehensive analyses suggest that CK may modulate the TCA cycle through the PI3K-AKT signaling pathways and inflammation-related MAPK and NF-κB signaling pathways. In vitro studies revealed that CK significantly inhibited LPS-induced inflammation and oxidative stress in BV2 cells, as well as reduced oxidative stress and neuronal apoptosis in HT22 cells. CONCLUSION These findings underscore the potential of CK as a therapeutic agent in alleviating AD pathology. This study offers new insights into CK's mechanisms, suggesting that its therapeutic effects may be achieved through the coordinated reduction of neuroinflammation, oxidative stress, and neuronal apoptosis across multiple pathways, collectively working to counteract AD pathology.
Collapse
Affiliation(s)
- Yong-Lin Liu
- National Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Nanchang, Jiangxi, 330096, PR China; Research and Development Department, Jiangzhong Pharmaceutical Co., Ltd., Nanchang, Jiangxi, 330103, PR China; School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, PR China.
| | - Sha Xu
- National Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Nanchang, Jiangxi, 330096, PR China; Research and Development Department, Jiangzhong Pharmaceutical Co., Ltd., Nanchang, Jiangxi, 330103, PR China.
| | - Xi Xu
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, PR China.
| | - Yuan Tang
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, PR China.
| | - Jian Shao
- National Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Nanchang, Jiangxi, 330096, PR China; Research and Development Department, Jiangzhong Pharmaceutical Co., Ltd., Nanchang, Jiangxi, 330103, PR China.
| | - Jie Chen
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, PR China.
| | - Yi-Guang Li
- National Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Nanchang, Jiangxi, 330096, PR China; Research and Development Department, Jiangzhong Pharmaceutical Co., Ltd., Nanchang, Jiangxi, 330103, PR China; School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, PR China.
| |
Collapse
|
2
|
Chen J, Xie J, Deng F, Cai J, Chen S, Song X, Xia S, Shen Q, Guo X, Tang Y. Expansion of peripheral cytotoxic CD4+ T cells in Alzheimer's disease: New insights from multi-omics evidence. Genomics 2025; 117:110976. [PMID: 39657893 DOI: 10.1016/j.ygeno.2024.110976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 11/19/2024] [Accepted: 12/04/2024] [Indexed: 12/12/2024]
Abstract
The significance of the adaptive immune response in Alzheimer's disease (AD) is increasingly recognized. We analyzed scRNA-Seq data from AD patients, revealing a notable rise in CD4 cytotoxic T cells (CD4-CTLs) in peripheral blood mononuclear cells (PBMCs), validated in vivo and in vitro. This rise correlates with cognitive decline in AD patients. We also identified transcription factors TBX21 and MYBL1 as key drivers of CD4-CTL expansion. Further analyses indicate these cells are terminally differentiated, showing clonal expansion, metabolic changes, and unique communication patterns. Mendelian randomization identified risk genes SRGN and ITGB1, suggesting their genetic regulation in CD4-CTLs may contribute to AD. To summarize, our findings characterize the expansion of CD4-CTLs in the PBMCs of AD patients, providing valuable understanding into the possible mechanisms involved in the expansion of CD4-CTLs in AD.
Collapse
Affiliation(s)
- Jiongxue Chen
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Jiatian Xie
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Fuyin Deng
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Jinhua Cai
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Sitai Chen
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Xingrong Song
- Department of Anesthesiology, Guangzhou Women and Children Medical Center, Guangzhou 510623, China
| | - Shangzhou Xia
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, California 90033, USA
| | - Qingyu Shen
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Xinying Guo
- Department of Anesthesiology, Guangzhou Women and Children Medical Center, Guangzhou 510623, China.
| | - Yamei Tang
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Brain Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Epigenetics and Gene Regulation of Malignant Tumors, Sun Yat-sen Memorial Hospital, Guangzhou, China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
3
|
Kostic M, Zivkovic N, Cvetanovic A, Basic J, Stojanovic I. Dissecting the immune response of CD4 + T cells in Alzheimer's disease. Rev Neurosci 2024:revneuro-2024-0090. [PMID: 39238424 DOI: 10.1515/revneuro-2024-0090] [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: 07/04/2024] [Accepted: 08/18/2024] [Indexed: 09/07/2024]
Abstract
The formation of amyloid-β (Aβ) plaques is a neuropathological hallmark of Alzheimer's disease (AD), however, these pathological aggregates can also be found in the brains of cognitively unimpaired elderly population. In that context, individual variations in the Aβ-specific immune response could be key factors that determine the level of Aβ-induced neuroinflammation and thus the propensity to develop AD. CD4+ T cells are the cornerstone of the immune response that coordinate the effector functions of both adaptive and innate immunity. However, despite intensive research efforts, the precise role of these cells during AD pathogenesis is still not fully elucidated. Both pathogenic and beneficial effects have been observed in various animal models of AD, as well as in humans with AD. Although this functional duality of CD4+ T cells in AD can be simply attributed to the vast phenotype heterogeneity of this cell lineage, disease stage-specific effect have also been proposed. Therefore, in this review, we summarized the current understanding of the role of CD4+ T cells in the pathophysiology of AD, from the aspect of their antigen specificity, activation, and phenotype characteristics. Such knowledge is of practical importance as it paves the way for immunomodulation as a therapeutic option for AD treatment, given that currently available therapies have not yielded satisfactory results.
Collapse
Affiliation(s)
- Milos Kostic
- Department of Immunology, Medical Faculty of Nis, University of Nis, Blvd. dr Zorana Djindjica 81, Nis, 18000, Serbia
| | - Nikola Zivkovic
- Department of Pathology, Medical Faculty of Nis, University of Nis, Blvd. dr Zorana Djindjica 81, Nis, 18000, Serbia
| | - Ana Cvetanovic
- Department of Oncology, Medical Faculty of Nis, University of Nis, Blvd. dr Zorana Djindjica 81, Nis, 18000, Serbia
| | - Jelena Basic
- Department of Biochemistry, Medical Faculty of Nis, University of Nis, Blvd. dr Zorana Djindjica 81, Nis, 18000, Serbia
| | - Ivana Stojanovic
- Department of Biochemistry, Medical Faculty of Nis, University of Nis, Blvd. dr Zorana Djindjica 81, Nis, 18000, Serbia
| |
Collapse
|
4
|
Carr LM, Mustafa S, Care A, Collins-Praino LE. More than a number: Incorporating the aged phenotype to improve in vitro and in vivo modeling of neurodegenerative disease. Brain Behav Immun 2024; 119:554-571. [PMID: 38663775 DOI: 10.1016/j.bbi.2024.04.023] [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: 05/22/2023] [Revised: 03/04/2024] [Accepted: 04/22/2024] [Indexed: 04/29/2024] Open
Abstract
Age is the number one risk factor for developing a neurodegenerative disease (ND), such as Alzheimer's disease (AD) or Parkinson's disease (PD). With our rapidly ageing world population, there will be an increased burden of ND and need for disease-modifying treatments. Currently, however, translation of research from bench to bedside in NDs is poor. This may be due, at least in part, to the failure to account for the potential effect of ageing in preclinical modelling of NDs. While ageing can impact upon physiological response in multiple ways, only a limited number of preclinical studies of ND have incorporated ageing as a factor of interest. Here, we evaluate the aged phenotype and highlight the critical, but unmet, need to incorporate aspects of this phenotype into both the in vitro and in vivo models used in ND research. Given technological advances in the field over the past several years, we discuss how these could be harnessed to create novel models of ND that more readily incorporate aspects of the aged phenotype. This includes a recently described in vitro panel of ageing markers, which could help lead to more standardised models and improve reproducibility across studies. Importantly, we cannot assume that young cells or animals yield the same responses as seen in the context of ageing; thus, an improved understanding of the biology of ageing, and how to appropriately incorporate this into the modelling of ND, will ensure the best chance for successful translation of new therapies to the aged patient.
Collapse
Affiliation(s)
- Laura M Carr
- School of Biomedicine, University of Adelaide, Adelaide, SA, Australia
| | - Sanam Mustafa
- School of Biomedicine, University of Adelaide, Adelaide, SA, Australia; Australian Research Council Centre of Excellence for Nanoscale Biophotonics, The University of Adelaide, Adelaide, SA, Australia; Davies Livestock Research Centre, The University of Adelaide, Roseworthy, SA, Australia
| | - Andrew Care
- School of Life Sciences, University of Technology Sydney, Sydney, NSW, Australia
| | - Lyndsey E Collins-Praino
- School of Biomedicine, University of Adelaide, Adelaide, SA, Australia; Australian Research Council Centre of Excellence for Nanoscale Biophotonics, The University of Adelaide, Adelaide, SA, Australia.
| |
Collapse
|
5
|
Whitson HE, Banks WA, Diaz MM, Frost B, Kellis M, Lathe R, Schmader KE, Spudich SS, Tanzi R, Garden G. New approaches for understanding the potential role of microbes in Alzheimer's disease. Brain Behav Immun Health 2024; 36:100743. [PMID: 38435720 PMCID: PMC10906156 DOI: 10.1016/j.bbih.2024.100743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 02/17/2024] [Accepted: 02/20/2024] [Indexed: 03/05/2024] Open
Abstract
Alzheimer's disease (AD) involves a complex pathological process that evolves over years, and its etiology is understood as a classic example of gene-environment interaction. The notion that exposure to microbial organisms may play some role in AD pathology has been proposed and debated for decades. New evidence from model organisms and -omic studies, as well as epidemiological data from the recent COVID-19 pandemic and widespread use of vaccines, offers new insights into the "germ hypothesis" of AD. To review new evidence and identify key research questions, the Duke/University of North Carolina (Duke/UNC) Alzheimer's Disease Research Center hosted a virtual symposium and workshop: "New Approaches for Understanding the Potential Role of Microbes in Alzheimer's disease." Discussion centered around the antimicrobial protection hypothesis of amyloid accumulation, and other mechanisms by which microbes could influence AD pathology including immune cell activation, changes in blood-brain barrier, or direct neurotoxicity. This summary of proceedings reviews the content presented in the symposium and provides a summary of major topics and key questions discussed in the workshop.
Collapse
Affiliation(s)
- Heather E. Whitson
- Duke Center for the Study of Aging and Human Development, Duke University School of Medicine, Busse Bldg Rm 3502, Durham, NC, 27710, USA
- Durham VA Medical Center, Geriatric Research Education and Clinical Center, 508 Fulton Street, Durham, NC, 27705, USA
| | - William A. Banks
- Veterans Affairs Puget Sound Health Care System, 1660 S Columbian Way, Seattle, WA, 98108, USA
| | - Monica M. Diaz
- Department of Neurology, University of North Carolina at Chapel Hill, 170 Manning Dr, CB 7025, Chapel Hill, NC, 27599, USA
| | - Bess Frost
- Barshop Institute for Longevity & Aging Studies, 4939 Charles Katz Rm 1041, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Manolis Kellis
- Computer Science and Artificial Intelligence Lab, Massachusetts Institute of Technology, 32 Vassar St., Cambridge, MA, 02139, USA
| | - Richard Lathe
- Division of Infection Medicine, University of Edinburgh Medical School, Edinburgh BioQuarter, Little France, Edinburgh, EH16 4SB, UK
| | - Kenneth E. Schmader
- Duke Center for the Study of Aging and Human Development, Duke University School of Medicine, Busse Bldg Rm 3502, Durham, NC, 27710, USA
- Durham VA Medical Center, Geriatric Research Education and Clinical Center, 508 Fulton Street, Durham, NC, 27705, USA
| | - Serena S. Spudich
- Department of Neurology, Yale University School of Medicine, 300 George Street, Room 8300, New Haven, CT, 06510, USA
| | - Rudolph Tanzi
- Genetics and Aging Research Unit, Massachusetts General Hospital, 114 16th Street, Charlestown, MA, 02129, USA
| | - Gwenn Garden
- University of North Carolina - Dept of Neurology, 170 Manning Drive, Campus Box 7025, Chapel Hill, NC, 27599-7025, USA
| |
Collapse
|
6
|
Tan H, Huang Y, Dong S, Bai Z, Chen C, Wu X, Chao M, Yan H, Wang S, Geng D, Gao F. A Chiral Nanocomplex for Multitarget Therapy to Alleviate Neuropathology and Rescue Alzheimer's Cognitive Deficits. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2303530. [PMID: 37635125 DOI: 10.1002/smll.202303530] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 07/23/2023] [Indexed: 08/29/2023]
Abstract
Alzheimer's disease (AD) is a severe neurodegenerative condition characterized by inflammation, beta-amyloid (Aβ) plaques, and neurodegeneration, which currently lack effective treatments. Chiral nanomaterials have emerged as a promising option for treating neurodegenerative disorders due to their high biocompatibility, strong sustained release ability, and specific enantiomer selectivity. The development of a stimulus-responsive chiral nanomaterial, UiO-66-NH2 @l-MoS2 QDs@PA-Ni (MSP-U), for the treatment of AD is reported. MSP-U is found to stimulate neural stem cell (NSCs) differentiation, promote in situ hydrogen (H2 ) production, and clear Aβ plaques. l-MoS2 QDs modified with l-Cysteine (l-Cys) effectively enhance the differentiation of NSCs into neurons through circularly polarized near-infrared radiation. Doped-phytic acid nickel (PA-Ni) improves the activity of l-MoS2 QDs in scavenging reactive oxygen species at the lesion site via photocatalytic H2 production. Loading l-MoS2 QDs with UiO-66 type metal oxide suppresses electron-hole recombination effect, thereby achieving rapid charge separation and improving transport of photogenerated electrons, leading to significantly improved H2 production efficiency. The photothermal effect of MSP-U also clears the generated Aβ plaques. In vivo evaluations show that MSP-U improves spatial cognition and memory, suggesting a promising potential candidate for the treatment of AD using chiral nanomaterials.
Collapse
Affiliation(s)
- Huarong Tan
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Jiangsu, 221002, P. R. China
- Department of Psychiatry, The Affiliated Hospital of Xuzhou Medical University, Jiangsu, 221002, P. R. China
| | - Yuqi Huang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Jiangsu, 221002, P. R. China
| | - Shuqing Dong
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Jiangsu, 221002, P. R. China
| | - Zetai Bai
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Jiangsu, 221002, P. R. China
| | - Cheng Chen
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Jiangsu, 221002, P. R. China
| | - Xiunan Wu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Jiangsu, 221002, P. R. China
| | - Minghao Chao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Jiangsu, 221002, P. R. China
| | - Hanrong Yan
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Jiangsu, 221002, P. R. China
| | - Shaoshen Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Jiangsu, 221002, P. R. China
| | - Deqin Geng
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Jiangsu, 221002, P. R. China
- Department of Psychiatry, The Affiliated Hospital of Xuzhou Medical University, Jiangsu, 221002, P. R. China
| | - Fenglei Gao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Jiangsu, 221002, P. R. China
- Department of Psychiatry, The Affiliated Hospital of Xuzhou Medical University, Jiangsu, 221002, P. R. China
| |
Collapse
|
7
|
Cousins O, Schubert JJ, Chandra A, Veronese M, Valkimadi P, Creese B, Khan Z, Arathimos R, Hampshire A, Rosenzweig I, Ballard C, Corbett A, Aasland D, Velayudhan L, O'Neill M, Collier D, Awais R, Sander K, Årstad E, Howes O, Turkheimer F, Hodges A. Microglial activation, tau and amyloid deposition in TREM2 p.R47H carriers and mild cognitive impairment patients: a multi-modal/multi-tracer PET/MRI imaging study with influenza vaccine immune challenge. J Neuroinflammation 2023; 20:272. [PMID: 37990275 PMCID: PMC10664604 DOI: 10.1186/s12974-023-02945-0] [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: 03/21/2023] [Accepted: 10/31/2023] [Indexed: 11/23/2023] Open
Abstract
BACKGROUND Microglia are increasingly understood to play an important role in the pathogenesis of Alzheimer's disease. The rs75932628 (p.R47H) TREM2 variant is a well-established risk factor for Alzheimer's disease. TREM2 is a microglial cell surface receptor. In this multi-modal/multi-tracer PET/MRI study we investigated the effect of TREM2 p.R47H carrier status on microglial activation, tau and amyloid deposition, brain structure and cognitive profile. METHODS We compared TREM2 p.R47H carriers (n = 8; median age = 62.3) and participants with mild cognitive impairment (n = 8; median age = 70.7). Participants underwent two [18F]DPA-714 PET/MRI scans to assess TSPO signal, indicative of microglial activation, before and after receiving the seasonal influenza vaccination, which was used as an immune stimulant. Participants also underwent [18F]florbetapir and [18F]AV1451 PET scans to assess amyloid and tau burden, respectively. Regional tau and TSPO signal were calculated for regions of interest linked to Braak stage. An additional comparison imaging healthy control group (n = 8; median age = 45.5) had a single [18F]DPA-714 PET/MRI. An expanded group of participants underwent neuropsychological testing, to determine if TREM2 status influenced clinical phenotype. RESULTS Compared to participants with mild cognitive impairment, TREM2 carriers had lower TSPO signal in Braak II (P = 0.04) and Braak III (P = 0.046) regions, despite having a similar burden of tau and amyloid. There were trends to suggest reduced microglial activation following influenza vaccine in TREM2 carriers. Tau deposition in the Braak VI region was higher in TREM2 carriers (P = 0.04). Furthermore, compared to healthy controls TREM2 carriers had smaller caudate (P = 0.02), total brain (P = 0.049) and white matter volumes (P = 0.02); and neuropsychological assessment revealed worse ADAS-Cog13 (P = 0.03) and Delayed Matching to Sample (P = 0.007) scores. CONCLUSIONS TREM2 p.R47H carriers had reduced levels of microglial activation in brain regions affected early in the Alzheimer's disease course and differences in brain structure and cognition. Changes in microglial response may underlie the increased Alzheimer's disease risk in TREM2 p.R47H carriers. Future therapeutic agents in Alzheimer's disease should aim to enhance protective microglial actions.
Collapse
Affiliation(s)
- Oliver Cousins
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 9RT, UK
| | - Julia J Schubert
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 9RT, UK
| | - Avinash Chandra
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 9RT, UK
| | - Mattia Veronese
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 9RT, UK
- Department of Information Engineering, University of Padua, 35131, Padua, Italy
| | - Polena Valkimadi
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 9RT, UK
| | - Byron Creese
- College of Medicine and Health, University of Exeter, Exeter, EX1 2HZ, UK
- Division of Psychology, Department of Life Sciences, Brunel University London, London, UB8 3PH, UK
| | - Zunera Khan
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 9RT, UK
| | - Ryan Arathimos
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 9RT, UK
| | - Adam Hampshire
- Faculty of Medicine, Imperial College London, London, SW7 2AZ, UK
| | - Ivana Rosenzweig
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 9RT, UK
| | - Clive Ballard
- College of Medicine and Health, University of Exeter, Exeter, EX1 2HZ, UK
| | - Anne Corbett
- College of Medicine and Health, University of Exeter, Exeter, EX1 2HZ, UK
| | - Dag Aasland
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 9RT, UK
| | - Latha Velayudhan
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 9RT, UK
| | | | | | - Ramla Awais
- Centre for Radiopharmaceutical Chemistry, University College London, London, WC1E 6BS, UK
| | - Kerstin Sander
- Centre for Radiopharmaceutical Chemistry, University College London, London, WC1E 6BS, UK
| | - Erik Årstad
- Centre for Radiopharmaceutical Chemistry, University College London, London, WC1E 6BS, UK
| | - Oliver Howes
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 9RT, UK
| | - Federico Turkheimer
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 9RT, UK
| | - Angela Hodges
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 9RT, UK.
| |
Collapse
|
8
|
Qi F, Zuo Z, Hu K, Wang R, Wu T, Liu H, Tang J, Wang Q, Xie Y, Tan L, Yang Y, Zhang X, Zheng J, Xu J, Yao Z, Wang S, Wu LJ, Guo K. VEGF-A in serum protects against memory impairment in APP/PS1 transgenic mice by blocking neutrophil infiltration. Mol Psychiatry 2023; 28:4374-4389. [PMID: 37280283 PMCID: PMC10827659 DOI: 10.1038/s41380-023-02097-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 04/17/2023] [Accepted: 04/27/2023] [Indexed: 06/08/2023]
Abstract
Activation of innate immunity in the brain is a prominent feature of Alzheimer's disease (AD). The present study investigated the regulation of innate immunity by wild-type serum injection in a transgenic AD mouse model. We found that treatment with wild-type mouse serum significantly reduced the number of neutrophils and microglial reactivity in the brains of APP/PS1 mice. Mimicking this effect, neutrophil depletion via Ly6G neutralizing antibodies resulted in improvements in AD brain functions. Serum proteomic analysis identified vascular endothelial growth factor-A (VEGF-A) and chemokine (C-X-C motif) ligand 1 (CXCL1) as factors enriched in serum samples, which are crucial for neutrophil migration and chemotaxis, leukocyte migration, and cell chemotaxis. Exogenous VEGF-A reversed amyloid β (Aβ)-induced decreases in cyclin-dependent kinase 5 (Cdk5) and increases in CXCL1 in vitro and blocked neutrophil infiltration into the AD brain. Endothelial Cdk5 overexpression conferred an inhibitory effect on CXCL1 and neutrophil infiltration, thereby restoring memory abilities in APP/PS1 mice. Our findings uncover a previously unknown link between blood-derived VEGF signaling and neutrophil infiltration and support targeting endothelial Cdk5 signaling as a potential therapeutic strategy for AD.
Collapse
Affiliation(s)
- Fangfang Qi
- Department of Anatomy and Physiology, Guangdong Province Key Laboratory of Brain Function and Disease, Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Editorial Department of Journal of Sun Yat-sen University, Guangzhou, 510080, China
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Zejie Zuo
- Department of Rehabilitation Medicine, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Kaishun Hu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Rui Wang
- Department of Anatomy and Physiology, Guangdong Province Key Laboratory of Brain Function and Disease, Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Tong Wu
- Department of Anatomy and Physiology, Guangdong Province Key Laboratory of Brain Function and Disease, Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Hao Liu
- Department of Anatomy and Physiology, Guangdong Province Key Laboratory of Brain Function and Disease, Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jiaoling Tang
- Department of Anatomy and Physiology, Guangdong Province Key Laboratory of Brain Function and Disease, Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Qingbo Wang
- Department of Anatomy and Physiology, Guangdong Province Key Laboratory of Brain Function and Disease, Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yufeng Xie
- Five-year Programs of Clinical Medicine in the 2017 grade, School of Medicine, Sun Yat-sen University, Shenzhen, 528406, China
| | - Liren Tan
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Yunjie Yang
- Department of Anatomy and Physiology, Guangdong Province Key Laboratory of Brain Function and Disease, Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xiaoran Zhang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jiaying Zheng
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Jie Xu
- Department of Anatomy and Physiology, Guangdong Province Key Laboratory of Brain Function and Disease, Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Zhibin Yao
- Department of Anatomy and Physiology, Guangdong Province Key Laboratory of Brain Function and Disease, Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Shengwen Wang
- Department of Neurosurgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA.
| | - Kaihua Guo
- Department of Anatomy and Physiology, Guangdong Province Key Laboratory of Brain Function and Disease, Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
9
|
Salas J, Morley JE, Hoft DF, Scherrer JF. Lower risk for COVID-19 hospitalization among patients in the United States with past vaccinations for herpes zoster and tetanus, diphtheria and pertussis. Prev Med Rep 2023; 35:102302. [PMID: 37441187 PMCID: PMC10290736 DOI: 10.1016/j.pmedr.2023.102302] [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: 02/16/2023] [Revised: 06/21/2023] [Accepted: 06/23/2023] [Indexed: 07/15/2023] Open
Abstract
Influenza, tetanus, diphtheria, and herpes zoster (HZ) vaccination received within 10 years of the COVID-19 pandemic have been associated with less severe COVID-19 infection. We expanded on this evidence to determine if a receiving two different vaccinations (i.e., HZ and tetanus, diphtheria, and pertussis (Tdap)) was associated with a lower risk for COVID-19 hospitalization. De-identified medical record data from a large mid-western health care system was used to determine if, compared to those with neither HZ or Tdap vaccination, patients with either HZ or Tdap and patients with both HZ and Tdap vaccination had lower risk for COVID-19 hospitalization between 4/1/2020 and 12/31/2020. Confounding was controlled using entropy balancing. Patients (n = 363,293) were 71.5 (±8.4) years of age, 57.8% female and 89.2% White race. Prior to controlling for confounding, as compared to patients without either vaccination, those that had either HZ or Tdap were significantly less likely to have a COVID-19 hospitalization (RR = 0.85; 95 %CI: 0.75-0.95). The risk for hospitalization decreased further among those with both HZ and Tdap vaccination (RR = 0.45; 95 %CI:0.28-0.71). After controlling for confounding, including healthy patient bias, receiving both vs. neither vaccinations remained significantly associated with a lower risk of COVID-19 hospitalization (RR = 0.48; 95 %CI: 0.26-0.90). Receiving both Tdap and HZ vaccination is associated with lower risk for COVID-19 hospitalization. Whether there is any benefit of past vaccination exposure in COVID-19 vaccinated patients should be investigated.
Collapse
Affiliation(s)
- Joanne Salas
- Department of Family and Community Medicine, Saint Louis University School of Medicine, 1008 S. Spring, St. Louis, MO 63110, United States
- Advanced HEAlth Data (AHEAD) Research Institute, Saint Louis University School of Medicine, 3545 Lafayette Ave, 4th Floor, St. Louis, MO 63104, United States
| | - John E. Morley
- Saint Louis University, School of Medicine, Department of Internal Medicine, Division of Geriatric Medicine, Saint Louis University, School of Medicine. 1402 South Grand Blvd, St. Louis, MO, United States
| | - Daniel F. Hoft
- Saint Louis University, School of Medicine, Department of Internal Medicine Division of Infectious Diseases, Allergy, and Immunology, Saint Louis, MO, United States
- Saint Louis University, Department of Molecular Microbiology & Immunology, Saint Louis, MO, United States
| | - Jeffrey F. Scherrer
- Department of Family and Community Medicine, Saint Louis University School of Medicine, 1008 S. Spring, St. Louis, MO 63110, United States
- Advanced HEAlth Data (AHEAD) Research Institute, Saint Louis University School of Medicine, 3545 Lafayette Ave, 4th Floor, St. Louis, MO 63104, United States
- Department of Psychiatry and Behavioral Neuroscience, Saint Louis University School of Medicine, 1438 South Grand Blvd., St. Louis, MO 63104, United States
| |
Collapse
|
10
|
Hjelholt AJ, Bergh C, Bhatt DL, Fröbert O, Kjolby MF. Pleiotropic Effects of Influenza Vaccination. Vaccines (Basel) 2023; 11:1419. [PMID: 37766096 PMCID: PMC10536538 DOI: 10.3390/vaccines11091419] [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] [Received: 07/06/2023] [Revised: 08/20/2023] [Accepted: 08/23/2023] [Indexed: 09/29/2023] Open
Abstract
Influenza vaccines are designed to mimic natural influenza virus exposure and stimulate a long-lasting immune response to future infections. The evolving nature of the influenza virus makes vaccination an important and efficacious strategy to reduce healthcare-related complications of influenza. Several lines of evidence indicate that influenza vaccination may induce nonspecific effects, also referred to as heterologous or pleiotropic effects, that go beyond protection against infection. Different explanations are proposed, including the upregulation and downregulation of cytokines and epigenetic reprogramming in monocytes and natural killer cells, imprinting an immunological memory in the innate immune system, a phenomenon termed "trained immunity". Also, cross-reactivity between related stimuli and bystander activation, which entails activation of B and T lymphocytes without specific recognition of antigens, may play a role. In this review, we will discuss the possible nonspecific effects of influenza vaccination in cardiovascular disease, type 1 diabetes, cancer, and Alzheimer's disease, future research questions, and potential implications. A discussion of the potential effects on infections by other pathogens is beyond the scope of this review.
Collapse
Affiliation(s)
- Astrid Johannesson Hjelholt
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Palle Juul-Jensens Boulevard 11, 8200 Aarhus N, Denmark; (O.F.); (M.F.K.)
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000 Aarhus, Denmark
- Department of Clinical Pharmacology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 11, 8200 Aarhus N, Denmark
| | - Cecilia Bergh
- Clinical Epidemiology and Biostatistics, School of Medical Sciences, Örebro University, S-701 82 Örebro, Sweden;
| | - Deepak L. Bhatt
- Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, One Gustave L. Levi Place, P.O. Box 1030, New York, NY 10029-6574, USA;
| | - Ole Fröbert
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Palle Juul-Jensens Boulevard 11, 8200 Aarhus N, Denmark; (O.F.); (M.F.K.)
- Department of Clinical Pharmacology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 11, 8200 Aarhus N, Denmark
- Department of Clinical Medicine, Faculty of Health, Aarhus University, Palle Juul-Jensens Boulevard 11, 8200 Aarhus N, Denmark
- Faculty of Health, Department of Cardiology, Örebro University, SE-701 82 Örebro, Sweden
| | - Mads Fuglsang Kjolby
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Palle Juul-Jensens Boulevard 11, 8200 Aarhus N, Denmark; (O.F.); (M.F.K.)
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000 Aarhus, Denmark
- Department of Clinical Pharmacology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 11, 8200 Aarhus N, Denmark
| |
Collapse
|
11
|
Hobson R, Levy SHS, Flaherty D, Xiao H, Ciener B, Reddy H, Singal C, Teich AF, Shneider NA, Bradshaw EM, Elyaman W. Clonal CD8 T cells in the leptomeninges are locally controlled and influence microglia in human neurodegeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.13.548931. [PMID: 37503131 PMCID: PMC10369982 DOI: 10.1101/2023.07.13.548931] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Recent murine studies have highlighted a crucial role for the meninges in surveilling the central nervous system (CNS) and influencing CNS inflammation. However, how meningeal immunity is altered in human neurodegeneration and its potential effects on neuroinflammation is understudied. In the present study, we performed single-cell analysis of the transcriptomes and T cell receptor repertoire of 72,576 immune cells from 36 postmortem human brain and leptomeninges tissues from donors with neurodegenerative diseases including amyotrophic lateral sclerosis, Alzheimer's disease, and Parkinson's disease. We identified the meninges as an important site of antigen presentation and CD8 T cell activation and clonal expansion and found that T cell activation in the meninges is a requirement for infiltration into the CNS. We further found that natural killer cells have the potential to negatively regulate T cell activation locally in the meninges through direct killing and are one of many regulatory mechanisms that work to control excessive neuroinflammation.
Collapse
|
12
|
Ecarnot F, Boccardi V, Calcagno A, Franceschi C, Fülop T, Itzhaki RF, Michel JP, Panza F, Rainero I, Solfrizzi V, Ticinesi A, Veronese N, Maggi S. Dementia, infections and vaccines: 30 years of controversy. Aging Clin Exp Res 2023; 35:1145-1160. [PMID: 37160649 PMCID: PMC10169152 DOI: 10.1007/s40520-023-02409-8] [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: 03/04/2023] [Accepted: 03/21/2023] [Indexed: 05/11/2023]
Abstract
This paper reports the proceedings of a virtual meeting convened by the European Interdisciplinary Council on Ageing (EICA), to discuss the involvement of infectious disorders in the pathogenesis of dementia and neurological disorders leading to dementia. We recap how our view of the infectious etiology of dementia has changed over the last 30 years in light of emerging evidence, and we present evidence in support of the implication of infection in dementia, notably Alzheimer's disease (AD). The bacteria and viruses thought to be responsible for neuroinflammation and neurological damage are reviewed. We then review the genetic basis for neuroinflammation and dementia, highlighting the genes that are currently the focus of investigation as potential targets for therapy. Next, we describe the antimicrobial hypothesis of dementia, notably the intriguing possibility that amyloid beta may itself possess antimicrobial properties. We further describe the clinical relevance of the gut-brain axis in dementia, the mechanisms by which infection can move from the intestine to the brain, and recent findings regarding dysbiosis patterns in patients with AD. We review the involvement of specific pathogens in neurological disorders, i.e. SARS-CoV-2, human immunodeficiency virus (HIV), herpes simplex virus type 1 (HSV1), and influenza. Finally, we look at the role of vaccination to prevent dementia. In conclusion, there is a large body of evidence supporting the involvement of various infectious pathogens in the pathogenesis of dementia, but large-scale studies with long-term follow-up are needed to elucidate the role that infection may play, especially before subclinical or clinical disease is present.
Collapse
Affiliation(s)
- Fiona Ecarnot
- EA3920, University of Franche-Comté, 25000, Besancon, France
- Department of Cardiology, University Hospital Besancon, 3-8 Boulevard Fleming, 25000, Besancon, France
| | - Virginia Boccardi
- Institute of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Santa Maria Della Misericordia Hospital, Piazzale Gambuli 1, 06132, Perugia, Italy
| | - Andrea Calcagno
- Unit of Infectious Diseases, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Claudio Franceschi
- Laboratory of Systems Medicine of Healthy Aging, Institute of Biology and Biomedicine and Institute of Information Technology, Mathematics and Mechanics, Department of Applied Mathematics, N. I. Lobachevsky State University, Nizhny Novgorod, Russia
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Tamas Fülop
- Department of Medicine, Geriatrics Division, Research Center on Aging, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC, J1H 5N4, Canada
| | - Ruth F Itzhaki
- Institute of Population Ageing, University of Oxford and Faculty of Life Sciences, University of Manchester, Manchester, UK
| | | | - Francesco Panza
- Unit of Research Methodology and Data Sciences for Population Health, National Institute of Gastroenterology "Saverio de Bellis", Research Hospital, Castellana Grotte, Bari, Italy
- Dipartimento Interdisciplinare di Medicina, Clinica Medica e Geriatria "Cesare Frugoni", University of Bari Aldo Moro, Bari, Italy
| | - Innocenzo Rainero
- Dementia Center, Department of Neuroscience "Rita Levi Montalcini", University of Torino, Turin, Italy
| | - Vincenzo Solfrizzi
- Dipartimento Interdisciplinare di Medicina, Clinica Medica e Geriatria "Cesare Frugoni", University of Bari Aldo Moro, Bari, Italy
| | - Andrea Ticinesi
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Geriatric-Rehabilitation Department, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Nicola Veronese
- Geriatrics Section, Department of Internal Medicine, University of Palermo, Palermo, Italy.
| | - Stefania Maggi
- National Research Council, Neuroscience Institute, Aging Branch, Padua, Italy
| |
Collapse
|
13
|
Lemaitre P, Tareen SHK, Pasciuto E, Mascali L, Martirosyan A, Callaerts‐Vegh Z, Poovathingal S, Dooley J, Holt MG, Yshii L, Liston A. Molecular and cognitive signatures of ageing partially restored through synthetic delivery of IL2 to the brain. EMBO Mol Med 2023; 15:e16805. [PMID: 36975362 PMCID: PMC10165365 DOI: 10.15252/emmm.202216805] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 03/08/2023] [Accepted: 03/10/2023] [Indexed: 03/29/2023] Open
Abstract
Cognitive decline is a common pathological outcome during aging, with an ill-defined molecular and cellular basis. In recent years, the concept of inflammaging, defined as a low-grade inflammation increasing with age, has emerged. Infiltrating T cells accumulate in the brain with age and may contribute to the amplification of inflammatory cascades and disruptions to the neurogenic niche observed with age. Recently, a small resident population of regulatory T cells has been identified in the brain, and the capacity of IL2-mediated expansion of this population to counter neuroinflammatory disease has been demonstrated. Here, we test a brain-specific IL2 delivery system for the prevention of neurological decline in aging mice. We identify the molecular hallmarks of aging in the brain glial compartments and identify partial restoration of this signature through IL2 treatment. At a behavioral level, brain IL2 delivery prevented the age-induced defect in spatial learning, without improving the general decline in motor skill or arousal. These results identify immune modulation as a potential path to preserving cognitive function for healthy aging.
Collapse
Affiliation(s)
- Pierre Lemaitre
- VIB Center for Brain and Disease ResearchLeuvenBelgium
- Department of Microbiology, Immunology and TransplantationKU LeuvenLeuvenBelgium
| | | | - Emanuela Pasciuto
- VIB Center for Brain and Disease ResearchLeuvenBelgium
- Department of Microbiology, Immunology and TransplantationKU LeuvenLeuvenBelgium
| | - Loriana Mascali
- VIB Center for Brain and Disease ResearchLeuvenBelgium
- Department of Microbiology, Immunology and TransplantationKU LeuvenLeuvenBelgium
| | - Araks Martirosyan
- VIB Center for Brain and Disease ResearchLeuvenBelgium
- Department of NeurosciencesKU LeuvenLeuvenBelgium
| | | | | | - James Dooley
- Immunology ProgrammeThe Babraham InstituteBabrahamUK
- Department of PathologyThe University of CambridgeCambridgeUK
| | - Matthew G Holt
- VIB Center for Brain and Disease ResearchLeuvenBelgium
- Department of NeurosciencesKU LeuvenLeuvenBelgium
- Instituto de Investigaçāo e Inovaçāo em Saúde (i3S)University of PortoPortoPortugal
| | - Lidia Yshii
- VIB Center for Brain and Disease ResearchLeuvenBelgium
- Department of Microbiology, Immunology and TransplantationKU LeuvenLeuvenBelgium
- Department of NeurosciencesKU LeuvenLeuvenBelgium
| | - Adrian Liston
- VIB Center for Brain and Disease ResearchLeuvenBelgium
- Department of Microbiology, Immunology and TransplantationKU LeuvenLeuvenBelgium
- Immunology ProgrammeThe Babraham InstituteBabrahamUK
- Department of PathologyThe University of CambridgeCambridgeUK
| |
Collapse
|
14
|
Demuth L, Ohm M, Michaelsen-Preusse K, Schulze K, Riese P, Guzmán CA, Korte M, Hosseini S. Influenza vaccine is able to prevent neuroinflammation triggered by H7N7 IAV infection. Front Pharmacol 2023; 14:1142639. [PMID: 37063291 PMCID: PMC10090407 DOI: 10.3389/fphar.2023.1142639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 03/21/2023] [Indexed: 03/31/2023] Open
Abstract
Influenza A virus (IAV) subtypes are a major cause of illness and mortality worldwide and pose a threat to human health. Although IAV infection is considered a self-limiting respiratory syndrome, an expanded spectrum of cerebral manifestations has been reported following IAV infection. Neurotropic IAVs, such as the H7N7 subtype, are capable of invading the central nervous system (CNS) and replicating in brain cells, resulting in microglia-induced neuroinflammation. Microglial cells, the brain’s resident immune cells, are instrumental in the inflammatory response to viral infection. While activation of microglia is important to initially contain the virus, excessive activation of these cells leads to neuronal damage. Previous studies have shown that acute and even long-term IAV-induced neuroinflammation leads to CNS damage. Therefore, the search for possible preventive or therapeutic strategies is of great importance. In this study, we investigated the potential effect of vaccination against acute neuroinflammation induced by H7N7 infection and subsequent neuronal damage in the hippocampus, a particularly vulnerable brain region, comparing young and aged mice. Immunosenescence is one of the striking pathophysiological changes during mammalian aging that leads to “inflammaging” and critically limits the protection by vaccines in the elderly. The results suggest that formalin-inactivated H7N7 vaccine has a preventive effect against the inflammatory responses in the periphery and also in the CNS after H7N7 infection. Cytokine and chemokine levels, increased microglial density, and cell volume after H7N7 infection were all attenuated by vaccination. Further structural analysis of microglial cells also revealed a change in branching complexity after H7N7 infection, most likely reflecting the neuroprotective effect of the vaccination. In addition, synapse loss was prevented in vaccinated mice. Remarkably, engulfment of post-synaptic compartments by microglia can be proposed as the underlying mechanism for spine loss triggered by H7N7 infection, which was partially modulated by vaccination. Although young mice showed better protection against neuroinflammation and the resulting deleterious neuronal effects upon vaccination, a beneficial role of the vaccine was also observed in the brains of older mice. Therefore, vaccination can be proposed as an important strategy to prevent neurological sequelae of H7N7 infection.
Collapse
Affiliation(s)
- Luisa Demuth
- Department of Cellular Neurobiology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
| | - Melanie Ohm
- Department of Cellular Neurobiology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
| | - Kristin Michaelsen-Preusse
- Department of Cellular Neurobiology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
| | - Kai Schulze
- Helmholtz Centre for Infection Research, Department of Vaccinology and Applied Microbiology, Braunschweig, Germany
| | - Peggy Riese
- Helmholtz Centre for Infection Research, Department of Vaccinology and Applied Microbiology, Braunschweig, Germany
| | - Carlos A. Guzmán
- Helmholtz Centre for Infection Research, Department of Vaccinology and Applied Microbiology, Braunschweig, Germany
| | - Martin Korte
- Department of Cellular Neurobiology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
- Helmholtz Centre for Infection Research, Neuroinflammation and Neurodegeneration Group, Braunschweig, Germany
| | - Shirin Hosseini
- Department of Cellular Neurobiology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
- Helmholtz Centre for Infection Research, Neuroinflammation and Neurodegeneration Group, Braunschweig, Germany
- *Correspondence: Shirin Hosseini,
| |
Collapse
|
15
|
Olson KE, Mosley RL, Gendelman HE. The potential for treg-enhancing therapies in nervous system pathologies. Clin Exp Immunol 2023; 211:108-121. [PMID: 36041453 PMCID: PMC10019130 DOI: 10.1093/cei/uxac084] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/28/2022] [Accepted: 08/30/2022] [Indexed: 11/13/2022] Open
Abstract
While inflammation may not be the cause of disease, it is well known that it contributes to disease pathogenesis across a multitude of peripheral and central nervous system disorders. Chronic and overactive inflammation due to an effector T-cell-mediated aberrant immune response ultimately leads to tissue damage and neuronal cell death. To counteract peripheral and neuroinflammatory responses, research is being focused on regulatory T cell enhancement as a therapeutic target. Regulatory T cells are an immunosuppressive subpopulation of CD4+ T helper cells essential for maintaining immune homeostasis. The cells play pivotal roles in suppressing immune responses to maintain immune tolerance. In so doing, they control T cell proliferation and pro-inflammatory cytokine production curtailing autoimmunity and inflammation. For nervous system pathologies, Treg are known to affect the onset and tempo of neural injuries. To this end, we review recent findings supporting Treg's role in disease, as well as serving as a therapeutic agent in multiple sclerosis, myasthenia gravis, Guillain-Barre syndrome, Parkinson's and Alzheimer's diseases, and amyotrophic lateral sclerosis. An ever-broader role for Treg in the control of neurologic disease has been shown for traumatic brain injury, stroke, neurotrophic pain, epilepsy, and psychiatric disorders. To such ends, this review serves to examine the role played by Tregs in nervous system diseases with a focus on harnessing their functional therapeutic role(s).
Collapse
Affiliation(s)
- Katherine E Olson
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA
| | - R L Mosley
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA
| |
Collapse
|
16
|
Sun H, Liu M, Liu J. Association of Influenza Vaccination and Dementia Risk: A Meta-Analysis of Cohort Studies. J Alzheimers Dis 2023; 92:667-678. [PMID: 36744343 DOI: 10.3233/jad-221036] [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: 02/05/2023]
Abstract
BACKGROUND Dementia is a critical global public health problem. Previous cohort studies have found that influenza vaccination can decrease the risk of dementia. OBJECTIVE This meta-analysis aimed to systematically examine the relationship between influenza vaccination and dementia risk. METHODS We searched PubMed, Embase, Web of Science, ScienceDirect, medRxiv, and bioRxiv for studies investigating dementia risk based on influenza vaccination status, up to September 14, 2022. Relative risks (RRs) and 95% confidence intervals (95% CIs) were pooled in the meta-analysis. Subgroup analyses and sensitivity analyses were conducted as well. RESULTS Of the 4,087 articles initially reviewed, 6 cohort studies were included in the final meta-analysis, and all eligible studies were at low risk of bias. There were 2,087,195 participants without dementia at baseline (mean age: 61.8-75.5 years, 57.05% males), and 149,804 (7.18%) cases of dementia occurred during 4.00-13.00 years of follow-up. Pooled analysis of adjusted RRs found that influenza vaccination could reduce dementia risk by 31% (RR = 0.69, 95% CI: 0.57-0.83). Subgroup analyses showed that in the study with a mean age of 75-80 years or 75%-100% males, the association was generally weakened compared with studies with a mean age of 60-75 years or 25%-50% males. The results were stable in the sensitivity analyses, and no publication bias was observed. CONCLUSION Influenza vaccination in older adults was markedly associated with a decreased risk of dementia. More mechanistic studies and epidemiological studies are needed to clarify the association between influenza vaccination and decreased dementia risk.
Collapse
Affiliation(s)
- Huimin Sun
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Haidian District, Beijing, China
| | - Min Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Haidian District, Beijing, China
| | - Jue Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Haidian District, Beijing, China.,Institute for Global Health and Development, Peking University, Haidian District, Beijing, China.,Global Center for Infectious Disease and Policy Research & Global Health and Infectious Diseases Group, Peking University, Beijing, China
| |
Collapse
|
17
|
Liu Y, Tan Y, Zhang Z, Li H, Yi M, Zhang Z, Hui S, Peng W. Neuroimmune mechanisms underlying Alzheimer's disease: Insights into central and peripheral immune cell crosstalk. Ageing Res Rev 2023; 84:101831. [PMID: 36565960 DOI: 10.1016/j.arr.2022.101831] [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: 08/24/2022] [Revised: 12/15/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
Alzheimer's disease (AD) is a highly life-threatening neurodegenerative disease. Dysregulation of the immune system plays a critical role in promoting AD, which has attracted extensive attention recently. Central and peripheral immune responses are involved in the pathogenesis of AD. Immune changes precede Aβ-associated senile plaque formation and tau-related neurofibrillary tangles, which are the recognised pathological features of AD. Therefore, elucidating immune-related mechanisms underlying the development of AD can help to prevent and treat AD at the source by blocking its progression before the development of pathological changes. To understand the specific pathogenesis of AD, it is important to examine the role of central and peripheral immunity in AD. This review summarises immune-related mechanisms underlying the pathogenesis of AD, focusing on the effect of various central and peripheral immune cells, and describes the possible crosstalk between central and peripheral immunity during the development of AD. This review provides novel insights into the treatment of AD and offers a new direction for immune-related research on AD in the future.
Collapse
Affiliation(s)
- Yuqing Liu
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, China; National Clinical Research Center for Metabolic Diseases, Changsha 410011, China.
| | - Yejun Tan
- School of Mathematics, University of Minnesota Twin Cities, Minneapolis, MN, USA.
| | - Zheyu Zhang
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, China; National Clinical Research Center for Metabolic Diseases, Changsha 410011, China.
| | - Hongli Li
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, China; National Clinical Research Center for Metabolic Diseases, Changsha 410011, China.
| | - Min Yi
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, China; National Clinical Research Center for Metabolic Diseases, Changsha 410011, China.
| | - Zhen Zhang
- YangSheng College of Traditional Chinese Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, Guizhou, China.
| | - Shan Hui
- Department of Geratology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410005, China.
| | - Weijun Peng
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, China; National Clinical Research Center for Metabolic Diseases, Changsha 410011, China.
| |
Collapse
|
18
|
Harris K, Ling Y, Bukhbinder AS, Chen L, Phelps KN, Cruz G, Thomas J, Kim Y, Jiang X, Schulz PE. The Impact of Routine Vaccinations on Alzheimer's Disease Risk in Persons 65 Years and Older: A Claims-Based Cohort Study using Propensity Score Matching. J Alzheimers Dis 2023; 95:703-718. [PMID: 37574727 PMCID: PMC10578243 DOI: 10.3233/jad-221231] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/05/2023] [Indexed: 08/15/2023]
Abstract
BACKGROUND Accumulating evidence suggests that adult vaccinations can reduce the risk of developing Alzheimer's disease (AD) and Alzheimer's disease related dementias. OBJECTIVE To compare the risk for developing AD between adults with and without prior vaccination against tetanus and diphtheria, with or without pertussis (Tdap/Td); herpes zoster (HZ); or pneumococcus. METHODS A retrospective cohort study was performed using Optum's de-identified Clinformatics® Data Mart Database. Included patients were free of dementia during a 2-year look-back period and were≥65 years old by the start of the 8-year follow-up period. We compared two similar cohorts identified using propensity score matching (PSM), one vaccinated and another unvaccinated, with Tdap/Td, HZ, or pneumococcal vaccines. We calculated the relative risk (RR) and absolute risk reduction (ARR) for developing AD. RESULTS For the Tdap/Td vaccine, 7.2% (n = 8,370) of vaccinated patients and 10.2% (n = 11,857) of unvaccinated patients developed AD during follow-up; the RR was 0.70 (95% CI, 0.68-0.72) and ARR was 0.03 (95% CI, 0.02-0.03). For the HZ vaccine, 8.1% (n = 16,106) of vaccinated patients and 10.7% (n = 21,417) of unvaccinated patients developed AD during follow-up; the RR was 0.75 (95% CI, 0.73-0.76) and ARR was 0.02 (95% CI, 0.02-0.02). For the pneumococcal vaccine, 7.92% (n = 20,583) of vaccinated patients and 10.9% (n = 28,558) of unvaccinated patients developed AD during follow-up; the RR was 0.73 (95% CI, 0.71-0.74) and ARR was 0.02 (95% CI, 0.02-0.03). CONCLUSION Several vaccinations, including Tdap/Td, HZ, and pneumococcal, are associated with a reduced risk for developing AD.
Collapse
Affiliation(s)
- Kristofer Harris
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Yaobin Ling
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Avram S. Bukhbinder
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
- Division of Pediatric Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Luyao Chen
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Kamal N. Phelps
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Gabriela Cruz
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jenna Thomas
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Yejin Kim
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Xiaoqian Jiang
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Paul E. Schulz
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
19
|
Vellecco V, Saviano A, Raucci F, Casillo GM, Mansour AA, Panza E, Mitidieri E, Femminella GD, Ferrara N, Cirino G, Sorrentino R, Iqbal AJ, d'Emmanuele di Villa Bianca R, Bucci M, Maione F. Interleukin-17 (IL-17) triggers systemic inflammation, peripheral vascular dysfunction, and related prothrombotic state in a mouse model of Alzheimer's disease. Pharmacol Res 2023; 187:106595. [PMID: 36470548 DOI: 10.1016/j.phrs.2022.106595] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/22/2022] [Accepted: 11/30/2022] [Indexed: 12/08/2022]
Abstract
Alzheimer's disease (AD) is one of the most prevalent forms of neurodegenerative disorders. Previously, we have shown that in vivo administration of an IL-17 neutralizing antibody (IL-17Ab) rescues amyloid-β-induced neuro-inflammation and memory impairment, demonstrating the pivotal role of IL-17 in AD-derived cognitive deficit. Recently, AD has been recognized as a more intriguing pathology affecting vascular networks and platelet function. However, not much is known about peripheral vascular inflammation and how pro-inflammatory circulating cells/mediators could affect peripheral vessels' function. This study aimed to evaluate whether IL-17Ab treatment could also impact peripheral AD features, such as systemic inflammation, peripheral vascular dysfunction, and related pro-thrombotic state in a non-genetic mouse model of AD. Mice were injected intracerebroventricularly with Aβ1-42 peptide (3 μg/3 μl). To evaluate the systemic/peripheral protective profile of IL-17Ab, we used an intranasal administration of IL-17Ab (1 μg/10 μl) at 5, 12, and 19 days after Aβ1-42 injection. Circulating Th17/Treg cells and related cyto-chemokines, haematological parameters, vascular/endothelial reactivity, platelets and coagulation function in mice were evaluated. IL-17Ab treatment ameliorates the systemic/peripheral inflammation, immunological perturbance, vascular/endothelial impairment and pro-thrombotic state, suggesting a key role for this cytokine in fostering inflammatory processes that characterize the multifaced aspects of AD.
Collapse
Affiliation(s)
- Valentina Vellecco
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy.
| | - Anella Saviano
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy.
| | - Federica Raucci
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy.
| | - Gian Marco Casillo
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy.
| | - Adel Abo Mansour
- Institute of Cardiovascular Sciences (ICVS), College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia.
| | - Elisabetta Panza
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy.
| | - Emma Mitidieri
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy.
| | - Grazia Daniela Femminella
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy.
| | - Nicola Ferrara
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; Istituti Clinici Scientifici ICS-Maugeri, Telese Terme, BN, Italy.
| | - Giuseppe Cirino
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy.
| | - Raffaella Sorrentino
- Department of Molecular Medicine and Medical Biotechnologies, School of Medicine, University of Naples, Federico II, Via Pansini, 5, 80131 Naples, Italy.
| | - Asif Jilani Iqbal
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy; Institute of Cardiovascular Sciences (ICVS), College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK.
| | | | - Mariarosaria Bucci
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy.
| | - Francesco Maione
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy.
| |
Collapse
|
20
|
Casares N, Alfaro M, Cuadrado-Tejedor M, Lasarte-Cia A, Navarro F, Vivas I, Espelosin M, Cartas-Cejudo P, Fernández-Irigoyen J, Santamaría E, García-Osta A, Lasarte JJ. Improvement of cognitive function in wild-type and Alzheimer´s disease mouse models by the immunomodulatory properties of menthol inhalation or by depletion of T regulatory cells. Front Immunol 2023; 14:1130044. [PMID: 37187754 PMCID: PMC10175945 DOI: 10.3389/fimmu.2023.1130044] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 04/13/2023] [Indexed: 05/17/2023] Open
Abstract
A complex network of interactions exists between the olfactory, immune and central nervous systems. In this work we intend to investigate this connection through the use of an immunostimulatory odorant like menthol, analyzing its impact on the immune system and the cognitive capacity in healthy and Alzheimer's Disease Mouse Models. We first found that repeated short exposures to menthol odor enhanced the immune response against ovalbumin immunization. Menthol inhalation also improved the cognitive capacity of immunocompetent mice but not in immunodeficient NSG mice, which exhibited very poor fear-conditioning. This improvement was associated with a downregulation of IL-1β and IL-6 mRNA in the brain´s prefrontal cortex, and it was impaired by anosmia induction with methimazole. Exposure to menthol for 6 months (1 week per month) prevented the cognitive impairment observed in the APP/PS1 mouse model of Alzheimer. Besides, this improvement was also observed by the depletion or inhibition of T regulatory cells. Treg depletion also improved the cognitive capacity of the APPNL-G-F/NL-G-F Alzheimer´s mouse model. In all cases, the improvement in learning capacity was associated with a downregulation of IL-1β mRNA. Blockade of the IL-1 receptor with anakinra resulted in a significant increase in cognitive capacity in healthy mice as well as in the APP/PS1 model of Alzheimer´s disease. These data suggest an association between the immunomodulatory capacity of smells and their impact on the cognitive functions of the animals, highlighting the potential of odors and immune modulators as therapeutic agents for CNS-related diseases.
Collapse
Affiliation(s)
- Noelia Casares
- Immunology and Immunotherapy Program, Center for Applied Medical Research (CIMA), University of Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- *Correspondence: Juan José Lasarte, ; Noelia Casares,
| | - María Alfaro
- Immunology and Immunotherapy Program, Center for Applied Medical Research (CIMA), University of Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Mar Cuadrado-Tejedor
- Gene Therapy for Neurological Disease Program, Center for Applied Medical Research (CIMA), University of Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain
| | - Aritz Lasarte-Cia
- Immunology and Immunotherapy Program, Center for Applied Medical Research (CIMA), University of Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Flor Navarro
- Immunology and Immunotherapy Program, Center for Applied Medical Research (CIMA), University of Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Isabel Vivas
- Department of Radiology, Clínica Universidad de Navarra, University of Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - María Espelosin
- Gene Therapy for Neurological Disease Program, Center for Applied Medical Research (CIMA), University of Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Paz Cartas-Cejudo
- Clinical Neuroproteomics Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Joaquín Fernández-Irigoyen
- Clinical Neuroproteomics Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Enrique Santamaría
- Clinical Neuroproteomics Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Ana García-Osta
- Gene Therapy for Neurological Disease Program, Center for Applied Medical Research (CIMA), University of Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Juan José Lasarte
- Immunology and Immunotherapy Program, Center for Applied Medical Research (CIMA), University of Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- *Correspondence: Juan José Lasarte, ; Noelia Casares,
| |
Collapse
|
21
|
Rosa-Gonçalves P, de Sousa LP, Maia AB, Ribeiro-Gomes FL, Gress CCTDL, Werneck GL, Souza DO, Almeida RF, Daniel-Ribeiro CT. Dynamics and immunomodulation of cognitive deficits and behavioral changes in non-severe experimental malaria. Front Immunol 2022; 13:1021211. [PMID: 36505414 PMCID: PMC9729266 DOI: 10.3389/fimmu.2022.1021211] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 11/04/2022] [Indexed: 11/25/2022] Open
Abstract
Data recently reported by our group indicate that stimulation with a pool of immunogens capable of eliciting type 2 immune responses can restore the cognitive and behavioral dysfunctions recorded after a single episode of non-severe rodent malaria caused by Plasmodium berghei ANKA. Here we explored the hypothesis that isolated immunization with one of the type 2 immune response-inducing immunogens, the human diphtheria-tetanus (dT) vaccine, may revert damages associated with malaria. To investigate this possibility, we studied the dynamics of cognitive deficits and anxiety-like phenotype following non-severe experimental malaria and evaluated the effects of immunization with both dT and of a pool of type 2 immune stimuli in reversing these impairments. Locomotor activity and long-term memory deficits were assessed through the open field test (OFT) and novel object recognition task (NORT), while the anxiety-like phenotype was assessed by OFT and light/dark task (LDT). Our results indicate that poor performance in cognitive-behavioral tests can be detected as early as the 12th day after the end of antimalarial treatment with chloroquine and may persist for up to 155 days post infection. The single immunization strategy with the human dT vaccine showed promise in reversal of long-term memory deficits in NORT, and anxiety-like behavior in OFT and LDT.
Collapse
Affiliation(s)
- Pamela Rosa-Gonçalves
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil,Centro de Pesquisa, Diagnóstico e Treinamento em Malária, Fiocruz and Secretaria de Vigilância em Saúde, Ministério da Saúde, Rio de Janeiro, Brazil
| | - Luciana Pereira de Sousa
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil,Centro de Pesquisa, Diagnóstico e Treinamento em Malária, Fiocruz and Secretaria de Vigilância em Saúde, Ministério da Saúde, Rio de Janeiro, Brazil
| | - Aline Barbosa Maia
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil,Centro de Pesquisa, Diagnóstico e Treinamento em Malária, Fiocruz and Secretaria de Vigilância em Saúde, Ministério da Saúde, Rio de Janeiro, Brazil
| | - Flávia Lima Ribeiro-Gomes
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil,Centro de Pesquisa, Diagnóstico e Treinamento em Malária, Fiocruz and Secretaria de Vigilância em Saúde, Ministério da Saúde, Rio de Janeiro, Brazil
| | - Caroline Cristhiani Tavares de Lima Gress
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil,Centro de Pesquisa, Diagnóstico e Treinamento em Malária, Fiocruz and Secretaria de Vigilância em Saúde, Ministério da Saúde, Rio de Janeiro, Brazil
| | - Guilherme Loureiro Werneck
- Departamento de Epidemiologia do Instituto de Medicina Social da Universidade do Estado do Rio de Janeiro and Instituto de Estudos de Saúde Coletiva da Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Diogo Onofre Souza
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Roberto Farina Almeida
- Departamento de Ciências Biológicas, Programa de Pós-Graduação em Ciências Biológicas, Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
| | - Cláudio Tadeu Daniel-Ribeiro
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil,Centro de Pesquisa, Diagnóstico e Treinamento em Malária, Fiocruz and Secretaria de Vigilância em Saúde, Ministério da Saúde, Rio de Janeiro, Brazil,*Correspondence: Cláudio Tadeu Daniel-Ribeiro,
| |
Collapse
|
22
|
Schattner A. Alzheimer's disease and increasing the uptake of influenza vaccination. Eur J Intern Med 2022; 105:121. [PMID: 35914994 DOI: 10.1016/j.ejim.2022.07.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 07/14/2022] [Accepted: 07/26/2022] [Indexed: 11/18/2022]
Affiliation(s)
- Ami Schattner
- The Faculty of Medicine, Hebrew University and Hadassah Medical School, Jerusalem 91120, Israel.
| |
Collapse
|
23
|
Ali E, Shaikh A. Influenza vaccination and dementia risk; an unanticipated benefit? Ann Med Surg (Lond) 2022; 80:104187. [PMID: 36045777 PMCID: PMC9422174 DOI: 10.1016/j.amsu.2022.104187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 07/10/2022] [Indexed: 12/04/2022] Open
Abstract
Alzheimer's disease (AD) characterized by cognitive decline and dementia has evolved into source of extreme concern globally, often associated with functional dependence and financial instability before progressing to complete degeneration of neural and motor skills. Despite multiple interventions being available, only few have been able to show clinical efficacy, others not meeting satisfactory efficacy endpoints as more options are being explored. According to various studies, influenza vaccines have shown clinical evidence in being effective against reduction in dementia risk. Multiple large-scale cohort studies are being conducted to test the effectiveness of vaccinations against dementia. Some of them have shown significant results, establishing a statistically significant relationship between vaccinations and a reduction in symptomatology in already diagnosed dementia patients. These vaccines offer lower-cost, low-risk mechanism of prevention of dementia with better outcomes than pre-existing vaccines. However, there is a need of more large-scale retrospective studies and randomized trials, with longer follow-ups, to be conducted to assess the safety and consistent efficacy of this strategy.
Collapse
|
24
|
Froggatt HM, Heaton NS. Nonrespiratory sites of influenza-associated disease: mechanisms and experimental systems for continued study. FEBS J 2022; 289:4038-4060. [PMID: 35060315 PMCID: PMC9300775 DOI: 10.1111/febs.16363] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 10/20/2021] [Accepted: 01/19/2022] [Indexed: 12/15/2022]
Abstract
The productive replication of human influenza viruses is almost exclusively restricted to cells in the respiratory tract. However, a key aspect of the host response to viral infection is the production of inflammatory cytokines and chemokines that are not similarly tissue restricted. As such, circulating inflammatory mediators, as well as the resulting activated immune cells, can induce damage throughout the body, particularly in individuals with underlying conditions. As a result, more holistic experimental approaches are required to fully understand the pathogenesis and scope of influenza virus-induced disease. This review summarizes what is known about some of the most well-appreciated nonrespiratory tract sites of influenza virus-induced disease, including neurological, cardiovascular, gastrointestinal, muscular and fetal developmental phenotypes. In the context of this discussion, we describe the in vivo experimental systems currently being used to study nonrespiratory symptoms. Finally, we highlight important future questions and potential models that can be used for a more complete understanding of influenza virus-induced disease.
Collapse
Affiliation(s)
- Heather M. Froggatt
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Nicholas S. Heaton
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
| |
Collapse
|
25
|
Bukhbinder AS, Ling Y, Hasan O, Jiang X, Kim Y, Phelps KN, Schmandt RE, Amran A, Coburn R, Ramesh S, Xiao Q, Schulz PE. Risk of Alzheimer's Disease Following Influenza Vaccination: A Claims-Based Cohort Study Using Propensity Score Matching. J Alzheimers Dis 2022; 88:1061-1074. [PMID: 35723106 PMCID: PMC9484126 DOI: 10.3233/jad-220361] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background: Prior studies have found a reduced risk of dementia of any etiology following influenza vaccination in selected populations, including veterans and patients with serious chronic health conditions. However, the effect of influenza vaccination on Alzheimer’s disease (AD) risk in a general cohort of older US adults has not been characterized. Objective: To compare the risk of incident AD between patients with and without prior influenza vaccination in a large US claims database. Methods: Deidentified claims data spanning September 1, 2009 through August 31, 2019 were used. Eligible patients were free of dementia during the 6-year look-back period and≥65 years old by the start of follow-up. Propensity-score matching (PSM) was used to create flu-vaccinated and flu-unvaccinated cohorts with similar baseline demographics, medication usage, and comorbidities. Relative risk (RR) and absolute risk reduction (ARR) were estimated to assess the effect of influenza vaccination on AD risk during the 4-year follow-up. Results: From the unmatched sample of eligible patients (n = 2,356,479), PSM produced a sample of 935,887 flu–vaccinated-unvaccinated matched pairs. The matched sample was 73.7 (SD, 8.7) years of age and 56.9% female, with median follow-up of 46 (IQR, 29–48) months; 5.1% (n = 47,889) of the flu-vaccinated patients and 8.5% (n = 79,630) of the flu-unvaccinated patients developed AD during follow-up. The RR was 0.60 (95% CI, 0.59–0.61) and ARR was 0.034 (95% CI, 0.033–0.035), corresponding to a number needed to treat of 29.4. Conclusion: This study demonstrates that influenza vaccination is associated with reduced AD risk in a nationwide sample of US adults aged 65 and older.
Collapse
Affiliation(s)
- Avram S Bukhbinder
- John P. and Katherine G. McGovern Medical School at UTHealth, Houston, TX, USA
| | - Yaobin Ling
- UTHealth School of Biomedical Informatics, Houston, TX, USA
| | | | - Xiaoqian Jiang
- UTHealth School of Biomedical Informatics, Houston, TX, USA
| | - Yejin Kim
- UTHealth School of Biomedical Informatics, Houston, TX, USA
| | - Kamal N Phelps
- John P. and Katherine G. McGovern Medical School at UTHealth, Houston, TX, USA
| | | | - Albert Amran
- John P. and Katherine G. McGovern Medical School at UTHealth, Houston, TX, USA
| | - Ryan Coburn
- John P. and Katherine G. McGovern Medical School at UTHealth, Houston, TX, USA
| | - Srivathsan Ramesh
- John P. and Katherine G. McGovern Medical School at UTHealth, Houston, TX, USA
| | - Qian Xiao
- UTHealth School of Public Health, Houston, TX, USA
| | - Paul E Schulz
- John P. and Katherine G. McGovern Medical School at UTHealth, Houston, TX, USA
| |
Collapse
|
26
|
Liston A, Dooley J, Yshii L. Brain-resident regulatory T cells and their role in health and disease. Immunol Lett 2022; 248:26-30. [PMID: 35697195 DOI: 10.1016/j.imlet.2022.06.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 06/03/2022] [Accepted: 06/09/2022] [Indexed: 01/01/2023]
Abstract
Regulatory T cells (Tregs) control inflammation and maintain immune homeostasis. The well-characterised circulatory population of CD4+Foxp3+ Tregs is effective at preventing autoimmunity and constraining the immune response, through direct and indirect restraint of conventional T cell activation. Recent advances in Treg cell biology have identified tissue-resident Tregs, with tissue-specific functions that contribute to the maintenance of tissue homeostasis and repair. A population of brain-resident Tregs, characterised as CD69+, has recently been identified in the healthy brain of mice and humans, with rapid population expansion observed under a number of neuroinflammatory conditions. During neuroinflammation, brain-resident Tregs have been proposed to control astrogliosis through the production of amphiregulin, polarize microglia into neuroprotective states, and restrain inflammatory responses by releasing IL-10. While protective effects for Tregs have been demonstrated in a number of neuroinflammatory pathologies, a clear demarcation between the role of circulatory and brain-resident Tregs has been difficult to achieve. Here we review the state-of-the-art for brain-resident Treg population, and describe their potential utilization as a therapeutic target across different neuroinflammatory conditions.
Collapse
Affiliation(s)
- Adrian Liston
- Immunology Programme, The Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT United Kingdom.
| | - James Dooley
- Immunology Programme, The Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT United Kingdom
| | - Lidia Yshii
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Leuven 3000, Belgium; KU Leuven, Department of Neurosciences, Leuven 3000, Belgium.
| |
Collapse
|
27
|
Saviano A, Raucci F, Casillo GM, Mansour AA, Piccolo V, Montesano C, Smimmo M, Vellecco V, Capasso G, Boscaino A, Summa V, Mascolo N, Iqbal AJ, Sorrentino R, Bianca RDDV, Bucci M, Brancaleone V, Maione F. Anti-inflammatory and immunomodulatory activity of Mangifera indica L. reveals the modulation of COX-2/mPGES-1 axis and Th17/Treg ratio. Pharmacol Res 2022; 182:106283. [PMID: 35662629 DOI: 10.1016/j.phrs.2022.106283] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 05/27/2022] [Accepted: 05/29/2022] [Indexed: 10/18/2022]
Abstract
In the context of inflammation and immunity, there are fragmented and observational studies relating to the pharmacological activity of Mangifera indica L. and its main active component, mangiferin. Therefore, we aimed to analyze the potential beneficial effects of this plant extract (MIE, 90% in mangiferin) in a mouse model of gouty arthritis, to allow the evaluation of cellular immune phenotypes and the biochemical mechanism/s beyond MIE activity. Gouty arthritis was induced by the intra-articular administration of MSU crystals (200μg 20μl-1), whereas MIE (0.1-10mgkg-1) or corresponding vehicle (DMSO/saline 1:3) were orally administrated concomitantly with MSU (time 0), 6 and 12h after the stimulus. Thereafter, knee joint score and oedema were evaluated in addition to western blot analysis for COX-2/mPGES-1 axis. Moreover, the analysis of pro/anti-inflammatory cyto-chemokines coupled with the phenotyping of the cellular infiltrate was performed. Treatment with MIE revealed a dose-dependent reduction in joint inflammatory scores with maximal inhibition observed at 10mgkg-1. MIE significantly reduced leukocyte infiltration and activation and the expression of different pro-inflammatory cyto-chemokines in inflamed tissues. Furthermore, biochemical analysis revealed that MIE modulated COX-2/mPGES-1 and mPGDS-1/PPARγ pathways. Flow cytometry analysis also highlighted a prominent modulation of inflammatory monocytes (CD11b+/CD115+/LY6Chi), and Treg cells (CD4+/CD25+/FOXP3+) after MIE treatment. Collectively, the results of this study demonstrate a novel function of MIE to positively affect the local and systemic inflammatory/immunological perturbance in the onset and progression of gouty arthritis.
Collapse
Affiliation(s)
- Anella Saviano
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Federica Raucci
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Gian Marco Casillo
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Adel Abo Mansour
- Institute of Cardiovascular Sciences (ICVS), College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK; Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Vincenzo Piccolo
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Camilla Montesano
- Department of Chemistry, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Martina Smimmo
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Valentina Vellecco
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | | | | | - Vincenzo Summa
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Nicola Mascolo
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Asif Jilani Iqbal
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy; Institute of Cardiovascular Sciences (ICVS), College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Raffaella Sorrentino
- Department of Molecular Medicine and Medical Biotechnologies, School of Medicine, University of Naples, Federico II, Via Pansini, 5, 80131 Naples, Italy
| | | | - Mariarosaria Bucci
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Vincenzo Brancaleone
- Department of Science, University of Basilicata, Via Dell'Ateneo Lucano, 85100, Potenza, Italy
| | - Francesco Maione
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy.
| |
Collapse
|
28
|
Wu X, Yang H, He S, Xia T, Chen D, Zhou Y, Liu J, Liu M, Sun Z. Adult Vaccination as a Protective Factor for Dementia: A Meta-Analysis and Systematic Review of Population-Based Observational Studies. Front Immunol 2022; 13:872542. [PMID: 35592323 PMCID: PMC9110786 DOI: 10.3389/fimmu.2022.872542] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 04/04/2022] [Indexed: 11/19/2022] Open
Abstract
Background Common vaccinations may have impacts on dementia risk, but current evidence is inconsistent. We therefore investigated the association between vaccinations and dementia risk by systematic review and meta-analysis approach. Methods We conducted an extensive search of PubMed, Embase, Cochrane Library, and Web of Science to identify studies that compared the risk of dementia in vaccinated versus unvaccinated populations. The adjusted hazard ratio (HR) and corresponding 95% confidence intervals (CIs) were pooled as measures. Results Of the 9124 records initially retrieved, 17 studies with 1857134 participants were included in our analysis. The overall pooled results showed that vaccinations were associated with a 35% lower dementia risk (HR=0.65, 95% CI: 0.60-0.71, Poverall effect < 0.001; I2 = 91.8%, Pheterogeneity<0.001). All types of vaccination were associated with a trend toward reduced dementia risk, with rabies (HR=0.43), tetanus & diphtheria & pertussis (Tdap) (HR=0.69), herpes zoster (HR=0.69), influenza (HR=0.74), hepatitis A (HR=0.78), typhoid (HR=0.80), and hepatitis B (HR=0.82) vaccinations being significant. Individuals with more full vaccination types and more annual influenza vaccinations were less likely to develop dementia. Gender and age had no effect on this association. Conclusion Routine adult vaccinations are associated with a significant reduction in dementia risk and may be an effective strategy for dementia prevention. Further research is needed to elucidate the causal effects of this association and the underlying mechanisms.
Collapse
Affiliation(s)
- Xinhui Wu
- Department of Geriatric, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Haixia Yang
- The General Hospital of Western Theater Command, Chengdu, China
| | - Sixian He
- The General Hospital of Western Theater Command, Chengdu, China
| | - Ting Xia
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Diang Chen
- Clinical Medicine Teaching Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yexin Zhou
- The First Clinical Medical College, Guangxi University of Chinese Medicine, Guangxi, China
| | - Jin Liu
- Xi'an Hospital of Traditional Chinese Medicine, Xi'an, China
| | - MengSi Liu
- Hengyang Medical School, University of South China, Hengyang, China
| | - Zhen Sun
- Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
29
|
Rosa-Gonçalves P, Ribeiro-Gomes FL, Daniel-Ribeiro CT. Malaria Related Neurocognitive Deficits and Behavioral Alterations. Front Cell Infect Microbiol 2022; 12:829413. [PMID: 35281436 PMCID: PMC8904205 DOI: 10.3389/fcimb.2022.829413] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 01/31/2022] [Indexed: 01/29/2023] Open
Abstract
Typical of tropical and subtropical regions, malaria is caused by protozoa of the genus Plasmodium and is, still today, despite all efforts and advances in controlling the disease, a major issue of public health. Its clinical course can present either as the classic episodes of fever, sweating, chills and headache or as nonspecific symptoms of acute febrile syndromes and may evolve to severe forms. Survivors of cerebral malaria, the most severe and lethal complication of the disease, might develop neurological, cognitive and behavioral sequelae. This overview discusses the neurocognitive deficits and behavioral alterations resulting from human naturally acquired infections and murine experimental models of malaria. We highlighted recent reports of cognitive and behavioral sequelae of non-severe malaria, the most prevalent clinical form of the disease worldwide. These sequelae have gained more attention in recent years and therapies for them are required and demand advances in the understanding of neuropathogenesis. Recent studies using experimental murine models point to immunomodulation as a potential approach to prevent or revert neurocognitive sequelae of malaria.
Collapse
Affiliation(s)
- Pamela Rosa-Gonçalves
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil
- Centro de Pesquisa, Diagnóstico e Treinamento em Malária, Fiocruz and Secretaria de Vigilância em Saúde, Ministério da Saúde, Rio de Janeiro, Brazil
- Laboratório de Biologia, campus Duque de Caxias, Colégio Pedro II, Duque de Caxias, Brazil
- *Correspondence: Pamela Rosa-Gonçalves,
| | - Flávia Lima Ribeiro-Gomes
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil
- Centro de Pesquisa, Diagnóstico e Treinamento em Malária, Fiocruz and Secretaria de Vigilância em Saúde, Ministério da Saúde, Rio de Janeiro, Brazil
| | - Cláudio Tadeu Daniel-Ribeiro
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil
- Centro de Pesquisa, Diagnóstico e Treinamento em Malária, Fiocruz and Secretaria de Vigilância em Saúde, Ministério da Saúde, Rio de Janeiro, Brazil
| |
Collapse
|
30
|
Veronese N, Demurtas J, Smith L, Michel JP, Barbagallo M, Bolzetta F, Noale M, Maggi S. Influenza vaccination reduces dementia risk: A systematic review and meta-analysis. Ageing Res Rev 2022; 73:101534. [PMID: 34861456 DOI: 10.1016/j.arr.2021.101534] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/23/2021] [Accepted: 11/27/2021] [Indexed: 12/25/2022]
Abstract
Animal models have indicated that influenza vaccination may prevent or delay the onset of dementia. However, the epidemiological evidence in human beings is still limited. Given this background, this systematic review and meta-analysis aimed to summarize the current state of the art of observational studies investigating the association between influenza vaccination and the risk of dementia. We searched Scopus and Pubmed/Medline until 24 September 2021 for studies investigating the risk of dementia by influenza vaccination status. After adjustment for potentially important confounding variables, data were reported as risk ratios (RRs) with 95% confidence intervals (CIs). Among 273 articles initially evaluated, five were included for a total of 292,157 older people free from dementia at baseline (mean age=75.5 ± 7.4 years; 46.8% females). All studies were of high quality. Over a mean follow-up of 9 years, influenza vaccination mitigated the risk of dementia (RR=0.97; 95%CI: 0.94-1.00; I2 =99%). This association held after adjustment for a mean of nine potential confounders (RR=0.71; 95%CI: 0.60-0.94; I2 =95.9%). In sensitivity analysis, removing one study from the adjusted analyses, the adjusted RR remained similar (RR= 0.67; 95%CI: 0.63-0.70), but the heterogeneity disappears (I2 =0%). In conclusion, influenza vaccination was associated with a significantly lower risk of dementia suggesting that the vaccination of older people against influenza may also aid in the prevention of dementia.
Collapse
|
31
|
Harkins AL, Kopec AL, Keeler AM. Regulatory T Cell Therapeutics for Neuroinflammatory Disorders. Crit Rev Immunol 2022; 42:1-27. [PMID: 37017285 PMCID: PMC11465901 DOI: 10.1615/critrevimmunol.2022045080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
A delicate balance of immune regulation exists in the central nervous system (CNS) that is often dysreg-ulated in neurological diseases, making them complicated to treat. With altered immune surveillance in the diseased or injured CNS, signals that are beneficial in the homeostatic CNS can be disrupted and lead to neuroinflammation. Recent advances in niche immune cell subsets have provided insight into the complicated cross-talk between the nervous system and the immune system. Regulatory T cells (Tregs) are a subset of T cells that are capable of suppressing effector T-cell activation and regulating immune tolerance, and play an important role in neuroprotection. Tregs have been shown to be effective therapies in a variety of immune-related disorders including, graft-versus-host disease (GVHD), type 1 diabetes (T1D), and inflammatory bowel disease (IBD), as well as within the CNS. Recently, significant advancements in engineering T cells, such as chimeric antigen receptor (CAR) T cells, have led to several approved therapies suggesting the safety and efficacy for similar engineered Treg therapies. Further, as understanding of the immune system's role in neuroinflammation has progressed, Tregs have recently become a potential therapeutic in the neurology space. In this review, we discuss Tregs and their evolving role as therapies for neuroinflammatory related disorders.
Collapse
Affiliation(s)
- Ashley L. Harkins
- Graduate Program in Neuroscience, Morningside Graduate School of Biomedical Sciences
- Horae Gene Therapy Center
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA
| | | | - Allison M. Keeler
- Graduate Program in Neuroscience, Morningside Graduate School of Biomedical Sciences
- Horae Gene Therapy Center
- NeuroNexus Institute, University of Massachusetts Chan Medical School, Worcester, MA
- Department of Pediatrics, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| |
Collapse
|
32
|
Zhang Y, Wang X, Yang X, Yang X, Xue J, Yang Y. Ganoderic Acid A To Alleviate Neuroinflammation of Alzheimer's Disease in Mice by Regulating the Imbalance of the Th17/Tregs Axis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:14204-14214. [PMID: 34798773 DOI: 10.1021/acs.jafc.1c06304] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Ganoderic acid A (GAA) is a kind of lanostane-type triterpenoid isolated from Ganoderma lucidum. Imbalance of the Th17/Tregs axis exists in the progress of neuroinflammation of Alzheimer's disease (AD). In this study, the alleviating neuroinflammatory effect of GAA on d-galactose mice was studied from the aspect of regulating the imbalance of the Th17/Tregs axis. The Morris water maze test was used to evaluate the cognitive ability of AD mice. Flow cytometry was used to detect the percentages of IL-17A, IL-17F, IL-21, IL-22, and CD4+CD25+Foxp3+ in peripheral blood. Transmission electron microscopy was used to assess the cerebral mitochondrial ultrastructure. Metabolomic analysis based on gas chromatography-mass spectrometry was used to evaluate the mitochondrial dysfunction metabolism. Western blot analysis was used to detect the protein expressions of cytokines secreted by Th17 cells and Treg cells in the brain. As the results show, GAA has an alleviating neuroinflammatory effect on AD mice via regulating the imbalance of the Th17/Tregs axis. The potential mechanism was related to inhibition of the JAK/STAT signaling pathway induced by Th17 cells and enhancement of the mitochondrial oxidative phosphorylation by regulating Treg cells, thereby improving mitochondrial dysfunction of AD mice.
Collapse
Affiliation(s)
- Yan Zhang
- School of Chemical and Pharmaceutical Engineering, Jilin Institute of Chemical Technology, Jilin 132022, P. R. China
| | - Xinyan Wang
- Graduate School, Jilin Institute of Chemical Technology, Jilin 132022, P. R. China
| | - Xiaomei Yang
- Nutritional Department, Jilin Medical University Affiliated Hospital, Jilin 132013, P. R. China
| | - Xiudong Yang
- School of Chemical and Pharmaceutical Engineering, Jilin Institute of Chemical Technology, Jilin 132022, P. R. China
| | - Jianfei Xue
- School of Chemical and Pharmaceutical Engineering, Jilin Institute of Chemical Technology, Jilin 132022, P. R. China
| | - Yanjun Yang
- School of Chemical and Pharmaceutical Engineering, Jilin Institute of Chemical Technology, Jilin 132022, P. R. China
| |
Collapse
|
33
|
Dai L, Shen Y. Insights into T-cell dysfunction in Alzheimer's disease. Aging Cell 2021; 20:e13511. [PMID: 34725916 PMCID: PMC8672785 DOI: 10.1111/acel.13511] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/22/2021] [Accepted: 10/22/2021] [Indexed: 12/11/2022] Open
Abstract
T cells, the critical immune cells of the adaptive immune system, are often dysfunctional in Alzheimer's disease (AD) and are involved in AD pathology. Reports highlight neuroinflammation as a crucial modulator of AD pathogenesis, and aberrant T cells indirectly contribute to neuroinflammation by secreting proinflammatory mediators via direct crosstalk with glial cells infiltrating the brain. However, the mechanisms underlying T‐cell abnormalities in AD appear multifactorial. Risk factors for AD and pathological hallmarks of AD have been tightly linked with immune responses, implying the potential regulatory effects of these factors on T cells. In this review, we discuss how the risk factors for AD, particularly Apolipoprotein E (ApoE), Aβ, α‐secretase, β‐secretase, γ‐secretase, Tau, and neuroinflammation, modulate T‐cell activation and the association between T cells and pathological AD hallmarks. Understanding these associations is critical to provide a comprehensive view of appropriate therapeutic strategies for AD.
Collapse
Affiliation(s)
- Linbin Dai
- Institute on Aging and Brain Disorders The First Affiliated Hospital of USTC Division of Life Sciences and Medicine University of Sciences and Technology of China Hefei China
- Neurodegenerative Disease Research Center University of Science and Technology of China Hefei China
- Hefei National Laboratory for Physical Sciences at the Microscale University of Science and Technology of China Hefei China
| | - Yong Shen
- Institute on Aging and Brain Disorders The First Affiliated Hospital of USTC Division of Life Sciences and Medicine University of Sciences and Technology of China Hefei China
- Neurodegenerative Disease Research Center University of Science and Technology of China Hefei China
- Hefei National Laboratory for Physical Sciences at the Microscale University of Science and Technology of China Hefei China
| |
Collapse
|
34
|
Wiemken TL, Salas J, Hoft DF, Jacobs C, Morley JE, Scherrer JF. Dementia risk following influenza vaccination in a large veteran cohort. Vaccine 2021; 39:5524-5531. [PMID: 34420785 DOI: 10.1016/j.vaccine.2021.08.046] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 07/28/2021] [Accepted: 08/12/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Two Taiwan based studies indicated influenza vaccinations are associated with lower risk for dementia in patient cohorts with chronic disease. We determined if such associations exist in a large, nationally distributed sample of U.S. patients not selected for chronic disease. METHODS Data was obtained from Veterans Health Administration medical records (9/1/2009 - 8/31/19). Eligible patients were ≥65 years of age and free of dementia for two years prior to enrollment through the end of the first influenza season (9/1/2009 to 3/1/2012). Competing risk models estimated the risk of dementia in those with influenza vaccination (n = 66,822) compared to those without vaccination (n = 56,925). Propensity scores and inverse probability of treatment weighting controlled for confounding. RESULTS On average, patients were 75.5 (±7.3) years of age, 3.8% were female and 91.6% were white race. After controlling for confounding, patients with influenza vaccination were significantly less likely to develop dementia compared to patients without vaccination (HR = 0.86; 95 %CI:0.83-0.88). Patients with 1, 2 or 3-5 vaccines vs. none had similar risks for dementia and patients with ≥ 6 influenza vaccines vs. none had a significant lower risk for dementia (HR = 0.88, 95 %CI: 0.83-0.94). CONCLUSIONS Repeated receipt of influenza vaccinations, compared to remaining unvaccinated, is associated with lower risk for dementia. This is consistent with the hypotheses that vaccinations may reduce risk of dementia by training the immune system and not by preventing specific infectious disease. If vaccines are identified as causative factors in reducing incident dementia, they offer an inexpensive, low-risk intervention with effects greater than any existing preventive measure.
Collapse
Affiliation(s)
- Timothy L Wiemken
- Saint Louis University, School of Medicine, Department of Internal Medicine, Division of Infectious Diseases, Allergy, and Immunology, Saint Louis, MO, United States; The Advanced HEAlth Data (AHEAD) Research Institute, Saint Louis University, School of Medicine, 1402 South Grand Blvd, St. Louis, MO, United States.
| | - Joanne Salas
- Department of Family and Community Medicine, Saint Louis University, School of Medicine, 1402 South Grand Blvd, St. Louis, MO, United States; Harry S. Truman Veterans Administration Medical Center, Columbia, MO, United States; The Advanced HEAlth Data (AHEAD) Research Institute, Saint Louis University, School of Medicine, 1402 South Grand Blvd, St. Louis, MO, United States
| | - Daniel F Hoft
- Saint Louis University, School of Medicine, Department of Internal Medicine, Division of Infectious Diseases, Allergy, and Immunology, Saint Louis, MO, United States; Saint Louis University, Department of Molecular Microbiology & Immunology, Saint Louis, MO, United States
| | - Christine Jacobs
- Department of Family and Community Medicine, Saint Louis University, School of Medicine, 1402 South Grand Blvd, St. Louis, MO, United States
| | - John E Morley
- Saint Louis University, School of Medicine, Department of Internal Medicine, Division of Geriatric Medicine, 1402 South Grand Blvd, St. Louis, MO, United States
| | - Jeffrey F Scherrer
- Department of Family and Community Medicine, Saint Louis University, School of Medicine, 1402 South Grand Blvd, St. Louis, MO, United States; Harry S. Truman Veterans Administration Medical Center, Columbia, MO, United States; The Advanced HEAlth Data (AHEAD) Research Institute, Saint Louis University, School of Medicine, 1402 South Grand Blvd, St. Louis, MO, United States
| |
Collapse
|
35
|
Illouz T, Madar R, Hirsh T, Biragyn A, Okun E. Induction of an effective anti-Amyloid-β humoral response in aged mice. Vaccine 2021; 39:4817-4829. [PMID: 34294479 DOI: 10.1016/j.vaccine.2021.07.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 06/20/2021] [Accepted: 07/08/2021] [Indexed: 01/03/2023]
Abstract
Aging-related decline in immune functions, termed immunosenescence, is a primary cause of reduced protective responses to vaccines in the elderly, due to impaired induction of cellular and humoral responses to new antigens (Ag), especially if the response is T cell dependent. The result is a more severe morbidity following infections, more prolonged and frequent hospitalization, and a higher mortality rate than in the general population. Therefore, there is an increasing need to develop vaccination strategies that overcome immunosenescence, especially for aging-related diseases such as Alzheimer's disease (AD). Here we report a new vaccination strategy harnessing memory-based immunity, which is less affected by aging. We found that aged C57BL/6 and 5xFAD mice exhibit a dramatic reduction in anti-Amyloid-β (Aβ) antibody (Ab) production. We aimed to reverse this process by inducing memory response at a young age. To this end, young mice were primed with the vaccine carrier Hepatitis B surface antigen (HBsAg). At an advanced age, these mice were immunized with an Aβ1-11 fused to HBsAg. This vaccination scheme elicited a markedly higher Aβ-specific antibody titer than vaccinating aged unprimed mice with the same construct. Importantly, this vaccine strategy more efficiently reduced cerebral Aβ levels and altered microglial phenotype. Overall, we provide evidence that priming with an exogenous Ag carrier can overcome impaired humoral responses to self-antigens in the elderly, paving the route for a potent immunotherapy to AD.
Collapse
Affiliation(s)
- Tomer Illouz
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan 5290002, Israel; The Paul Feder Laboratory on Alzheimer's Disease Research, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Ravit Madar
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan 5290002, Israel; The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel; The Paul Feder Laboratory on Alzheimer's Disease Research, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Tamir Hirsh
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan 5290002, Israel; The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel; The Paul Feder Laboratory on Alzheimer's Disease Research, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Arya Biragyn
- Immunoregulation Section, Laboratory of Molecular Biology and Immunology, NIA, NIH, MD 21224, USA
| | - Eitan Okun
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan 5290002, Israel; The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel; The Paul Feder Laboratory on Alzheimer's Disease Research, Bar-Ilan University, Ramat Gan 5290002, Israel.
| |
Collapse
|
36
|
Shi Y, Guo M, Yang W, Liu S, Zhu B, Yang L, Yang C, Liu C. Is SARS-CoV-2 vaccination safe and effective for elderly individuals with neurodegenerative diseases? Expert Rev Vaccines 2021; 20:375-383. [PMID: 33787439 PMCID: PMC8054494 DOI: 10.1080/14760584.2021.1911653] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Introduction Coronavirus Disease 2019 (COVID-19) poses a substantial threat to the lives of the elderly, especially those with neurodegenerative diseases, and vaccination against viral infections is recognized as an effective measure to reduce mortality. However, elderly patients with neurodegenerative diseases often suffer from abnormal immune function and take multiple medications, which may complicate the role of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines. Currently, there is no expert consensus on whether SARS-CoV-2 vaccines are suitable for patients with neurodegenerative diseases. Areas covered We searched Pubmed to conduct a systematic review of published studies, case reports, reviews, meta-analyses, and expert guidelines on the impact of SARS-CoV-2 on neurodegenerative diseases and the latest developments in COVID-19 vaccines. We also summarized the interaction between vaccines and age-related neurodegenerative diseases. The compatibility of future SARS-CoV-2 vaccines with neurodegenerative diseases is discussed. Expert opinion Vaccines enable the body to produce immunity by activating the body’s immune response. The pathogenesis and treatment of neurodegenerative diseases is complex, and these diseases often involve abnormal immune function, which can substantially affect the safety and effectiveness of vaccines. In short, this article provides recommendations for the use of vaccine candidates in patients with neurodegenerative diseases.
Collapse
Affiliation(s)
- Yan Shi
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing China
| | - Minna Guo
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing China
| | - Wenjing Yang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing China
| | - Shijiang Liu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing China
| | - Bin Zhu
- Department of Critical Care Medicine, The Third Affiliated Hospital of Soochow University, Changzhou China
| | - Ling Yang
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou China
| | - Chun Yang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing China
| | - Cunming Liu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing China
| |
Collapse
|
37
|
Perspective: Treatment for Disease Modification in Chronic Neurodegeneration. Cells 2021; 10:cells10040873. [PMID: 33921342 PMCID: PMC8069143 DOI: 10.3390/cells10040873] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 03/31/2021] [Accepted: 04/09/2021] [Indexed: 02/07/2023] Open
Abstract
Symptomatic treatments are available for Parkinson's disease and Alzheimer's disease. An unmet need is cure or disease modification. This review discusses possible reasons for negative clinical study outcomes on disease modification following promising positive findings from experimental research. It scrutinizes current research paradigms for disease modification with antibodies against pathological protein enrichment, such as α-synuclein, amyloid or tau, based on post mortem findings. Instead a more uniform regenerative and reparative therapeutic approach for chronic neurodegenerative disease entities is proposed with stimulation of an endogenously existing repair system, which acts independent of specific disease mechanisms. The repulsive guidance molecule A pathway is involved in the regulation of peripheral and central neuronal restoration. Therapeutic antagonism of repulsive guidance molecule A reverses neurodegeneration according to experimental outcomes in numerous disease models in rodents and monkeys. Antibodies against repulsive guidance molecule A exist. First clinical studies in neurological conditions with an acute onset are under way. Future clinical trials with these antibodies should initially focus on well characterized uniform cohorts of patients. The efficiency of repulsive guidance molecule A antagonism and associated stimulation of neurogenesis should be demonstrated with objective assessment tools to counteract dilution of therapeutic effects by subjectivity and heterogeneity of chronic disease entities. Such a research concept will hopefully enhance clinical test strategies and improve the future therapeutic armamentarium for chronic neurodegeneration.
Collapse
|
38
|
Wang C, Cai X, Wang R, Zhai S, Zhang Y, Hu W, Zhang Y, Wang D. Neuroprotective effects of verbascoside against Alzheimer's disease via the relief of endoplasmic reticulum stress in Aβ-exposed U251 cells and APP/PS1 mice. J Neuroinflammation 2020; 17:309. [PMID: 33070776 PMCID: PMC7570123 DOI: 10.1186/s12974-020-01976-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 10/01/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Endoplasmic reticulum (ER) stress is involved in the progression of Alzheimer's disease (AD). Verbascoside (VB), an active phenylethanoid glycoside that was first isolated from Verbascum sinuatum (the wavyleaf mullein), possesses anti-inflammatory, antioxidative, and anti-apoptotic effects. The purpose of this study was to elucidate the beneficial effects of VB in amyloid β (Aβ)1-42-damaged human glioma (U251) cells and in APPswe/PSEN1dE9 transgenic (APP/PS1) mice. METHODS U251 cells were co-incubated with 10 μM of Aβ1-42 and treated with VB. The protective effects of VB were investigated by using 3-(4,5-dimethylthiazole-2-yl)-2,5-diphenyl tetrazolium bromide assay, flow cytometry, fluorescence staining, and transmission electron microscopy. APP/PS1 transgenic mice were treated for 6 weeks with VB. Learning and memory were evaluated using a Morris water maze test. Immunohistochemistry, terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end labeling, thioflavin-S staining, and proteomics analysis were performed to study the potential neuroprotective mechanism. Enzyme-linked immunosorbent assays and western blot were performed to analyze altered protein levels of brain lysates in APP/PS1 mice and/or Aβ1-42-damaged U251 cells. RESULTS In Aβ1-42-damaged U251 cells, VB significantly improved cell viability, inhibited apoptosis, reduced calcium accumulation and the intracellular concentrations of reactive oxygen species, and improved the morphology of mitochondria and ER. In APP/PS1 mice, 6-week administration of VB significantly improved memory and cognition. VB inhibited apoptosis, reduced the deposition of Aβ, reduced the formation of neurofibrillary tangles formed by hyperphosphorylated tau protein, and downregulated the expression levels of 4-hydroxynonenal and mesencephalic astrocyte-derived neurotrophic factor in the brains of APP/PS1 mice. Proteomics analysis of mouse hippocampus suggested that the neuroprotective effect of VB may be related to the reduction of ER stress. This was indicated by the fact that VB inhibited the three branches of the unfolded protein response, thereby attenuating ER stress and preventing apoptosis. CONCLUSIONS The results confirmed that VB possesses significant neuroprotective effects, which are related to the reduction of ER stress. These findings support the status of VB as a potentially effective treatment for AD and warrant further research.
Collapse
Affiliation(s)
- Chunyue Wang
- School of Life Sciences, Jilin University, Changchun, 130012 China
| | - Xueying Cai
- School of Life Sciences, Jilin University, Changchun, 130012 China
| | - Ruochen Wang
- School of Life Sciences, Jilin University, Changchun, 130012 China
| | - Siyu Zhai
- School of Life Sciences, Jilin University, Changchun, 130012 China
| | - Yongfeng Zhang
- School of Life Sciences, Jilin University, Changchun, 130012 China
| | - Wenji Hu
- School of Life Sciences, Jilin University, Changchun, 130012 China
| | - Yizhi Zhang
- Department of Neurology, The Second Hospital of Jilin University, Changchun, 130041 China
| | - Di Wang
- School of Life Sciences, Jilin University, Changchun, 130012 China
| |
Collapse
|